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:

Metabolic remodelling during early mouse embryo development

Abstract

During early mammalian embryogenesis, changes in cell growth and proliferation depend on strict genetic and metabolic instructions. However, our understanding of metabolic reprogramming and its influence on epigenetic regulation in early embryo development remains elusive. Here we show a comprehensive metabolomics profiling of key stages in mouse early development and the two-cell and blastocyst embryos, and we reconstructed the metabolic landscape through the transition from totipotency to pluripotency. Our integrated metabolomics and transcriptomics analysis shows that while two-cell embryos favour methionine, polyamine and glutathione metabolism and stay in a more reductive state, blastocyst embryos have higher metabolites related to the mitochondrial tricarboxylic acid cycle, and present a more oxidative state. Moreover, we identify a reciprocal relationship between α-ketoglutarate (α-KG) and the competitive inhibitor of α-KG-dependent dioxygenases, l-2-hydroxyglutarate (l-2-HG), where two-cell embryos inherited from oocytes and one-cell zygotes display higher l-2-HG, whereas blastocysts show higher α-KG. Lastly, increasing 2-HG availability impedes erasure of global histone methylation markers after fertilization. Together, our data demonstrate dynamic and interconnected metabolic, transcriptional and epigenetic network remodelling during early mouse embryo development.

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

Access options

Buy this article

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

Fig. 1: Metabolomics profiling of two-cell and blastocyst-stage embryos.
Fig. 2: Genomic and metabolic analysis reveals dynamic metabolic network integrated in the developmental TF network in early embryogenesis.
Fig. 3: A decrease of l-2-HG facilitates global erasure of histone methylation during early embryo development.
Fig. 4: l-2-HG supplementation impedes early embryo development which is rescuable by α-KG.
Fig. 5: L2hgdh deficiency impedes H3K4me3 methylation erasure during mouse early embryo development.

Similar content being viewed by others

Data availability

RNA-seq data have been deposited in the NCBI Gene Expression Omnibus under accession code GSE181648. Previously published RNA-seq data that were reanalysed here are available under accessions GSE45719, GSE98150 and GSE33923. Published ChIP–seq data for TFs are available under accession GSE11431. Published ATAC–seq data were downloaded from GSE66390. Source data are provided with this paper. The remaining data that support the findings of this study and uncropped versions of blots are available from the corresponding authors upon reasonable request.

Code availability

All the analysis in this study was made based on custom Perl (v5.30.0), Python (v2.7.16) and R (v4.0.2) codes and is available upon reasonable request.

References

  1. Zhang, J. et al. Metabolism in pluripotent stem cells and early mammalian development. Cell Metab. 27, 332–338 (2018).

    Article  CAS  PubMed  Google Scholar 

  2. Chronopoulou, E. & Harper, J. C. IVF culture media: past, present and future. Hum. Reprod. Update 21, 39–55 (2015).

    Article  CAS  PubMed  Google Scholar 

  3. Conaghan, J., Handyside, A. H., Winston, R. M. & Leese, H. J. Effects of pyruvate and glucose on the development of human preimplantation embryos in vitro. J. Reprod. Fertil. 99, 87–95 (1993).

    Article  CAS  PubMed  Google Scholar 

  4. Brinster, R. L. Studies on the development of mouse embryos in vitro. The effect of energy source. J. Exp. Zool. 158, 59–68 (1965).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  5. Brown, J. J. & Whittingham, D. G. The roles of pyruvate, lactate and glucose during preimplantation development of embryos from F1 hybrid mice in vitro. Development 112, 99–105 (1991).

    Article  CAS  PubMed  Google Scholar 

  6. Gardner, D. K. Changes in requirements and utilization of nutrients during mammalian preimplantation embryo development and their significance in embryo culture. Theriogenology 49, 83–102 (1998).

    Article  CAS  PubMed  Google Scholar 

  7. Gardner, D. K. & Lane, M. Culture and selection of viable blastocysts: a feasible proposition for human IVF? Hum. Reprod. Update 3, 367–382 (1997).

    Article  CAS  PubMed  Google Scholar 

  8. Houghton, F. D., Thompson, J. G., Kennedy, C. J. & Leese, H. J. Oxygen consumption and energy metabolism of the early mouse embryo. Mol. Reprod. Dev. 44, 476–485 (1996).

    Article  CAS  PubMed  Google Scholar 

  9. Leese, H. J. Metabolism of the preimplantation embryo: 40 years on. Reproduction 143, 417–427 (2012).

    Article  CAS  PubMed  Google Scholar 

  10. Xue, Z. et al. Genetic programs in human and mouse early embryos revealed by single-cell RNA sequencing. Nature 500, 593–597 (2013).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  11. Wu, J. et al. The landscape of accessible chromatin in mammalian preimplantation embryos. Nature 534, 652–657 (2016).

    Article  CAS  PubMed  Google Scholar 

  12. Lu, F. et al. Establishing chromatin regulatory landscape during mouse preimplantation development. Cell 165, 1375–1388 (2016).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  13. Dahl, J. A. et al. Broad histone H3K4me3 domains in mouse oocytes modulate maternal-to-zygotic transition. Nature 537, 548–552 (2016).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  14. Zhang, B. et al. Allelic reprogramming of the histone modification H3K4me3 in early mammalian development. Nature 537, 553–557 (2016).

    Article  CAS  PubMed  Google Scholar 

  15. Wang, C. et al. Reprogramming of H3K9me3-dependent heterochromatin during mammalian embryo development. Nat. Cell Biol. 20, 620–631 (2018).

    Article  CAS  PubMed  Google Scholar 

  16. Macfarlan, T. S. et al. Embryonic stem cell potency fluctuates with endogenous retrovirus activity. Nature 487, 57–63 (2012).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  17. Nichols, J. & Smith, A. Naive and primed pluripotent states. Cell Stem Cell 4, 487–492 (2009).

    Article  CAS  PubMed  Google Scholar 

  18. Smith, A. Formative pluripotency: the executive phase in a developmental continuum. Development 144, 365–373 (2017).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  19. Teslaa, T. & Teitell, M. A. Pluripotent stem cell energy metabolism: an update. EMBO J. 34, 138–153 (2015).

    Article  CAS  PubMed  Google Scholar 

  20. Zhou, W. et al. HIF1α induced switch from bivalent to exclusively glycolytic metabolism during ESC-to-EpiSC/hESC transition. EMBO J. 31, 2103–2116 (2012).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  21. Chandrasekaran, S. et al. Comprehensive mapping of pluripotent stem cell metabolism using dynamic genome-scale network modeling. Cell Rep. 21, 2965–2977 (2017).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  22. Sone, M. et al. Hybrid cellular metabolism coordinated by Zic3 and Esrrb synergistically enhances induction of naive pluripotency. Cell Metab. 25, 1103–1117 (2017).

    Article  CAS  PubMed  Google Scholar 

  23. Carbognin, E., Betto, R. M., Soriano, M. E., Smith, A. G. & Martello, G. Stat3 promotes mitochondrial transcription and oxidative respiration during maintenance and induction of naive pluripotency. EMBO J. 35, 618–634 (2016).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  24. Zhang, J. et al. LIN28 regulates stem cell metabolism and conversion to primed pluripotency cell stem cell article LIN28 regulates stem cell metabolism and conversion to primed pluripotency. Cell Stem Cell 19, 66–80 (2016).

    Article  CAS  PubMed  Google Scholar 

  25. Scognamiglio, R. et al. Myc depletion induces a pluripotent dormant state mimicking diapause. Cell 164, 668–680 (2016).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  26. Bulut-Karslioglu, A. et al. Inhibition of mTOR induces a paused pluripotent state. Nature 540, 119–123 (2016).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  27. Xu, R., Li, C., Liu, X. & Gao, S. Insights into epigenetic patterns in mammalian early embryos. Protein Cell https://doi.org/10.1007/s13238-020-00757-z (2020).

  28. Xia, W. & Xie, W. Rebooting the epigenomes during mammalian early embryogenesis. Stem Cell Reports https://doi.org/10.1016/j.stemcr.2020.09.005 (2020).

  29. Eckersley-Maslin, M. A., Alda-Catalinas, C. & Reik, W. Dynamics of the epigenetic landscape during the maternal-to-zygotic transition. Nat. Rev. Mol. Cell Biol. 19, 436–450 (2018).

    Article  CAS  PubMed  Google Scholar 

  30. Burton, A. et al. Heterochromatin establishment during early mammalian development is regulated by pericentromeric RNA and characterized by non-repressive H3K9me3. Nat. Cell Biol. 22, 767–778 (2020).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  31. Jukam, D., Shariati, S. A. M. & Skotheim, J. M. Zygotic genome activation in vertebrates. Dev. Cell 42, 316–332 (2017).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  32. Xu, W. et al. Oncometabolite 2-hydroxyglutarate is a competitive inhibitor of alpha-ketoglutarate-dependent dioxygenases. Cancer Cell 19, 17–30 (2011).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  33. Fu, X., Djekidel, M. N. & Zhang, Y. A transcriptional roadmap for 2C-like-to-pluripotent state transition. Sci. Adv. 6, eaay5181 (2020).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  34. Deng, Q., Ramskold, D., Reinius, B. & Sandberg, R. Single-cell RNA-seq reveals dynamic, random monoallelic gene expression in mammalian cells. Science 343, 193–196 (2014).

    Article  CAS  PubMed  Google Scholar 

  35. Birsoy, K. et al. An essential role of the mitochondrial electron transport chain in cell proliferation is to enable aspartate synthesis. Cell 162, 540–551 (2015).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  36. Zhao, Y. et al. In vivo monitoring of cellular energy metabolism using SoNar, a highly responsive sensor for NAD+/NADH redox state. Nat. Protoc. 11, 1345–1359 (2016).

    Article  CAS  PubMed  Google Scholar 

  37. Chen, X. et al. Integration of external signaling pathways with the core transcriptional network in embryonic stem cells. Cell 133, 1106–1117 (2008).

    Article  CAS  PubMed  Google Scholar 

  38. Lu, C. et al. IDH mutation impairs histone demethylation and results in a block to cell differentiation. Nature 483, 474–478 (2012).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  39. Carey, B. W., Finley, L. W., Cross, J. R., Allis, C. D. & Thompson, C. B. Intracellular alpha-ketoglutarate maintains the pluripotency of embryonic stem cells. Nature 518, 413–416 (2015).

    Article  CAS  PubMed  Google Scholar 

  40. Intlekofer, A. M. et al. Hypoxia induces production of l-2-hydroxyglutarate. Cell Metab. 22, 304–311 (2015).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  41. Intlekofer, A. M. et al. l-2-hydroxyglutarate production arises from noncanonical enzyme function at acidic pH. Nat. Chem. Biol. 13, 494–500 (2017).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  42. Steinert, E. M., Vasan, K. & Chandel, N. S. Mitochondrial metabolism regulation of T cell-mediated Immunity. Annu. Rev. Immunol. 39, 395–416 (2021).

    Article  CAS  PubMed  Google Scholar 

  43. Shim, E. H. et al. l-2-hydroxyglutarate: an epigenetic modifier and putative oncometabolite in renal cancer. Cancer Discov. 4, 1290–1298 (2014).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  44. Amankulor, N. M. et al. Mutant IDH1 regulates the tumor-associated immune system in gliomas. Genes Dev. 31, 774–786 (2017).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  45. Terunuma, A. et al. MYC-driven accumulation of 2-hydroxyglutarate is associated with breast cancer prognosis. J. Clin. Invest. 124, 398–412 (2014).

    Article  CAS  PubMed  Google Scholar 

  46. Gross, S. et al. Cancer-associated metabolite 2-hydroxyglutarate accumulates in acute myelogenous leukemia with isocitrate dehydrogenase 1 and 2 mutations. J. Exp. Med. 207, 339–344 (2010).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  47. Bunse, L. et al. Suppression of antitumor T cell immunity by the oncometabolite (R)-2-hydroxyglutarate. Nat. Med. 24, 1192–1203 (2018).

    Article  CAS  PubMed  Google Scholar 

  48. Tyrakis, P. A. et al. S-2-hydroxyglutarate regulates CD8+ T lymphocyte fate. Nature 540, 236–241 (2016).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  49. Fu, X. et al. 2-hydroxyglutarate inhibits ATP synthase and mTOR signaling. Cell Metab. 22, 508–515 (2015).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  50. Oldham, W. M., Clish, C. B., Yang, Y. & Loscalzo, J. Hypoxia-mediated increases in l-2-hydroxyglutarate coordinate the metabolic response to reductive stress. Cell Metab. 22, 291–303 (2015).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  51. Ye, D., Guan, K. L. & Xiong, Y. Metabolism, activity, and targeting of d- and l-2-hydroxyglutarates. Trends Cancer 4, 151–165 (2018).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  52. Agathocleous, M. et al. Ascorbate regulates haematopoietic stem cell function and leukaemogenesis. Nature 549, 476–481 (2017).

    Article  PubMed  PubMed Central  Google Scholar 

  53. Nasr-Esfahani, M. H. & Johnson, M. H. Quantitative analysis of cellular glutathione in early preimplantation mouse embryos developing in vivo and in vitro. Hum. Reprod. 7, 1281–1290 (1992).

    Article  CAS  PubMed  Google Scholar 

  54. Dumollard, R., Ward, Z., Carroll, J. & Duchen, M. R. Regulation of redox metabolism in the mouse oocyte and embryo. Development 134, 455–465 (2007).

    Article  CAS  PubMed  Google Scholar 

  55. Liu, X. et al. Distinct features of H3K4me3 and H3K27me3 chromatin domains in pre-implantation embryos. Nature 537, 558–562 (2016).

    Article  CAS  PubMed  Google Scholar 

  56. Nagaraj, R. et al. Nuclear localization of mitochondrial TCA cycle enzymes as a critical step in mammalian zygotic genome activation. Cell 168, 210–223 (2017).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  57. Vander Heiden, M. G., Cantley, L. C. & Thompson, C. B. Understanding the Warburg effect: the metabolic requirements of cell proliferation. Science 324, 1029–1033 (2009).

    Article  CAS  Google Scholar 

  58. Orkin, S. H. et al. The transcriptional network controlling pluripotency in ES cells. Cold Spring Harb. Symp. Quant. Biol. 73, 195–202 (2008).

    Article  CAS  PubMed  Google Scholar 

  59. Ng, H. H. & Surani, M. A. The transcriptional and signalling networks of pluripotency. Nat. Cell Biol. 13, 490–496 (2011).

    Article  CAS  PubMed  Google Scholar 

  60. DeNicola, G. M. et al. NRF2 regulates serine biosynthesis in non-small cell lung cancer. Nat. Genet. 47, 1475–1481 (2015).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  61. Huang, F. et al. Inosine monophosphate dehydrogenase dependence in a subset of small cell lung cancers. Cell Metab. 28, 369–382 (2018).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  62. Kim, J. et al. CPS1 maintains pyrimidine pools and DNA synthesis in KRAS/LKB1-mutant lung cancer cells. Nature 546, 168–172 (2017).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  63. Piskounova, E. et al. Oxidative stress inhibits distant metastasis by human melanoma cells. Nature 527, 186–191 (2015).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  64. Li, X. K. et al. Arginine deficiency is involved in thrombocytopenia and immunosuppression in severe fever with thrombocytopenia syndrome. Sci. Transl. Med. https://doi.org/10.1126/scitranslmed.aat4162 (2018).

  65. Liu, X. et al. Regulation of mitochondrial biogenesis in erythropoiesis by mTORC1-mediated protein translation. Nat. Cell Biol. 19, 626–638 (2017).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  66. Nakada, Y. et al. Hypoxia induces heart regeneration in adult mice. Nature 541, 222–227 (2017).

    Article  CAS  PubMed  Google Scholar 

  67. Park, J. S. et al. Mechanical regulation of glycolysis via cytoskeleton architecture. Nature 578, 621–626 (2020).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  68. Zheng, J. Y. et al. A readily 16O-/18O isotopically paired chiral derivatization approach for the quantification of 2-HG metabolic panel by liquid chromatography–tandem mass spectrometry. Anal. Chim. Acta 1077, 174–182 (2019).

    Article  CAS  PubMed  Google Scholar 

  69. Cheng, Q. Y. et al. Sensitive determination of onco-metabolites of d- and l-2-hydroxyglutarate enantiomers by chiral derivatization combined with liquid chromatography/mass spectrometry analysis. Sci. Rep. 5, 15217 (2015).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  70. Picelli, S. et al. Full-length RNA-seq from single cells using Smart-seq2. Nat. Protoc. 9, 171–181 (2014).

    Article  CAS  PubMed  Google Scholar 

  71. Bolger, A. M., Marc, L. & Bjoern, U. Trimmomatic: a flexible trimmer for Illumina sequence data. Bioinformatics 30, 2114–2120 (2014).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  72. Daehwan, K., Ben, L. & Salzberg, S. L. HISAT: a fast spliced aligner with low memory requirements. Nat. Methods 12, 357–360 (2015).

    Article  CAS  Google Scholar 

  73. Mihaela, P. et al. StringTie enables improved reconstruction of a transcriptome from RNA-seq reads. Nat. Biotechnol. 33, 290–295 (2015).

    Article  CAS  Google Scholar 

  74. Yu, G., Wang, L. G., Han, Y. & He, Q. Y. clusterProfiler: an R package for comparing biological themes among gene clusters. OMICS 16, 284–287 (2012).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  75. Ramirez, F. et al. deepTools2: a next generation web server for deep-sequencing data analysis. Nucleic Acids Res. 44, W160–W165 (2016).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  76. Kim, E. et al. MouseNet v2: a database of gene networks for studying the laboratory mouse and eight other model vertebrates. Nucleic Acids Res. 44, D848–D854 (2016).

    Article  CAS  PubMed  Google Scholar 

  77. Yu, H. et al. NetMiner—an ensemble pipeline for building genome-wide and high-quality gene co-expression network using massive-scale RNA-seq samples. PLoS ONE 13, e0192613 (2018).

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  78. Shen, F., Cheek, C. & Chandrasekaran, S. Dynamic network modeling of stem cell metabolism. Methods Mol. Biol. 1975, 305–320 (2019).

    Article  CAS  PubMed  Google Scholar 

  79. Chong, J. et al. MetaboAnalyst 4.0: towards more transparent and integrative metabolomics analysis. Nucleic Acids Res. 46, W486–w494 (2018).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

Download references

Acknowledgements

We thank J. Sheng, L. Shen, D. Ye, Y. Ni, X. Huang, M. Guan, Y. Yang, C. Navdeep and A. Intlekofer for helpful discussion and sharing of facilities. We thank G. Daley and M. Teitell for their long-term support. J. Zhang is supported by the National Key Research and Development Program of China (2018YFA0107100). Z.H. is supported by grants from National Natural Science Foundation of China (92057209). J. Zhang is also supported by the National Key Research and Development Program of China (2018YFA0107103 and 2018YFC1005002), the National Natural Science Foundation projects of China (31871453 and 91857116), Zhejiang Innovation Team grant (2019R01004) and the Zhejiang Natural Science Foundation projects of China (LR19C120001). Z.H. is also supported by grants from National Key R&D Program of China (2019YFA0802102), Tsinghua-Peking Center for Life Sciences (100084) and Beijing Frontier Research Center for Biological Structure.

Author information

Authors and Affiliations

Authors

Contributions

J. Zhang and Z.H. designed and supervised the study. J. Zhao, K.Y., L.Z., Y.X., L.C., Z.S., Y.Z., Y.Q., S.J., H.P., M.Z. and J.C. performed the experiments. J. Zhao and L.Z. performed sample preparation and metabolomics results data analysis. K.Y. and Z.H. developed the metabolomics method, performed the metabolomics experiments and data analysis. H.-Y.F., J. Zhang, Z.H., C.Z., C.C., W.T. and D.-W.L. performed the bioinformatics analysis. J. Zhang, Z.H., H.L., W.X., H.-Y.F., D.Z., X.F., S.C. and Y.Z. contributed to writing and discussing the manuscript.

Corresponding authors

Correspondence to Zeping Hu or Jin Zhang.

Ethics declarations

Competing interests

The authors declare no competing interests.

Additional information

Peer review information Nature Metabolism thanks Navdeep Chandel and the other, anonymous, reviewer(s) for their contribution to the peer review of this work. Primary Handling Editors: Ashley Castellanos-Jankiewicz; Pooja Jha.

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

Extended data

Extended Data Fig. 1 Metabolomics sample gradient titration, and metabolomics profiling for in vivo derived embryos and in vitro cultured embryo stem cells.

(a, b) Metabolites from 2.5 K to 80 K mouse embryonic stem cells or 15 to 240 zygotes were extracted and titrated for targeted metabolomics. Heatmap showing relative metabolite abundance normalized with MetaboAnalyst 4.0. Zy: zygote. (c) Representative metabolite SAM was shown for the correlation between the ES cell number used and relative mass spectrometry intensity obtained. R is the Pearson correlation coefficient. Data in c are from n = 3 biological replicates. Data are mean ± SAM. SAM:S-Adenosyl-methionine. (d) A plot showing R2 and -log (p-value) of each detected metabolite in the ES cell titration experiment. The significance level or p-value of Pearson correlation coefficients was obtained using cor.test function in R. (e) Representative metabolites were shown for correlation between the number of embryos and relative mass spectrometry intensity obtained. R is the correlation coefficient. (f) Flow cytometry showing tdTomato positive 2C-like cells (2CLC) and GFP positive ES cells (ESC). (g) The GSH/GSSG ratio in 2-cell and BC embryos. Data in g are from n = 3 biological replicates; Data are mean ± SEM. Statistical significance was determined by two-tailed unpaired t-test. BC: blastocyst. (h) Relative methionine levels (signal peak areas normalized to total metabolites) in oocytes and 2 C embryos. Data in h are from n = 3 biological replicates; Data are mean ± SEM. Statistical significance was determined by two-tailed unpaired t-test. (i) The SAM/SAH ratio in 2-cell and BC embryos. Data in i are from n = 3 biological replicates; Data are mean ± SEM. Statistical significance was determined by two-tailed paired t-test.

Source data

Extended Data Fig. 2 Analysis of metabolites and the corresponding metabolic enzyme genes in the TCA cycle in 2-cell and blastocyst embryos.

(a-e) Abundance of metabolites in the TCA cycle. Data in a-e are from n = 3 biological replicates; Data are mean ± SEM. Statistical significance was determined by two-tailed unpaired t-test. (f) The TCA cycle pathway with differential metabolites and genes indicated by the arrows. (g-m) Expression of the TCA cycle metabolic genes in 2-cell and ICM of blastocyst. Data in g-m are from n = 4 biological replicates; Data are mean ± SEM. Statistical significance was determined by two-tailed unpaired t-test. ICM: Inner Cell Mass.

Source data

Extended Data Fig. 3 Abundance of metabolites and expression the corresponding metabolic enzyme genes in the purine metabolism pathway in 2-cell and blastocyst embryos.

(a-j) Abundance of metabolites in the purine metabolism pathway. Data in a-j are from n = 3 biological replicates; Data are mean ± SEM. Statistical significance was determined by two-tailed unpaired t-test. (k) The purine metabolism pathways with differential metabolites and genes indicated by the arrows. (l-n) Expression of the purine metabolism genes in the 2-cell embryos and blastocyst ICM. Data in l-n are from n = 4 biological replicates; Data are mean ± SEM. Statistical significance was determined by two-tailed unpaired t-test.

Source data

Extended Data Fig. 4 Metabolite abundance and metabolic gene expression in one carbon metabolism and redox metabolism-related pathways in 2-cell and blastocyst embryos.

(a-g) Abundance of metabolites in one carbon metabolism, glutathione metabolism and polyamine metabolism pathways. Data in a-g are from n = 3 biological replicates; Data are mean ± SEM. Statistical significance was determined by two-tailed unpaired t-test. (h) The above metabolism pathways with differential metabolites and genes indicated by the arrows. (i-j) Expression of the above pathway metabolism genes in the 2-cell embryos and ICM of blastocyst. Data in i-j are from n = 4 biological replicates; Data are mean ± SEM. Statistical significance was determined by two-tailed unpaired t-test.

Source data

Extended Data Fig. 5 Metabolic network analysis and metabolomics analysis with 2CLCs.

(a, b) Metabolic network of 2 C and BC embryos constructed from metabolomics data as the method before21. Deletions of metabolic enzyme genes were simulated, and the differential sensitivity analysis was presented in b. (c) The PCA analysis of metabolomics from 2-cell, blastocyst (BC) embryos, 2-cell like cells (2CLC) and ES cells. (d) Schematics for obtaining the tdTomato+ 2CLCs from the 2 C transcription factor Dux inducible ES cell line (Dox-iDux ESC) and the tdTomato- ESCs. (e) Flow cytometry analysis showing the induced tdTomato+ 2CLCs upon addition of 1 μg/ml doxycycline(dox) for 24 hours. (f) qRT-PCR showing higher expression of 2 C genes in 2CLCs obtained above compared with ESCs. Data in f are from n = 3 biological replicates; Data are mean ± SEM. Statistical significance was determined by two-way(ANOVA) with Sidak’s multiple comparisons post-test. (g) The PCA analysis of metabolomics from 2-cell embryos, blastocyst embryos (BC), 2CLCs from the inducible Dux line or 2CLC (iDux), and ESCs. (h) Heatmap of the metabolomics analysis of the 2CLC and ESC samples. (i) Relative abundance of GSH, GSSG, and spermidine from the 2CLC and ESC samples described above. Data in i are from n = 3 biological replicates; Data are mean ± SEM. Statistical significance was determined by two-way ANOVA with Sidak’s multiple comparisons post-test. (j) The ratio of GSH/GSSG from the 2CLC and ESC samples described above. Data in j are from n = 3 biological experiments; Data are mean ± SEM. Statistical significance was determined by two-tailed unpaired t-test.

Source data

Extended Data Fig. 6 Single cell RNA-seq analysis, epigenetics analysis of metabolic genes, and anabolic metabolism analysis during pre-implantation embryo development.

(a) Genome-wide KEGG pathway analysis showing dynamic gene expression in different developmental stages using publicly available single cell RNA-seq data34. Blast: blastocyst. KEGG secondary category pathways are shown. (b) All metabolic genes are clustered into 6 clusters (C1-C6) by k-means analysis using single-cell RNA-seq data as in a. (c) Enriched KEGG terms for Cluster 1 (blastocyst stage), Cluster 5 (2-cell stage) and Cluster 6 (zygote stage) as defined in b. (d) GSEA analysis of the TCA cycle and Oxidative Phosphorylation between 2CLCs (tomato positive) and ESCs (tomato negative). (e) Boxplots showing expression levels of all OxPhos genes in different mouse embryo development stages analyzed from the same bulk RNA-seq data as in a. Data in e are from n = 4 biological replicates; In e, the center line is the median, the bottom of the box is the 25th percentile boundary, the top of the box the 75th, and the whiskers define the bounds of the data that are not considered outliers, with outliers defined as greater/lesser than ± 1.5 x IQR, where IQR = inter quartile range.v. Statistical significance was determined by Wilcox signed rank test. (f) Left: Representative images showing the NADH/NAD + ratio in the developing embryos. Scale bar: 50 µm. Right: Normalized F405 nm/F488 nm of different stages of embryos. Data are mean ± SEM. (g) Boxplots showing expression levels of all Ribosomal genes in different mouse embryo development stages analyzed from the same bulk RNA-seq data as above. Data in e are from n = 4 biological replicates; In g, the center line is the median, the bottom of the box is the 25th percentile boundary, the top of the box the 75th, and the whiskers define the bounds of the data that are not considered outliers, with outliers defined as greater/lesser than ± 1.5 x IQR, where IQR = inter quartile range.v. Statistical significance was determined by Wilcox signed rank test. (h) Translation activity of different stage embryos stained with OP-Puromycin. Representative images of full z-series confocal max projections of embryos are shown. Data in h are from n = 2 biological experiments with similar results; Scale bar: 50 μm. (i) Analysis of publicly available ATAC-seq data for the developmental stage-specific metabolic gene clusters defined in Fig. 2b. (j) Schematic diagram for constructing transcription factor-metabolic genes (TF-MG) regulatory network in each developmental stage. (k) Heatmap showing gene expression patterns of a few hub TFs identified in early embryo development as in Fig. 2d. (l) Gene regulatory networks between TFs or epigenetic factors and metabolic genes for 2-cell, four-cell, 8-cell/morula embryos and ICM in blastocyst embryos.

Source data

Extended Data Fig. 7 Absolute concentration of L-2-HG, and concentration titration for treating embryos.

(a) The extracted-ion chromatogram of L-2-HG and D-2-HG standards (concentration 100 nM). It shows clear separation between these two enantiomers. (b) The absolute concentration of L-2-HG and D-2-HG in MII, zygote, 2-cell and blastocyst embryos. (c) Reported absolute concentration of 2-HG in various cell lines, tumor and tissues. (d) Schematic illustration of Tunel experimental approach. Zygotes were collected 16 hours after injection of hCG and cultured in KSOM with different concentration L-2-HG. (e) Tunel assay was performed at different stage embryos with different concentration. Embryos were cultured in KSOM with L-2-HG at 0 mM, 0.15 mM, 0.3 mM, 0.45 mM and 0.6 mM. Representative images of full z-series confocal max projections of embryos in three independent experiments are shown. The scale bar is 50 µm.

Source data

Extended Data Fig. 8 Expression of Ldh, Mdh and L2hgdh/D2hgdh mRNA and their protein during early embryo development.

(a) mRNA expression of Ldh, Mdh and L2hgdh/D2hgdh genes from RNA-seq results of early embryos. Data are from n = 4 biological replicates; Data are mean ± SEM. (b) qRT-PCR of Ldhb expression in early embryos. Data are from n = 2 biological experiments with similar results; Data are mean ± SEM. (c) Western blotting of LDHB and L2HGHD in early embryos. Equal microgram of protein from protein lysate extracted from equal number of embryos were used for loading.

Source data

Supplementary information

Reporting Summary

Supplementary Table 1

Sensitivity of reaction.

Supplementary Table 2

qPCR primers.

Supplementary Table 3

siRNA sequence.

Supplementary Data 1–3

2C and BC metabolism analysis, 2C (idux+), ES cell metabolism analysis and gene transcripts per million reads of RNA-seq with 2-HG and α-KG.

Source data

Source Data Fig. 1

Statistical source data.

Source Data Fig. 3

Statistical source data.

Source Data Fig. 4

Statistical source data.

Source Data Fig. 5

Statistical source data.

Source Data Extended Data Fig. 1

Statistical source data.

Source Data Extended Data Fig. 2

Statistical source data.

Source Data Extended Data Fig. 3

Statistical source data.

Source Data Extended Data Fig. 4

Statistical source data.

Source Data Extended Data Fig. 5

Statistical source data.

Source Data Extended Data Fig. 6

Statistical source data.

Source Data Extended Data Fig. 7

Statistical source data.

Source Data Extended Data Fig. 8

Statistical source data.

Source Data Extended Data Fig. 8

Unprocessed western blots and/or gels.

Rights and permissions

Reprints and permissions

About this article

Check for updates. Verify currency and authenticity via CrossMark

Cite this article

Zhao, J., Yao, K., Yu, H. et al. Metabolic remodelling during early mouse embryo development. Nat Metab 3, 1372–1384 (2021). https://doi.org/10.1038/s42255-021-00464-x

Download citation

  • Received:

  • Accepted:

  • Published:

  • Issue Date:

  • DOI: https://doi.org/10.1038/s42255-021-00464-x

This article is cited by

Search

Quick links

Nature Briefing: Translational Research

Sign up for the Nature Briefing: Translational Research newsletter — top stories in biotechnology, drug discovery and pharma.

Get what matters in translational research, free to your inbox weekly. Sign up for Nature Briefing: Translational Research