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.

  • Letter
  • Published:

Glis1 facilitates induction of pluripotency via an epigenome–metabolome–epigenome signalling cascade

An Author Correction to this article was published on 08 October 2020

This article has been updated

Abstract

Somatic cell reprogramming provides insight into basic principles of cell fate determination, which remain poorly understood. Here we show that the transcription factor Glis1 induces multi-level epigenetic and metabolic remodelling in stem cells that facilitates the induction of pluripotency. We find that Glis1 enables reprogramming of senescent cells into pluripotent cells and improves genome stability. During early phases of reprogramming, Glis1 directly binds to and opens chromatin at glycolytic genes, whereas it closes chromatin at somatic genes to upregulate glycolysis. Subsequently, higher glycolytic flux enhances cellular acetyl-CoA and lactate levels, thereby enhancing acetylation (H3K27Ac) and lactylation (H3K18la) at so-called ‘second-wave’ and pluripotency gene loci, opening them up to facilitate cellular reprogramming. Our work highlights Glis1 as a powerful reprogramming factor, and reveals an epigenome–metabolome–epigenome signalling cascade that involves the glycolysis-driven coordination of histone acetylation and lactylation in the context of cell fate determination.

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: Glis1 activates glycolysis genes and suppresses the somatic cell state during somatic cell reprogramming.
Fig. 2: Glycolytic genes are downstream targets of Glis1 during somatic cell reprogramming.
Fig. 3: Glis1 switches the metabolic state during somatic cell reprogramming.
Fig. 4: Glis1 increases histone H3K27Ac and H3K18la at pluripotency loci, opening and activating pluripotent genes.

Similar content being viewed by others

Data availability

The data supporting the findings of this study are available from the corresponding author on reasonable request. RNA-seq and ChIP–seq data are available in the Gene Expression Omnibus database under the accession number GSE131692. ATAC–seq raw sequence data reported in this paper have been deposited in the Genome Sequence Archive49 at the Beijing Institute of Genomics (BIG) Data Center50, BIG, Chinese Academy of Sciences, under accession number CRA002469, which is publicly accessible at https://bigd.big.ac.cn/gsa. Source data for Figs. 14 and Extended Data Figs. 16 are presented within the paper. Source data are provided with this paper.

Change history

References

  1. Takahashi, K. & Yamanaka, S. Induction of pluripotent stem cells from mouse embryonic and adult fibroblast cultures by defined factors. Cell 126, 663–676 (2006).

    Article  CAS  PubMed  Google Scholar 

  2. Takahashi, K. et al. Induction of pluripotent stem cells from adult human fibroblasts by defined factors. Cell 131, 861–872 (2007).

    Article  CAS  PubMed  Google Scholar 

  3. Zhao, Y. et al. A XEN-like state bridges somatic cells to pluripotency during chemical reprogramming. Cell 163, 1678–1691 (2015).

    Article  CAS  PubMed  Google Scholar 

  4. Polo, J. M. et al. A molecular roadmap of reprogramming somatic cells into iPS cells. Cell 151, 1617–1632 (2012).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  5. Sardina, J. L. et al. Transcription factors drive Tet2-mediated enhancer demethylation to reprogram cell fate. Cell Stem Cell 23, 727–741 e9 (2018).

    Article  CAS  PubMed  Google Scholar 

  6. Li, D. et al. Chromatin accessibility dynamics during iPSC reprogramming. Cell Stem Cell 21, 819–833 e6 (2017).

    Article  CAS  PubMed  Google Scholar 

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

    Article  CAS  PubMed  Google Scholar 

  8. Folmes, C. D. et al. Somatic oxidative bioenergetics transitions into pluripotency-dependent glycolysis to facilitate nuclear reprogramming. Cell Metab. 14, 264–271 (2011).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  9. Mathieu, J. et al. Hypoxia-inducible factors have distinct and stage-specific roles during reprogramming of human cells to pluripotency. Cell Stem Cell 14, 592–605 (2014).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  10. Sone, M. et al. Hybrid cellular metabolism coordinated by Zic3 and Esrrb synergistically enhances induction of naive pluripotency. Cell Metab. 25, 1103–1117 e6 (2017).

    Article  CAS  PubMed  Google Scholar 

  11. Wang, L. et al. Fatty acid synthesis is critical for stem cell pluripotency via promoting mitochondrial fission. EMBO J. 36, 1330–1347 (2017).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  12. Wu, Y. et al. Srebp-1 Interacts with c-Myc to enhance somatic cell reprogramming. Stem Cells 34, 83–92 (2016).

    Article  CAS  PubMed  Google Scholar 

  13. Wu, Y. et al. Phospholipid remodeling is critical for stem cell pluripotency by facilitating mesenchymal-to-epithelial transition. Sci. Adv. 5, eaax7525 (2019).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  14. Li, R. et al. A mesenchymal-to-epithelial transition initiates and is required for the nuclear reprogramming of mouse fibroblasts. Cell Stem Cell 7, 51–63 (2010).

    Article  CAS  PubMed  Google Scholar 

  15. Liu, J. et al. The oncogene c-Jun impedes somatic cell reprogramming. Nat. Cell Biol. 17, 856–867 (2015).

    Article  CAS  PubMed  Google Scholar 

  16. Liu, X. et al. Sequential introduction of reprogramming factors reveals a time-sensitive requirement for individual factors and a sequential EMT-MET mechanism for optimal reprogramming. Nat. Cell Biol. 15, 829–838 (2013).

    Article  CAS  PubMed  Google Scholar 

  17. Takahashi, K. et al. Induction of pluripotency in human somatic cells via a transient state resembling primitive streak-like mesendoderm. Nat. Commun. 5, 3678 (2014).

    Article  CAS  PubMed  Google Scholar 

  18. Li, X. et al. Direct reprogramming of fibroblasts via a chemically induced XEN-like state. Cell Stem Cell 21, 264–273 e7 (2017).

    Article  CAS  PubMed  Google Scholar 

  19. Moussaieff, A. et al. Glycolysis-mediated changes in acetyl-CoA and histone acetylation control the early differentiation of embryonic stem cells. Cell Metab. 21, 392–402 (2015).

    Article  CAS  PubMed  Google Scholar 

  20. TeSlaa, T. et al. alpha-Ketoglutarate accelerates the initial differentiation of primed human pluripotent stem cells. Cell Metab. 24, 485–493 (2016).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  21. Mathieu, J. & Ruohola-Baker, H. Metabolic remodeling during the loss and acquisition of pluripotency. Development 144, 541–551 (2017).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  22. Ryall, J. G., Cliff, T., Dalton, S. & Sartorelli, V. Metabolic reprogramming of stem cell epigenetics. Cell Stem Cell 17, 651–662 (2015).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  23. Zhang, D. et al. Metabolic regulation of gene expression by histone lactylation. Nature 574, 575–580 (2019).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  24. Yoshioka, N. et al. Efficient generation of human iPSCs by a synthetic self-replicative RNA. Cell Stem Cell 13, 246–254 (2013).

    Article  CAS  PubMed  Google Scholar 

  25. Maekawa, M. et al. Direct reprogramming of somatic cells is promoted by maternal transcription factor Glis1. Nature 474, 225–229 (2011).

    Article  CAS  PubMed  Google Scholar 

  26. Kim, Y. S. et al. Identification of Glis1, a novel Gli-related, Kruppel-like zinc finger protein containing transactivation and repressor functions. J. Biol. Chem. 277, 30901–30913 (2002).

    Article  CAS  PubMed  Google Scholar 

  27. Stadtfeld, M., Maherali, N., Breault, D. T. & Hochedlinger, K. Defining molecular cornerstones during fibroblast to iPS cell reprogramming in mouse. Cell Stem Cell 2, 230–240 (2008).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  28. Wei, Z. et al. Klf4 organizes long-range chromosomal interactions with the oct4 locus in reprogramming and pluripotency. Cell Stem Cell 13, 36–47 (2013).

    Article  CAS  PubMed  Google Scholar 

  29. Morris, J. P. T. et al. alpha-Ketoglutarate links p53 to cell fate during tumour suppression. Nature 573, 595–599 (2019).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

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

    Article  CAS  PubMed  Google Scholar 

  31. Wellen, K. E. et al. ATP-citrate lyase links cellular metabolism to histone acetylation. Science 324, 1076–1080 (2009).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  32. Zhuang, Q. et al. NCoR/SMRT co-repressors cooperate with c-MYC to create an epigenetic barrier to somatic cell reprogramming. Nat. Cell Biol. 20, 400–412 (2018).

    Article  CAS  PubMed  Google Scholar 

  33. Tie, F. et al. CBP-mediated acetylation of histone H3 lysine 27 antagonizes Drosophila polycomb silencing. Development 136, 3131–3141 (2009).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  34. Le, R. et al. Enhanced telomere rejuvenation in pluripotent cells reprogrammed via nuclear transfer relative to induced pluripotent stem cells. Cell Stem Cell 14, 27–39 (2014).

    Article  CAS  PubMed  Google Scholar 

  35. Zhang, W. et al. Lactate is a natural suppressor of RLR signaling by targeting MAVS. Cell 178, 176–189 e15 (2019).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  36. Flores, A. et al. Lactate dehydrogenase activity drives hair follicle stem cell activation. Nat. Cell Biol. 19, 1017–1026 (2017).

    Article  CAS  PubMed  Google Scholar 

  37. Gaffney, D. O. et al. Nonenzymatic lysine lactoylation of glycolytic enzymes. Cell Chem. Biol. 27, 206–213 e6 (2020).

    Article  CAS  PubMed  Google Scholar 

  38. Zhao, T. et al. Single-cell RNA-Seq reveals dynamic early embryonic-like programs during chemical reprogramming. Cell Stem Cell 23, 31–45 e7 (2018).

    Article  CAS  PubMed  Google Scholar 

  39. Trokovic, R., Weltner, J. & Otonkoski, T. Generation of iPSC line HEL24.3 from human neonatal foreskin fibroblasts. Stem Cell Res. 15, 266–268 (2015).

    Article  CAS  PubMed  Google Scholar 

  40. Chen, Z., Zhao, T. & Xu, Y. The genomic stability of induced pluripotent stem cells. Protein Cell 3, 271–277 (2012).

    Article  PubMed  PubMed Central  Google Scholar 

  41. Yoshihara, M., Hayashizaki, Y. & Murakawa, Y. Genomic instability of iPSCs: challenges towards their clinical applications. Stem Cell Rev. Rep. 13, 7–16 (2017).

    Article  CAS  PubMed  Google Scholar 

  42. Ying, Z. et al. Transient activation of mitoflashes modulates nanog at the early phase of somatic cell reprogramming. Cell Metab. 23, 220–226 (2016).

    Article  CAS  PubMed  Google Scholar 

  43. Wu, D. Y., Zhang, X. & Miao, Y. L. Reprogramming of aged cells into pluripotent stem cells by nuclear transfer. Methods Mol. Biol. 2045, 271–281 (2019).

    Article  CAS  PubMed  Google Scholar 

  44. Skamagki, M. et al. ZSCAN10 expression corrects the genomic instability of iPSCs from aged donors. Nat. Cell Biol. 19, 1037–1048 (2017).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  45. Vadnais, C. et al. Autocrine activation of the Wnt/beta-catenin pathway by CUX1 and GLIS1 in breast cancers. Biol. Open 3, 937–946 (2014).

    Article  PubMed  PubMed Central  Google Scholar 

  46. Urner, F. & Sakkas, D. Characterization of glycolysis and pentose phosphate pathway activity during sperm entry into the mouse oocyte. Biol. Reprod. 60, 973–978 (1999).

    Article  CAS  PubMed  Google Scholar 

  47. Comizzoli, P., Urner, F., Sakkas, D. & Renard, J. P. Up-regulation of glucose metabolism during male pronucleus formation determines the early onset of the s phase in bovine zygotes. Biol. Reprod. 68, 1934–1940 (2003).

    Article  CAS  PubMed  Google Scholar 

  48. Li, B. et al. Fructose-1,6-bisphosphatase opposes renal carcinoma progression. Nature 513, 251–255 (2014).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  49. Wang, Y. et al. GSA: Genome Sequence Archive. Genomics Proteomics Bioinformatics 15, 14–18 (2017).

    Article  PubMed  PubMed Central  Google Scholar 

  50. National Genomics Data Center Members and Partners. Database Resources of the National Genomics Data Center in 2020. Nucleic Acids Res. 48, D24–D33 (2020).

Download references

Acknowledgements

We thank J. Chen for providing pMXs-Glis1 plasmids. We also thank all the members in X.L.’s laboratory. This work was financially supported by the National Key Research and Development Program of China (grant no. 2018YFA0107100), the Strategic Priority Research Program of the Chinese Academy of Sciences (grant no. XDA16030505), the National Key Research and Development Program of China (grant nos. 2017YFA0106300, 2017YFA0102900, 2017YFC1001602, 2019YFA09004500, 2016YFA0100300), the National Natural Science Foundation projects of China (grant nos. U1601227, 31631163001, 31701281, 31701106, 31801168, 31900614, 31970709, 81901275), the Key Research Program of Frontier Sciences, Chinese Academy of Sciences (CAS) (grant no. QYZDB-SSW-SMC001), CAS STS Program KFJ-STS-QYZD-125, Guangzhou Health Care and Cooperative Innovation Major Project (grant no. 201704020218), Guangdong Province Science and Technology Program (grant nos. 2017B020230005, 2017A020215056, 2017B030314056, 2018A030313825, 2018GZR110103002, 2020A1515011200, 2020A1515010919, 2020A1515011410), Guangzhou Science and Technology Program (grant nos. 201707010178, 201807010067, 202002030277), a grant from Yangtse River Scholar Bonus Schemes (to X.L.) and CAS Youth Innovation Promotion Association (to K.C.).

Author information

Authors and Affiliations

Authors

Contributions

X.L. conceived, designed and supervised the project. L.L., K.C. and Y.W. designed and performed the experiments and analysed data. G.X., B.M., M.C., Zihuang Liu, Zijian Liu and Z.H. participated in plasmids construction. Q.L., Y.Z., J.Q., M.G., H.Y. and D.Z. participated in iPSC generation and ChIP experiments. T.W., G.P. and J.N. participated in ChIP–seq, RNA-seq and ATAC–seq data analysis. D.P. gave suggestions. C.Z., J.Z. and D.Y. participated in metabolome analysis. X.L., L.L., Y.W. and K.C. wrote the manuscript.

Corresponding author

Correspondence to Xingguo Liu.

Ethics declarations

Competing interests

The authors declare no competing interests.

Additional information

Peer review information Primary Handling Editor: Christoph Schmitt.

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

Extended data

Extended Data Fig. 1 Pluripotency identification of iPSCs derived from senescent somatic cells.

a, OG2 MEFs were infected with retroviral SKO plus Flag or Glis1. GFP+ colonies were counted on day12, day14, day16, day18, day20. Data are presented as the mean ± S.D. (n = 3, each group).Group differences are analyzed by the two-tailed Student’s t test. b, OG2 MEFs were infected with retroviral SKO plus Flag or Glis1. (left): GFP colony pictures on day35 during reprogramming; (right): GFP+ colonies were counted on the indicated days during reprogramming. Data are presented as the mean ± S.D. (n = 3, each group).Group differences are analyzed by the two-tailed Student’s t test. c, Bright and β-gal staining images of P2-MEFs, P7-MEFs, P8-Flag-MEFs and P8-Glis1 expressed stably MEFs (P8-Glis1-MEFs) (a senescent cell was circled in red in MEFs). Scale bar: 100 μm. One replicate (n = 1, each group) was used for bright and β-gal staining images analysis. n = 3 independent experiments were repeated with similar results (a–b). d, β-gal positive cells counting for P2-MEFs, P7- MEFs (left); P8-Flag-MEFs, P8-Glis1-MEFs (right). Data are presented as the mean ± S.D. (P2-MEFs, n=52; P7- MEFs, n=58; P8-Flag-MEFs, n=58; P8-Glis1-MEFs, n=75 fields, each group).Group differences are analyzed by the two-tailed Student’s t test. e, Cell proliferation capacity analysis of P2-MEFs, P7-MEFs (left); P8-Flag-MEFs, P8-Glis1-MEFs (right) by Edu. Data are presented as the mean ± S.D. (n = 3, each group).Group differences are analyzed by the two-tailed Student’s t test. f, RT-qPCR analysis for detecting the expression of p16, p21 and p53 in P8-Flag-MEFs or P8-Glis1-MEFs. Data are presented as the mean ± S.D. (n = 3, each group).Group differences are analyzed by the two-tailed Student’s t test. g, Western blot analysis of P16, P21 and P53 upon Glis1 overexpression in P8-Flag-MEFs or P8-Glis1-MEFs (n = 1 independent experiments). h, Oct4-GFP images and immunofluorescence staining for the indicated pluripotency markers (red) for P7-iPSCs and P8-iPSCs clones derived from OG2 MEFs (n = 6 clonies for each group). Scale bar: 50 μm (left); RT-qPCR showing endogenous expression of indicated pluripotency genes in MEFs, mouse embryonic stem cells (R1) and P7-iPSCs and P8-iPSCs clones (right).

Source data

Extended Data Fig. 2 ChIP-Seq analysis of Glis1 during somatic cell reprogramming.

a, DNA integrity assessment of P10 control-iPSCs and Glis1-iPSCs by comet assay. The experiment were performed twice, and the data are represented as mean ± S.E.M, (Ctrl, n=134; Glis1, n=187 cell nucleus). Group differences are analyzed by the two-tailed Student’s t test. b, RT-qPCR analysis for detecting the expression of endogenous Glis1 during SKO-mediated reprogramming. Data are presented as the mean ± S.D. (n = 3, each group).Group differences are analyzed by the two-tailed Student’s t test. c, RT-qPCR analysis for detecting the Glis1 knockdown efficiency in MEFs relative to shLuc control. Data are presented as the mean ± S.D. (n = 3, each group).Group differences are analyzed by the two-tailed Student’s t test. d, OG2 MEFs were infected with retroviral SKO and shRNAs against Luciferase (shLuc) or Glis1 (shGlis1). GFP+ colonies were counted on day 20. Data are presented as the mean ± S.D. (n = 3, each group).Group differences are analyzed by the two-tailed Student’s t test. e, Schematic maps of Glis1 truncations used in this study; numbers are for amino acids. f, OG2 MEFs were infected with retroviral SKO plus Glis1 wild-type or deletion mutants. GFP+ colonies were counted on day 20. Data are presented as the mean ± S.D. (n = 3, each group).Group differences are analyzed by the two-tailed Student’s t test. g, RT-qPCR analysis for detecting the Glis1 knockdown efficiency in MEFs relative to shLuc control in DOX-inducible knockdown system. Data are presented as the mean ± S.D. (n =4, each group).Group differences are analyzed by the two-tailed Student’s t test. h, OG2 MEFs were infected with retroviral SKO and DOX-inducible knockdown of Glis1. DOX was added immediately after infection until the indicated day or added from indicated day until day 20. The reprogramming efficiencies were compared with DOX-free control. Data are presented as the mean ± S.D. (n = 3, each group).Group differences are analyzed by the two-tailed Student’s t test. i, OG2 MEFs were infected with retroviral SKO and DOX-inducible Glis1. DOX was added immediately after infection until the indicated day or added from indicated day until day 20. The reprogramming efficiencies were compared with DOX-free control. Data are presented as the mean ± S.D. (n = 3, each group).Group differences are analyzed by the two-tailed Student’s t test. j, The Glis1-N-HA-tag enhanced iPSCs generation equally well as non-tag Glis1. GFP+ colonies were determined on day 20. Data are presented as the mean ± S.D. (n = 3, each group).Group differences are analyzed by the two-tailed Student’s t test. k, GO term analysis of Glis1 peak position to nearest genes by ChIP-seq. 1 replicate was used for Go term analysis in Glis1 ChIP-seq assay. n = 2 independent experiments were repeated with similar results (ac). l, Genome view of Glis1 binding at indicated gene loci by ChIP-seq. Scale bar, 5 kb. n = 2 independent experiments were repeated with similar results (kj). Pie chart indicating the number of genes upregulated, downregulated or unchanged upon Glis1 overexpression on day 8. 1 replicate was used for RNA-seq analysis. n = 2 independent experiments were repeated with similar results.

Source data

Extended Data Fig. 3 Glis1 suppressed somatic state genes during somatic cell reprogramming.

a, The binding of Glis1 on glycolytic process gene loci was determined by ChIP–qPCR. TF-NC represents negative control. Data are presented as the mean ± S.D. (n = 3, each group).Group differences are analyzed by the two-tailed Student’s t test. b, Time courses of qPCR analyses of somatic state gene expression levels by SKO plus Flag and SKO plus Glis1. Data are presented as the mean ± S.D. (Setbp1, Col11a1 n=3; other genes, n=4 each group).Group differences are analyzed by the two-tailed Student’s t test. c, RT-qPCR analysis for detecting the glycolysis genes knockdown efficiency in MEFs.

Source data

Extended Data Fig. 4 Glis1 did not suppress OCR during somatic cell reprogramming.

a, Mitochondrial stress test using Seahorse XF24 analyzer on day 8. SKO plus Flag as a control compared to SKO plus Glis1. Flag as the control to all group. Data are presented as the mean ± S.D. (n = 3, each group). b, Oxygen consumption rates (OCR) of mitochondrial maximal respiration quantified from the mitochondrial-stress test in (a) are shown. Data are presented as the mean ± S.D. (n = 3, each group).Group differences are analyzed by the two-tailed Student’s t test. c, Mitochondrial DNA copy numbers in MEFs with SKO plus Glis1 or SKO plus Flag. Data are presented as the mean ± S.D. (n = 3, each group). Group differences are analyzed by the two-tailed Student’s t test. d, Mitochondrial stress test using Seahorse XF24 analyzer on day 8. SKO plus shLuc as a control compared to SKO plus shGlis1, Flag as a control to all groups. Data are presented as the mean ± S.D. (n = 3, each group). e, Oxygen consumption rates (OCR) of mitochondrial maximal respiration quantified from the mitochondrial stress test in (d) are shown. Data are presented as the mean ± S.D. (n = 3, each group). Group differences are analyzed by the two-tailed Student’s t test. f, Flow cytometry analysis of SSEA1 in MEFs transduced with SKO in reprogramming on day3. (n = 3, each group). g, SSEA1- cells were infected with retroviral Flag or Glis1. GFP+ colonies were counted on day 20. Data are presented as the mean ± S.D. (n = 3, each group). Group differences are analyzed by the two-tailed Student’s t test. h, Mitochondrial stress test using Seahorse XF24 analyzer on day 8. SSEA1- plus Flag as a control compared to SSEA1- plus Glis1. Data are presented as the mean ± S.D. (n = 3, each group). i, Oxygen consumption rates (OCR) of mitochondrial maximal respiration quantified from the mitochondrial stress test in (h) are shown. Data are presented as the mean ± S.D. (n = 3, each group). Group differences are analyzed by the two-tailed Student’s t test. j, Glycolysis stress test of iPSCs by Seahorse XF24 analyzer. The resultant iPSCs of SKO plus Flag as a control compared to SKO plus Glis1. Data are presented as the mean ± S.D. (n = 3, each group). k, Extracellular acidification rates (ECAR) quantified from the glycolysis stress test in (j) are shown. Data are presented as the mean ± S.D. (n = 3, each group). Group differences are analyzed by the two-tailed Student’s t test. l, Mitochondrial stress test of iPSCs by Seahorse XF24 analyzer. The resultant iPSCs of SKO plus Flag as a control compared to SKO plus Glis1. Data are presented as the mean ± S.D. (n = 3, each group). m, Oxygen consumption rates (OCR) of mitochondrial maximal respiration quantified from the mitochondrial stress test in l, are shown. Data are presented as the mean ± S.D. (n = 3, each group). Group differences are analyzed by the two-tailed Student’s t test. n, Relative concentration of pentose phosphate pathway (PPP) metabolites and nucleotide-related molecules. Data are presented as the mean ± S.D. (n = 3, each group). Group differences are analyzed by the two-tailed Student’s t test. o, Relative band densities of H3K27Ac upon Glis1 overexpression and knockdown were quantified using Image J. Data are presented as the mean ± S.D. (n = 3, each group). Group differences are analyzed by the two-tailed Student’s t test. p, Western blot analysis of Pan Kla and H3K18la after 20 mM lactate supplementation and knockdown of Ldha. Data are presented as the mean ± S.D. n = 3 independent experiments were repeated with similar results. q, Relative band densities of Pan Kla and H3K18la upon Glis1 overexpression and knockdown were quantified using Image J. Data are presented as the mean ± S.D. (n = 3, each group). Group differences are analyzed by the two-tailed Student’s t test.

Source data

Extended Data Fig. 5 Glis1 enhances reprogramming by glycolysis-driven acetyl-CoA production.

(a) Western blot analysis of H3K27me3, H3K9me3 after Glis1 overexpression or knockdown on day 8 during somatic reprogramming. (n = 1, each group). (b) The effect of acetate on cell proliferation at different concentration. Data are presented as the mean ± S.D. (n = 3, each group). Group differences are analyzed by the two-tailed Student’s t test. (c) The effect of acetate on the level of acetyl-CoA at different concentrations. Data are presented as the mean ± S.D. (n = 3, each group). Group differences are analyzed by the two-tailed Student’s t test. (d) RT-qPCR analysis for detecting the Acly knockdown efficiency in MEFs. (e) OG2 MEFs were infected with retroviral SKO plus shLuc or shAcly addition of acetate. GFP+ colonies were counted on day 20. Data are presented as the mean ± S.D. (n = 3, each group). Group differences are analyzed by the two-tailed Student’s t test. (f) OG2 MEFs were infected with retroviral SKO plus Flag or Glis1 addition of VPA (0.5 mM). GFP+ colonies were counted on day 20. Data are presented as the mean ± S.D. (n = 3, each group). Group differences are analyzed by the two-tailed Student’s t test.(g) OG2 MEFs were infected with retroviral SKO plus shLuc or shGlis1 addition of VPA (0.5 mM). GFP+ colonies were counted on day 20. Data are presented as the mean ± S.D. (n = 3, each group). Group differences are analyzed by the two-tailed Student’s t test. (h) OG2 MEFs were infected with retroviral SKO plus Flag or Glis1 addition of TSA (1 nM). GFP+ colonies were counted on day 20. Data are presented as the mean ± S.D. (n = 3, each group). Group differences are analyzed by the two-tailed Student’s t test. (i) OG2 MEFs were infected with retroviral SKO plus shLuc or shGlis1 addition of TSA (1 nM). GFP+ colonies were counted on day 20. Data are presented as the mean ± S.D. (n = 3, each group). Group differences are analyzed by the two-tailed Student’s t test.

Source data

Extended Data Fig. 6 Glis1 increased histone lactylation and acetylation at Sall4 and Mycn loci promoting their activation.

(a) Genome views of H3K27Ac tag density at Sall4 and Mycn; red box on the tracks indicates H3K27Ac peaks. Scale bar, 2 kb. For IGV analysis, 1 replicate (n = 1, each group) was used for H3K27Ac analysis in ChIP-seq assay. n = 2 independent experiments were repeated with similar results.(b) ChIP–qPCR analysis of H3K27Ac at Sall4 and Mycn on day 8 in MEFs transduced with SKO plus Flag or Glis1. Data are presented as the mean ± S.D. (n = 3, each group). Group differences are analyzed by the two-tailed Student’s t test. (c) ChIP–qPCR analysis of H3K27Ac at Oct4, Sall4 and Mycn on day 8 in MEFs transduced with SKO plus shLuc or shGlis1. Data are presented as the mean ± S.D. (n = 3, each group). Group differences are analyzed by the two-tailed Student’s t test. (d) ChIP–qPCR analysis of Pan Kla and H3K18la at Oct4, Sall4 and Mycn on day 8 in MEFs transduced with SKO plus shLuc or shGlis1. Data are presented as the mean ± S.D. (Pan kla, n = 3; H3K18la, n=3–4 each group). Group differences are analyzed by the two-tailed Student’s t test.(e) ChIP–qPCR analysis of P300 at Oct4, Sall4, Mycn, Setbp1, Col1a1 and Col5a1 on day 8 in MEFs transduced with Flag or SKO plus Flag. Data are presented as the mean ± S.D. (n = 3, each group). Group differences are analyzed by the two-tailed Student’s t test. (f) Time course of qPCR analysis of endo-Oct4, Sall4, Mycn in MEFs transduced with SKO plus Flag or Glis1. Data are presented as the mean ± S.D. (n = 3, each group). Group differences are analyzed by the two-tailed Student’s t test. (g) The number of peaks defined in each of the OC/CO categories. Two replicate (n = 2, each group) was used for ATAC assay. (h) Time courses of qPCR analyses of T and Eomes in MEFs infected with retroviral SKO plus Flag or Glis1. Data are presented as the mean ± S.D. (n = 3, each group). Group differences are analyzed by the two-tailed Student’s t test.

Source data

Supplementary information

Supplementary Information

Supplementary Tables 1–4 and Fig. 1

Reporting Summary

Source data

Source Data Fig. 1

Statistical Source Data

Source Data Fig. 2

Statistical Source Data

Source Data Fig. 3

Statistical Source Data

Source Data Fig. 3

Unprocessed western blots

Source Data Fig. 4

Statistical Source Data

Source Data Extended Data Fig. 1

Statistical Source Data

Source Data Extended Data Fig. 1

Unprocessed Western Blots

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. 4

Unprocessed Western Blots

Source Data Extended Data Fig. 5

Statistical Source Data

Source Data Extended Data Fig. 5

Unprocessed Western Blots

Source Data Extended Data Fig. 6

Statistical Source Data

Rights and permissions

Reprints and permissions

About this article

Check for updates. Verify currency and authenticity via CrossMark

Cite this article

Li, L., Chen, K., Wang, T. et al. Glis1 facilitates induction of pluripotency via an epigenome–metabolome–epigenome signalling cascade. Nat Metab 2, 882–892 (2020). https://doi.org/10.1038/s42255-020-0267-9

Download citation

  • Received:

  • Accepted:

  • Published:

  • Issue Date:

  • DOI: https://doi.org/10.1038/s42255-020-0267-9

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