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 control of primed human pluripotent stem cell fate and function by the miR-200c–SIRT2 axis

Abstract

A hallmark of cancer cells is the metabolic switch from oxidative phosphorylation (OXPHOS) to glycolysis, a phenomenon referred to as the ‘Warburg effect’, which is also observed in primed human pluripotent stem cells (hPSCs). Here, we report that downregulation of SIRT2 and upregulation of SIRT1 is a molecular signature of primed hPSCs and that SIRT2 critically regulates metabolic reprogramming during induced pluripotency by targeting glycolytic enzymes including aldolase, glyceraldehyde-3-phosphate dehydrogenase, phosphoglycerate kinase, and enolase. Remarkably, knockdown of SIRT2 in human fibroblasts resulted in significantly decreased OXPHOS and increased glycolysis. In addition, we found that miR-200c-5p specifically targets SIRT2, downregulating its expression. Furthermore, SIRT2 overexpression in hPSCs significantly affected energy metabolism, altering stem cell functions such as pluripotent differentiation properties. Taken together, our results identify the miR-200c–SIRT2 axis as a key regulator of metabolic reprogramming (Warburg-like effect), via regulation of glycolytic enzymes, during human induced pluripotency and pluripotent stem cell function.

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

Figure 1: SIRT2 downregulation and SIRT1 upregulation is a molecular signature of human pluripotency.
Figure 2: SIRT2 regulates acetylation and enzymatic activity of glycolytic enzymes.
Figure 3: Acetylation status of K322 regulates AldoA activity.
Figure 4: SIRT2 influences metabolism and cell survival of hPSCs.
Figure 5: SIRT2 influences metabolism during early in vitro differentiation of hESCs.
Figure 6: SIRT2KD facilitates metabolic reprogramming in fibroblasts during the induced pluripotency.
Figure 7: SIRT2 influences somatic nuclear reprogramming through metabolic changes.
Figure 8: miR-200c directly targets SIRT2.

Similar content being viewed by others

Accession codes

Accessions

Gene Expression Omnibus

References

  1. Warburg, O., Wind, F. & Negelein, E. The metabolism of tumors in the body. J. Gen. Physiol. 8, 519–530 (1927).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  2. Warburg, O. On the origin of cancer cells. Science 123, 309–314 (1956).

    Article  CAS  PubMed  Google Scholar 

  3. Rafalski, V. A., Mancini, E. & Brunet, A. Energy metabolism and energy-sensing pathways in mammalian embryonic and adult stem cell fate. J. Cell Sci. 125, 5597–5608 (2012).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  4. Ito, K. & Suda, T. Metabolic requirements for the maintenance of self-renewing stem cells. Nat. Rev. Mol. Cell Biol. 15, 243–256 (2014).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  5. Burgess, R. J., Agathocleous, M. & Morrison, S. J. Metabolic regulation of stem cell function. J. Intern. Med. 276, 12–24 (2014).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  6. Zhang, J., Nuebel, E., Daley, G. Q., Koehler, C. M. & Teitell, M. A. Metabolic regulation in pluripotent stem cells during reprogramming and self-renewal. Cell Stem Cell 11, 589–595 (2012).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  7. Folmes, C. D., Dzeja, P. P., Nelson, T. J. & Terzic, A. Metabolic plasticity in stem cell homeostasis and differentiation. Cell Stem Cell 11, 596–606 (2012).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  8. Zhang, J. et al. UCP2 regulates energy metabolism and differentiation potential of human pluripotent stem cells. EMBO J. 30, 4860–4873 (2011).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  9. 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 

  10. Varum, S. et al. Energy metabolism in human pluripotent stem cells and their differentiated counterparts. PLoS ONE 6, e20914 (2011).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  11. 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 

  12. 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 

  13. Panopoulos, A. D. et al. The metabolome of induced pluripotent stem cells reveals metabolic changes occurring in somatic cell reprogramming. Cell Res. 22, 168–177 (2012).

    Article  CAS  PubMed  Google Scholar 

  14. Kim, S. C. et al. Substrate and functional diversity of lysine acetylation revealed by a proteomics survey. Mol. Cell 23, 607–618 (2006).

    Article  CAS  PubMed  Google Scholar 

  15. Zhao, S. et al. Regulation of cellular metabolism by protein lysine acetylation. Science 327, 1000–1004 (2010).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  16. Wang, Q. et al. Acetylation of metabolic enzymes coordinates carbon source utilization and metabolic flux. Science 327, 1004–1007 (2010).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  17. Guarente, L. The logic linking protein acetylation and metabolism. Cell Metab. 14, 151–153 (2011).

    Article  CAS  PubMed  Google Scholar 

  18. Guarente, L. & Franklin, H. Epstein Lecture: Sirtuins, aging, and medicine. N. Engl. J. Med. 364, 2235–2244 (2011).

    Article  CAS  PubMed  Google Scholar 

  19. Finkel, T., Deng, C. X. & Mostoslavsky, R. Recent progress in the biology and physiology of sirtuins. Nature 460, 587–591 (2009).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  20. Gatenby, R. A. & Gillies, R. J. Why do cancers have high aerobic glycolysis? Nat. Rev. Cancer 4, 891–899 (2004).

    Article  CAS  PubMed  Google Scholar 

  21. 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 

  22. Prigione, A. et al. HIF1α modulates cell fate reprogramming through early glycolytic shift and upregulation of PDK1-3 and PKM2. Stem Cells 32, 364–376 (2014).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  23. North, B. J., Marshall, B. L., Borra, M. T., Denu, J. M. & Verdin, E. The human Sir2 ortholog, SIRT2, is an NAD + -dependent tubulin deacetylase. Mol. Cell 11, 437–444 (2003).

    Article  CAS  PubMed  Google Scholar 

  24. Fathi, A. et al. Comprehensive gene expression analysis of human embryonic stem cells during differentiation into neural cells. PLoS ONE 6, e22856 (2011).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  25. Assou, S. et al. A meta-analysis of human embryonic stem cells transcriptome integrated into a web-based expression atlas. Stem Cells 25, 961–973 (2007).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  26. Li, Z. et al. Functional and transcriptional characterization of human embryonic stem cell-derived endothelial cells for treatment of myocardial infarction. PLoS ONE 4, e8443 (2009).

    Article  PubMed  PubMed Central  Google Scholar 

  27. Masaki, H. et al. Heterogeneity of pluripotent marker gene expression in colonies generated in human iPS cell induction culture. Stem Cell Res. 1, 105–115 (2007).

    Article  CAS  PubMed  Google Scholar 

  28. Barrett, T. et al. NCBI GEO: archive for functional genomics data sets–update. Nucleic Acids Res. 41, D991–D995 (2013).

    Article  CAS  PubMed  Google Scholar 

  29. Calvanese, V. et al. Sirtuin 1 regulation of developmental genes during differentiation of stem cells. Proc. Natl Acad. Sci. USA 107, 13736–13741 (2010).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  30. Lee, Y. L. et al. Sirtuin 1 facilitates generation of induced pluripotent stem cells from mouse embryonic fibroblasts through the miR-34a and p53 pathways. PLoS ONE 7, e45633 (2012).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  31. Si, X. et al. Activation of GSK3β by Sirt2 is required for early lineage commitment of mouse embryonic stem cell. PLoS ONE 8, e76699 (2013).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  32. Marchler-Bauer, A. et al. CDD: NCBI’s conserved domain database. Nucleic Acids Res. 43, D222–D226 (2015).

    Article  CAS  PubMed  Google Scholar 

  33. Gamblin, S. J. et al. Activity and specificity of human aldolases. J. Mol. Biol. 219, 573–576 (1991).

    Article  CAS  PubMed  Google Scholar 

  34. Zhang, J. et al. Measuring energy metabolism in cultured cells, including human pluripotent stem cells and differentiated cells. Nat. Protoc. 7, 1068–1085 (2012).

    Article  CAS  PubMed  Google Scholar 

  35. Park, K. S. et al. Transcription elongation factor Tcea3 regulates the pluripotent differentiation potential of mouse embryonic stem cells via the Lefty1-Nodal-Smad2 pathway. Stem Cells 31, 282–292 (2013).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  36. Ivanova, N. et al. Dissecting self-renewal in stem cells with RNA interference. Nature 442, 533–538 (2006).

    CAS  PubMed  Google Scholar 

  37. Mu, W. L. et al. Sox2 deacetylation by Sirt1 is involved in mouse somatic reprogramming. Stem Cells 33, 2135–2147 (2015).

    Article  CAS  PubMed  Google Scholar 

  38. Miranda, K. C. et al. A pattern-based method for the identification of MicroRNA binding sites and their corresponding heteroduplexes. Cell 126, 1203–1217 (2006).

    Article  CAS  PubMed  Google Scholar 

  39. Suh, M. R. et al. Human embryonic stem cells express a unique set of microRNAs. Dev. Biol. 270, 488–498 (2004).

    Article  CAS  PubMed  Google Scholar 

  40. Wang, G. et al. Critical regulation of miR-200/ZEB2 pathway in Oct4/Sox2-induced mesenchymal-to-epithelial transition and induced pluripotent stem cell generation. Proc. Natl Acad. Sci. USA 110, 2858–2863 (2013).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  41. Gomes, P., Outeiro, T. F. & Cavadas, C. Emerging role of Sirtuin 2 in the regulation of mammalian metabolism. Trends Pharmacol. Sci. 36, 756–768 (2015).

    Article  CAS  PubMed  Google Scholar 

  42. Xiong, Y. & Guan, K. L. Mechanistic insights into the regulation of metabolic enzymes by acetylation. J. Cell Biol. 198, 155–164 (2012).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  43. Xu, Y. et al. Oxidative stress activates SIRT2 to deacetylate and stimulate phosphoglycerate mutase. Cancer Res. 74, 3630–3642 (2014).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  44. Hallows, W. C., Yu, W. & Denu, J. M. Regulation of glycolytic enzyme phosphoglycerate mutase-1 by Sirt1 protein-mediated deacetylation. J. Biol. Chem. 287, 3850–3858 (2012).

    Article  CAS  PubMed  Google Scholar 

  45. Tsusaka, T. et al. Deacetylation of phosphoglycerate mutase in its distinct central region by SIRT2 down-regulates its enzymatic activity. Genes Cells 19, 766–777 (2014).

    Article  CAS  PubMed  Google Scholar 

  46. Li, T. Glyceraldehyde-3-phosphate dehydrogenase is activated by lysine 254 acetylation in response to glucose signal. J. Biol. Chem. 289, 3775–3785 (2014).

    Article  CAS  PubMed  Google Scholar 

  47. Somers, A. et al. Generation of transgene-free lung disease-specific human induced pluripotent stem cells using a single excisable lentiviral stem cell cassette. Stem Cells 28, 1728–1740 (2010).

    CAS  PubMed  PubMed Central  Google Scholar 

  48. 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 

  49. Kislinger, T. et al. Global survey of organ and organelle protein expression in mouse: combined proteomic and transcriptomic profiling. Cell 125, 173–186 (2006).

    Article  CAS  PubMed  Google Scholar 

  50. Zhou, J. Y. et al. Galectin-3 is a candidate biomarker for amyotrophic lateral sclerosis: discovery by a proteomics approach. J. Proteome Res. 9, 5133–5141 (2010).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  51. Kim, D. et al. Generation of human induced pluripotent stem cells by direct delivery of reprogramming proteins. Cell Stem Cell 4, 472–476 (2009).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

Download references

Acknowledgements

This work was supported by NIH grants (NS084869, NS070577, and GM101420) and the National Research Foundation of Korea (2011-0030043). The authors are grateful to members of the Kim laboratory for critical discussions.

Author information

Authors and Affiliations

Authors

Contributions

Y.C. and M.-J.H.: concept and design, collection and/or assembly of data, data analysis and interpretation, and writing. L.G., H.-J.C. and K.-S.K.: concept and design, data analysis and interpretation, and writing. J.Z., A.B., J.H.J., Y.J. and H.-C.J.: collection and/or assembly of data. C.-H.K., B.-G.K., R.L. and C.R.K.: data analysis and interpretation.

Corresponding author

Correspondence to Kwang-Soo Kim.

Ethics declarations

Competing interests

K.-S.K. is co-founder of NurrON Pharmaceuticals and the remaining authors have no financial conflict to disclose.

Integrated supplementary information

Supplementary Figure 1 Warburg-like effect in hESCs and hiPSCs compared to hDFs.

(a) Human ESCs (H9) and hiPSCs cultured under feeder-free condition were stained with specific antibodies against pluripotency markers (e.g., Oct4, Nanog, SSEA4, and TRA1-60) along with Hoechst nuclear staining. Scale bar, 100 μm. (b) Representative pictures of hESCs and hiPSCs. Scale bar, 100 μm. (c) In vitro spontaneous differentiation of hESCs and hiPSCs by culturing in serum-free ITSFn medium for 7 days. Immunostaining images (first and second row panels) show lineage specific markers for ectoderm (Otx2), mesoderm (Brachyury; B-T), and endoderm (Sox17). Scale bar, 100 μm. (d) Intracellular ATP levels were significantly lower in hiPSCs and hESCs than in the original fibroblasts. (mean ± s.e.m., n=3 biologically independent experiments, p < 0.005, two-tailed unpaired Student’s t-test). (e) Mitochondrial bioenergetics of parental hDFs and hiPSCs as well as hESCs assessed by Seahorse XF analyzer. (mean ± s.d., n=4 biologically independent experiments). (f) Expression levels of glucose transporters (GLUTs) including GLUT1 to GLUT7 in hDFs and hiPSCs as well as hESCs. (mean ± s.e.m., n=3 biologically independent experiments, p < 0.05; p < 0.01; p < 0.005; p < 0.001, one-way ANOVA with Bonferroni post-test). (g) Immunoprecipitation of hDF and hESCs proteins using antibodies against acetyl-Lys, followed by LC-MS/MS analyses to identify acetylated proteins. Statistics source data are in Supplementary Table 9. Unprocessed original scans of blots (d,g) are shown in Supplementary Fig. 9.

Supplementary Figure 2 CID spectra for the acetylated proteins shown in Supplementary Fig. 1g and Supplementary Table 1.

Peptides for tubulin, Fructose-bisphosphate aldolase, glyceraldehyde-3-phosphate dehydrogenase, phosphoglycerate kinase1, enolase, pyruvate kinase isozymes M1/M2 and ATP synthase were detected via combination of IP and LC-MS/MS analyses. IP was performed with anti-acetyl-Lys antibody.

Supplementary Figure 3 Meta-analysis of Sirtuin family expression in hESCs.

(a) Representative data showing SIRT2 expression changes between different cells. SIRT2 downregulation was observed in hPSCs compared to differentiated cells and original fibroblasts. (bf) Mean value scatter plot of expression levels of SIRT3 (b), SIRT4 (c), SIRT5, (d) SIRT6 (e), and SIRT7 (f) in hESC lines (n=25) and normal somatic cell lines (n=15) using results from a Database search (http://www.nextbio.com). All cell lines information is shown in Supplementary Table 5. (mean ± s.e.m., two-tailed unpaired Student’s t-test).

Supplementary Figure 4 Characterization of inducible SIRT2-GFP H9 hESCs.

(a) Plasmid map of doxycycline (Dox) inducible SIRT2-EGFP overexpression vector. (b) Expression levels of pluripotency markers in wild-type hESCs, hDFs, and SIRT2-GFP hESCs with or without Dox. (mean ± s.d., n=3 biologically independent experiments, p < 0.005, one-way ANOVA with Bonferroni post-test). (c) Effects of TGF-β on spontaneous differentiation of SIRT2-GFP hESCs. Cells were maintained in hESC culture conditions with or without TGF-β. TGF-β was added for 4 days and cells were immunostained for expression of the endodermal marker Sox17; Scale bar, 100 μm. (d) Expression levels of glycolytic enzymes in SIRT2-GFP hESCs with or without Dox analyzed by qRT-PCR. (mean ± s.d., n=3 biologically independent experiments, p < 0.005, two-tailed unpaired Student’s t-test). Statistics source data are in Supplementary Table 9.

Supplementary Figure 5 Effects of altered SIRT2 expression on acetylation of AldoA.

(ad) Detection of AldoA K111 (a, b) and K322 (c,d) acetylation by mass spectrometry analysis. Symbol @ indicates the acetylation site. (e) Myc-tagged AldoA, AldoAK111Q, and AldoAK322Q were each expressed in 293T cells. AldoA proteins were purified by IP with Myc antibody, and specific activity for AldoA was determined. (mean ± s.d., n=3 biologically independent experiments, p < 0.005, one-way ANOVA with Bonferroni post-test). (f) Myc-tagged AldoA, AldoAK111R, and AldoAK322R were each expressed in 293T cells co-expressing SIRT2 shRNA (SIRT2KD). AldoA proteins were purified by IP with Myc antibody and specific activity for AldoA was determined. (mean ± s.d., n=3 biologically independent experiments, p < 0.005, one-way ANOVA with Bonferroni post-test). Statistics source data are in Supplementary Table 9.

Supplementary Figure 6 Metabolic and functional characterization of hPSC lines following SIRT2 overexpression.

(a,c,e) Glycolytic bioenergetics of wild type (Mock) and inducible SIRT2-GFP cell lines from H7 hESCs (a) and two iPSC lines (c,e) with or without Dox were assessed by XF analyzer. (mean ± s.d., n=3 biologically independent experiments). (b,d,f) Basal glycolytic rate, glycolytic capacity and glycolytic reserve of mock and SIRT2OE from H7 hESCs (b) and two iPSC lines (d,f) with or without Dox are shown in (a, c, e), respectively. (mean ± s.d., n=3 biologically independent experiments, p < 0.05; p < 0.01, one-way ANOVA with Bonferroni post-test). (g) OCRs for two hESC lines (H9 and H7) and the hiPSC-1 line with or without Dox are shown. 1: Mock w/o Dox, 2: Mock with Dox, 3: SIRT2OE w/o Dox, 4: SIRT2OE with Dox. (mean ± s.d., n=3 biologically independent experiments, p < 0.05; p < 0.005, one-way ANOVA with Bonferroni post-test). (h) Cell proliferation of mock and SIRT2OE from H7 hESCs and two independent iPSC lines (hiPSC-1 and hiPSC-2) with or without Dox were analyzed by determining cell numbers every 2 days under ESC culture conditions. (mean ± s.d., n=3 biologically independent experiments, p < 0.01; p < 0.005, two-way ANOVA with Bonferroni post-test). Statistics source data are in Supplementary Table 9.

Supplementary Figure 7 SIRT2 influences metabolic signatures of early differentiation potential of hiPSCs.

(ac) Inducible SIRT2OE hiPSC-1 cells with or without Dox were induced to differentiate spontaneously by culturing in serum-free ITSFn medium for up to 4 days, and gene expression levels of pluripotency markers (Oct4, Nanog, and Rex1) (a), early-differentiation markers (Pax6, Brachyury (B-T), and Sox17) (b) and SIRT2 (c) were determined by qRT-PCR. (mean ± s.d., n=3 biologically independent experiments, p < 0.05, one-way ANOVA with Bonferroni post-test). (d) Glycolytic bioenergetics of mock and SIRT2OE hiPSC-1 cells with or without Dox were assessed using the Seahorse XF analyzer. (mean ± s.d., n=3 biologically independent experiments, p < 0.05, one-way ANOVA with Bonferroni post-test). (e) Extracellular lactate production of mock and SIRT2OE hiPSC-1 cells with or without Dox. (mean ± s.d., n=3 biologically independent experiments, p < 0.05; p < 0.01, one-way ANOVA with Bonferroni post-test). (f) Heatmaps depicting gene expression levels of markers representing ectoderm (Pax6, Map2, GFAP and AADC), endoderm (Foxa2, Sox17, AFP, CK8 and CK18), and mesoderm (Msx1 and B-T) in wild type (Mock) and inducible SIRT2OE hiPSC lines including hiPSC-1 and hiPSC-2 with or without Dox for up to 12 days under differentiation conditions. (n=2 biologically independent experiments). 1: Mock w/o Dox, 2: Mock with Dox, 3: SIRT2OE w/o Dox, 4: SIRT2OE with Dox. Statistics source data are in Supplementary Table 9.

Supplementary Figure 8 Effects of altered SIRT1 expression on metabolic reprogramming and iPSC generation.

(a) Plasmid map of doxycycline (Dox) inducible SIRT1-EGFP overexpression vector. (b) OCR for hDFs infected with wild type (Mock) or inducible SIRT1-GFP (SIRT1OE) with or without Dox at day 3 post transfection. (mean ± s.d., n=3 biologically independent experiments). (c,d) OCR/ECAR ratio (c), and relative OCR changes after FCCP injection (d) from Mock and SIRT1OE with or without Dox are shown in (b). (mean ± s.d., n=3 biologically independent experiments). (e,f) Effects of SIRT1KD or OE on iPSC generation. Upper: Efficiency of SIRT1KD or OE was confirmed by western blotting with anti-SIRT1 antibody. Lower: Representative pictures of AP-positive colonies at day 14 post-transduction. (mean ± s.e.m., n=3 biologically independent experiments, p < 0.005, two-tailed unpaired Student’s t-test). (g,h) OCR in hDF infected with Y4 and/or SIRT1OE at 3 (g) or 6 (h) days after transfection. (mean ± s.d., n=3 biologically independent experiments). Statistics source data are in Supplementary Table 9. Unprocessed original scans of blots (e,f) are shown in Supplementary Fig. 9.

Supplementary information

Supplementary Information

Supplementary Information (PDF 4966 kb)

Supplementary Table 1

Supplementary Information (XLSX 13 kb)

Supplementary Table 2

Supplementary Information (XLSX 11 kb)

Supplementary Table 3

Supplementary Information (XLSX 12 kb)

Supplementary Table 4

Supplementary Information (XLSX 16 kb)

Supplementary Table 5

Supplementary Information (XLSX 14 kb)

Supplementary Table 6

Supplementary Information (XLSX 9 kb)

Supplementary Table 7

Supplementary Information (XLSX 9 kb)

Supplementary Table 8

Supplementary Information (XLSX 11 kb)

Supplementary Table 9

Supplementary Information (XLSX 342 kb)

Rights and permissions

Reprints and permissions

About this article

Check for updates. Verify currency and authenticity via CrossMark

Cite this article

Cha, Y., Han, MJ., Cha, HJ. et al. Metabolic control of primed human pluripotent stem cell fate and function by the miR-200c–SIRT2 axis. Nat Cell Biol 19, 445–456 (2017). https://doi.org/10.1038/ncb3517

Download citation

  • Received:

  • Accepted:

  • Published:

  • Issue Date:

  • DOI: https://doi.org/10.1038/ncb3517

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