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:

Control of intestinal stem cell function and proliferation by mitochondrial pyruvate metabolism

Abstract

Most differentiated cells convert glucose to pyruvate in the cytosol through glycolysis, followed by pyruvate oxidation in the mitochondria. These processes are linked by the mitochondrial pyruvate carrier (MPC), which is required for efficient mitochondrial pyruvate uptake. In contrast, proliferative cells, including many cancer and stem cells, perform glycolysis robustly but limit fractional mitochondrial pyruvate oxidation. We sought to understand the role this transition from glycolysis to pyruvate oxidation plays in stem cell maintenance and differentiation. Loss of the MPC in Lgr5-EGFP-positive stem cells, or treatment of intestinal organoids with an MPC inhibitor, increases proliferation and expands the stem cell compartment. Similarly, genetic deletion of the MPC in Drosophila intestinal stem cells also increases proliferation, whereas MPC overexpression suppresses stem cell proliferation. These data demonstrate that limiting mitochondrial pyruvate metabolism is necessary and sufficient to maintain the proliferation of intestinal stem cells.

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: MPC expression is low in intestinal stem cells and increases following differentiation.
Figure 2: Drosophila MPC regulates intestinal stem cell proliferation.
Figure 3: MPC deletion in the Lgr5-EGFP compartment increases intestinal stem cell maintenance and proliferation.
Figure 4: In vitro loss of the MPC increases stem cell function and organoid formation.
Figure 5: In vitro loss of the MPC alters protein expression and metabolism.
Figure 6: In vitro inhibition of MPC activity increases organoid formation.

Similar content being viewed by others

Accession codes

Primary accessions

Gene Expression Omnibus

References

  1. Warburg, O. On respiratory impairment in cancer cells. Science 124, 269–270 (1956).

    CAS  PubMed  Google Scholar 

  2. 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  Google Scholar 

  3. Sullivan, L. B. et al. Supporting aspartate biosynthesis is an essential function of respiration in proliferating cells. Cell 162, 552–563 (2015).

    Article  CAS  Google Scholar 

  4. Flavell, R. B. Mitochondrion as a multifunctional organelle. Nature 230, 504–506 (1971).

    Article  CAS  Google Scholar 

  5. Martínez-Reyes, I. et al. TCA cycle and mitochondrial membrane potential are necessary for diverse biological functions. Mol. Cell 61, 199–209 (2016).

    Article  Google Scholar 

  6. Simsek, T. et al. The distinct metabolic profile of hematopoietic stem cells reflects their location in a hypoxic niche. Cell Stem Cell 7, 380–390 (2010).

    Article  CAS  Google Scholar 

  7. 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  Google Scholar 

  8. Stringari, C. et al. Metabolic trajectory of cellular differentiation in small intestine by Phasor Fluorescence Lifetime Microscopy of NADH. Sci. Rep. 2, 568 (2012).

    Article  Google Scholar 

  9. Fan, Y.-Y. et al. A bioassay to measure energy metabolism in mouse colonic crypts, organoids, and sorted stem cells. Am. J. Physiol. Gastrointest. Liver Physiol. 309, G1–G9 (2015).

    Article  CAS  Google Scholar 

  10. Rodríguez-Colman, M. J. et al. Interplay between metabolic identities in the intestinal crypt supports stem cell function. Nature 543, 424–427 (2017).

    Article  Google Scholar 

  11. Berger, E. et al. Mitochondrial function controls intestinal epithelial stemness and proliferation. Nat. Commun. 7, 13171 (2016).

    Article  Google Scholar 

  12. Bricker, D. K. et al. A mitochondrial pyruvate carrier required for pyruvate uptake in yeast, Drosophila, and humans. Science 337, 96–100 (2012).

    Article  CAS  Google Scholar 

  13. Herzig, S. et al. Identification and functional expression of the mitochondrial pyruvate carrier. Science 337, 93–96 (2012).

    Article  CAS  Google Scholar 

  14. Schell, J. C. et al. A role for the mitochondrial pyruvate carrier as a repressor of the Warburg effect and colon cancer cell growth. Mol. Cell 56, 400–413 (2014).

    Article  CAS  Google Scholar 

  15. Sato, T. et al. Single Lgr5 stem cells build crypt-villus structures in vitro without a mesenchymal niche. Nature 459, 262–265 (2009).

    Article  CAS  Google Scholar 

  16. Sato, T. & Clevers, H. Primary mouse small intestinal epithelial cell cultures. Methods Mol. Biol. 945, 319–328 (2013).

    Article  Google Scholar 

  17. Uhlen, M. et al. Towards a knowledge-based Human Protein Atlas. Nat. Biotechnol. 28, 1248–1250 (2010).

    Article  CAS  Google Scholar 

  18. Uhlen, M., Pontén, F. & Lindskog, C. Charting the human proteome: understanding disease using a tissue-based atlas. Science 347, 1274–1274 (2015).

    Article  Google Scholar 

  19. Jiang, H. & Edgar, B. A. Intestinal stem cell function in Drosophila and mice. Curr. Opin. Genet. Dev. 22, 354–360 (2012).

    Article  CAS  Google Scholar 

  20. Li, H. & Jasper, H. Gastrointestinal stem cells in health and disease: from flies to humans. Dis. Model Mech. 9, 487–499 (2016).

    Article  Google Scholar 

  21. Dutta, D. et al. Regional cell-specific transcriptome mapping reveals regulatory complexity in the adult Drosophila midgut. Cell Rep. 12, 346–358 (2015).

    Article  CAS  Google Scholar 

  22. Buchon, N. et al. Morphological and molecular characterization of adult midgut compartmentalization in Drosophila. Cell Rep. 3, 1725–1738 (2013).

    Article  CAS  Google Scholar 

  23. Wu, J. S. & Luo, L. A protocol for mosaic analysis with a repressible cell marker (MARCM) in Drosophila. Nat. Protoc. 1, 2583–2589 (2006).

    Article  CAS  Google Scholar 

  24. Micchelli, C. A. & Perrimon, N. Evidence that stem cells reside in the adult Drosophila midgut epithelium. Nature 439, 475–479 (2006).

    Article  CAS  Google Scholar 

  25. Ohlstein, B. & Spradling, A. The adult Drosophila posterior midgut is maintained by pluripotent stem cells. Nature 439, 470–474 (2006).

    Article  CAS  Google Scholar 

  26. Li, H. et al. Drosophila larvae synthesize the putative oncometabolite L-2-hydroxyglutarate during normal developmental growth. Proc. Natl Acad. Sci. USA 114, 1353–1358 (2017).

    Article  CAS  Google Scholar 

  27. Micchelli, C. A. Whole-mount immunostaining of the adult Drosophila gastrointestinal tract. Methods 68, 273–279 (2014).

    Article  CAS  Google Scholar 

  28. Vigueira, P. A. et al. Mitochondrial pyruvate carrier 2 hypomorphism in mice leads to defects in glucose-stimulated insulin secretion. Cell Rep. 7, 2042–2053 (2014).

    Article  CAS  Google Scholar 

  29. Vacanti, N. M. et al. Regulation of substrate utilization by the mitochondrial pyruvate carrier. Mol. Cell 56, 425–435 (2014).

    Article  CAS  Google Scholar 

  30. Bender, T., Pena, G. & Martinou, J.-C. Regulation of mitochondrial pyruvate uptake by alternative pyruvate carrier complexes. EMBO J. 34, 911–924 (2015).

    Article  CAS  Google Scholar 

  31. Yang, C. et al. Glutamine oxidation maintains the TCA cycle and cell survival during impaired mitochondrial pyruvate transport. Mol. Cell 56, 414–424 (2014).

    Article  CAS  Google Scholar 

  32. Yin, X. et al. Niche-independent high-purity cultures of Lgr5 + intestinal stem cells and their progeny. Nat. Methods 11, 106–112 (2014).

    Article  CAS  Google Scholar 

  33. Aires, C. C. P. et al. Pyruvate uptake is inhibited by valproic acid and metabolites in mitochondrial membranes. FEBS Lett. 582, 3359–3366 (2008).

    Article  CAS  Google Scholar 

  34. Benavides, J., Martin, A., Ugarte, M. & Valdivieso, F. Inhibition by valproic acid of pyruvate uptake by brain mitochondria. Biochem. Pharmacol. 31, 1633–1636 (1982).

    Article  CAS  Google Scholar 

  35. Dailey, M. J. Nutrient-induced intestinal adaption and its effect in obesity. Physiol. Behav. 136, 74–78 (2014).

    Article  CAS  Google Scholar 

  36. Beyaz, S. et al. High-fat diet enhances stemness and tumorigenicity of intestinal progenitors. Nature 531, 53–58 (2016).

    Article  CAS  Google Scholar 

  37. Cao, F. et al. Transcriptional and functional profiling of human embryonic stem cell-derived cardiomyocytes. PLoS ONE 3, e3474 (2008).

    Article  Google Scholar 

  38. Chen, B.-Z. et al. Identification of microRNAs expressed highly in pancreatic islet-like cell clusters differentiated from human embryonic stem cells. Cell Biol. Int. 35, 29–37 (2011).

    Article  CAS  Google Scholar 

  39. Ralston, A. et al. Gata3 regulates trophoblast development downstream of Tead4 and in parallel to Cdx2. Development 137, 395–403 (2010).

    Article  CAS  Google Scholar 

  40. Muntean, A. G. et al. The PAF complex synergizes with MLL fusion proteins at HOX loci to promote leukemogenesis. Cancer Cell 17, 609–621 (2010).

    Article  CAS  Google Scholar 

  41. Keller, M. A. et al. Transcriptional regulatory network analysis of developing human erythroid progenitors reveals patterns of coregulation and potential transcriptional regulators. Physiol. Genomics 28, 114–128 (2006).

    Article  CAS  Google Scholar 

  42. Tateno, H. et al. Glycome diagnosis of human induced pluripotent stem cells using lectin microarray. J. Biol. Chem. 286, 20345–20353 (2011).

    Article  CAS  Google Scholar 

  43. Nishino, K. et al. DNA methylation dynamics in human induced pluripotent stem cells over time. PLoS Genet. 7, e1002085 (2011).

    Article  CAS  Google Scholar 

  44. Saito, S. et al. Possible linkages between the inner and outer cellular states of human induced pluripotent stem cells. BMC Syst. Biol. 5 (Suppl. 1), S17 (2011).

  45. Wang, X.-M. et al. The gene expression profiles of induced pluripotent stem cells from individuals with childhood cerebral adrenoleukodystrophy are consistent with proposed mechanisms of pathogenesis. Stem Cell Res. Ther. 3, 39 (2012).

    Article  Google Scholar 

  46. Flores, A. et al. Lactate dehydrogenase activity drives hair follicle stem cell activation. Nat. Cell Biol. http://dx.doi.org/10.1038/ncb3575 (2017).

  47. Camarda, R. et al. Inhibition of fatty acid oxidation as a therapy for MYC-overexpressing triple-negative breast cancer. Nat. Med. 22, 427–432 (2016).

    Article  CAS  Google Scholar 

  48. Boroughs, L. K. & DeBerardinis, R. J. Metabolic pathways promoting cancer cell survival and growth. Nat. Cell Biol. 17, 351–359 (2015).

    Article  CAS  Google Scholar 

  49. Edmunds, L. R. et al. c-Myc programs fatty acid metabolism and dictates acetyl-CoA abundance and fate. J. Biol. Chem. 289, 25382–25392 (2014).

    Article  CAS  Google Scholar 

  50. Pate, K. T. et al. Wnt signaling directs a metabolic program of glycolysis and angiogenesis in colon cancer. EMBO J. 33, 1454–1473 (2014).

    Article  CAS  Google Scholar 

  51. Gerstein, M. B. et al. Architecture of the human regulatory network derived from ENCODE data. Nature 489, 91–100 (2012).

    Article  CAS  Google Scholar 

  52. Wang, J. et al. Factorbook.org: a Wiki-based database for transcription factor-binding data generated by the ENCODE consortium. Nucleic Acids Res. 41, D171–D176 (2013).

    Article  CAS  Google Scholar 

  53. Wang, J. et al. Sequence features and chromatin structure around the genomic regions bound by 119 human transcription factors. Genome Res. 22, 1798–1812 (2012).

    Article  CAS  Google Scholar 

  54. Karolchik, D. et al. The UCSC Genome Browser database: 2014 update. Nucleic Acids Res. 42, D764–D770 (2014).

    Article  CAS  Google Scholar 

  55. McCool, K. W., Xu, X., Singer, D. B., Murdoch, F. E. & Fritsch, M. K. The role of histone acetylation in regulating early gene expression patterns during early embryonic stem cell differentiation. J. Biol. Chem. 282, 6696–6706 (2007).

    Article  CAS  Google Scholar 

  56. Roostaee, A., Benoit, Y. D., Boudjadi, S. & Beaulieu, J.-F. Epigenetics in intestinal epithelial cell renewal. J. Cell. Physiol. 231, 2361–2367 (2016).

    Article  CAS  Google Scholar 

  57. Golob, J. L., Paige, S. L., Muskheli, V., Pabon, L. & Murry, C. E. Chromatin remodeling during mouse and human embryonic stem cell differentiation. Dev. Dyn. 237, 1389–1398 (2008).

    Article  CAS  Google Scholar 

  58. Barker, N. et al. Identification of stem cells in small intestine and colon by marker gene Lgr5. Nature 449, 1003–1007 (2007).

    Article  CAS  Google Scholar 

  59. Gray, L. R. et al. Hepatic mitochondrial pyruvate carrier 1 is required for efficient regulation of gluconeogenesis and whole-body glucose homeostasis. Cell Metab. 22, 669–681 (2015).

    Article  CAS  Google Scholar 

  60. Moolenbeek, C. & Ruitenberg, E. J. The ‘Swiss roll’: a simple technique for histological studies of the rodent intestine. Lab. Anim. 15, 57–59 (1981).

    Article  CAS  Google Scholar 

  61. Daniels, R. W., Rossano, A. J., Macleod, G. T. & Ganetzky, B. Expression of multiple transgenes from a single construct using viral 2A peptides in Drosophila. PLoS ONE 9, e100637 (2014).

    Article  Google Scholar 

  62. Sato, T. et al. Paneth cells constitute the niche for Lgr5 stem cells in intestinal crypts. Nature 469, 415–418 (2011).

    Article  CAS  Google Scholar 

  63. Ootani, A. et al. Sustained in vitro intestinal epithelial culture within a Wnt-dependent stem cell niche. Nat. Med. 15, 701–706 (2009).

    Article  CAS  Google Scholar 

  64. Simmini, S. et al. Transformation of intestinal stem cells into gastric stem cells on loss of transcription factor Cdx2. Nat. Commun. 5, 5728 (2014).

    Article  CAS  Google Scholar 

  65. Weber, K., Mock, U., Petrowitz, B., Bartsch, U. & Fehse, B. Lentiviral gene ontology (LeGO) vectors equipped with novel drug-selectable fluorescent proteins: new building blocks for cell marking and multi-gene analysis. Gene Ther. 17, 511–520 (2010).

    Article  CAS  Google Scholar 

Download references

Acknowledgements

We thank B. Edgar (University of Utah, USA) for stocks and reagents, C. Micchelli (Washington University School of Medicine, USA) for providing the Notch RNAi line, K. Beebe for helpful advice and comments on the Drosophila intestinal studies, G. Lam for establishing the Drosophila MPC overexpression strain, O. Yilmaz and D. Sabatini for assistance and insight into intestinal stem cell metabolism, D. Tantin for critiques and comments, members of the Rutter laboratory for assistance and advice, J. O’Shea, R. Orbus and C. DeHeer for assistance with NanoString, W. Swiatek for mouse assistance, ARUP Institute for Clinical and Experimental Pathology, and S. R. Tripp and E. Hammond for histology; L. Nikolova at the University of Utah Electron Microscopy Core Laboratory performed electron microscopy; mass spectrometry analysis was performed at the Mass Spectrometry and Proteomics Core Facility at the University of Utah. Mass spectrometry equipment was obtained through NCRR Shared Instrumentation Grant no. 1 S10 RR020883-01, 1 S10 RR025532-01A1, NIH 1 S10OD021505-01 (J.E.C.) and the Diabetes and Metabolism Center at the University of Utah. This study was conducted with support from the Biorepository and Molecular Pathology Shared Resource supported by the Cancer Center Support Grant awarded to the Huntsman Cancer Institute by the National Cancer Institute of the National Institutes of Health. Nanostring transcript analysis utilized the Molecular Diagnostics Section of the Biorepository and Molecular Pathology Shared Resource and was supported by the National Cancer Institute of the National Institutes of Health under Award Number P30CA042014 (the content is solely the responsibility of the authors and does not necessarily represent the official views of the NIH). J. Marvin at the University of Utah Flow Cytometry Facility carried out flow sorting (National Cancer Institute through Award Number 5P30CA042014-24, National Center for Research Resources of the National Institutes of Health under Award Number 1S10RR026802-01). Funding was also provided by HHMI (J.R.), Treadwell (J.R.) and RO1GM094232 (to J.R. and C.S.T.). J.C.S. was supported by an NIH Developmental Biology Training Grant (5T32 HD07491) and a University of Utah Graduate Research Fellowship. D.R.W. was supported by a University of Utah Graduate Research Fellowship.

Author information

Authors and Affiliations

Authors

Contributions

Conceptualization, J.C.S., D.R.W., C.S.T. and J.R.; methodology, J.C.S., D.R.W., C.B., H.Z., P.W., J.T., A.F., J.M., L.K.S., C.S.E., K.A.O., D.D., P.K., M.P.B., D.Y.L., J.E.C., H.R.C., W.E.L., C.S.T. and J.R.; investigation, J.C.S., D.R.W., C.B., H.Z., P.W., J.T., A.F., J.M., L.K.S., C.S.E., R.M., D.D. and P.K.; formal analysis, J.C.S., D.R.W., C.B., P.W., T.C.W., R.M., L.J., R.J.D. and J.E.C.; writing—original draft, review and editing, J.C.S., D.R.W., C.S.T. and J.R.; funding acquisition, C.S.T. and J.R.; resources, D.Y.L., J.C., C.S.T. and J.R.; supervision, C.S.T. and J.R.

Corresponding author

Correspondence to Jared Rutter.

Ethics declarations

Competing interests

The authors declare no competing financial interests.

Integrated supplementary information

Supplementary Figure 1 Additional images of flow gating and IHC in mouse and human.

(a) Gating startegy for sorting of Lgr5-EGFP positive stem cells for transcript abundance. Top showing final gating for collection of GFP-low, GFP-mid, and GFP-high cells for transcript abundance. Bottom showing full gating strategy exclusion of cell debris and purification of singlet cells. (b) IHC of VDAC1 and MPC1 throughout the mouse small intestine from proximal to distal showing staining of crypt base and differentiating transient amplifying cells. (scale bar = 20 μm). (c) Human Protein Atlas (http://www.proteinatlas.org) images of duodenum, small intestine, and colon stained for TFAM, NRF1, PDK1, and CK20 (scale bar = 20 μm (b), 100 μm (c)).

Supplementary Figure 2 Drosophila MPC regulates stem cell proliferation.

(a) esg-GAL4 used to target RNAi as indicated or MPC overexpression (o/e) under infected conditions (scale bar = 50 μm). pHH3 + cells was quantified per intestine. (Control n = 10 (no infection) Control (with infection) n = 10, MPC O/E n = 10, LDH RNAi n = 10 MPC RNAi n = 10 PDH RNAi n = 10) (b) Western blot analysis to detect dMPC1 protein in control animals, dMPC1 mutants, HSP70-GAL4 controls, and HSP70 > UAS-dMPC1-dMPC2 transgenic animals following a 30-minute heat treatment and four hour recovery. Data are mean ± s.e.m. P < 0.05, P < 0.01, P < 0.001. All p values were calculated using Student’s t-test. Unprocessed western blots are provided in Supplementary Figure 7.

Supplementary Figure 3 MPC1 allele and additional quantification of in vivo phenotype.

(a) Schematic of MPC1 allele used for inducible knockout. (b) Close-up images of villi tip from MPC1 knockout stained for H&E, alcian blue, MPC1, VDAC and CK20 (scale bar = 20 μm). (c) Body weight for control and MPC1 knockout animals 30 days post-treatment. (n = 5 male control and knockout, n = 6 female control, = 3 knockout) (d) Total crypt + villi height and crypt height from individual regions of the small intestine. (n = 9 control, = 10 knockout for crypt + villi, n = 10 control, = 9 knockout for crypt height from each region) (e) Quantification of Alcian Blue positive cells per area of the proximal small intestine. (n = 10 control and knockout) (f) Quantification fo BrdU positive cells per crypt of proximal small intestine. (n = 9 control, = 10 knockout) (g) Images of Olfm4 and Lgr5 in situ in control and MPC1 knockout. (scale bar = 50μm) (h) Additional quantification of in situ staining. (n = 9 control, n = 10 knockout) (i) Quantification of Lrig1 IHC staining in control and MPC1 knockout proximal small intestine. (n = 9 control, n = 10 knockout) Data are mean ± s.e.m. P < 0.05, P < 0.01, P < 0.001. All p values were calculated using Student’s t-test.

Supplementary Figure 4 Additional characterization of in vito Mpc1 Lgr5-KO.

(a) Flow cytometry profile for organoids from control and MPC1 knockout cultures showing GFP positivity. (b) Long-term cultures retain increased growth ability throughout extended passaging (over 2 months) (n = 15 control and knockout, scale bar = 200 μm) (c) Control and MPC1 knockout organoid western blots for MPC1 and MPC2 and pyruvate dehydrogenase (phospho 293 and total). (d) Transcript abundance of MPC1 knockout organoids showing stem and differentiation marks (n = 3). (e) Quantification of specific Acetyl Histone H3 marks in control and knockout organoids (paired control set to 1.0) (n = 8 organoids). (f) Steady state metabolite abundance for control and MPC1 knockout organoids (n = 6 control and knockout). Data are mean ± s.e.m. P < 0.05, P < 0.01, P < 0.001. All p values were calculated using Student’s t-test. Unprocessed western blots are provided in Supplementary Figure 7.

Supplementary Figure 5 Effects of UK-5099 in maintaining organoids.

(a) Organoid formation from different regions of small intestine and colon with treatment conditions (n = 6 per condition, p values by t-test with respect to wENR for each location). (b) Loss of passaging efficiency in crypts maintained in wENR compared to UK-5099. When identical cell numbers are passaged normally, wENR treated are more likely to fail to passage. Images are of multiple matrigel/organoid spots of identical size (5uL) on a 6 well plate tiled to make one image (organoids from n = 3 wells were counted and number per well used for quantification, scale bar = 2000 μm). (c) Formation of hyperintense lumens in crypt organoids grown in wENR compared to UK-5099 3 days after changing media to remove CHIR99021 and Valproic acid (n = 69 total organoids from 6 individual wells). (d) Dose curves for individual drugs in supporting organoid formation from crypts (n = 8 wENR, all other conditions n = 6, p value by t-test with respect to wENR). Data are mean ± s.e.m. P < 0.05, P < 0.01, P < 0.001. All p values were calculated using Student’s t-test.

Supplementary Figure 6 Effects of UK-5099 and MPC over expression.

(a) Flux profiling of U-C13 Glucose incorporation into Citrate for tMEF and C2C12 cell lines treated with UK-5099 or Valproic Acid over 4 hours (n = 3 per condition per timepoint). (b) Representative images of infected Lgr5-EGFP (green) organoids with either Empty Vector-mCherry (in red) or MPC1-iRFP (in blue) and MPC2 mCherry (in red). Traces below represent intensity of each color over the length of the line outlined by white dotted line in the image. (n = 4 organoids with at least 15 infected cells per organoid counted). (c) ENCODE track for Myc and associated transcription factors localized to the Mpc1 promoter. (d) Model where crypt homeostasis involves low pyruvate oxidation at the base which increases during differentiation. MPC1 loss leads to reduced pyruvate oxidation, increased glycolysis, increased proliferation, and an expansion of the stem cell compartment within the crypt. Data are mean ± s.e.m. P < 0.05, P < 0.01, P < 0.001. All p values were calculated using Student’s t-test.

Supplementary information

Supplementary Information

Supplementary Information (PDF 23221 kb)

Supplementary Information

Life Sciences Reporting Summary (PDF 77 kb)

Supplementary Table 1

Supplementary Information (XLS 26 kb)

Supplementary Table 2

Supplementary Information (XLS 22 kb)

Supplementary Table 3

Supplementary Information (XLS 21 kb)

Supplementary Table 4

Supplementary Information (XLS 22 kb)

Supplementary Table 5

Supplementary Information (XLSX 42 kb)

Supplementary Table 6

Supplementary Information (XLSX 20 kb)

Supplementary Table 7

Supplementary Information (XLSX 57 kb)

Rights and permissions

Reprints and permissions

About this article

Check for updates. Verify currency and authenticity via CrossMark

Cite this article

Schell, J., Wisidagama, D., Bensard, C. et al. Control of intestinal stem cell function and proliferation by mitochondrial pyruvate metabolism. Nat Cell Biol 19, 1027–1036 (2017). https://doi.org/10.1038/ncb3593

Download citation

  • Received:

  • Accepted:

  • Published:

  • Issue Date:

  • DOI: https://doi.org/10.1038/ncb3593

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