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:

FOXK1 and FOXK2 regulate aerobic glycolysis

Abstract

Adaptation to the environment and extraction of energy are essential for survival. Some species have found niches and specialized in using a particular source of energy, whereas others—including humans and several other mammals—have developed a high degree of flexibility1. A lot is known about the general metabolic fates of different substrates but we still lack a detailed mechanistic understanding of how cells adapt in their use of basic nutrients2. Here we show that the closely related fasting/starvation-induced forkhead transcription factors FOXK1 and FOXK2 induce aerobic glycolysis by upregulating the enzymatic machinery required for this (for example, hexokinase-2, phosphofructokinase, pyruvate kinase, and lactate dehydrogenase), while at the same time suppressing further oxidation of pyruvate in the mitochondria by increasing the activity of pyruvate dehydrogenase kinases 1 and 4. Together with suppression of the catalytic subunit of pyruvate dehydrogenase phosphatase 1 this leads to increased phosphorylation of the E1α regulatory subunit of the pyruvate dehydrogenase complex, which in turn inhibits further oxidation of pyruvate in the mitochondria—instead, pyruvate is reduced to lactate. Suppression of FOXK1 and FOXK2 induce the opposite phenotype. Both in vitro and in vivo experiments, including studies of primary human cells, show how FOXK1 and/or FOXK2 are likely to act as important regulators that reprogram cellular metabolism to induce aerobic glycolysis.

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: Regulation and metabolism in 3T3‐L1 adipocytes and L6 myotubes overexpressing or knocked down for FOXK1 or FOXK2.
Fig. 2: FOXK1 and FOXK2 induce aerobic glycolysis and lactate production.
Fig. 3: FOXK1 and FOXK2 regulate glycolysis.
Fig. 4: Regulation of aerobic glycolysis and lactate production by FOXK1 and FOXK2 in vivo and in primary human cells.

Similar content being viewed by others

Data availability

RNA-seq datasets are available at the Gene Expression Omnibus (GEO) website under accession number GSE114258. Proteomics data for 3T3-L1 cells overexpressing or knocked down for FOXK1 and FOXK2 can be found in Supplementary Tables 14. Source Data for animal experiments can be found in the online version of the paper. Uncropped images of immunoblots can be found in Supplementary Fig. 1. All other data are available from the corresponding authors upon reasonable request.

References

  1. Campbell, B. Human Evolution (Aldine Transactions, 2009).

  2. Goodpaster, B. H. & Sparks, L. M. Metabolic flexibility in health and disease. Cell Metab. 25, 1027–1036 (2017).

    Article  CAS  Google Scholar 

  3. Palm, W. & Thompson, C. B. Nutrient acquisition strategies of mammalian cells. Nature 546, 234–242 (2017).

    Article  CAS  ADS  Google Scholar 

  4. Thacker, S. V., Nickel, M. & DiGirolamo, M. Effects of food restriction on lactate production from glucose by rat adipocytes. Am. J. Physiol. 253, E336–E342 (1987).

    CAS  PubMed  Google Scholar 

  5. DiGirolamo, M., Newby, F. D. & Lovejoy, J. Lactate production in adipose tissue: a regulated function with extra-adipose implications. FASEB J. 6, 2405–2412 (1992).

    Article  CAS  Google Scholar 

  6. Wu, P. et al. Starvation and diabetes increase the amount of pyruvate dehydrogenase kinase isoenzyme 4 in rat heart. Biochem. J. 329, 197–201 (1998).

    Article  CAS  Google Scholar 

  7. Jeoung, N. H. et al. Role of pyruvate dehydrogenase kinase isoenzyme 4 (PDHK4) in glucose homoeostasis during starvation. Biochem. J. 397, 417–425 (2006).

    Article  CAS  Google Scholar 

  8. Hui, S. et al. Glucose feeds the TCA cycle via circulating lactate. Nature 551, 115–118 (2017).

    Article  ADS  Google Scholar 

  9. Holness, M. J. & Sugden, M. C. Regulation of pyruvate dehydrogenase complex activity by reversible phosphorylation. Biochem. Soc. Trans. 31, 1143–1151 (2003).

    Article  CAS  Google Scholar 

  10. 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  ADS  Google Scholar 

  11. Han, H., Pappin, D. J., Ross, P. L. & McLuckey, S. A. Electron transfer dissociation of iTRAQ labeled peptide ions. J. Proteome Res. 7, 3643–3648 (2008).

    Article  CAS  Google Scholar 

  12. Kamburov, A., Cavill, R., Ebbels, T. M., Herwig, R. & Keun, H. C. Integrated pathway-level analysis of transcriptomics and metabolomics data with IMPaLA. Bioinformatics 27, 2917–2918 (2011).

    Article  CAS  Google Scholar 

  13. Kim, J. W. & Dang, C. V. Cancer’s molecular sweet tooth and the Warburg effect. Cancer Res. 66, 8927–8930 (2006).

    Article  CAS  Google Scholar 

  14. Cederbaum, S. D. et al. Sensitivity to carbohydrate in a patient with familial intermittent lactic acidosis and pyruvate dehydrogenase deficiency. Pediatr. Res. 10, 713–720 (1976).

    Article  CAS  Google Scholar 

  15. Bonnefont, J. P. et al. α-ketoglutarate dehydrogenase deficiency presenting as congenital lactic acidosis. J. Pediatr. 121, 255–258 (1992).

    Article  CAS  Google Scholar 

  16. Jackson, C. B. et al. Mutations in SDHD lead to autosomal recessive encephalomyopathy and isolated mitochondrial complex II deficiency. J. Med. Genet. 51, 170–175 (2014).

    Article  CAS  Google Scholar 

  17. Raimundo, N. et al. Differential metabolic consequences of fumarate hydratase and respiratory chain defects. Biochim. Biophys. Acta 1782, 287–294 (2008).

    Article  CAS  Google Scholar 

  18. Mookerjee, S. A., Nicholls, D. G. & Brand, M. D. Determining maximum glycolytic capacity using extracellular flux measurements. PLoS ONE 11, e0152016 (2016).

    Article  Google Scholar 

  19. Zhang, S., Hulver, M. W., McMillan, R. P., Cline, M. A. & Gilbert, E. R. The pivotal role of pyruvate dehydrogenase kinases in metabolic flexibility. Nutr. Metab. (Lond.) 11, 10 (2014).

    Article  Google Scholar 

  20. Sugden, M. C. & Holness, M. J. Mechanisms underlying regulation of the expression and activities of the mammalian pyruvate dehydrogenase kinases. Arch. Physiol. Biochem. 112, 139–149 (2006).

    Article  CAS  Google Scholar 

  21. Roche, T. E. et al. Distinct regulatory properties of pyruvate dehydrogenase kinase and phosphatase isoforms. Prog. Nucleic Acid Res. Mol. Biol. 70, 33–75 (2001).

    Article  CAS  Google Scholar 

  22. Subhash, S. & Kanduri, C. GeneSCF: a real-time based functional enrichment tool with support for multiple organisms. BMC Bioinformatics 17, 365 (2016).

    Article  Google Scholar 

  23. Cao, Y., Rathmell, J. C. & Macintyre, A. N. Metabolic reprogramming towards aerobic glycolysis correlates with greater proliferative ability and resistance to metabolic inhibition in CD8 versus CD4 T cells. PLoS ONE 9, e104104 (2014).

    Article  ADS  Google Scholar 

  24. He, L. et al. mTORC1 promotes metabolic reprogramming by the suppression of GSK3-dependent Foxk1 phosphorylation. Mol. Cell 70, 949–960e944 (2018).

    Article  CAS  Google Scholar 

  25. Gladden, L. B. Lactate metabolism: a new paradigm for the third millennium. J. Physiol. (Lond.) 558, 5–30 (2004).

    Article  CAS  Google Scholar 

  26. Brooks, G. A. Cell-cell and intracellular lactate shuttles. J. Physiol. (Lond.) 587, 5591–5600 (2009).

    Article  CAS  Google Scholar 

  27. Pellerin, L. et al. Evidence supporting the existence of an activity-dependent astrocyte-neuron lactate shuttle. Dev. Neurosci. 20, 291–299 (1998).

    Article  CAS  Google Scholar 

  28. Smith, D. et al. Lactate: a preferred fuel for human brain metabolism in vivo. J. Cereb. Blood Flow Metab. 23, 658–664 (2003).

    Article  CAS  Google Scholar 

  29. Cederberg, A., Grände, M., Rhedin, M., Peng, X. R. & Enerbäck, S. In vitro differentiated adipocytes from a Foxc2 reporter knock-in mouse as screening tool. Transgenic Res. 18, 889–897 (2009).

    Article  CAS  Google Scholar 

  30. Silver, D. P. & Livingston, D. M. Self-excising retroviral vectors encoding the Cre recombinase overcome Cre-mediated cellular toxicity. Mol. Cell 8, 233–243 (2001).

    Article  CAS  Google Scholar 

  31. Ohsumi, J. et al. Troglitazone prevents the inhibitory effects of inflammatory cytokines on insulin-induced adipocyte differentiation in 3T3-L1 cells. Endocrinology 135, 2279–2282 (1994).

    Article  CAS  Google Scholar 

  32. Lee, S. et al. Berberine inhibits rat vascular smooth muscle cell proliferation and migration in vitro and improves neointima formation after balloon injury in vivo: Berberine improves neointima formation in a rat model. Atherosclerosis 186, 29–37 (2006).

    Article  CAS  Google Scholar 

  33. Bartesaghi, S. et al. Thermogenic activity of UCP1 in human white fat-derived beige adipocytes. Mol. Endocrinol. 29, 130–139 (2015).

    Article  Google Scholar 

  34. Pike Winer, L. S. & Wu, M. Rapid analysis of glycolytic and oxidative substrate flux of cancer cells in a microplate. PLoS ONE 9, e109916 (2014).

    Article  ADS  Google Scholar 

  35. Shi, J. & Kandror, K. V. Study of glucose uptake in adipose cells. Methods Mol. Biol. 456, 307–315 (2008).

    Article  Google Scholar 

  36. Rune, A. et al. Evidence against a sexual dimorphism in glucose and fatty acid metabolism in skeletal muscle cultures from age-matched men and post-menopausal women. Acta Physiol. 197, 207–215 (2009).

    Article  CAS  Google Scholar 

  37. Al-Khalili, L. et al. Insulin action in cultured human skeletal muscle cells during differentiation: assessment of cell surface GLUT4 and GLUT1 content. Cell. Mol. Life Sci. 60, 991–998 (2003).

    Article  CAS  Google Scholar 

  38. Jensen, T. C., Crosson, S. M., Kartha, P. M. & Brady, M. J. Specific desensitization of glycogen synthase activation by insulin in 3T3-L1 adipocytes. Connection between enzymatic activation and subcellular localization. J. Biol. Chem. 275, 40148–40154 (2000).

    Article  CAS  Google Scholar 

  39. Zeng, X. et al. Lysine-specific demethylase 1 promotes brown adipose tissue thermogenesis via repressing glucocorticoid activation. Genes Dev. 30, 1822–1836 (2016).

    Article  CAS  Google Scholar 

  40. Kazak, L. et al. A creatine-driven substrate cycle enhances energy expenditure and thermogenesis in beige fat. Cell 163, 643–655 (2015).

    Article  CAS  Google Scholar 

  41. Kim, D., Langmead, B. & Salzberg, S. L. HISAT: a fast spliced aligner with low memory requirements. Nat. Methods 12, 357–360 (2015).

    Article  CAS  Google Scholar 

  42. Harrow, J. et al. GENCODE: producing a reference annotation for ENCODE. Genome Biol. 7 (Suppl. 1), S4 (2006).

    Article  Google Scholar 

  43. Lassmann, T., Hayashizaki, Y. & Daub, C. O. SAMStat: monitoring biases in next generation sequencing data. Bioinformatics 27, 130–131 (2011).

    Article  CAS  Google Scholar 

  44. Liao, Y., Smyth, G. K. & Shi, W. featureCounts: an efficient general purpose program for assigning sequence reads to genomic features. Bioinformatics 30, 923–930 (2014).

    Article  CAS  Google Scholar 

  45. Robinson, M. D., McCarthy, D. J. & Smyth, G. K. edgeR: a Bioconductor package for differential expression analysis of digital gene expression data. Bioinformatics 26, 139–140 (2010).

    Article  CAS  Google Scholar 

  46. Pandey, R. R. et al. Kcnq1ot1 antisense noncoding RNA mediates lineage-specific transcriptional silencing through chromatin-level regulation. Mol. Cell 32, 232–246 (2008).

    Article  CAS  Google Scholar 

  47. Kwon, A. T., Arenillas, D. J., Worsley Hunt, R. & Wasserman, W. W. oPOSSUM-3: advanced analysis of regulatory motif over-representation across genes or ChIP-seq datasets. G3 (Bethesda) 2, 987–1002 (2012).

    Article  CAS  Google Scholar 

  48. Lakso, M. et al. Efficient in vivo manipulation of mouse genomic sequences at the zygote stage. Proc. Natl Acad. Sci. USA 93, 5860–5865 (1996).

    Article  CAS  ADS  Google Scholar 

  49. Liu, P., Jenkins, N. A. & Copeland, N. G. A highly efficient recombineering-based method for generating conditional knockout mutations. Genome Res. 13, 476–484 (2003).

    Article  CAS  Google Scholar 

  50. Ishikawa, M., Hosoi, R., Kobayashi, K., Nishimura, T. & Inoue, O. Rolipram depresses [(3)H]2-deoxyglucose uptake in mouse brain and heart in vivo. Eur. J. Nucl. Med. Mol. Imaging 29, 1212–1215 (2002).

    Article  CAS  Google Scholar 

  51. Kalland, M. E., Oberprieler, N. G., Vang, T., Taskén, K. & Torgersen, K. M. T cell-signaling network analysis reveals distinct differences between CD28 and CD2 costimulation responses in various subsets and in the MAPK pathway between resting and activated regulatory T cells. J. Immunol. 187, 5233–5245 (2011).

    Article  CAS  Google Scholar 

  52. Tadesse, F. G. et al. Unpredicted phenotypes of two mutants of the TcR DMF5. J. Immunol. Methods 425, 37–44 (2015).

    Article  CAS  Google Scholar 

  53. Ogilvie, R. W. & Feeback, D. L. A metachromatic dye-ATPase method for the simultaneous identification of skeletal muscle fiber types I, IIA, IIB and IIC. Stain Technol. 65, 231–241 (1990).

    Article  CAS  Google Scholar 

  54. Hauner, H., Skurk, T. & Wabitsch, M. Cultures of human adipose precursor cells. Methods Mol. Biol. 155, 239–247 (2001).

    CAS  PubMed  Google Scholar 

Download references

Acknowledgements

We thank G. Petersson for technical support and T. Gnad for help with glucose uptake measurements in human adipocytes; R. Palmer, B. Hallberg and J. Tetteh Siaw for support and advice regarding cancer cell lines; and J.-O. Jansson for supplying adipose tissue from Ob/Ob mice. S.E. is supported by the Swedish Research Council (2014–2516), The Knut and Alice Wallenberg Foundation, Sahlgrenska’s University Hospital (LUA-ALF), and Novo Nordisk Foundation. K.T. is supported by the Norwegian Cancer Society.

Author information

Authors and Affiliations

Authors

Contributions

V.S., H.M., W.Z., S.B., S.S., M.H., M.J.B., D.N., M.B., M.E.L., M.P.J., S.H.-B., T.M. and C.K. designed and executed experiments. A.P., J.N. and S.H-B. performed experiments with human adipocytes and myocytes. S.B., X.-R.P. and H.P. performed bioenergetics experiments. S.S. performed bioinformatics analysis of RNA-seq and proteomic datasets. K.T. and H.F. performed experiments with human T cells. S.E. designed experiments, established collaborations and wrote the manuscript with valuable input from all authors.

Corresponding author

Correspondence to Sven Enerbäck.

Ethics declarations

Competing interests

The authors declare no competing interests.

Additional information

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

Extended data figures and tables

Extended Data Fig. 1 Metabolic profile induced by FOXK1 or FOXK2.

a, Glucose uptake in 3T3-L1 adipocytes (on day 11) in response to doxycycline-induced expression of FOXK1 or FOXK2, with or without 100 nM insulin. Cells were differentiated and expression of FOXK1 or FOXK2 was induced by doxycycline at indicated days (d) during differentiation; n = 4. b, Glycerol release from 3T3-L1 adipocytes with knockdown of Foxk1 or Foxk2 compared with empty vector or shGFP; n = 4. c, Incorporation of glucose into glycogen in L6 myotubes in response to doxycycline-induced expression of FOXK1 or FOXK2; n = 3. d, Foxk2 mRNA levels in MEF adipocytes derived from wild type mice or mice lacking one Foxk2 allele (Foxk2fl/+-cre) or both Foxk2 alleles (Foxk2fl/fl-cre). e, Glucose uptake in MEF adipocytes from wild-type, Foxk2fl/+-cre or Foxk2fl/fl-cre mice; n = 4. f, Western blot of GLUT4 and GLUT1 in 3T3‐L1 preadipocytes (d = 0) and adipocytes (d = 7 and d = 11) in response to overexpression of FOXK1 or FOXK2. FOXK1 and FOXK2 were identified by their Flag tags. gj, Glucose uptake induced by FOXK1 or FOXK2. Comparison of glucose uptake measurements using [3H]-2-deoxyglucose or [14C]-glucose; n = 4. Experiments repeated three times. k, Glucose depletion from tissue culture medium by 3T3-L1 adipocytes after 48 h incubation. n = 3 biological independent samples; experiments repeated three times. Data shown are averages of three independent replicas. l, m, Expression of FOXK1 and FOXK2 in WAT from wild-type or Ob/Ob mice. Retroperitoneal WAT: wild-type n = 9, Ob/Ob n = 7; gonadal WAT: wild-type n = 10, Ob/Ob n = 7) n, Leptin levels in 3T3-L1 adipocytes overexpressing or knocked down for FOXK1 or FOXK2; n = 3. o, Rate of glucose uptake and lactate secretion into tissue culture medium of 3T3-L1 adipocytes during 48 h with induced expression of FOXK1 or FOXK2. p, Lactate production rate of 3T3-L1 adipocytes during 48 h with overexpression of FOXK1 or FOXK2. q, Glucose depletion rate of 3T3-L1 adipocytes during 48 h with overexpression of FOXK1 or FOXK2. oq, n = 3 biological independent samples, experiments were repeated three times. Data shown are an average of three independent replicas. This set of experiments (n = 3 and repeated 3 times) was performed twice with similar results. Throughout figure, experiments replicated at least three times with the exception of c (twice) and ln (once). Representative experiments are shown if not indicated otherwise. Unpaired two-sided Student’s t-test; ***P < 0.001, ** P < 0.01, *P < 0.05. For gel source data, see Supplementary Fig. 1.

Source Data

Extended Data Fig. 2 Characterization of FOXK1 and FOXK2 and metabolic changes induced by FOXK1 and FOXK2 in adipocytes and muscle cells.

a, FOXK1 and FOXK2 showing amino acid sequence similarities, forkhead associated domain (FHA), winged helix domain (WHD) and nuclear localization signal (NL). Cross-species amino acid comparison shows ~90% similarity between human and mouse for FOXK1, and ~95% for FOXK2. b, c, Relative mRNA levels for Foxk1 (b) and Foxk2 (c) in the SVF and in mature adipocytes (Ad). Experiment performed once, n = 3. d, Glycerol release in 3T3‐L1 adipocytes with increased expression of FOXK1 or FOXK2 with or without 20 μM isoprenaline, n = 6. e, Glucose uptake in differentiating C2C12 myocytes (on day 8) in response to doxycycline-induced expression of FOXK1 or FOXK2 initiated at indicated days (d) during differentiation, n = 6. A representative experiment out of three (d) or two (e) are shown. Data shown as mean ± s.d., n shows biological independent experiments. Unpaired two-sided Student’s t-test; ***P < 0.001, **P < 0.01, *P < 0.05.

Source Data

Extended Data Fig. 3 Knockdown efficiency of shFoxk1-1, shFoxk1-2, shFoxk2-1 and shFoxk2-2, and complementation test of FOXK1 and FOXK2.

3T3-L1 adipocytes were transduced to express shRNA targeting Foxk1 and Foxk2. a, Proteins and RNA were prepared and protein levels were determined by western blot using specific antibodies against FOXK1, FOXK2 and β-actin. b, mRNA levels were determined using quantitative real-time PCR, n = 3. Experiment performed once. 3T3-L1 adipocytes were transduced to overexpress FOXK1, FOXK2, shFoxk1 and shFoxk2 in the combinations listed. c, 2-DG uptake was determined under basal and insulin-stimulated conditions (100 nM), n = 4. d, Lactate secreted into medium, n = 4. c, d, Representative experiments out of two are shown. e, Knockdown efficiency and selectivity of shPdk1 and shPdk4 tested in 3T3-L1 adipocytes using quantitative real-time PCR, n = 3. Experiment performed once. f, g, Lactate production in 3T3-L1 adipocytes transduced with shRNA targeting Pdk1 or Pdk4 either alone or in combination with FOXK1 or FOXK2 expression vectors. n = 3. Representative experiments out of two are shown. Data shown as mean ± s.d., n shows biological independent experiments. Unpaired two-sided Student’s t-test; ***P < 0.001, **P < 0.01, *P < 0.05. For gel source data, see Supplementary Fig. 1.

Extended Data Fig. 4 Bioenergetic changes induced by FOXK2 in 3T3-L1 adipocytes, FOXK2-induced 2-DG uptake under several inhibitor treatments, and role of FOXK1 and FOXK2 in T cell activation.

a, Glycolytic analysis of 3T3-L1 upon FOXK2 overexpression (FOXK2) or suppression (shFoxk2) compared to empty vector and shCtrl controls, respectively. Data are averages of four independent experiments, n = 12. b, Left, kinetic ECAR response of 3T3-L1 adipocytes overexpressing FOXK2 to glucose (10 mM), oligomycin (1 µM) and 2-DG (100 mM). A representative experiment out of three is shown, n = 3. Right, dissection of glycolysis and glycolytic capacity in 3T3-L1 adipocytes overexpressing FOXK2. Data are averages of four independent experiments, n = 4. The assay medium was substrate-free base medium supplemented with 2 mM glutamine. c, 2-DG uptake by 3T3-L1 adipocytes under basal conditions with overexpression of FOXK2 or an empty vector control after treatment with salirasib 25 μM for 2 h and rapamycin 100 nM for 16 h or 2 h. Representative experiment out of two is shown, n = 3. dg, Human peripheral blood CD3+ T cells from 3 donors (n = 3) were transduced with Flag-tagged FOXK1 or FOXK2 vectors. Transduced cells were incubated in X-vivo 15 medium supplemented with 5% human serum and IL-2 for 3 days before Ki-67 staining and analysis by flow cytometry. Cells were gated on CD4 or CD8 and Flag-tag (FOXK1 or FOXK2 overexpression) before expression of the proliferation marker Ki-67 was analysed. Experiments were performed three times. Data shown as mean ± s.d. Unpaired two-sided Student’s t-test; ***P < 0.001, **P < 0.01, *P < 0.05.

Extended Data Fig. 5 FOXK1 and FOXK2 regulate proteins involved in the glycolytic pathway.

ak, Western blot experiments for proteins involved in the glycolytic pathway in 3T3‐L1 adipocytes overexpressing or knocked down for FOXK1 or FOXK2. a, HK2; b, PFKM; c, PKM‐M2; d, ALDOA; e, LDHA; f, MCT1; g, PDK1; h, PDK4; i, PDP1; j, GLUD1. k, Phosphorylation of Ser293, Ser232, and Ser300 of the E1α regulatory subunit using specific antisera. Lower parts show PDC complex after immunoprecipitation by PDH Immunocapture Kit and silver staining. Representative experiments from independent replicate samples are shown. l, Schematic view of FOXK1 and FOXK2 regulation sites in the glycolytic pathway. For gel source data, see Supplementary Fig. 1.

Extended Data Fig. 6 Regulation of Foxk1 and Foxk2 at the transcriptome and proteome levels.

a, Correlation between expression profiles of 3T3-L1 adipocytes overexpressing or knocked down for FOXK1 or FOXK2. x- and y-axes denote log2-transformed expression in RPKM (reads per kilobase of transcript per million). The correlation and P values are derived from Pearson’s correlation test. Precise P values are presented from Pearson’s correlation tests. b, c, Overlaps (b) between genes upregulated and downregulated by FOXK1 and FOXK2 knockdown (left) and overexpression (right) profiles and heat maps (c) showing logFC values of overlapping genes from FOXK1 and FOXK2 knockdown (left) and overexpression (right) profiles. d, Commonly deregulated genes or proteins from RNA-seq (FOXK1, n = 3 and FOXK2, n = 2) and proteome (FOXK1, n = 4 and FOXK2, n = 2) profiles of knockdown and overexpression. The P values for overlaps were calculated using hypergeometric test by assuming all expressed protein-coding genes from this study as background (n = 17,038). e, f, Gene enrichment (biological process) analysis of commonly deregulated genes from FOXK1 and FOXK2 overexpression or knockdown in RNA-seq profiles (e) and proteome profiles (f). Axes denote –log10P from the GeneSCF tool. The Fisher’s exact test P values obtained using GeneSCF functional enrichment tool. g, Regulation of glycolytic pathway genes or proteins in RNA-seq and proteome profiles. RNA-seq data for overexpression or knockdown experiments depicted here were performed and analysed in three biological replicas (FOXK1, n = 3) or two biological replicas (FOXK2, n = 2). Proteomics data for overexpression or knockdown experiments depicted here were performed and analysed in four biological replicas (FOXK1, n = 4) or two biological replicas (FOXK2, n = 2).

Extended Data Fig. 7 ChIP–PCR validation of FOXK1 and FOXK2 target genes.

ai, Schematic of predicted binding sites and real-time PCR results for FOXK1 and FOXK2 target genes: Hk2 (a), Pfkm (b), Aldoa (c), Pkm (d), Mct1 (e), Pdk1 (f), Pdk4 (g), Pdp1 (h), and Glud1 (i). Mct1 site #2 is not a FOXK1/FOXK2 binding site and serves as a negative control. Input and ChIP cycle threshold (Ct) values were normalized separately to empty vector control as 1. a, Hk2 ChIP–PCR, n = 2 for control input and FOXK2 ChIP, n = 3 for others; b, Pfkm ChIP-PCR, n = 2 for control input, n = 3 for others; c, Aldoa ChIP–PCR, sites #1 and #3, n = 2 for FOXK1 ChIP, n = 3 for others, site #2, n = 3; d, Pkm ChIP–PCR, n = 2 for control input, n = 3 for others; e, Mct1 ChIP–PCR, site #1, n = 2 for control input, n = 3 for others, site #2, n = 2 for ChIPs, n = 3 for inputs; f, Pdk1 ChIP–PCR, n = 2 for FOXK1 ChIP, n = 3 for others; g, Pdk4 ChIP–PCR, site #1, n = 2 for control and FOXK2 ChIP, n = 3 for others, site #2, n = 2 for control ChIP, n = 3 for others; h, Pdp1 ChIP–PCR, n = 2 for ChIPs, n = 3 for inputs; i, Glud1 ChIP-PCR, n = 3. Experiments replicated once. Unpaired two-sided Student’s t-test, ***P < 0.001, **P < 0.01, *P < 0.05.

Extended Data Table 1 Unbiased molecular pathway analysis
Extended Data Table 2 Proteomic data expressed in fold change
Extended Data Table 3 PCR primer list for ChIP–PCR

Supplementary information

Supplementary Information

This file contains Supplementary Note 1.

Reporting Summary

Supplementary Figure

Supplementary Figure 1. This file contains gel source data shown in this study. Red rectangle parts were shown in the extended data figures. Primary antibodies used are indicated besides the images.

Supplementary Figure

Supplementary Figure 2. T-cells flow cytometry gating strategy.

Supplementary Table

This file contains Supplementary Table 1. Proteomics data of 3T3-L1 cells with control and FOXK1 over-expression (OE). Paired Two-sided T-test was performed and there was no adjustments/p-value corrections made.

Supplementary Table

This file contains Supplementary Table 2. Proteomics data of 3T3-L1 cells with control and Foxk1 knock-down with shRNA (KD). Paired Two-sided T-test was performed and there was no adjustments/p-value corrections made.

Supplementary Table

This file contains Supplementary Table 3. Proteomics data of 3T3-L1 cells with control and FOXK2 over-expression (OE). Paired Two-sided T-test was performed and there was no adjustments/p-value corrections made.

Supplementary Table

This file contains Supplementary Table 4. Proteomics data of 3T3-L1 cells with control and Foxk2 knock-down with shRNA (KD). Paired Two-sided T-test was performed and there was no adjustments/p-value corrections made.

Source data

Rights and permissions

Reprints and permissions

About this article

Check for updates. Verify currency and authenticity via CrossMark

Cite this article

Sukonina, V., Ma, H., Zhang, W. et al. FOXK1 and FOXK2 regulate aerobic glycolysis. Nature 566, 279–283 (2019). https://doi.org/10.1038/s41586-019-0900-5

Download citation

  • Received:

  • Accepted:

  • Published:

  • Issue Date:

  • DOI: https://doi.org/10.1038/s41586-019-0900-5

This article is cited by

Comments

By submitting a comment you agree to abide by our Terms and Community Guidelines. If you find something abusive or that does not comply with our terms or guidelines please flag it as inappropriate.

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