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:

AKT methylation by SETDB1 promotes AKT kinase activity and oncogenic functions

Abstract

Aberrant activation of AKT disturbs the proliferation, survival and metabolic homeostasis of various human cancers. Thus, it is critical to understand the upstream signalling pathways governing AKT activation. Here, we report that AKT undergoes SETDB1-mediated lysine methylation to promote its activation, which is antagonized by the Jumonji-family demethylase KDM4B. Notably, compared with wild-type mice, mice harbouring non-methylated mutant Akt1 not only exhibited reduced body size but were also less prone to carcinogen-induced skin tumours, in part due to reduced AKT activation. Mechanistically, the interaction of phosphatidylinositol (3,4,5)-trisphosphate with AKT facilitates its interaction with SETDB1 for subsequent AKT methylation, which in turn sustains AKT phosphorylation. Pathologically, genetic alterations, including SETDB1 amplification, aberrantly promote AKT methylation to facilitate its activation and oncogenic functions. Thus, AKT methylation is an important step, synergizing with PI3K signalling to control AKT activation. This suggests that targeting SETDB1 signalling could be a potential therapeutic strategy for combatting hyperactive AKT-driven cancers.

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: AKT methylation promotes its activity and oncogenic functions.
Fig. 2: Methylation-deficient Akt1 knock-in mice display reduced body size and weight and are resistant to chemical carcinogen-induced skin tumorigenesis in vivo.
Fig. 3: SETDB1 methylates AKT on K140 and K142 to promote its kinase activity.
Fig. 4: Oncogenic function of SETDB1 depends on the activation of AKT.
Fig. 5: SETDB1-mediated methylation of AKT synergizes with PI3K to activate AKT.
Fig. 6: KMD4B demethylates AKT to inhibit AKT kinase activity.
Fig. 7: Deficiency of SETDB1 inhibits AKT kinase activity and oncogenic function.

Similar content being viewed by others

Data availability

The MS-based screening data generated in this study have been deposited in ProteomeXchange under the accession code PXD011657. The SETDB1, EZH2, PTEN, EGFR, PIK3CA and AKT1 genetic alterations in TCGA datasets were integrated from the cBioPortal database (www.cbioportal.org), with the query of genes “ESET”, “EZH2”, “PTEN”, “EGFR”, “PIK3CA” and “AKT1” for both mutation and copy number alterations (CNAs) in different cancer types, such as BRCA and melanoma. Information regarding each cancer study is as follows: one dataset for BRCA (TCGA, provisional); one dataset for melanoma (TCGA, provisional). The source data for Figs. 1e,f,h,j–m, 2b,c,g,h, 3n,p,q, 4c,f,h,j,l,n, 6m–o and 7b,d,j,l,n, and Supplementary Figs. 1l,m, 2e,f, 3o, 4c–f,h and 7e,g,i have been provided as Supplementary Table 2. All other data supporting the findings of this study are available from the corresponding author upon reasonable request.

References

  1. Jones, P. A., Issa, J. P. & Baylin, S. Targeting the cancer epigenome for therapy. Nat. Rev. Genet. 17, 630–641 (2016).

    Article  CAS  Google Scholar 

  2. You, J. S. & Jones, P. A. Cancer genetics and epigenetics: two sides of the same coin? Cancer Cell 22, 9–20 (2012).

    Article  CAS  Google Scholar 

  3. Glaser, K. B. HDAC inhibitors: clinical update and mechanism-based potential. Biochem. Pharmacol. 74, 659–671 (2007).

    Article  CAS  Google Scholar 

  4. Fahy, J., Jeltsch, A. & Arimondo, P. B. DNA methyltransferase inhibitors in cancer: a chemical and therapeutic patent overview and selected clinical studies. Expert Opin. Ther. Pat. 22, 1427–1442 (2012).

    Article  CAS  Google Scholar 

  5. Delmore, J. E. et al. BET bromodomain inhibition as a therapeutic strategy to target c-Myc. Cell 146, 904–917 (2011).

    Article  CAS  Google Scholar 

  6. Kelly, T. K., De Carvalho, D. D. & Jones, P. A. Epigenetic modifications as therapeutic targets. Nat. Biotechnol. 28, 1069–1078 (2010).

    Article  CAS  Google Scholar 

  7. Daigle, S. R. et al. Potent inhibition of DOT1L as treatment of MLL-fusion leukemia. Blood 122, 1017–1025 (2013).

    Article  CAS  Google Scholar 

  8. Biggar, K. K. & Li, S. S. Non-histone protein methylation as a regulator of cellular signalling and function. Nat. Rev. Mol. Cell Biol. 16, 5–17 (2015).

    Article  CAS  Google Scholar 

  9. Chuikov, S. et al. Regulation of p53 activity through lysine methylation. Nature 432, 353–360 (2004).

    Article  CAS  Google Scholar 

  10. Guo, A. et al. Immunoaffinity enrichment and mass spectrometry analysis of protein methylation. Mol. Cell. Proteomics 13, 372–387 (2014).

    Article  CAS  Google Scholar 

  11. Saddic, L. A. et al. Methylation of the retinoblastoma tumor suppressor by SMYD2. J. Biol. Chem. 285, 37733–37740 (2010).

    Article  CAS  Google Scholar 

  12. Vanhaesebroeck, B. & Alessi, D. R. The PI3K–PDK1 connection: more than just a road to PKB. Biochem. J. 346, 561–576 (2000).

    CAS  PubMed  PubMed Central  Google Scholar 

  13. Manning, B. D. & Toker, A. AKT/PKB signaling: navigating the network. Cell 169, 381–405 (2017).

    Article  CAS  Google Scholar 

  14. Ozes, O. N. et al. NF-kappaB activation by tumour necrosis factor requires the Akt serine-threonine kinase. Nature 401, 82–85 (1999).

    Article  CAS  Google Scholar 

  15. Yang, W. L. et al. The E3 ligase TRAF6 regulates Akt ubiquitination and activation. Science 325, 1134–1138 (2009).

    Article  CAS  Google Scholar 

  16. Liu, P. et al. Cell-cycle-regulated activation of Akt kinase by phosphorylation at its carboxyl terminus. Nature 508, 541–545 (2014).

    Article  CAS  Google Scholar 

  17. Guo, J. et al. pVHL suppresses kinase activity of Akt in a proline-hydroxylation-dependent manner. Science 353, 929–932 (2016).

    Article  CAS  Google Scholar 

  18. Mazur, P. K. et al. SMYD3 links lysine methylation of MAP3K2 to Ras-driven cancer. Nature 510, 283–287 (2014).

    Article  CAS  Google Scholar 

  19. Yoshioka, Y. et al. SMYD3-mediated lysine methylation in the PH domain is critical for activation of AKT1. Oncotarget 7, 75023–75037 (2016).

    Article  Google Scholar 

  20. Cong, L. et al. Multiplex genome engineering using CRISPR/Cas systems. Science 339, 819–823 (2013).

    Article  CAS  Google Scholar 

  21. Cho, H., Thorvaldsen, J. L., Chu, Q., Feng, F. & Birnbaum, M. J. Akt1/PKBalpha is required for normal growth but dispensable for maintenance of glucose homeostasis in mice. J. Biol. Chem. 276, 38349–38352 (2001).

    Article  CAS  Google Scholar 

  22. Nasti, T. H. et al. A murine model for the development of melanocytic nevi and their progression to melanoma. Mol. Carcinog. 55, 646–658 (2016).

    Article  CAS  Google Scholar 

  23. Gao, H. et al. Akt/PKB interacts with the histone H3 methyltransferase SETDB1 and coordinates to silence gene expression. Mol. Cell. Biochem. 305, 35–44 (2007).

    Article  CAS  Google Scholar 

  24. Liu, T. et al. Histone methyltransferase SETDB1 maintains survival of mouse spermatogonial stem/progenitor cells via PTEN/AKT/FOXO1 pathway. Biochim. Biophys. Acta 1860, 1094–1102 (2017).

    Article  CAS  Google Scholar 

  25. Cha, T. L. et al. Akt-mediated phosphorylation of EZH2 suppresses methylation of lysine 27 in histone H3. Science 310, 306–310 (2005).

    Article  CAS  Google Scholar 

  26. Cerami, E. et al. The cBio cancer genomics portal: an open platform for exploring multidimensional cancer genomics data. Cancer Discov. 2, 401–404 (2012).

    Article  Google Scholar 

  27. Schultz, D. C., Ayyanathan, K., Negorev, D., Maul, G. G. & Rauscher, F. J. 3rd SETDB1: a novel KAP-1-associated histone H3, lysine 9-specific methyltransferase that contributes to HP1-mediated silencing of euchromatic genes by KRAB zinc-finger proteins. Genes Dev. 16, 919–932 (2002).

    Article  CAS  Google Scholar 

  28. Ceol, C. J. et al. The histone methyltransferase SETDB1 is recurrently amplified in melanoma and accelerates its onset. Nature 471, 513–517 (2011).

    Article  CAS  Google Scholar 

  29. Macgregor, S. et al. Genome-wide association study identifies a new melanoma susceptibility locus at 1q21.3. Nat. Genet. 43, 1114–1118 (2011).

    Article  CAS  Google Scholar 

  30. Fei, Q. et al. Histone methyltransferase SETDB1 regulates liver cancer cell growth through methylation of p53. Nat. Commun. 6, 8651 (2015).

    Article  CAS  Google Scholar 

  31. Wong, C. M. et al. Up-regulation of histone methyltransferase SETDB1 by multiple mechanisms in hepatocellular carcinoma promotes cancer metastasis. Hepatology 63, 474–487 (2016).

    Article  CAS  Google Scholar 

  32. Carpten, J. D. et al. A transforming mutation in the pleckstrin homology domain of AKT1 in cancer. Nature 448, 439–444 (2007).

    Article  CAS  Google Scholar 

  33. Cao, J. et al. MC1R is a potent regulator of PTEN after UV exposure in melanocytes. Mol. Cell 51, 409–422 (2013).

    Article  CAS  Google Scholar 

  34. Dankort, D. et al. Braf(V600E) cooperates with Pten loss to induce metastatic melanoma. Nat. Genet. 41, 544–552 (2009).

    Article  CAS  Google Scholar 

  35. Garraway, L. A. et al. Integrative genomic analyses identify MITF as a lineage survival oncogene amplified in malignant melanoma. Nature 436, 117–122 (2005).

    Article  CAS  Google Scholar 

  36. Cantley, L. C. The phosphoinositide 3-kinase pathway. Science 296, 1655–1657 (2002).

    Article  CAS  Google Scholar 

  37. Alessi, D. R. et al. Characterization of a 3-phosphoinositide-dependent protein kinase which phosphorylates and activates protein kinase Balpha. Curr. Biol. 7, 261–269 (1997).

    Article  CAS  Google Scholar 

  38. Stephens, L. et al. Protein kinase B kinases that mediate phosphatidylinositol 3,4,5-trisphosphate-dependent activation of protein kinase B. Science 279, 710–714 (1998).

    Article  CAS  Google Scholar 

  39. Franke, T. F., Kaplan, D. R., Cantley, L. C. & Toker, A. Direct regulation of the Akt proto-oncogene product by phosphatidylinositol-3,4-bisphosphate. Science 275, 665–668 (1997).

    Article  CAS  Google Scholar 

  40. Chan, C. H. et al. The Skp2-SCF E3 ligase regulates Akt ubiquitination, glycolysis, herceptin sensitivity, and tumorigenesis. Cell 149, 1098–1111 (2012).

    Article  CAS  Google Scholar 

  41. Cederquist, C. T. et al. Systemic insulin sensitivity is regulated by GPS2 inhibition of AKT ubiquitination and activation in adipose tissue. Mol. Metab. 6, 125–137 (2017).

    Article  CAS  Google Scholar 

  42. Whetstine, J. R. et al. Reversal of histone lysine trimethylation by the JMJD2 family of histone demethylases. Cell 125, 467–481 (2006).

    Article  CAS  Google Scholar 

  43. Luo, J., Manning, B. D. & Cantley, L. C. Targeting the PI3K–Akt pathway in human cancer: rationale and promise. Cancer Cell 4, 257–262 (2003).

    Article  CAS  Google Scholar 

  44. Hennessy, B. T., Smith, D. L., Ram, P. T., Lu, Y. & Mills, G. B. Exploiting the PI3K/AKT pathway for cancer drug discovery. Nat. Rev. Drug. Discov. 4, 988–1004 (2005).

    Article  CAS  Google Scholar 

  45. Ryu, H. et al. ESET/SETDB1 gene expression and histone H3 (K9) trimethylation in Huntington’s disease. Proc. Natl Acad. Sci. USA 103, 19176–19181 (2006).

    Article  CAS  Google Scholar 

  46. Chen, W. S. et al. Growth retardation and increased apoptosis in mice with homozygous disruption of the Akt1 gene. Genes Dev. 15, 2203–2208 (2001).

    Article  CAS  Google Scholar 

  47. DuPage, M., Dooley, A. L. & Jacks, T. Conditional mouse lung cancer models using adenoviral or lentiviral delivery of Cre recombinase. Nat. Protoc. 4, 1064–1072 (2009).

    Article  CAS  Google Scholar 

  48. Downward, J. Mechanisms and consequences of activation of protein kinase B/Akt. Curr. Opin. Cell Biol. 10, 262–267 (1998).

    Article  CAS  Google Scholar 

  49. Wang, G. H. et al. SETDB1-mediated methylation of Akt promotes its K63-linked ubiquitination and activation leading to tumorigenesis. Nat. Cell Biol. https://doi.org/10.1038/s41556-018-0266-1 (2019).

  50. Liu, S. et al. Setdb1 is required for germline development and silencing of H3K9me3-marked endogenous retroviruses in primordial germ cells. Genes Dev. 28, 2041–2055 (2014).

    Article  CAS  Google Scholar 

  51. Li, H. et al. The histone methyltransferase SETDB1 and the DNA methyltransferase DNMT3A interact directly and localize to promoters silenced in cancer cells. J. Biol. Chem. 281, 19489–19500 (2006).

    Article  CAS  Google Scholar 

  52. Sun, Q. Y. et al. SETDB1 accelerates tumourigenesis by regulating the WNT signalling pathway. J. Pathol. 235, 559–570 (2015).

    Article  CAS  Google Scholar 

  53. Ding, X. et al. Epigenetic activation of AP1 promotes squamous cell carcinoma metastasis. Sci. Signal. 6, ra28 (2013).

    Article  Google Scholar 

  54. Kawazu, M. et al. Histone demethylase JMJD2B functions as a co-factor of estrogen receptor in breast cancer proliferation and mammary gland development. PLoS ONE 6, e17830 (2011).

    Article  CAS  Google Scholar 

  55. Allen, J. J. et al. A semisynthetic epitope for kinase substrates. Nat. Methods 4, 511–516 (2007).

    Article  CAS  Google Scholar 

  56. Shi, X. et al. Modulation of p53 function by SET8-mediated methylation at lysine 382. Mol. Cell 27, 636–646 (2007).

    Article  CAS  Google Scholar 

  57. Huang, J. et al. p53 is regulated by the lysine demethylase LSD1. Nature 449, 105–108 (2007).

    Article  CAS  Google Scholar 

  58. Breitkopf, S. B., Yuan, M., Helenius, K. P., Lyssiotis, C. A. & Asara, J. M. Triomics analysis of imatinib-treated myeloma cells connects kinase inhibition to RNA processing and decreased lipid biosynthesis. Anal. Chem. 87, 10995–11006 (2015).

    Article  CAS  Google Scholar 

Download references

Acknowledgements

The authors thank B. North, J. Zhang, F. Dang and other Wei Lab members for critical reading of the manuscript, as well as members of the Pandofi and Toker Laboratory for helpful discussions. The authors thank H. Okada (Kindai University of Medicine) for the generation of Kdm4bflox/flox MEFs. W.G. is supported by K99CA207867 from the National Cancer Institute. W.W. is a LLS research scholar. This work was supported in part by the NIH grant CA177910 (to W.W. and A.T.). The MS work was partially supported by NIH grants P01CA120964 (to J.A.) and P30CA006516 (to J.A.)

Author information

Authors and Affiliations

Authors

Contributions

J.G. designed and performed most of the experiments with assistance from X.D., B.L., W.G., P.L. W.W., P.P.P. and Y.S. A.T. supervised the study. J.G., N.Z. and J.Z. performed the revision. A.G. performed the IAP-LC-MS/MS screen. M.Y. and J.M.A. performed the MS work. J.G. and W.W. wrote the manuscript. All authors commented on the manuscript.

Corresponding author

Correspondence to Wenyi Wei.

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.

Integrated supplementary information

Supplementary Figure 1 Mass spectrometry-based non-biased approach to identify Akt methylation.

a, Tri-methylation sites and proteins were identified from OVCAR5 cells with the IAP-LC-MS approach (also see Table S1). This screen was repeated twice. b, Immunoblot (IB) analysis of Akt immunoprecipitates (IP) products and whole cell lysates (WCL) derived from OVCAR5 and A375 cells treated with/without methyltransferase inhibitors DZneP (5 μM) 12 hrs before harvesting. c, A schematic graph represents the methylation occurring in Akt1 identified by MS. Where indicated, mono-, di- and tri-methylation of Akt1 were respectively. d, IB analysis of HA-IP products and WCL derived from HEK293 cells transfected with indicated constructs. e,f, IB analysis of WCL derived from cells generated from pancreas or lung tissues of Smyd3-knockout mice (Kras;p53 and Kras;p53;Smyd3-/-). g, HEK293 cells were transfected with indicated constructs, and stimulated with IGF (100 ng/ml) after serum-starvation, for GST pull-down assays and IB analysis. h-j, AKT1/2-/--DLD1 cells were lentivirally infected with indicated constructs. Cells were serum-starved and stimulated with insulin (0.1 μM) (h-i) or EGF (10 ng/ml) (j) before harvesting for IB analysis. k, Akt in vitro kinase assays were performed with bacterially purified recombinant His-GSK3β as the substrate, and the recombinant Akt1 immunopurified from HEK293 cells as the source of kinase. l,m, Cells generated in (i) were subjected to glucose uptake (l) and lactate production (m) assays. The experiments were performed twice independently with three repeats, exhibited similar results. The relative glucose or lactate levels derived from two independent experiments were plotted in (l,m). n,o, Cells generated in (i) were subcutaneously injected into nude mice, and the tumors were monitored (n) and dissected. The tumor lysates were subjected for IB analysis (o). p, A sequence alignment of nuclear acids between WT and K140/142R mutant form of AKT1 indicates the strategy for generating the AKT1-K140/142R knock-in mutation. (q) The gel image illustrates the Sac II-digesting genomic DNAs derived from HEK293 knock-in cells. Corresponding DNAs were subjected for sequencing (r). s-u, Cells generated in (s) were serum-starved for IB analysis in different time periods after EGF (10 ng/ml) (t) or IGF (100 ng/ml) (u) stimulation. Statistical source data for l and m are shown in Supplementary Table 2. All Western-blots above were performed twice independently with similar results. Scanned images of unprocessed blots are shown in Supplementary Fig. 8.

Supplementary Figure 2 Depletion of SETDB1 decreases Akt kinase activity.

a, A sequence alignment of nuclear acids between WT and K140/142R mutant form of Akt1 indicates the strategy for generating the Akt1-K140/142R knock-in (KI) mice. As indicated, TGG sequence in WT-Akt1 was selected as the sgRNA PAM sequence. b,c, The gel image illustrates the Sac II-digesting genomic DNAs derived from litters selected from knock-in mice, and the corresponding DNAs were subjected for sequencing (c). d-f, Frequency of genotypes produced from Akt1 KI/WT mouse intercrosses (d). Numbers in parentheses indicate the expected number by Mendelian ratio. Growth of Akt1WT versus Akt1KI/KI female and male mice. Data were shown as mean+/-s.e.m. (male, n = 26 mice in Akt1WT group and n = 18 mice in Akt1KI/KI group; female, n = 19 mice in Akt1WT group and n = 14 mice in Akt1KI/KI group). g,h, The mice derived from the same litter were imaged at age of 4 weeks old, and were euthanized and their organs were dissected (g) and weighed, and further subjected to H&E and IHC staining (h). Scale bar, 50 μm. The experiment in h was performed twice, independently, with similar results. i, IB analysis of IP and WCL derived from HEK293 cells transfected with indicated constructs. j, Genomic alterations of SETDB1 are mutually exclusive with the alterations of the PI3K/Akt pathway-related genes (PTEN, EGFR, PIK3CA and AKT1) from the TCGA database including melanoma and breast cancer. k, A schematic illustration of the potential regulation of Akt1 with SETDB1 or EZH2, respectively. Where indicated, SETDB1 is the upstream regulator of Akt capable of methylating Akt, whereas EZH2 has been reported to be the downstream substrate of Akt, which can be phosphorylated by Akt. l,m, IB analysis of GST pulldown and WCL derived from HEK293 cells transfected with indicated constructs. Statistical source data for e and f are shown in Supplementary Table 2. All Western-blots above were performed twice independently with similar results. Scanned images of unprocessed blots are shown in Supplementary Fig. 8.

Supplementary Figure 3 Depletion of SETDB1 decreases Akt kinase activity.

a-c, IB analysis of GST pull-down (a,b) or IP (c) products and WCL derived from HEK293T cells transfected with indicated constructs. d, IB analysis of cell fractionations separated from HEK293, DLD1 and A375 cells, respectively. e-g, IB analysis of dot blot (e) and IP products derived from HEK293 cells transfected with the indicated constructs (f,g). h, In vitro methylation assays were performed with IP Flag-SETDB1 derived from HEK293T cells as the source of methyltransferase, and the commercially purified His-Akt1 as the substrate. i-k, IB analysis of IP products, GST pull-down and WCL derived from HBL cells lentivirally infected with shRNAs against SETDB1 (i), or derived from HEK293 cells transfected with the indicated constructs (j,k). l-o, IB analysis of Setdb1 conditional knockout MEFs treated with or without 4-OHT (500 nM) in different time points (l,m). Resulting cells were subjected to colony formation assays (n). The experiment was performed twice independently with three repeats, and exhibit similar results (n). Representative images were shown in n and relative colony numbers derived from two independent experiments were plotted in o. Source data for o are shown in Supplementary Table 2. All Western-blots above were performed twice independently with similar results. Scanned images of unprocessed blots are shown in Supplementary Fig. 8.

Supplementary Figure 4 Depletion of SETDB1 decreases Akt oncogenic functions.

a-e, IB assays of WCL derived from OVCAR5 (a) and DLD1 (b) cells infected with shRNAs against SETDB1. Resulting cells were subjected for colony formation and soft agar assays (c-e, top panel). The experiment was performed twice independently with three repeats, and exhibit similar results (c-e). Representative images were shown in (c-e, top panel) and relative colony numbers derived from two independent experiments were plotted in (c-e, bottom panel). f, The glucose uptake was measured with FACS as mentioned in Material section. The experiment was performed twice independently with three repeats, and exhibit similar results (f). Relative glucose levels derived from two independent experiments were plotted in f. g,h, IB analysis of WCL derived from A375 cells infected with lentivirus against SETDB1, and sequentially infected with retrovirus encoding myr-Akt1 (g). Resulting cells were subjected to colony formation assays (h, bottom panel). The experiment was performed twice independently with three repeats, and exhibit similar results (h). Representative images were shown in (h, top panel) and relative colony numbers derived from two independent experiments were plotted in (h, bottom panel). i-k, IB analysis of HA- or Akt1-IP as well as WCL derived from A375 or HEK293 cells transfected with the indicated constructs. Resulting cells were serum-starved for 18 hrs and stimulated with IGF (30 ng/ml) (i,k) or insulin (0.03 μM) (j) in different time points before harvest for IB analysis. l,m, IB analysis of IP products and WCL derived from HEK293 cells transfected with the indicated constructs treated with (l) or without (m) PI3K inhibitors. Source data for c, d, e, f and h are shown in Supplementary Table 2. All Western-blots above were performed twice independently with similar results. Scanned images of unprocessed blots are shown in Supplementary Fig. 8.

Supplementary Figure 5 PIP3 enhances the interaction of SETDB1 with Akt1 PH motif.

a-c, A cartoon illustration of the workflow describing the method for in vitro binding assays (a). Briefly, 293T cells transfected with different Akt1 encoding constructs were harvested for GST pull-down assay after treatment with the PI3K inhibitor, LY294002. The pull-down Akt1 was eluted from the beads (b) and incubated with flag-bead immunoprecipitated SETDB1 (c) in the presence/absence of PIP3 (20 μM) for 8 hrs in 4 degrees. Then the flag-beads were washed 3 times and subjected for IB analysis. d,e, His-Akt1 was purified from insect cells and eluted (d), then subjected to in vitro binding assay in the presence or absence of PIP3, and detected with IB analysis (e). f, HCT116-PTEN-/- and counterpart cells were starved and stimulated with IGF (100 ng/ml), then subjected to IB analysis. g,h, IB analysis of GST pull-down and WCL derived from HEK293 cells transfected with the indicated constructs. i-k, IB analysis of cell fractionations separated from SETDB1 knockdown A375 cells (i), AKT1K140/142R-edited and parental HEK293 cells (j) and Setdb1-deleted MEFs (k). Where indicated, MEFs were treated with 4-OHT (500 nM) or vehicle for 48 hrs, and resulting cells were serum-starved for 12 hrs and insulin-stimulated in different time points before harvest. l,m, IB analysis of WCL derived from HeLa cells transfected with C-terminal GFP-fusion Akt1. Cells were serum-starved 12 hrs and stimulated with or without insulin for 30 minutes, then subjected to DAPI staining and detected with fluorescence microscope. The nuclear DNA was stained with DAPI. Scale bar, 20 μm. The experiment in m was performed twice, independently, with similar results (m). n, A schematic illustration of the proposed model in which full Akt activation is achieved by a coordinated action of the SETDB1-mediated Akt methylation events and the canonical PI3K-PDK1 dependent kinase cascades leading to Akt phosphorylation. o-r, IB analysis of GST pull-down (o,q), his-tagged ubiquitination (p,r) and WCL derived from HEK293 cells transfected with indicated encoding constructs. All Western-blots above were performed twice independently with similar results. Scanned images of unprocessed blots are shown in Supplementary Fig. 8.

Supplementary Figure 6 KDM4B demethylates Akt1.

a, IB analyses of GST pull-down and WCL derived from HEK293 cells transfected with the indicated constructs. b-e, Coomassie staining image to illustrate mammalian purified GST-KDM4 (b) and bacterially purified GST-KDM4B catalytic domains (d), which were used for in vitro demethylation assays with Akt1 methylated peptides (c) or cell purified HA-Akt1 (insulin treated before harvesting) (e) as substrates. f-l, IB analyses of GST pull-down and WCL derived from HEK293 cells transfected with the indicated constructs. m-o, IB analysis of WCL derived from A375 cells (m), OVCAR5 cells (n) or HEK293 cells (o) lentivirally infected with shRNAs against KDM4B. p, IB analysis of WCL derived from AKT1K140/142R-edited HEK293 cells infected with lentivirus against KDM4B. q,r, IB analysis of cell fractionations separated from HEK293 cells infected with lentiviruses against KDM4B. s, IB analysis of cell fractionations separated from HEK293 cells infected with shRNA against KDM4B. t, HEK293 cells were transfected with indicated constructs and serum starved for 12 hrs. Resulting cells were stimulated with insulin (0.1 μM) in different time points before harvesting for GST pull-down and IB analysis. All Western-blots above were performed twice independently with similar results. Scanned images of unprocessed blots are shown in Supplementary Fig. 8.

Supplementary Figure 7 Inhibition of SETDB1 by Mithramycin A reduces colony formation in vitro and tumor growth in vivo.

a, A schematic illustration of the proposed model for the molecular mechanism underlying the tight regulation of Akt kinase activity and oncogenic functions by the synergy of SETDB1 and PI3K signaling axis. Where indicated, SETDB1 inhibitors were highlighted to combat tumors by reprogramming epigenome and repressing Akt oncogenic signaling. b, IB analysis of GST pull-down and WCL derived from DLD1 cells transfected with indicated constructs treated with different doses of Mithramycin A for 72 hrs before harvest. c-e, IB analysis of WCL derived from DLD1-AKT1/2-/- infected with WT or K140/142R-Akt1 lentivirus post treatment of different doses of Mithramycin A for 72 hrs (c). The resulting cells were subjected to colony formation assays (d). The experiment was performed twice independently with three repeats, and exhibit similar results (d). Representative images were shown in (d) and relative colony numbers derived from two independent experiments were plotted in (e). (e). f-i, Mithramycin A treatment retards in vivo tumorigenesis of xenografted DLD1 cells. When the tumors of xenografted DLD1 cells reached 100 mm3, the mice were treated with Mithramycin A (0.2 mg/kg) or PBS (as a negative control). Tumor sizes were monitored (f,g) and tumor mass were weighed and presented (h,i). Error bars are mean ± s.e.m, n = 8 mice. P value were calculated using two-way ANOVA analysis (g) or two-tailed unpaired Student’s t test (i). j, IB analysis of tumor lysates derived from the mice bearing A375 xenografted tumors treated with Mithramycin A or PBS. k, A schematic illustration of the proposed model for the molecular mechanisms underlying the tight regulation of Akt kinase activity and oncogenic functions by the SETDB1-KDM4B signaling axis. Where indicated, SETDB1 distributes both in nucleus and cytoplasm to repress gene expressions and promote oncogenic functions by methylating histone substrate H3K9 and non-histone substrate Akt, respectively. Conversely, KDM4B could de-methylate H3K9, Akt-K140/142 and other substrates to integratedly exert its physiological or pathological functions. Detailed statistical tests were described in Methods. Statistical source data for e,g and i are shown in Supplementary Table 2. All Western-blots above were performed twice independently with similar results. Scanned images of unprocessed blots are shown in Supplementary Fig. 8.

Supplementary Figure 8

Unprocessed scans of key blots.

Supplementary information

Supplementary Information

Supplementary Figures 1–8, and legends for Supplementary Tables 1 and 2

Reporting Summary

Supplementary Table 1

The list of lysine trimethylated peptides derived from IAP-LC-MS/MS based high throughput screening from OVCAR5 cells

Supplementary Table 2

Statistics source data

Rights and permissions

Reprints and permissions

About this article

Check for updates. Verify currency and authenticity via CrossMark

Cite this article

Guo, J., Dai, X., Laurent, B. et al. AKT methylation by SETDB1 promotes AKT kinase activity and oncogenic functions. Nat Cell Biol 21, 226–237 (2019). https://doi.org/10.1038/s41556-018-0261-6

Download citation

  • Received:

  • Accepted:

  • Published:

  • Issue Date:

  • DOI: https://doi.org/10.1038/s41556-018-0261-6

This article is cited by

Search

Quick links

Nature Briefing: Cancer

Sign up for the Nature Briefing: Cancer newsletter — what matters in cancer research, free to your inbox weekly.

Get what matters in cancer research, free to your inbox weekly. Sign up for Nature Briefing: Cancer