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:

SIRT1 deacetylates WEE1 and sensitizes cancer cells to WEE1 inhibition

Abstract

The cell-cycle checkpoint kinase WEE1 is emerging as a therapeutic target for cancer treatment. However, how its catalytic activity is regulated remains poorly understood, and reliable biomarkers for predicting response to WEE1 inhibitor remain to be identified. Here we identify an evolutionarily conserved segment surrounding its Lys177 residue that inhibits WEE1 activity through an intermolecular interaction with the catalytic kinase domain. Upon DNA damage, CHK1-dependent phosphorylation of WEE1 at Ser642 primes GCN5-mediated acetylation at Lys177, resulting in dissociation of the inhibitory segment from the kinase domain and subsequent activation of WEE1 and cell-cycle checkpoints. Conversely, SIRT1 associates with and deacetylates WEE1, which maintains it in an inactive state. Consequently, SIRT1 deficiency induces WEE1 hyperacetylation and activation, rendering cancer cells resistant to WEE1 inhibition. These results suggest that SIRT1 expression level and abundance of WEE1 Lys177 acetylation in tumor cells can serve as useful biomarkers for predicting WEE1 inhibitor sensitivity or resistance.

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: SIRT1 expression levels are negatively correlated with the IC50 values of MK-1775.
Fig. 2: SIRT1 interacts with and deacetylates WEE1.
Fig. 3: WEE1 is acetylated at Lys177.
Fig. 4: Acetylation at Lys177 directly activates WEE1 kinase activity.
Fig. 5: Phosphorylation by CHK1 stimulates WEE1 acetylation and activation.
Fig. 6: Acetylation of WEE1 releases the intermolecular inhibition of its kinase domain.

Similar content being viewed by others

Data availability

The authors declare that data supporting the findings of this study are available within the article, the accompanying source data files and the Supplementary Information. The pharmacogenomics dataset of 1,001 human cancer cell lines were obtained from the ArrayExpress database via the accession code E-MTAB-3610. Source data are provided with this paper.

References

  1. O’Connor, M. J. Targeting the DNA damage response in cancer. Mol. Cell 60, 547–560 (2015).

    Article  PubMed  Google Scholar 

  2. Castedo, M. et al. Cell death by mitotic catastrophe: a molecular definition. Oncogene 23, 2825–2837 (2004).

    Article  CAS  PubMed  Google Scholar 

  3. Matheson, C. J., Backos, D. S. & Reigan, P. Targeting WEE1 kinase in cancer. Trends Pharmacol. Sci. 37, 872–881 (2016).

    Article  CAS  PubMed  Google Scholar 

  4. Qiu, Z., Oleinick, N. L. & Zhang, J. ATR/CHK1 inhibitors and cancer therapy. Radiother. Oncol. 126, 450–464 (2018).

    Article  CAS  PubMed  Google Scholar 

  5. Weber, A. M. & Ryan, A. J. ATM and ATR as therapeutic targets in cancer. Pharmacol. Ther. 149, 124–138 (2015).

    Article  CAS  PubMed  Google Scholar 

  6. Fang, Y. et al. Sequential therapy with PARP and WEE1 inhibitors minimizes toxicity while maintaining efficacy. Cancer Cell 35, 851–867 e7 (2019).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  7. Pfister, S. X. et al. Inhibiting WEE1 selectively kills histone H3K36me3-deficient cancers by dNTP starvation. Cancer Cell 28, 557–568 (2015).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  8. Cleary, J. M., Aguirre, A. J., Shapiro, G. I. & D’Andrea, A. D. Biomarker-guided development of DNA repair inhibitors. Mol. Cell 78, 1070–1085 (2020).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  9. Hirai, H. et al. Small-molecule inhibition of Wee1 kinase by MK-1775 selectively sensitizes p53-deficient tumor cells to DNA-damaging agents. Mol. Cancer Ther. 8, 2992–3000 (2009).

    Article  CAS  PubMed  Google Scholar 

  10. Rajeshkumar, N. V. et al. MK-1775, a potent Wee1 inhibitor, synergizes with gemcitabine to achieve tumor regressions, selectively in p53-deficient pancreatic cancer xenografts. Clin. Cancer Res. 17, 2799–2806 (2011).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  11. Parker, L. L. & Piwnica-Worms, H. Inactivation of the p34cdc2-cyclin B complex by the human WEE1 tyrosine kinase. Science 257, 1955–1957 (1992).

    Article  CAS  PubMed  Google Scholar 

  12. Watanabe, N., Broome, M. & Hunter, T. Regulation of the human WEE1Hu CDK tyrosine 15-kinase during the cell cycle. EMBO J. 14, 1878–1891 (1995).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  13. Russell, P. & Nurse, P. Negative regulation of mitosis by wee1+, a gene encoding a protein kinase homolog. Cell 49, 559–567 (1987).

    Article  CAS  PubMed  Google Scholar 

  14. McGowan, C. H. & Russell, P. Human Wee1 kinase inhibits cell division by phosphorylating p34cdc2 exclusively on Tyr15. EMBO J. 12, 75–85 (1993).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  15. Beck, H. et al. Regulators of cyclin-dependent kinases are crucial for maintaining genome integrity in S phase. J. Cell Biol. 188, 629–638 (2010).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  16. Beck, H. et al. Cyclin-dependent kinase suppression by WEE1 kinase protects the genome through control of replication initiation and nucleotide consumption. Mol. Cell. Biol. 32, 4226–4236 (2012).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  17. Moiseeva, T. N., Qian, C., Sugitani, N., Osmanbeyoglu, H. U. & Bakkenist, C. J. WEE1 kinase inhibitor AZD1775 induces CDK1 kinase-dependent origin firing in unperturbed G1- and S-phase cells. Proc. Natl Acad. Sci. USA 116, 23891–23893 (2019).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  18. Heijink, A. M. et al. A haploid genetic screen identifies the G1/S regulatory machinery as a determinant of Wee1 inhibitor sensitivity. Proc. Natl Acad. Sci. USA 112, 15160–15165 (2015).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  19. Kreahling, J. M. et al. MK1775, a selective Wee1 inhibitor, shows single-agent antitumor activity against sarcoma cells. Mol. Cancer Ther. 11, 174–182 (2012).

    Article  CAS  PubMed  Google Scholar 

  20. Guertin, A. D. et al. Preclinical evaluation of the WEE1 inhibitor MK-1775 as single-agent anticancer therapy. Mol. Cancer Ther. 12, 1442–1452 (2013).

    Article  CAS  PubMed  Google Scholar 

  21. Bridges, K. A. et al. MK-1775, a novel Wee1 kinase inhibitor, radiosensitizes p53-defective human tumor cells. Clin. Cancer Res. 17, 5638–5648 (2011).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  22. Tominaga, Y., Li, C., Wang, R. H. & Deng, C. X. Murine Wee1 plays a critical role in cell cycle regulation and pre-implantation stages of embryonic development. Int J. Biol. Sci. 2, 161–170 (2006).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  23. Bukhari, A. B. et al. Inhibiting Wee1 and ATR kinases produces tumor-selective synthetic lethality and suppresses metastasis. J. Clin. Invest. 129, 1329–1344 (2019).

    Article  PubMed  PubMed Central  Google Scholar 

  24. Ruiz, S. et al. A genome-wide CRISPR screen identifies CDC25A as a determinant of sensitivity to ATR inhibitors. Mol. Cell 62, 307–313 (2016).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  25. Young, L. A. et al. Differential activity of ATR and WEE1 inhibitors in a highly sensitive subpopulation of DLBCL linked to replication stress. Cancer Res. 79, 3762–3775 (2019).

    Article  CAS  PubMed  Google Scholar 

  26. De Witt Hamer, P. C., Mir, S. E., Noske, D., Van Noorden, C. J. & Wurdinger, T. WEE1 kinase targeting combined with DNA-damaging cancer therapy catalyzes mitotic catastrophe. Clin. Cancer Res. 17, 4200–4207 (2011).

    Article  PubMed  Google Scholar 

  27. Geenen, J. J. J. & Schellens, J. H. M. Molecular pathways: targeting the protein kinase Wee1 in cancer. Clin. Cancer Res. 23, 4540–4544 (2017).

    Article  CAS  PubMed  Google Scholar 

  28. Iorio, F. et al. A landscape of pharmacogenomic interactions in cancer. Cell 166, 740–754 (2016).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  29. Chen, G. et al. Suppression of Sirt1 sensitizes lung cancer cells to WEE1 inhibitor MK-1775-induced DNA damage and apoptosis. Oncogene 36, 6863–6872 (2017).

    Article  CAS  PubMed  Google Scholar 

  30. Chen, X. et al. High levels of SIRT1 expression enhance tumorigenesis and associate with a poor prognosis of colorectal carcinoma patients. Sci. Rep. 4, 7481 (2014).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  31. Neumann, H., Peak-Chew, S. Y. & Chin, J. W. Genetically encoding Nε-acetyllysine in recombinant proteins. Nat. Chem. Biol. 4, 232–234 (2008).

    Article  CAS  PubMed  Google Scholar 

  32. Ryu, Y. & Schultz, P. G. Efficient incorporation of unnatural amino acids into proteins in Escherichia coli. Nat. Methods 3, 263–265 (2006).

    Article  CAS  PubMed  Google Scholar 

  33. Lee, J., Kumagai, A. & Dunphy, W. G. Positive regulation of Wee1 by Chk1 and 14-3-3 proteins. Mol. Biol. Cell 12, 551–563 (2001).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  34. O’Connell, M. J., Raleigh, J. M., Verkade, H. M. & Nurse, P. Chk1 is a wee1 kinase in the G2 DNA damage checkpoint inhibiting cdc2 by Y15 phosphorylation. EMBO J. 16, 545–554 (1997).

    Article  PubMed  PubMed Central  Google Scholar 

  35. Katayama, K., Fujita, N. & Tsuruo, T. Akt/protein kinase B-dependent phosphorylation and inactivation of WEE1Hu promote cell cycle progression at G2/M transition. Mol. Cell. Biol. 25, 5725–5737 (2005).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  36. Kawabe, T. G2 checkpoint abrogators as anticancer drugs. Mol. Cancer Ther. 3, 513–519 (2004).

    Article  CAS  PubMed  Google Scholar 

  37. Wang, J. et al. Acetylation of XPF by TIP60 facilitates XPF-ERCC1 complex assembly and activation. Nat. Commun. 11, 786 (2020).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  38. Jumper, J. et al. Highly accurate protein structure prediction with AlphaFold. Nature 596, 583–589 (2021).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  39. Nivon, L. G., Moretti, R. & Baker, D. A Pareto-optimal refinement method for protein design scaffolds. PLoS One 8, e59004 (2013).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  40. Leaver-Fay, A. et al. ROSETTA3: an object-oriented software suite for the simulation and design of macromolecules. Methods Enzymol. 487, 545–574 (2011).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  41. Marze, N. A., Roy Burman, S. S., Sheffler, W. & Gray, J. J. Efficient flexible backbone protein–protein docking for challenging targets. Bioinformatics 34, 3461–3469 (2018).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  42. Pettersen, E. F. et al. UCSF Chimera—a visualization system for exploratory research and analysis. J. Comput. Chem. 25, 1605–1612 (2004).

    Article  CAS  PubMed  Google Scholar 

Download references

Acknowledgements

We thank all members of the Liu group for insightful discussions. This work was supported by National Key Research and Development Program of China (2022YFA1302800 and 2021YFA1101000 to T.L.), National Natural Science Foundation of China (31961160725, 31730021, and 31971220 to J.H., 32270769, 31970664, and 31822031 to T.L., 81970956 to M.W.), Zhejiang Science Foundation for Distinguished Young Scholars (LR18C070001 to T.L.), Fok Ying Tung Education Foundation, and Fundamental Research Funds for the Zhejiang Provincial Universities (2021XZZX039 to J.H.).

Author information

Authors and Affiliations

Authors

Contributions

X.Z., Q.S., H.X., and L.S. performed the cell biology, biochemistry, and xenograft experiments; F.Y. assisted with computational modeling; D.Z., B.W., S.L., J.H., and M.W. participated in experimental design and data analysis. T.L. designed and supervised the project and wrote the manuscript.

Corresponding author

Correspondence to Ting Liu.

Ethics declarations

Competing interests

The authors declare no competing interests.

Peer review

Peer review information

Nature Chemical Biology thanks Gordon Chan, Zhenkun Lou and the other, anonymous, reviewer(s) for their contribution to the peer review of this work.

Additional information

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

Extended data

Extended Data Fig. 1 Correlation between the IC50 values of MK-1775 and SIRTs mRNA levels.

af, Two-sided P-values were determined using the relation between estimated coefficient and Student’s t distribution.

Extended Data Fig. 2 SIRT1 sensitizes cancer cells to WEE1 inhibitor MK-1775.

(a) Expression analysis of SIRT1 in different human cancer cell lines. (b–d) Knockdown of SIRT1 causes MK-1775 resistance. Cells infected with lentiviruses expressing control or SIRT1 shRNAs were treated with indicated doses of MK-1775 (0 nM, 50 nM, 100 nM, 150 nM, 300 nM, 600 nM, 800 nM, 1000 nM, 1200 nM, and 1500 nM for BxPC-3 cells; 0 nM, 50 nM, 100 nM, 150 nM, 300 nM, and 600 nM for PANC-1 and Capan-1 cells) for 48 hr or mock untreated. Cells were then shifted to fresh medium and permitted to grow for an additional 14 days before staining. Results shown are means of three independent experiments and were presented as means ± SEM. P-values were determined by two-way ANOVA. (e) Overexpression of SIRT1 sensitizes cells to MK-1775. BxPC-3 cells stably expressing Flag-tagged wild-type SIRT1 or the catalytic-inactive mutant were treated with indicated doses of MK-1775 (0 nM, 50 nM, 100 nM, 150 nM, 300 nM, and 600 nM) for 48 hr or mock untreated. Cells were then shifted to fresh medium and permitted to grow for an additional 14 days before staining. Results shown are means of three independent experiments and were presented as means ± SEM. P-values were determined by two-way ANOVA. (f–h) Cells infected with lentiviruses expressing control or SIRT1 shRNAs were treated with indicated doses of MK-1775 (0 nM, 50 nM, 100 nM, 150 nM, 300 nM, and 600 nM) for 48 hr or mock untreated. Cells were then shifted to fresh medium and permitted to grow for an additional 14 days before staining. Results shown are means of three independent experiments and were presented as means ± SEM. P-values were determined by two-way ANOVA.

Source data

Extended Data Fig. 3 SIRT1 depletion sensitizes cells to CPT.

MDA-MB-231 cells infected with lentiviruses expressing control or SIRT1 shRNAs were treated with indicated doses of CPT. Cells were then shifted to fresh medium and permitted to grow for an additional 14 days before staining. Results shown are means of three independent experiments and were presented as means ± SEM. P-values were determined by two-way ANOVA.

Source data

Extended Data Fig. 4 WEE1 is acetylated by GCN5 and deacetylated by SIRT1.

(a) SIRT1 interacts with WEE1. HEK293T cells were transfected with the indicated plasmids for 24 hr. Cell lysates were then prepared and western blot analysis was carried out as indicated. (b) SIRT1 directly interacts with WEE1 in vitro. GST-SIRT1 and MBP-WEE1 were expressed and purified from E. coli and subjected to MBP pull-down assays. (c–e) Association of endogenous SIRT1 with WEE1. Cells were irradiated with 40 J/m2 UV and allowed to recover for the indicated time. Cell lysates were then prepared and western blot analysis was carried out as indicated. (f) GCN5 acetylates WEE1 in vivo. HEK293T cells were transfected with the indicated plasmids for 24 hr. Cell lysates were then prepared and western blot analysis was carried out as indicated. (g) HEK293T cells were transfected with the indicated plasmids for 24 hr. Cell lysates were then prepared and western blot analysis was carried out as indicated. (h) WEE1 forms a complex with GCN5. HEK293T cells were transfected with the indicated plasmids for 24 hr. Cell lysates were then prepared and western blot analysis was carried out as indicated. (i) Association of endogenous WEE1 with GCN5. MDA-MB-231 cells were irradiated with 40 J/m2 UV and allowed to recover for the indicated time. Cell lysates were then prepared and western blot analysis was carried out as indicated. (j) WEE1 directly interacts with GCN5 in vitro. GST-GCN5 and MBP-WEE1 were expressed and purified from E. coli and subjected to MBP pull-down assays. (k) SIRT1 mediates WEE1 deacetylation. HEK293T cells were transfected with the indicated plasmids for 24 hr. Cell lysates were then prepared and western blot analysis was carried out as indicated. Data in A-K were independently replicated at least three times, with similar results.

Source data

Extended Data Fig. 5 WEE1 is acetylated at Lys177.

(a) The sequence of the region containing the acetylation site of WEE1 was aligned with those from various species. (b) Characterization of the anti-AcK177-WEE1 antibody by a dot blot assay. Various amounts of unacetylated or acetylated WEE1-K177 peptides were spotted onto nitrocellulose membrane and immunoblotted with the anti-AcK177-WEE1 antibody. (c) HEK293T cells were transfected with the indicated plasmids for 24 hr. Cell lysates were then prepared and western blot analysis was carried out as indicated. (d) HEK293T cells were transfected with the indicated plasmids for 24 hr. Cells were then left untreated or irradiated with 40 J/m2 UV and allowed to recover for 4 hr. Cell lysates were then prepared and western blot analysis was carried out as indicated. (e) HEK293T cells transfected with the indicated plasmids were left untreated or irradiated with 40 J/m2 UV and allowed to recover for 4 hr. Cell lysates were then prepared and western blot analysis was carried out as indicated. (f) U2OS cells were left untreated or treated with HU (4 mM), CPT (1 μM), or IR (10 Gy) for 4 hr. Cell lysates were then prepared and western blot analysis was carried out as indicated.

Source data

Extended Data Fig. 6 Acetylation is required for UV-induced CDK1 Tyr15 phosphorylation.

(a) WEE1 acetylation is required for G2/M checkpoint activation following DNA damage. A U2OS cell line to express siRNA-resistant SFB-WEE1 under the control of a tetracycline-inducible promoter was generated. The resulting cells were transfected with the indicated siRNAs and then treated with doxycycline (1 μg/mL) to induce the expression of WEE1. 8 hr later, cells were irradiated with 10 J/m2 UV and allowed to recover for 4 hr. Percentages of mitotic cells were determined by FACS analysis. Results shown are means of three independent experiments and were presented as means ± SEM. P-values were determined by two-tailed Student’s t-test. (b, c) Knockdown of GCN5 sensitizes cells to MK-1775. Results shown are means of three independent experiments and were presented as means ± SEM. P-values were determined by two-way ANOVA. (d, e) SIRT1 depletion delays mitotic entry and induces cell apoptosis. Percentages of mitotic and apoptotic cells were determined by FACS analysis. Results shown are means of three independent experiments and were presented as means ± SEM. P-values were determined by two-tailed Student’s t-test. (f) HEK293T cells were transfected with the indicated plasmids for 24 hr. Cell lysates were then prepared and western blot analysis was carried out as indicated. (g) SFB-WEE1 immunopurified from HEK293T cells were incubated with SFB-CDK1 in a kinase reaction mixture in the presence or absence of MK-1775 (10 μM). (h, i) SIRT1 sensitizes cells to MK-1775 by deacetylating WEE1. A MDA-MB-231 cell line to express siRNA-resistant SFB-WEE1 under the control of a tetracycline-inducible promoter was generated. The resulting cells were infected with lentiviruses expressing control or SIRT1 shRNAs and then transfected with the indicated siRNAs. 36 hr post-transfection, cells were treated with doxycycline (1 μg/mL for 24 hr) and indicated doses of MK-1775 for 48 hr. Results shown are means of three independent experiments and were presented as means ± SEM (I). P-values were determined by two-way ANOVA. Knockdown efficiency was confirmed by western blotting (H). Data in F-G were independently replicated at least three times, with similar results.

Source data

Extended Data Fig. 7 Acetylation at Lys177 directly activates WEE1.

(a) A schematic demonstrating the generation of site-specific acetylated recombinant WEE1 protein with an acetyl-lysine tRNA, which incorporates the acetyl-lysine on the amber codon. (b) Bacterially purified His-WEE1 or His-WEE1Lys177-Ac was incubated with SFB-CDK1 in a kinase reaction mixture. After reaction, proteins were resolved by SDS-PAGE and subjected to immunoblotting with anti-pY15-CDK1 antibody. Data in B were independently replicated at least three times, with similar results.

Source data

Extended Data Fig. 8 CHK1 promotes WEE1 phosphorylation at Ser642.

(a) U2OS cells were irradiated with 40 J/m2 UV and allowed to recover for the indicated time. (b) U2OS cells were pre-treated with CHK1 inhibitor MK-8776 (2 μM) or ATR inhibitor VE-821 (10 μM) for 1 hr prior to UV irradiation. (c) A U2OS cell line to express SFB-WEE1 under the control of a tetracycline-inducible promoter was generated. The resulting cells transfected with the indicated siRNAs were treated with doxycycline to induce the expression of WEE1. 8 hr later, cells were irradiated with 10 J/m2 UV and allowed to recover for 4 hr. Percentages of mitotic cells were determined by FACS analysis. Results shown are means of three independent experiments and were presented as means ± SEM. P-values were determined by two-tailed Student’s t-test. (d) A MDA-MB-231 cell line to express SFB-WEE1 under the control of a tetracycline-inducible promoter was generated. The resulting cells were transfected with the indicated siRNAs and then treated with doxycycline (24 hr) and indicated doses of MK-1775 for 48 hr. Results shown are means of three independent experiments and were presented as means ± SEM. P-values were determined by two-way ANOVA. (e) MDA-MB-231 cells were either synchronized in G1/S phase by a double thymidine (Thd) block or in mitosis using nocodazole (Noc) treatment. Cells were then released into the cell cycle and collected at the indicated time points. (f) MDA-MB-231 cells were synchronized in G1/S phase. Cells were then released into the cell cycle and collected at the indicated time points. (g) MDA-MB-231 cells were synchronized in G1/S phase. Cells were then treated with CHK1 inhibitor MK-8776 (2 μM), released into the cell cycle, and collected at the indicated time points. (h) MDA-MB-231 cells were either synchronized in G1/S phase or in mitosis. Cells were then released into the cell cycle and collected at the indicated time points. The co-immunoprecipitated GCN5/SIRT1 levels were normalized with immunoprecipitated WEE1 levels expressed as the normalized ratio (shown below the blots). Data in A-B, E-H were independently replicated at least three times, with similar results.

Source data

Extended Data Fig. 9 Aberrant WEE1 kinase activity induces cell apoptosis.

(a) A U2OS cell line to express siRNA-resistant SFB-WEE1 under the control of a tetracycline-inducible promoter was generated. The resulting cells were transfected with the indicated siRNAs and then treated with doxycycline (1 μg/mL) to induce the expression of WEE1. 48 hr later, percentages of apoptotic cells were determined by FACS analysis. Results shown are means of three independent experiments and were presented as means ± SEM. P-values were determined by two-tailed Student’s t-test. (b) HEK293T cells were transfected with the indicated plasmids for 24 hr. Cells were then fixed, permeabilized, and stained with anti-Flag antibody. Scale bar: 10 μm. Data in B were independently replicated at least three times, with similar results.

Source data

Extended Data Fig. 10 Acetylation relieves the inhibitory interaction between the KIM and the catalytic kinase domain.

(a) WEE1 self-associates in an intermolecular fashion. HEK293T cells transfected with the indicated plasmids were irradiated with 40 J/m2 UV and allowed to recover for 4 hr. Cell lysates were then prepared and western blot analysis was carried out as indicated. (b) WEE1 exists primarily as a dimer in non-irradiated cells. U2OS cells were irradiated with 40 J/m2 UV and allowed to recover for the indicated time. Cell lysates were then prepared and western blot analysis was carried out as indicated. (c) Mutation of Asp405 to Ala on WEE1-CK impairs its interaction with WEE1-N. Bacterially purified GST-WEE1-N or GST-WEE1-N-K177Q was incubated with amylose resin conjugated with MBP-WEE1-CK, MBP-WEE1-CK-E402A, MBP-WEE1-CK-D405A, or MBP-WEE1-CK-D426A. Proteins retained on the amylose resin were immunoblotted with anti-GST antibody. (d) Mutation of Asp405 to Ala leads to constitutive activation of WEE1. Bacterially purified MBP-WEE1-WT, MBP-WEE1-K177Q, or MBP-WEE1-D405A was incubated with SFB-CDK1 in a kinase reaction mixture. After reaction, proteins were resolved by SDS-PAGE and subjected to immunoblotting. (e) Bacterially purified MBP-WEE1-N, MBP-WEE1-N-K177R, or MBP-WEE1-N-K177Q was mixed with SFB-WEE1 immunopurified from HEK293T cells that were left untreated or irradiated with 40 J/m2 UV and allowed to recover for 4 hr in a kinase reaction buffer. The mixture was then incubated with SFB-CDK1 in the presence of 0.3 mM ATP. After reaction, proteins were resolved by SDS-PAGE and subjected to immunoblotting. (f) Bacterially purified MBP-WEE1-N, MBP-WEE1-N-K177R, or MBP-WEE1-N-K177Q was mixed with MBP-WEE1-CK in a kinase reaction buffer. The mixture was then incubated with SFB-CDK1 in the presence of 0.3 mM ATP. After reaction, proteins were resolved by SDS-PAGE and subjected to immunoblotting. Data in A-F were independently replicated at least three times, with similar results.

Source data

Supplementary information

Source data

Source Data Fig. 1

Statistical source data.

Source Data Fig. 1

Unprocessed western blots.

Source Data Fig. 2

Unprocessed western blots.

Source Data Fig. 3

Unprocessed western blots.

Source Data Fig. 4

Unprocessed western blots.

Source Data Fig. 5

Unprocessed western blots.

Source Data Fig. 6

Unprocessed western blots.

Source Data Extended Data Fig. 2

Unprocessed western blots.

Source Data Extended Data Fig. 2

Statistical source data.

Source Data Extended Data Fig. 3

Statistical source data.

Source Data Extended Data Fig. 3

Unprocessed western blots.

Source Data Extended Data Fig. 4

Unprocessed western blots.

Source Data Extended Data Fig. 5

Unprocessed western blots.

Source Data Extended Data Fig. 6

Statistical source data.

Source Data Extended Data Fig. 6

Unprocessed western blots.

Source Data Extended Data Fig. 7

Unprocessed western blots.

Source Data Extended Data Fig. 8

Unprocessed western blots.

Source Data Extended Data Fig. 8

Statistical source data.

Source Data Extended Data Fig. 9

Statistical source data.

Source Data Extended Data Fig. 10

Unprocessed western blots.

Rights and permissions

Springer Nature or its licensor (e.g. a society or other partner) holds exclusive rights to this article under a publishing agreement with the author(s) or other rightsholder(s); author self-archiving of the accepted manuscript version of this article is solely governed by the terms of such publishing agreement and applicable law.

Reprints and permissions

About this article

Check for updates. Verify currency and authenticity via CrossMark

Cite this article

Zhu, X., Su, Q., Xie, H. et al. SIRT1 deacetylates WEE1 and sensitizes cancer cells to WEE1 inhibition. Nat Chem Biol 19, 585–595 (2023). https://doi.org/10.1038/s41589-022-01240-y

Download citation

  • Received:

  • Accepted:

  • Published:

  • Issue Date:

  • DOI: https://doi.org/10.1038/s41589-022-01240-y

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