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:

Nuclear GSK3β promotes tumorigenesis by phosphorylating KDM1A and inducing its deubiquitylation by USP22

Abstract

Emerging evidence has shown that GSK3β plays oncogenic roles in multiple tumour types; however, the underlying mechanisms remain largely unknown. Here, we show that nuclear GSK3β is responsible for the accumulation of the histone demethylase KDM1A and critically regulates histone H3K4 methylation during tumorigenesis. GSK3β phosphorylates KDM1A Ser683 upon priming phosphorylation of KDM1A Ser687 by CK1α. Phosphorylation of KDM1A induces its binding with and deubiquitylation by USP22, leading to KDM1A stabilization. GSK3β- and USP22-dependent KDM1A stabilization is required for the demethylation of histone H3K4, thereby repressing BMP2, CDKN1A and GATA6 transcription, which results in cancer stem cell self-renewal and glioblastoma tumorigenesis. In human glioblastoma specimens, KDM1A levels are correlated with nuclear GSK3β and USP22 levels. Furthermore, a GSK3 inhibitor, tideglusib, sensitizes tumour xenografts to chemotherapy in mice via KDM1A downregulation and improves survival. Our findings demonstrate that nuclear GSK3β- and USP22-mediated KDM1A stabilization is essential for glioblastoma tumorigenesis.

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: GSK3β stabilizes KDM1A by decreasing its ubiquitylation.
Figure 2: GSK3β phosphorylates KDM1A Ser683 after the phosphorylation priming by CK1α.
Figure 3: USP22 is a deubiquitylase of KDM1A.
Figure 4: GSK3β promotes the binding and deubiquitylation of KDM1A by USP22.
Figure 5: GSK3β and USP22 repress KDM1A target genes and are required for the maintenance of GSCs.
Figure 6: The GSK3β–USP22–KDM1A axis is required for gliomagenesis of GSCs and associated with the grade of glioma malignance in human glioma specimens.
Figure 7: The GSK3 inhibitor tideglusib attenuates GSC self-renewal and enhances the effect of TMZ on GSC proliferation.
Figure 8: Tideglusib attenuates the tumour-initiating ability of GSCs and sensitizes GSC-derived xenografts to TMZ.

Similar content being viewed by others

References

  1. Cohen, P. & Frame, S. The renaissance of GSK3. Nat. Rev. Mol. Cell Biol. 2, 769–776 (2001).

    CAS  PubMed  Google Scholar 

  2. Doble, B. W. & Woodgett, J. R. GSK-3: tricks of the trade for a multi-tasking kinase. J. Cell Sci. 116, 1175–1186 (2003).

    CAS  PubMed  PubMed Central  Google Scholar 

  3. Hoeflich, K. P. et al. Requirement for glycogen synthase kinase-3β in cell survival and NF-κB activation. Nature 406, 86–90 (2000).

    CAS  Google Scholar 

  4. Farago, M. et al. Kinase-inactive glycogen synthase kinase 3β promotes Wnt signaling and mammary tumorigenesis. Cancer Res. 65, 5792–5801 (2005).

    CAS  PubMed  Google Scholar 

  5. Wang, Z. et al. GSK-3 promotes conditional association of CREB and its coactivators with MEIS1 to facilitate HOX-mediated transcription and oncogenesis. Cancer Cell 17, 597–608 (2010).

    CAS  PubMed  PubMed Central  Google Scholar 

  6. Naito, S. et al. Glycogen synthase kinase-3β: a prognostic marker and a potential therapeutic target in human bladder cancer. Clin. Cancer Res. 16, 5124–5132 (2010).

    CAS  PubMed  Google Scholar 

  7. Aberle, H., Bauer, A., Stappert, J., Kispert, A. & Kemler, R. β-catenin is a target for the ubiquitin-proteasome pathway. EMBO J. 16, 3797–3804 (1997).

    CAS  PubMed  PubMed Central  Google Scholar 

  8. Sears, R. et al. Multiple Ras-dependent phosphorylation pathways regulate Myc protein stability. Genes Dev. 14, 2501–2514 (2000).

    CAS  PubMed  PubMed Central  Google Scholar 

  9. Tang, Q. L. et al. Glycogen synthase kinase-3β, NF-κB signaling, and tumorigenesis of human osteosarcoma. J. Natl Cancer Inst. 104, 749–763 (2012).

    CAS  PubMed  PubMed Central  Google Scholar 

  10. Miyashita, K. et al. Potential therapeutic effect of glycogen synthase kinase 3β inhibition against human glioblastoma. Clin. Cancer Res. 15, 887–897 (2009).

    CAS  PubMed  Google Scholar 

  11. Ougolkov, A. V. et al. Aberrant nuclear accumulation of glycogen synthase kinase-3β in human pancreatic cancer: association with kinase activity and tumor dedifferentiation. Clin. Cancer Res. 12, 5074–5081 (2006).

    CAS  PubMed  PubMed Central  Google Scholar 

  12. Ougolkov, A. V., Bone, N. D., Fernandez-Zapico, M. E., Kay, N. E. & Billadeau, D. D. Inhibition of glycogen synthase kinase-3 activity leads to epigenetic silencing of nuclear factor κB target genes and induction of apoptosis in chronic lymphocytic leukemia B cells. Blood 110, 735–742 (2007).

    CAS  PubMed  PubMed Central  Google Scholar 

  13. Reddiconto, G. et al. Targeting of GSK3β promotes imatinib-mediated apoptosis in quiescent CD34 + chronic myeloid leukemia progenitors, preserving normal stem cells. Blood 119, 2335–2345 (2012).

    CAS  PubMed  Google Scholar 

  14. Esteller, M. Cancer epigenomics: DNA methylomes and histone-modification maps. Nat. Rev. Genet. 8, 286–298 (2007).

    CAS  PubMed  Google Scholar 

  15. Hill, E. V. et al. Glycogen synthase kinase-3 controls IL-10 expression in CD4(+) effector T-cell subsets through epigenetic modification of the IL-10 promoter. Eur. J. Immunol. 45, 1103–1115 (2015).

    CAS  PubMed  PubMed Central  Google Scholar 

  16. Shi, Y. J. et al. Regulation of LSD1 histone demethylase activity by its associated factors. Mol. Cell 19, 857–864 (2005).

    CAS  PubMed  Google Scholar 

  17. Wang, Y. et al. LSD1 is a subunit of the NuRD complex and targets the metastasis programs in breast cancer. Cell 138, 660–672 (2009).

    CAS  PubMed  Google Scholar 

  18. Lv, S. et al. LSD1 is required for chromosome segregation during mitosis. Eur. J. Cell Biol. 89, 557–563 (2010).

    CAS  PubMed  Google Scholar 

  19. Kauffman, E. C. et al. Role of androgen receptor and associated lysine-demethylase coregulators, LSD1 and JMJD2A, in localized and advanced human bladder cancer. Mol. Carcinog. 50, 931–944 (2011).

    CAS  PubMed  PubMed Central  Google Scholar 

  20. Hayami, S. et al. Overexpression of LSD1 contributes to human carcinogenesis through chromatin regulation in various cancers. Int. J. Cancer 128, 574–586 (2011).

    CAS  PubMed  Google Scholar 

  21. Harris, W. J. et al. The histone demethylase KDM1A sustains the oncogenic potential of MLL-AF9 leukemia stem cells. Cancer Cell 21, 473–487 (2012).

    CAS  PubMed  Google Scholar 

  22. Sareddy, G. R. et al. KDM1 is a novel therapeutic target for the treatment of gliomas. Oncotarget 4, 18–28 (2013).

    PubMed  Google Scholar 

  23. Suva, M. L. et al. Reconstructing and reprogramming the tumor-propagating potential of glioblastoma stem-like cells. Cell 157, 580–594 (2014).

    CAS  PubMed  PubMed Central  Google Scholar 

  24. Gong, A. H. et al. FoxM1 drives a feed-forward STAT3-activation signaling loop that promotes the self-renewal and tumorigenicity of glioblastoma stem-like cells. Cancer Res. 75, 2337–2348 (2015).

    CAS  PubMed  PubMed Central  Google Scholar 

  25. Rena, G., Bain, J., Elliott, M. & Cohen, P. D4476, a cell-permeant inhibitor of CK1, suppresses the site-specific phosphorylation and nuclear exclusion of FOXO1a. EMBO Rep. 5, 60–65 (2004).

    CAS  PubMed  Google Scholar 

  26. Adamo, A. et al. LSD1 regulates the balance between self-renewal and differentiation in human embryonic stem cells. Nat. Cell Biol. 13, 652–659 (2011).

    CAS  PubMed  Google Scholar 

  27. Sun, G. et al. Histone demethylase LSD1 regulates neural stem cell proliferation. Mol. Cell. Biol. 30, 1997–2005 (2010).

    CAS  PubMed  PubMed Central  Google Scholar 

  28. Dominguez, J. M. et al. Evidence for irreversible inhibition of glycogen synthase kinase-3β by tideglusib. J. Biol. Chem. 287, 893–904 (2012).

    CAS  PubMed  Google Scholar 

  29. Bolos, M., Fernandez, S. & Torres-Aleman, I. Oral administration of a GSK3 inhibitor increases brain insulin-like growth factor I levels. J. Biol. Chem. 285, 17693–17700 (2010).

    CAS  PubMed  PubMed Central  Google Scholar 

  30. Sereno, L. et al. A novel GSK-3β inhibitor reduces Alzheimer’s pathology and rescues neuronal loss in vivo. Neurobiol. Dis. 35, 359–367 (2009).

    CAS  PubMed  Google Scholar 

  31. Chen, J. et al. A restricted cell population propagates glioblastoma growth after chemotherapy. Nature 488, 522–526 (2012).

    CAS  PubMed  PubMed Central  Google Scholar 

  32. Liu, C. et al. Control of β-catenin phosphorylation/degradation by a dual-kinase mechanism. Cell 108, 837–847 (2002).

    CAS  PubMed  Google Scholar 

  33. Gregory, M. A., Qi, Y. & Hann, S. R. Phosphorylation by glycogen synthase kinase-3 controls c-myc proteolysis and subnuclear localization. J. Biol. Chem. 278, 51606–51612 (2003).

    CAS  PubMed  Google Scholar 

  34. Shin, S., Wolgamott, L., Yu, Y., Blenis, J. & Yoon, S. O. Glycogen synthase kinase (GSK)-3 promotes p70 ribosomal protein S6 kinase (p70S6K) activity and cell proliferation. Proc. Natl Acad. Sci. USA 108, E1204–E1213 (2011).

    PubMed  Google Scholar 

  35. Kotliarova, S. et al. Glycogen synthase kinase-3 inhibition induces glioma cell death through c-MYC, nuclear factor-κB, and glucose regulation. Cancer Res. 68, 6643–6651 (2008).

    CAS  PubMed  PubMed Central  Google Scholar 

  36. Beurel, E. & Jope, R. S. Differential regulation of STAT family members by glycogen synthase kinase-3. J. Biol. Chem. 283, 21934–21944 (2008).

    CAS  PubMed  PubMed Central  Google Scholar 

  37. Park, B. H., Qiang, L. & Farmer, S. R. Phosphorylation of C/EBPβ at a consensus extracellular signal-regulated kinase/glycogen synthase kinase 3 site is required for the induction of adiponectin gene expression during the differentiation of mouse fibroblasts into adipocytes. Mol. Cell. Biol. 24, 8671–8680 (2004).

    CAS  PubMed  PubMed Central  Google Scholar 

  38. Foltz, D. R., Santiago, M. C., Berechid, B. E. & Nye, J. S. Glycogen synthase kinase-3β modulates notch signaling and stability. Curr. Biol. 12, 1006–1011 (2002).

    CAS  PubMed  Google Scholar 

  39. Kubic, J. D., Mascarenhas, J. B., Iizuka, T., Wolfgeher, D. & Lang, D. GSK-3 promotes cell survival, growth, and PAX3 levels in human melanoma cells. Mol. Cancer Res. 10, 1065–1076 (2012).

    CAS  PubMed  PubMed Central  Google Scholar 

  40. Schaeffer, V. et al. Binding of OTULIN to the PUB domain of HOIP controls NF-κB signaling. Mol. Cell 54, 349–361 (2014).

    CAS  PubMed  Google Scholar 

  41. Elliott, P. R. et al. Molecular basis and regulation of OTULIN-LUBAC interaction. Mol. Cell 54, 335–348 (2014).

    CAS  PubMed  PubMed Central  Google Scholar 

  42. Wu, Y. et al. The deubiquitinase USP28 stabilizes LSD1 and confers stem-cell-like traits to breast cancer cells. Cell Rep. 5, 224–236 (2013).

    CAS  PubMed  PubMed Central  Google Scholar 

  43. Zhang, X. Y. et al. The putative cancer stem cell marker USP22 is a subunit of the human SAGA complex required for activated transcription and cell-cycle progression. Mol. Cell 29, 102–111 (2008).

    PubMed  PubMed Central  Google Scholar 

  44. Lin, Z. et al. USP22 antagonizes p53 transcriptional activation by deubiquitinating Sirt1 to suppress cell apoptosis and is required for mouse embryonic development. Mol. Cell 46, 484–494 (2012).

    CAS  PubMed  Google Scholar 

  45. Tolosa, E. et al. A phase 2 trial of the GSK-3 inhibitor tideglusib in progressive supranuclear palsy. Mov. Disord. 29, 470–478 (2014).

    CAS  PubMed  Google Scholar 

  46. Persano, L. et al. BMP2 sensitizes glioblastoma stem-like cells to Temozolomide by affecting HIF-1α stability and MGMT expression. Cell Death Differ. 3, e412 (2012).

    CAS  Google Scholar 

  47. Kamnasaran, D., Qian, B., Hawkins, C., Stanford, W. L. & Guha, A. GATA6 is an astrocytoma tumor suppressor gene identified by gene trapping of mouse glioma model. Proc. Natl Acad. Sci. USA 104, 8053–8058 (2007).

    CAS  PubMed  Google Scholar 

  48. Ligon, K. L. et al. Olig2-regulated lineage-restricted pathway controls replication competence in neural stem cells and malignant glioma. Neuron 53, 503–517 (2007).

    CAS  PubMed  PubMed Central  Google Scholar 

  49. Hirose, Y., Berger, M. S. & Pieper, R. O. p53 effects both the duration of G2/M arrest and the fate of temozolomide-treated human glioblastoma cells. Cancer Res. 61, 1957–1963 (2001).

    CAS  PubMed  Google Scholar 

  50. Schonberg, D. L. et al. Preferential iron trafficking characterizes glioblastoma stem-like cells. Cancer Cell 28, 441–455 (2015).

    CAS  PubMed  PubMed Central  Google Scholar 

  51. Hu, Y. & Smyth, G. K. ELDA: extreme limiting dilution analysis for comparing depleted and enriched populations in stem cell and other assays. J. Immunol. Methods 347, 70–78 (2009).

    CAS  PubMed  Google Scholar 

  52. Zhang, N. et al. FoxM1 promotes β-catenin nuclear localization and controls Wnt target-gene expression and glioma tumorigenesis. Cancer Cell 20, 427–442 (2011).

    CAS  PubMed  PubMed Central  Google Scholar 

  53. Shembade, N. & Harhaj, E. W. Elucidating dynamic protein-protein interactions and ubiquitination in NF-κB signaling pathways. Methods Mol. Biol. 1280, 283–295 (2015).

    CAS  PubMed  Google Scholar 

  54. Li, M. et al. Deubiquitination of p53 by HAUSP is an important pathway for p53 stabilization. Nature 416, 648–653 (2002).

    CAS  PubMed  Google Scholar 

Download references

Acknowledgements

We thank J. Munch in MD Anderson’s Department of Scientific Publications for editing the manuscript. We thank Y. Shi (Boston Children’s Hospital) for providing the Flag–KDM1A plasmid; J. Yang (Texas Children’s Cancer Center) for providing the USP2, USP4, USP5, USP8, USP13, USP14, USP16, USP18, USP21, USP25, USP26, USP28, USP29, USP30, USP36, USP39, USP46 and USP48 expression plasmids; H.-K. Lin (MD Anderson Cancer Center) for providing the USP1, USP3, USP11, USP15, USP22 and CYLD expression plasmids; and X. He (Boston Children’s Hospital) for providing β-catenin+/+ and β-catenin−/− MEFs. This work was supported in part by US National Cancer Institute grants R01CA157933, R01CA182684, R01CA152309, P50CA127001, R01CA195651 and CA16672 (Cancer Center Support Grant).

Author information

Authors and Affiliations

Authors

Contributions

S.H. and A.Z. conceived the project and designed the study; A.Z. and K.L. performed most of the experiments under the supervision of S.H.; S.Z. and Y.C. assisted in some in vitro experiments; J.X. and Z.W. assisted in the mouse experiments; N.Z., K.D.A., K.X. and J.R.W. provided reagents and conceptual advice; S.H. and A.Z. wrote the manuscript. All authors discussed the results and commented on the manuscript.

Corresponding author

Correspondence to Suyun Huang.

Ethics declarations

Competing interests

The authors declare no competing financial interests.

Integrated supplementary information

Supplementary Figure 1 GSK3β stabilizes KDM1A by inhibiting the proteasome degradation pathway.

(a) KDM1A protein levels were detected in GSK3β knockout (GSK3β+/+) mouse embryonic fibroblasts (MEFs), GSK3β knockout (GSK3β−/−) MEFs, and GSK3β−/− MEFs with reconstituted expression of GSK3β. (b) Effect of GSK3β knockdown on KDM1A expression. GSC11 cells were transfected with GSK3β siRNAs, and KDM1A expression was detected by immunoblotting. (c) Effect of GSK3α knockdown on KDM1A expression. KDM1A protein levels were detected in GSC11 cells after transfection of GSK3α siRNAs. (d) KDM1A mRNA levels were detected in GSK3β+/+ and GSK3β−/− MEFs. GAPDH was used as an internal control. (e) KDM1A protein levels were detected in β-catenin+/+ and β-catenin−/− MEFs. (f) KDM1A is regulated by the ubiquitin-proteasome pathway. HS683 cells were treated with cycloheximide (CHX) with or without the proteasome inhibitor MG132 for the indicated times and KDM1A levels were detected by immunoblotting. (g) 293T cells were transfected with Flag-KDM1A and the constitutively active form of GSK3β (GSK3β-CA) or the control vector, and then treated with CHX as indicated. Expression of KDM1A was detected by immunoblotting. Fold change of western blot bands was determined as described in Fig. 1b and shown. Source data of Supplementary Fig. 1d can be found in Supplementary Table 3. Uncropped images of blots are shown in Supplementary Fig. 8.

Supplementary Figure 2 GSK3β phosphorylates KDM1A serine 683 after priming phosphorylation by CKIα.

(a) The amino acid sequence of KDM1A shows the sites potentially targeted by GSK3β. (b) Immunofluorescence assays were performed in HFU-251 MG and GSC11 cells using antibodies against KDM1A and GSK3β. Scale bar, 50 μM. (c) Effects of mutations of various Ser/Thr residues on the ubiquitination of KDM1A. 293T cells were transfected with HA-Ubi, Myc-GSK3β, and wild-type Flag-KDM1A or various mutants with serine/threonine-to-alanine mutations. Cells were treated with MG132 for 4 h, harvested, and then lysed for immunoprecipitation (IP) analysis. (d) 293T cells were transfected with wild-type Flag-KDM1A (WT) or mutant with serine 683-to-alanine (S683A) or aspartic acid (S683D) mutation. Cells were treated with CHX for the indicated times. Band intensity of KDM1A from western blots was quantified as described in Fig. 1b (mean ± s.d., n = 3 independent experiments, two-tailed Student’s t-test). P < 0.01. (e) GSC11 or HFU-251 MG cells were treated with the CKI inhibitor D4476 or D4476 plus LiCl for 6 h. Cell lysates were subjected to immunoblotting analysis. Fold change of western blot bands was determined as described in Fig. 1b and shown. (f) Reciprocal interaction between Flag-KDM1A and HA-CK1α. 293T cells were transfected with Flag-KDM1A and HA-CKIα, HA-CKIδ, or HA-CKIɛ plasmids. Cell lysates were immunoprecipitated with the HA-tag (left panel) or Flag-tag antibody (right panel) and then analysed by immunoblotting using the indicated primary antibodies. Blots were incubated with a HRP-conjugated secondary antibody (light chain specific). (g) Reciprocal interaction of endogenous KDM1A with CK1α. GSC11 cell lysates were immunoprecipitated using antibodies against CKIα, CKIδ, and CKIɛ (left panel), or KDM1A (right panel). The immunoprecipitates were subjected to immunoblotting analysis using the indicated primary antibodies and a HRP-conjugated secondary antibody (light chain specific). Inputs correspond to 2% total cell lysates. (h) HF-U251 MG cells were transfected two independent siRNAs against CKIα, CKIδ, or CKIɛ, and then the expression of KDM1A was detected by western blotting. Uncropped images of blots are shown in Supplementary Fig. 8.

Supplementary Figure 3 USP22 deubiquitinates and stabilizes KDM1A.

(a) Effect of different deubiquitinating enzymes (DUBs) on the expression of KDM1A. 293T cells were transfected with different DUBs and then lysed for immunoblotting to detect KDM1A expression. (b) Effect of various DUBs on the ubiquitination of KDM1A. 293T cells were transfected with Flag-KDM1A, Myc-Ubi, and USP15, USP21, USP22, or USP28 and then treated with MG132 for 6 h before harvest. Cell lysates were immunoprecipitated using an anti-KDM1A antibody and then subjected to immunoblotting analysis using the indicated antibodies. (c,d) 293T cells were transfected with Flag-KDM1A, Myc-Ubi, and HA-USP22 or Flag-USP28. Cell lysates were immunoprecipitated using antibodies against Myc-tag (c) or KDM1A (d). (e) Knockdown of USP22 does not affect KDM1A mRNA level. GSC11 cell were transfected with control siRNA or USP22 siRNA and KDM1A mRNA were detected by real-time PCR. Values were normalized to that in control (mean ± s.e.m., n = 3 independent experiments, two-tailed Student’s t-test). GAPDH mRNA was used as an internal control. P > 0.05. (f) 293T cells were transfected with HA-USP22 or Flag-USP28 plasmid and then treated with 50 μg ml−1 CHX for the indicated times. Cell lysates were subjected to immunoblotting analysis as indicated. (g) The expression levels of nuclear KDM1A, USP22 and GSK3β in Fig. 3f were determined by quantification of the intensity of western blot bands, using the Lamin B for normalization and the results are expressed as level relative to NHA-E6/E7 cells. Then the correlation of nuclear KDM1A/USP22 and KDM1A/GSK3β levels in different cell lines was analysed (mean ± s.e.m., n = 3 independent experiments, Pearson correlation test). (h) Immunofluorescence assay was used to analyse the co-localization of KDM1A and USP22 in GSC11 cells. Scale bar, 20 μm. Uncropped images of blots are shown in Supplementary Fig. 8.

Supplementary Figure 4 GSK3β promotes the binding of KDM1A with USP22.

293T cells were transfected with Flag-KDM1A, HA-USP22, and GSK3β-CA or GSK3β-KD and then treated with MG132 for 6 h before harvest. Cell lysates were immunoprecipitated with antibodies against (a) HA or (b) KDM1A and then analysed by immunoblotting using the indicated antibodies. Uncropped images of blots are shown in Supplementary Fig. 8.

Supplementary Figure 5 GSK3β and USP22 promote stem cell self-renewal through KDM1A.

(a) ChIP assays were performed in GSC11 cells transfected with control shRNA, shRNA targeting KDM1A, GSK3β or USP22 using an antibody against KDM1A and primers in the promoter regions of BMP2, CDKNA1, and GATA6. ACTB promoter was used as a negative control. Values are the percentage to input (mean ± s.e.m., n = 3 independent experiments, two-tailed Student’s t-test). P < 0.01. (b) KDM1A expression was detected by immunoblotting in GSC11 cells stably expressing two individual shRNAs targeting KDM1A. (c) Primary (1st) and secondary (2nd) neurosphere formation were assessed in GSC11 and GSC20 cells using two different KDM1A shRNA. Scale bar, 500 μm. (d) Effects of KDM1A knockdown on the expression of stem cell self-renewal and differentiation markers. GSC11 and GSC20 cells expressing KDM1A shRNAs were analysed by immunoblotting using the indicated antibodies. (e) Immunofluorescence assays were performed to detect the expression of self-renewal and differentiation markers in GSC11 cells expressing KDM1A shRNAs. Scale bar, 100 μm. (f) GSC11 cells overexpressing KDM1A shRNAs were reconstituted by the expression of shRNA-resistant KDM1A (ShR), and neurosphere formation was assessed. Scale bar, 500 μm. (g) USP22 knockdown inhibits neurosphere formation through KDM1A. GSC11 cells overexpressing USP22 shRNAs were reconstituted by the expression of wild-type KDM1A, and neurosphere formation was assessed. Scale bar, 500 μm. (h) GSC11 cells stably expressing two different shRNAs targeting GSK3β were analysed by immunoblotting. (i) mRNA levels of KDM1A in GSC11 cells expressing sh-control, sh-GSK3β, sh-GSK3β + KDM1A S683D, and sh-GSK3β + KDM1A S683A were analysed by real-time PCR (mean ± s.e.m., n = 3 independent experiments, two-tailed Student’s t-test). (j) The secondary neurosphere formation efficiency (spheres/cells plated) of GSC11 cells stably expressing the indicated shRNAs or proteins were calculated (mean ± s.e.m., n = 3 independent experiments, two-tailed Student’s t-test). P < 0.05. Uncropped images of blots are shown in Supplementary Fig. 8.

Supplementary Figure 6 Immunohistochemical analysis of GSK3β and KDM1A in mouse brain tumours derived from GSC11 cells expressing Sh-Control, Sh-GSK3β, ShGSK3β + KDM1A S683D, or Sh-GSK3β + KDM1A S683A.

Insets: high magnification images corresponding to the areas marked by yellow dot lines. Scale bar for H&E staining, 50 μm; Scale bar for IHC, 25 μm.

Supplementary Figure 7 Tideglusib inhibits GSC self-renewal and tumour formation.

(a) Effect of tideglusib on Tau phosphorylation. GSC11 cells were treated with the indicated concentrations of tideglusib for 6 h (left panel) or with tideglusib at a final concentration of 2.5 μM for the indicated times (right panel) in the presence of MG132. Cell lysates were analysed by western blotting using antibodies against Tau or phospho-Tau (Ser396). Fold change of western blot bands was determined as described in Fig. 1b and shown. (b) Effect of tideglusib on the expression of stem cell self-renewal and differentiation markers. GSC11 and GSC20 cells were treated with 5 μM tideglusib for 48 h, and Oct4, CD133, and Tuj-1 expression was analysed by immunofluorescence. Scale bar, 50 μm. (c) NHAs were treated with 5 μM tideglusib for the indicated times, and cell viability was analysed using XTT assays. (d) Schedule for animal treatment. For treatment with TMZ (20 mg kg−1 d−1) or tideglusib (25 mg kg−1 d−1) alone, mice were intraperitoneally injected every other day. For combinatorial treatment, mice received injections of TMZ or tideglusib on alternating days for 30 d. Source data of Supplementary Fig. 7c can be found in Supplementary Table 3. Uncropped images of blots are shown in Supplementary Fig. 8.

Supplementary information

Supplementary Information

Supplementary Information (PDF 2194 kb)

Supplementary Table 1

Supplementary Information (XLSX 11 kb)

Supplementary Table 2

Supplementary Information (XLSX 11 kb)

Supplementary Table 3

Supplementary Information (XLSX 12 kb)

Rights and permissions

Reprints and permissions

About this article

Check for updates. Verify currency and authenticity via CrossMark

Cite this article

Zhou, A., Lin, K., Zhang, S. et al. Nuclear GSK3β promotes tumorigenesis by phosphorylating KDM1A and inducing its deubiquitylation by USP22. Nat Cell Biol 18, 954–966 (2016). https://doi.org/10.1038/ncb3396

Download citation

  • Received:

  • Accepted:

  • Published:

  • Issue Date:

  • DOI: https://doi.org/10.1038/ncb3396

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