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:

PTENα and PTENβ promote carcinogenesis through WDR5 and H3K4 trimethylation

An Author Correction to this article was published on 15 November 2019

This article has been updated

Abstract

PTENα and PTENβ are two longer translational variants of phosphatase and tensin homolog (PTEN) messenger RNA. Their expressional regulations and functions in carcinogenesis remain largely unknown. Here, we demonstrate that, in contrast with the well-established tumour-suppressive role of canonical PTEN, PTENα and PTENβ promote tumourigenesis by directly interacting with the histone H3 lysine 4 (H3K4) presenter WDR5 to promote H3K4 trimethylation and maintain a tumour-promoting signature. We also show that USP9X and FBXW11 bind to the amino-terminal extensions of PTENα/β, and respectively deubiquitinate and ubiquitinate lysines 235 and 239 in PTENα to regulate PTENα/β stability. In accordance, USP9X promotes tumourigenesis and FBXW11 suppresses tumourigenesis through PTENα/β. Taken together, our results indicate that the Pten gene is a double-edged sword for carcinogenesis, and reinterpretation of the importance of the Pten gene in carcinogenesis is warranted.

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: PTENα and PTENβ present expression inconsistent with that of PTEN protein in liver cancer and are regulated by USP9X.
Fig. 2: USP9X binds NTE to stabilize PTENα/β.
Fig. 3: FBXW11 degrades PTENα/β by inducing ubiquitination.
Fig. 4: PTENα/β promotes tumourigenesis.
Fig. 5: USP9X promotes tumourigenesis by stabilizing PTENα/β, while FBXW11 suppresses it by and degrading PTENα/β.
Fig. 6: PTENα/β maintains a tumour-promoting signature.
Fig. 7: PTENα/β and WDR5 interact with each other.
Fig. 8: PTENα and PTENβ promote tumourigenesis through the WDR5–H3K4me3 axis.

Similar content being viewed by others

Data availability

ChIP-Seq data that support the findings of this study have been deposited in the GEO under accession codes GSE135478 and GSE126982. RNA-Seq data that support the findings of this study have been deposited in the GEO under accession codes GSE135477 and GSE126983. Previously published RNA-Seq data that were reanalysed here are available under accession code GSE14520. Mass spectrometry data (Fig. 1e and Supplementary Table 3) have been deposited in ProteomeXchange with primary accession code PXD013196. The human LIHC, pancreatic cancer and breast cancer data were derived from the TCGA Research Network (http://cancergenome.nih.gov/). Source data are available online for Figs. 1–8 and Extended Data Figs. 1–8. All other data supporting the findings of this study are available from the corresponding author on reasonable request.

Change history

  • 15 November 2019

    An amendment to this paper has been published and can be accessed via a link at the top of the paper.

References

  1. Maehama, T. & Dixon, J. E. The tumor suppressor, PTEN/MMAC1, dephosphorylates the lipid second messenger, phosphatidylinositol 3,4,5-trisphosphate. J. Biol. Chem. 273, 13375–13378 (1998).

    Article  CAS  PubMed  Google Scholar 

  2. Lee, Y. R., Chen, M. & Pandolfi, P. P. The functions and regulation of the PTEN tumour suppressor: new modes and prospects. Nat. Rev. Mol. Cell Biol. 19, 547–562 (2018).

    Article  CAS  PubMed  Google Scholar 

  3. Naderali, E. et al. Regulation and modulation of PTEN activity. Mol. Biol. Rep. 45, 2869–2881 (2018).

    Article  CAS  PubMed  Google Scholar 

  4. Song, M. S. et al. Nuclear PTEN regulates the APC-CDH1 tumor-suppressive complex in a phosphatase-independent manner. Cell 144, 187–199 (2011).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  5. Shen, S. M. et al. AIF inhibits tumor metastasis by protecting PTEN from oxidation. EMBO Rep. 16, 1563–1580 (2015).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  6. Shen, S. M. et al. Nuclear PTEN safeguards pre-mRNA splicing to link Golgi apparatus for its tumor suppressive role. Nat. Commun. 9, 2392 (2018).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  7. Hopkins, B. D. et al. A secreted PTEN phosphatase that enters cells to alter signaling and survival. Science 341, 399–402 (2013).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  8. Liang, H. et al. PTENβ is an alternatively translated isoform of PTEN that regulates rDNA transcription. Nat. Commun. 8, 14771 (2017).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  9. Malaney, P., Uversky, V. N. & Dave, V. The PTEN long N-tail is intrinsically disordered: increased viability for PTEN therapy. Mol. Biosyst. 9, 2877–2888 (2013).

    Article  CAS  PubMed  Google Scholar 

  10. Chalhoub, N. & Baker, S. J. PTEN and the PI3-kinase pathway in cancer. Annu. Rev. Pathol. 4, 127–150 (2009).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  11. Zhang, B. et al. 42,573 cases of hepatectomy in China: a multicenter retrospective investigation. Sci. China Life Sci. 61, 660–670 (2018).

    Article  CAS  PubMed  Google Scholar 

  12. Liang, H. et al. PTENα, aPTEN isoform translated through alternative initiation, regulates mitochondrial function and energy metabolism. Cell Metab. 19, 836–848 (2014).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  13. Cai, J. et al. CK1α suppresses lung tumour growth by stabilizing PTEN and inducing autophagy. Nat. Cell Biol. 20, 465–478 (2018).

    Article  CAS  PubMed  Google Scholar 

  14. Theard, D. et al. USP9x-mediated deubiquitination of EFA6 regulates de novo tight junction assembly. EMBO J. 29, 1499–1509 (2010).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  15. Trotman, L. C. et al. Ubiquitination regulates PTEN nuclear import and tumor suppression. Cell 128, 141–156 (2007).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  16. Li, X. et al. USP9X regulates centrosome duplication and promotes breast carcinogenesis. Nat. Commun. 8, 14866 (2017).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  17. Schwickart, M. et al. Deubiquitinase USP9X stabilizes MCL1 and promotes tumour cell survival. Nature 463, 103–107 (2010).

    Article  CAS  PubMed  Google Scholar 

  18. Chen, Z. et al. USP9X deubiquitinates ALDH1A3 and maintains mesenchymal identity in glioblastoma stem cells. J. Clin. Invest. 130, 2043–2055 (2019).

    Article  Google Scholar 

  19. Li, L. et al. The deubiquitinase USP9X promotes tumor cell survival and confers chemoresistance through YAP1 stabilization. Oncogene 37, 2422–2431 (2018).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  20. Li, G. et al. PTENα regulates mitophagy and maintains mitochondrial quality control. Autophagy 14, 1742–1760 (2018).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  21. Song, M. S. et al. The deubiquitinylation and localization of PTEN are regulated by a HAUSP-PML network. Nature 455, 813–817 (2008).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  22. Zhou, L. et al. The significance of Notch1 compared with Notch3 in high metastasis and poor overall survival in hepatocellular carcinoma. PLoS ONE 8, e57382 (2013).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  23. Hu, L., Xue, F., Shao, M., Deng, A. & Wei, G. Aberrant expression of Notch3 predicts poor survival for hepatocellular carcinomas. Biosci. Trends 7, 152–156 (2013).

    PubMed  Google Scholar 

  24. Giovannini, C., Bolondi, L. & Gramantieri, L. Targeting Notch3 in hepatocellular carcinoma: molecular mechanisms and therapeutic perspectives. Int. J. Mol. Sci. 18, e56 (2016).

    Article  CAS  PubMed  Google Scholar 

  25. Zhou, J. X. et al. Association between high levels of Notch3 expression and high invasion and poor overall survival rates in pancreatic ductal adenocarcinoma. Oncol. Rep. 36, 2893–2901 (2016).

    Article  CAS  PubMed  Google Scholar 

  26. Cai, H. et al. LncRNA HOTAIR acts as competing endogenous RNA to control the expression of Notch3 via sponging miR-613 in pancreatic cancer. Oncotarget 8, 32905–32917 (2017).

    PubMed  PubMed Central  Google Scholar 

  27. Aburjania, Z. et al. The role of Notch3 in cancer. Oncologist 23, 900–911 (2018).

    Article  PubMed  PubMed Central  Google Scholar 

  28. Ali, S. A., Justilien, V., Jamieson, L., Murray, N. R. & Fields, A. P. Protein kinase Cι drives a NOTCH3-dependent stem-like phenotype in mutant KRAS lung adenocarcinoma. Cancer Cell 29, 367–378 (2016).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  29. Jeong, J. Y. et al. MicroRNA-136 inhibits cancer stem cell activity and enhances the anti-tumor effect of paclitaxel against chemoresistant ovarian cancer cells by targeting Notch3. Cancer Lett. 386, 168–178 (2017).

    Article  CAS  PubMed  Google Scholar 

  30. Zheng, Y. et al. A rare population of CD24+ITGB4+Notchhi cells drives tumor propagation in NSCLC and requires Notch3 for self-renewal. Cancer Cell 24, 59–74 (2013).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  31. Pastrana, E., Silva-Vargas, V. & Doetsch, F. Eyes wide open: a critical review of sphere-formation as an assay for stem cells. Cell Stem Cell 8, 486–498 (2011).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  32. Zampieri, M. et al. The epigenetic factor BORIS/CTCFL regulates the NOTCH3 gene expression in cancer cells. Biochim. Biophys. Acta 1839, 813–825 (2014).

    Article  CAS  PubMed  Google Scholar 

  33. Yang, G. et al. Integrated analysis of gene expression and methylation profiles of novel pancreatic cancer cell lines with highly metastatic activity. Sci. China Life Sci. 62, 791–806 (2019).

    Article  PubMed  Google Scholar 

  34. Schuettengruber, B., Martinez, A. M., Iovino, N. & Cavalli, G. Trithorax group proteins: switching genes on and keeping them active. Nat. Rev. Mol. Cell Biol. 12, 799–814 (2011).

    Article  CAS  PubMed  Google Scholar 

  35. Dou, Y. et al. Regulation of MLL1 H3K4 methyltransferase activity by its core components. Nat. Struct. Mol. Biol. 13, 713–719 (2006).

    Article  CAS  PubMed  Google Scholar 

  36. Jiang, H. et al. Role for Dpy-30 in ES cell-fate specification by regulation of H3K4 methylation within bivalent domains. Cell 144, 513–525 (2011).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  37. Patel, A., Dharmarajan, V., Vought, V. E. & Cosgrove, M. S. On the mechanism of multiple lysine methylation by the human mixed lineage leukemia protein-1 (MLL1) core complex. J. Biol. Chem. 284, 24242–24256 (2009).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  38. Ernst, P. & Vakoc, C. R. WRAD: enabler of the SET1-family of H3K4 methyltransferases. Brief Funct. Genomics 11, 217–226 (2012).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  39. Xue, H. et al. Structural basis of nucleosome recognition and modification by MLL methyltransferases. Nature 573, 445–449 (2019).

    Article  CAS  PubMed  Google Scholar 

  40. Thomas, L. R. et al. Interaction with WDR5 promotes target gene recognition and tumorigenesis by MYC. Mol. Cell 58, 440–452 (2015).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  41. Wang, L. et al. PTEN-L is a novel protein phosphatase for ubiquitin dephosphorylation to inhibit PINK1-Parkin-mediated mitophagy. Cell Res. 28, 787–802 (2018).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  42. Murtaza, M., Jolly, L. A., Gecz, J. & Wood, S. A. La FAM fatale: USP9X in development and disease. Cell Mol. Life Sci. 72, 2075–2089 (2015).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  43. Nielsen, C. P., Jernigan, K. K., Diggins, N. L., Webb, D. J. & MacGurn, J. A. USP9X deubiquitylates DVL2 to regulate WNT pathway specification. Cell Rep. 28, 1074–1089.e5 (2019).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  44. Shang, Z. et al. USP9X-mediated deubiquitination of B-cell CLL/lymphoma 9 potentiates Wnt signaling and promotes breast carcinogenesis. J. Biol. Chem. 294, 9844–9857 (2019).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  45. Chang, H. et al. MiR-182 promotes cell proliferation by suppressing FBXW7 and FBXW11 in non-small cell lung cancer. Am. J. Transl. Res. 10, 1131–1142 (2018).

    CAS  PubMed  PubMed Central  Google Scholar 

  46. Li, J. et al. Cbx4 governs HIF-1α to potentiate angiogenesis of hepatocellular carcinoma by its SUMO E3 ligase activity. Cancer Cell 25, 118–131 (2014).

    Article  CAS  PubMed  Google Scholar 

  47. Fedchenko, N. & Reifenrath, J. Different approaches for interpretation and reporting of immunohistochemistry analysis results in the bone tissue—a review. Diagn. Pathol. 9, 221 (2014).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  48. Alleva, E. & Santucci, D. Guide for the care and use of laboratory animals. Ethology 103, 1072–1073 (1997).

    Google Scholar 

  49. U.S. Office of Science and Technology Policy. Laboratory animal welfare; U.S. government principles for the utilization and care of vertebrate animals used in testing, research and training; notice. Fed. Regist. 50, 20864-20865 (1985).

  50. Cao, L. et al. Sphere-forming cell subpopulations with cancer stem cell properties in human hepatoma cell lines. BMC Gastroenterol. 11, 71 (2011).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  51. Konermann, S. et al. Genome-scale transcriptional activation by an engineered CRISPR–Cas9 complex. Nature 517, 583–588 (2015).

    Article  CAS  PubMed  Google Scholar 

Download references

Acknowledgements

We thank Y. Yin at Peking University for providing the materials described in the Methods. This work was supported by the National Key Research Program of China (NO2015CB910403) and the National Natural Science Foundation (81830091, 91853206, 81430061 and 81972583) and its innovative group support (number 81721004), as well as the Fundamental Research Funds for the Central Universities.

Author information

Authors and Affiliations

Authors

Contributions

S.-M.S., C.Z., M.-K.G. and S.-S.D. performed most of the experiments. L.X. performed the LC−MS/MS analyses. P.H., N.Z. and S.Y. conducted partial experiments. Y.J. performed the bioinformatics analysis. Q.X. collected patient samples. Y.Y., J.-K.Z. and J.-X.Y. provided constructive comments and discussion. G.-Q.C. and S.-M.S. designed and supervised the entire project and prepared the manuscript.

Corresponding authors

Correspondence to Shao-Ming Shen or Guo-Qiang Chen.

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

Extended Data Fig. 1 Inconsistent expressional patterns of PTEN and PTENα/β in liver cancer and potential regulators.

(A) Genomic alterations of PTEN gene in TCGA cohorts analyzed by The cBioPortal for Cancer Genomics (http://www.cbioportal.org/). Arrowhead points to the liver cancer cohort. (B) Representative images of the IHC staining of PTENα/β (top) and PTEN (bottom) in the sections of SMMC-7721xenografts with or without depletion of PTENα/β (PTENα/βKO-1) and PTEN (PTEN3KO) respectively (n=1 experiment). Scale bar, 100 μm. (C) Numbers of samples in 9 indicated subgroups divided according to PTEN and PTENα/β expressions in tumor tissues compared to paired adjacent tissues. (D) Representative images of the IHC staining of indicated proteins in the serial sections of tumor tissue (T) and paired adjacent normal tissue (N) isolated from one liver cancer patient (n=1 experiment). Scale bar, 100 μm. (E) Western blot analysis and quantification of indicated proteins in 293T, Hela and MEF treated with 50 μg/ml CHX for different time points. (F) Western blot analysis of indicated proteins in 293T cells transduced by shRNAs as indicated. (G) qRT-PCR analysis of PTEN mRNA in 293T cells with gNS or gUSP9X infection. (H) Analysis of the correlation between USP9X and PTENα/β in human liver cancer samples with decreased PTEN in tumor tissues compared to paired adjacent normal tissues. Samples were divided into 6 subgroups according to Figure 1J. Data were analyzed by Pearson’s χ2 test and the P value is shown. The experiments in E, F were repeated three times independently with the similar results, and one representative result was shown. For G, data represent means with bar as S.E.M. of three independent experiments; two-tailed unpaired t-test. NC, negative control.

Source data

Source data

Extended Data Fig. 2 USP9X deubiquitinates and stabilizes PTENα/β.

(A) Western blot analysis with anti-Flag or anti-HA antibody in the input and immunoprecipitates of 293T cells co-transfected with HA-tagged PTENβ and Flag-tagged USP9X or its four fragments as shown in the top schematics. (B) Schematics of N-terminal S-tag fused PTEN isoforms and derivatives. (C, D) 293T and Hela cells with or without USP9X knockdown by shRNA were treated with 50 μg/ml CHX for different time points (C) or 10 μM MG132 for 6 h (D), followed by Western blot analysis of indicated proteins. (E, F) In vivo ubiquitination assay in 293T cells transfected with the indicated plasmids was followed by Western blot analysis with antibody against total ubiquitin (E) and antibodies against different types of ubiquitin chain (F). All experiments were repeated at least three times independently with the similar results and one representative result was shown respectively. NC, negative control; EV, empty vector.

Source data

Extended Data Fig. 3 FBXW11 ubiquitinates and destabilizes PTENα/β.

(A) 293T cells were transduced by lentiviruses encoding shRNAs targeting indicated genes, followed by Western blot for PTEN and PTENα/β with β-actin as loading control. Protein bands were quantified and the ratio of PTEN or PTENα/β versus β-actin was respectively calculated and normalized with shNC cells. The shadowy area presents ratio range of 0.67 to 1.5. (B) The genomic locations of gRNAs targeting FBXW11 (top). The Sanger sequencing analysis of genomic DNA from 293T cells transduced by lentivirus encoding gRNAs targeting FBXW11, along with gNS (bottom). Purple arrowheads indicate the start sites of perturbed genomic sequence. (C) qRT-PCR analysis of PTEN mRNA in SMMC-7721 and 293T cells with or without FBXW11 depletion by CRISPR-CAS9. (D) The abundances of PTENα/β and PTEN relative to loading control according to the experiments in Figure 3D were respectively quantified (n=3 independent biological samples). (E-G) In vivo ubiquitination assay of Flag-tagged PTEN, PTENα and PTENβ in 293T cells without (E) or with (G) co-transfected of Myc-TR-TUBEs, followed by Western blot analysis with antibody against total ubiquitin (E, G) and antibodies against different types of ubiquitin chain (F). (H) 293T and SMMC-7721 cells with or without FBXW11 over-expression were treated with 10 μM MG132 for 6 h, followed by Western blot analysis of indicated proteins. Experiments in E-H were repeated three times independently with similar results and one representative result was shown. For C, D, data represent means with bar as S.E.M. of three independent experiments; two-tailed unpaired t-test (C) and two-way ANOVA (D). NC, negative control; NS, non-specific control; EV, empty vector.

Source data

Source data

Extended Data Fig. 4 Clinical significance of PTEN and PTENα/β, and their specific depletion by CRISPR-CAS9.

(A-F) Patients with liver cancer were divided into two groups respectively with equal/increased (n=49 samples) and decreased PTEN (n=39 samples) (A-C) or equal/increased (n= 63 samples) and decreased PTENα/β (n=25 samples) (D-F) in tumor tissues compared to paired adjacent normal tissues. Scatterplot analysis of tumor size (A, D), percentage of tumor stage (B, E), and Kaplan-Meier plot of overall survival (C, F) between the two groups were shown. Data represent means with bar as S.E.M. (A, D), and were analyzed using Mann–Whitney U test (two-sided) (A, D), Pearson’s χ2 test (B, E), and log-rank test (two-sided) (C, F). (G) Patients with liver cancer were divided into two groups respectively with equal/increased and decreased PTEN in tumor tissues compared to paired adjacent normal tissues, with the latter being further divided into two subgroups respectively with equal/increased and decreased PTENα/β. Kaplan-Meier plot of overall survival between the three groups were shown. Data were analyzed using log-rank test (two-sided), and P value is shown. (H) A schematic illustrating the two gRNAs respectively designed to target all three isoforms of PTEN (gPTEN3) and only the long isoforms of PTEN (gPTENα/β). (I, J) Western blot analysis of indicated proteins in 293T (I) and MCF-7 (J) cells transduced by lentiviruses encoding CAS9 and gPTEN3 or gPTENα/β. Experiments in I, J were repeated three times independently with the similar results, and one representative image was shown. NS, non-specific control.

Source data

Source data

Extended Data Fig. 5 PTENα/β is crucial for the tumor-promoting role of USP9X.

(A) Comparison of USP9X expression between tumor tissues and matched non-tumor tissues in LIHC and BRCA cohorts from TCGA. Paired two-tailed Student’s t-test. (B) Kaplan-Meier plot of overall survival of patients with high or low expression of USP9X in the tumor tissues of BRCA cohort from TCGA. Log-rank test (two-sided). (C-E) A DOX-inducible expression system encoding wild-type or mutant PTENα/β was introduced into SMMC-7721 USP9XKO cells, followed by Western blot analysis showing induction of indicated proteins by DOX administration (C). These cells were subcutaneously injected into nude mice (1.5×106 cells per mouse, n=5 mice for each group), followed by administration with or without 0.25 mg/kg DOX in feed immediately after injection. Tumors were measured at different days (D), and harvested and weighed on day 23 (E). (F-K) Western blot analysis of indicated proteins in two PTENα/β-knockout SMMC-7721 clones, PTENα/βKO-1 (F-H) and PTENα/βKO-2 (I-K), with gNS or gUSP9X infection (F, I). These cells were subcutaneously injected into nude mice (3×106 cells per mouse, n=8 or 10 mice). Tumors were measured at different days (G, J), and harvested and weighed on day 17 (H, K). (L, M) Western blot analysis of indicated proteins in 293T (L) and MiaPaCa2 cells (M) with or without USP9X depletion. Experiments in C, F, I, L, M and all animal experiments were respectively repeated three times and at least twice independently, with similar results, and result from one experiment was shown. For D, E, G, H, J, K data represent means with bar as S.E.M.; two-way ANOVA for D, G, J; two-tailed unpaired t-test for E, H, K. NC, negative control; NS, non-specific control.

Source data

Source data

Extended Data Fig. 6 Identification of PTENα/β-regulated genes and investigation of the role of NOTCH3 in tumorigenesis.

(A) Schematics of PTENα and PTENβ derivatives with a C-terminal GFP-tag. (B) The workflow for identifying PTENα/β-regulated (activated or inhibited) genes in SMMC-7721 cells. G1-4, group 1-4. (C, D) Cell type-independent PTENα/β-activated genes were identified by cross matching PTENα/β-activated genes identified in (B) with genes down-regulated by PTENα/β depletion in MiaPaCa2 cells. Workflow (C) and Venn diagram reporting the number of genes (D) are shown. (E-G) Western blot analysis of indicated proteins in SMMC-7721 cells with gNS or gNOTCH3 infection (E). These cells were subcutaneously injected into nude mice (3×106 cells per mouse, n=8 mice for each group). Tumors were measured at different days (F), and harvested and weighed on day 17 (G). RNA-seq experiment was performed on two independent samples for each cell line in SMMC-7721 and one sample for each cell line in MiaPaCa2. The source data was provided as GSE126983. E-G were repeated twice with the similar results, and result from one experiment was shown. For F, G, data represent means with bar as S.E.M.; two-way ANOVA for F; two-tailed unpaired t-test for G. NS, non-specific control.

Source data

Source data

Extended Data Fig. 7 PTENα/β and WDR5 interact with each other and regulate a similar subset of genes.

(A, B) Western blot analysis of indicated proteins (A) and qRT-PCR analysis of NOTCH paralogs (B) in MiaPaCa2 cells with gNS, gWDR5 or gRBBP5 infection. (C) qRT-PCR analysis of indicated genes in MiaPaCa2 cells gNS, gWDR5 or gRBBP5 infection. (D) Bacterially expressed PTEN and PTENα derivatives with an N-terminal S-tag were incubated with GST-tagged WDR5, followed by S-tag pulldown and Western blot analysis of indicated proteins. (E) Western blot analysis of indicated proteins in the immunoprecipitates of GFP-tagged proteins transfected in 293T cells. For B, C, data represent means with bar as S.E.M. of three independent experiments; two-tailed unpaired t-test. All other experiments were repeated three times independently with similar results, and result from one experiment was shown. NS, non-specific control.

Source data

Source data

Extended Data Fig. 8 PTENα/β promotes tumorigenesis through the WDR5-H3K4me3 axis.

(A) Example tracks of WDR5 and PTENα/β co-occupancy at the NOTCH3 (top) and SLC12A5 (bottom) loci in SMMC-7721 cells. (B) ChIP-qRT-PCR analysis of PTENα/β occupation on the promoters of indicated genes in SMMC-7721 cells with or without depletion of PTENα/β by CRISPR-CAS9, with IgG used as negative control (n=3 biologically independent experiments). (C) Immunofluorescent staining of H3K4me3 together with re-staining of DAPI in SMMC-7721 cells with gNS and gPTENα/β infection. Scale bars, 50 μm. (D) Bacterially expressed N-terminal-S-tagged PTEN and PTENα derivatives were incubated with GST-tagged WDR5, followed by S-tag pulldown and Western blot analysis. (E-G) Western blot analysis of indicated proteins in SMMC-7721 PTEN3KO cells transduced by PTENα or PTENαΔ116-148 (E). These cells were subcutaneously injected into nude mice (1.5×106 cells per mouse, n=5 mice for each group). Tumors were measured at different days (F), and harvested and weighed on day 21 (G). (H-J) Two WDR5-depleted SMMC-7721 clones (WDR5KO-1 and WDR5KO-2) along with parental cells were transduced by PTENα or EV, followed by Western blot analysis of indicated proteins (H). These cells were subcutaneously injected into nude mice (1.5×106 cells per mouse, n=7 mice for each group). Tumors were measured at different days (I), and harvested and weighed on day 17 (J). All animal experiments were repeated at least twice, and other experiments except for A were repeated three times independently, with the similar results, and result from one experiment was shown. For B, F, G, I, J data represent means with bar as S.E.M.; two-tailed unpaired t-test for B, G, J; two-way ANOVA for F, I. NS, non-specific control; EV, empty vector.

Source data

Source data

Supplementary information

Reporting Summary

Supplementary Table 1

Information on samples from patients with liver cancer (cohort 1). Supplementary Table 2 Information on microarrays of samples from patients with liver cancer (cohort 2). Supplementary Table 3 N173 and PTENα interacting proteins, including three sections, respectively, with N173, PTENα and both interacting proteins. Supplementary Table 4 PTENα/β-regulated genes. Supplementary Table 5 Plasmids used in this study. Supplementary Table 6 Target sequences of the shRNAs, gRNAs for CRISPR–Cas9 and gRNAs for CRISPR–dCas9 used in this study. Supplementary Table 7 Antibodies and reagents used in this study. Supplementary Table 8 Primers used in this study.

Source data

Source Data Fig. 1

Statistical Source Data

Source Data Fig. 1

Unprocessed Western Blots and/or gels

Source Data Fig. 2

Unprocessed Western Blots and/or gels

Source Data Fig. 3

Statistical Source Data

Source Data Fig. 3

Unprocessed Western Blots and/or gels

Source Data Fig. 4

Statistical Source Data

Source Data Fig. 4

Unprocessed Western Blots and/or gels

Source Data Fig. 5

Statistical Source Data

Source Data Fig. 5

Unprocessed Western Blots and/or gels

Source Data Fig. 6

Statistical Source Data

Source Data Fig. 6

Unprocessed Western Blots and/or gels

Source Data Fig. 7

Statistical Source Data

Source Data Fig. 7

Unprocessed Western Blots and/or gels

Source Data Fig. 8

Statistical Source Data

Source Data Fig. 8

Unprocessed Western Blots and/or gels

Source Data Extended Data Fig. 1

Statistical Source Data

Source Data Extended Data Fig. 1

Unprocessed Western Blots and/or gels

Source Data Extended Data Fig. 2

Unprocessed Western Blots and/or gels

Source Data Extended Data Fig. 3

Statistical Source Data

Source Data Extended Data Fig. 3

Unprocessed Western Blots and/or gels

Source Data Extended Data Fig. 4

Statistical Source Data

Source Data Extended Data Fig. 4

Unprocessed Western Blots and/or gels

Source Data Extended Data Fig. 5

Statistical Source Data

Source Data Extended Data Fig. 5

Unprocessed Western Blots and/or gels

Source Data Extended Data Fig. 6

Statistical Source Data

Source Data Extended Data Fig. 6

Unprocessed Western Blots and/or gels

Source Data Extended Data Fig. 7

Statistical Source Data

Source Data Extended Data Fig. 7

Unprocessed Western Blots and/or gels

Source Data Extended Data Fig. 8

Statistical Source Data

Source Data Extended Data Fig. 8

Unprocessed Western Blots and/or gels

Rights and permissions

Reprints and permissions

About this article

Check for updates. Verify currency and authenticity via CrossMark

Cite this article

Shen, SM., Zhang, C., Ge, MK. et al. PTENα and PTENβ promote carcinogenesis through WDR5 and H3K4 trimethylation. Nat Cell Biol 21, 1436–1448 (2019). https://doi.org/10.1038/s41556-019-0409-z

Download citation

  • Received:

  • Accepted:

  • Published:

  • Issue Date:

  • DOI: https://doi.org/10.1038/s41556-019-0409-z

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