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:

Deubiquitylase OTUD3 regulates PTEN stability and suppresses tumorigenesis

Abstract

PTEN is one of the most frequently mutated tumour suppressors and reduction in PTEN protein stability also plays a role in tumorigenesis. Although several ubiquitin ligases for PTEN have been identified, the deubiquitylase for de-polyubiquitylation and stabilization of PTEN is less defined. Here, we report OTUD3 as a deubiquitylase of PTEN. OTUD3 interacts with, de-polyubiquitylates and stabilizes PTEN. Depletion of OTUD3 leads to the activation of Akt signalling, induction of cellular transformation and cancer metastasis. OTUD3 transgenic mice exhibit higher levels of the PTEN protein and are less prone to tumorigenesis. Reduction of OTUD3 expression, concomitant with decreased PTEN abundance, correlates with human breast cancer progression. Furthermore, we identified loss-of-function OTUD3 mutations in human cancers, which either abolish OTUD3 catalytic activity or attenuate the interaction with PTEN. These findings demonstrate that OTUD3 is an essential regulator of PTEN and that the OTUD3–PTEN signalling axis plays a critical role in tumour suppression.

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: The deubiquitylase OTUD3 stabilizes PTEN.
Figure 2: OTUD3 de-polyubiquitylates PTEN.
Figure 3: OTUD3 regulates cytoplasmic PTEN and inhibits Akt signalling.
Figure 4: OTUD3 suppresses tumorigenesis.
Figure 5: OTUD3 transgenic mice exhibit higher PTEN expression and are less prone to tumorigenesis.
Figure 6: OTUD3 E86K mutant loses DUB function and gains oncogene function.
Figure 7: A subset of OTUD3 mutations in human cancer attenuate the ability to interact with and stabilize PTEN.
Figure 8: Positive correlation between OTUD3 and PTEN in human breast cancer.

Similar content being viewed by others

References

  1. Li, J. et al. PTEN, a putative protein tyrosine phosphatase gene mutated in human brain, breast, and prostate cancer. Science 275, 1943–1947 (1997).

    Article  CAS  PubMed  Google Scholar 

  2. Steck, P. A. et al. Identification of a candidate tumour suppressor gene, MMAC1, at chromosome 10q23.3 that is mutated in multiple advanced cancers. Nat. Genet. 15, 356–362 (1997).

    Article  CAS  PubMed  Google Scholar 

  3. Liaw, D. et al. Germline mutations of the PTEN gene in Cowden disease, an inherited breast and thyroid cancer syndrome. Nat. Genet. 16, 64–67 (1997).

    Article  CAS  PubMed  Google Scholar 

  4. Marsh, D. J. et al. Germline mutations in PTEN are present in Bannayan-Zonana syndrome. Nat. Genet. 16, 333–334 (1997).

    Article  CAS  PubMed  Google Scholar 

  5. Di Cristofano, A., Pesce, B., Cordon-Cardo, C. & Pandolfi, P. P. Pten is essential for embryonic development and tumour suppression. Nat. Genet. 19, 348–355 (1998).

    Article  CAS  PubMed  Google Scholar 

  6. Podsypanina, K. et al. Mutation of Pten/Mmac1 in mice causes neoplasia in multiple organ systems. Proc. Natl Acad. Sci. USA 96, 1563–1568 (1999).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  7. Suzuki, A. et al. High cancer susceptibility and embryonic lethality associated with mutation of the PTEN tumor suppressor gene in mice. Curr. Biol. 8, 1169–1178 (1998).

    Article  CAS  PubMed  Google Scholar 

  8. 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 

  9. Song, M. S., Salmena, L. & Pandolfi, P. P. The functions and regulation of the PTEN tumour suppressor. Nat. Rev. Mol. Cell Biol. 13, 283–296 (2012).

    Article  CAS  PubMed  Google Scholar 

  10. Shen, W. H. et al. Essential role for nuclear PTEN in maintaining chromosomal integrity. Cell 128, 157–170 (2007).

    Article  CAS  PubMed  Google Scholar 

  11. 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 

  12. Wang, X. et al. NEDD4-1 is a proto-oncogenic ubiquitin ligase for PTEN. Cell 128, 129–139 (2007).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  13. Maddika, S. et al. WWP2 is an E3 ubiquitin ligase for PTEN. Nat. Cell Biol. 13, 728–733 (2011).

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  14. Ahmed, S. F. et al. The chaperone-assisted E3 ligase C terminus of Hsc70-interacting protein (CHIP) targets PTEN for proteasomal degradation. J. Biol. Chem. 287, 15996–16006 (2012).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  15. Van Themsche, C., Leblanc, V., Parent, S. & Asselin, E. X-linked inhibitor of apoptosis protein (XIAP) regulates PTEN ubiquitination, content, and compartmentalization. J. Biol. Chem. 284, 20462–20466 (2009).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  16. Lee, J. T. et al. RFP-mediated ubiquitination of PTEN modulates its effect on AKT activation. Cell Res. 23, 552–564 (2013).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  17. Alimonti, A. et al. Subtle variations in Pten dose determine cancer susceptibility. Nat. Genet. 42, 454–458 (2010).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  18. Garcia-Cao, I. et al. Systemic elevation of PTEN induces a tumor-suppressive metabolic state. Cell 149, 49–62 (2012).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  19. Carracedo, A., Alimonti, A. & Pandolfi, P. P. PTEN level in tumor suppression: how much is too little? Cancer Res. 71, 629–633 (2011).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  20. Kruse, J. P. & Gu, W. Modes of p53 regulation. Cell 137, 609–622 (2009).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  21. Komander, D., Clague, M. J. & Urbe, S. Breaking the chains: structure and function of the deubiquitinases. Nat. Rev. Mol. Cell Biol. 10, 550–563 (2009).

    Article  CAS  PubMed  Google Scholar 

  22. 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 

  23. Sacco, J. J. et al. The deubiquitylase Ataxin-3 restricts PTEN transcription in lung cancer cells. Oncogene 33, 4265–4272 (2014).

    Article  CAS  PubMed  Google Scholar 

  24. Zhang, J. et al. Deubiquitylation and stabilization of PTEN by USP13. Nat. Cell Biol. 15, 1486–1494 (2013).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  25. Guy, C. T., Cardiff, R. D. & Muller, W. J. Induction of mammary tumors by expression of polyomavirus middle T oncogene: a transgenic mouse model for metastatic disease. Mol. Cell. Biol. 12, 954–961 (1992).

    CAS  PubMed  PubMed Central  Google Scholar 

  26. Maglione, J. E. et al. Transgenic Polyoma middle-T mice model premalignant mammary disease. Cancer Res. 61, 8298–8305 (2001).

    CAS  PubMed  Google Scholar 

  27. 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  PubMed  Google Scholar 

  28. Gao, J. et al. Integrative analysis of complex cancer genomics and clinical profiles using the cBioPortal. Sci. Signal. 6, pl1 (2013).

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  29. Perren, A. et al. Immunohistochemical evidence of loss of PTEN expression in primary ductal adenocarcinomas of the breast. Am. J. Pathol. 155, 1253–1260 (1999).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  30. Bose, S. et al. Reduced expression of PTEN correlates with breast cancer progression. Hum. Pathol. 33, 405–409 (2002).

    Article  CAS  PubMed  Google Scholar 

  31. FitzGerald, M. G. et al. Germline mutations in PTEN are an infrequent cause of genetic predisposition to breast cancer. Oncogene 17, 727–731 (1998).

    Article  CAS  PubMed  Google Scholar 

  32. Feilotter, H. E. et al. Analysis of the 10q23 chromosomal region and the PTEN gene in human sporadic breast carcinoma. Br. J. Cancer 79, 718–723 (1999).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  33. Rhei, E. et al. Mutation analysis of the putative tumor suppressor gene PTEN/MMAC1 in primary breast carcinomas. Cancer Res. 57, 3657–3659 (1997).

    CAS  PubMed  Google Scholar 

  34. Ali, I. U., Schriml, L. M. & Dean, M. Mutational spectra of PTEN/MMAC1 gene: a tumor suppressor with lipid phosphatase activity. J. Natl Cancer Inst. 91, 1922–1932 (1999).

    Article  CAS  PubMed  Google Scholar 

  35. Everett, R. D. et al. A novel ubiquitin-specific protease is dynamically associated with the PML nuclear domain and binds to a herpesvirus regulatory protein. EMBO J. 16, 1519–1530 (1997).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  36. Kayagaki, N. et al. DUBA: a deubiquitinase that regulates type I interferon production. Science 318, 1628–1632 (2007).

    Article  CAS  PubMed  Google Scholar 

  37. Hymowitz, S. G. & Wertz, I. E. A20: from ubiquitin editing to tumour suppression. Nat. Rev. Cancer 10, 332–341 (2010).

    Article  CAS  PubMed  Google Scholar 

  38. Nakada, S. et al. Non-canonical inhibition of DNA damage-dependent ubiquitination by OTUB1. Nature 466, 941–946 (2010).

    Article  CAS  PubMed  Google Scholar 

  39. Hu, H. et al. OTUD7B controls non-canonical NF-κB activation through deubiquitination of TRAF3. Nature 494, 371–374 (2013).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  40. Keusekotten, K. et al. OTULIN antagonizes LUBAC signaling by specifically hydrolyzing Met1-linked polyubiquitin. Cell 153, 1312–1326 (2013).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  41. Fiil, B. K. et al. OTULIN restricts Met1-linked ubiquitination to control innate immune signaling. Mol. Cell 50, 818–830 (2013).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  42. Rivkin, E. et al. The linear ubiquitin-specific deubiquitinase gumby regulates angiogenesis. Nature 498, 318–324 (2013).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  43. Mevissen, T. E. et al. OTU deubiquitinases reveal mechanisms of linkage specificity and enable ubiquitin chain restriction analysis. Cell 154, 169–184 (2013).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  44. Georgescu, M. M. et al. Stabilization and productive positioning roles of the C2 domain of PTEN tumor suppressor. Cancer Res. 60, 7033–7038 (2000).

    CAS  PubMed  Google Scholar 

  45. Torres, J. & Pulido, R. The tumor suppressor PTEN is phosphorylated by the protein kinase CK2 at its C terminus. Implications for PTEN stability to proteasome-mediated degradation. J. Biol. Chem. 276, 993–998 (2001).

    Article  CAS  PubMed  Google Scholar 

  46. Vazquez, F., Ramaswamy, S., Nakamura, N. & Sellers, W. R. Phosphorylation of the PTEN tail regulates protein stability and function. Mol. Cell. Biol. 20, 5010–5018 (2000).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  47. Raftopoulou, M., Etienne-Manneville, S., Self, A., Nicholls, S. & Hall, A. Regulation of cell migration by the C2 domain of the tumor suppressor PTEN. Science 303, 1179–1181 (2004).

    Article  CAS  PubMed  Google Scholar 

  48. Okahara, F., Ikawa, H., Kanaho, Y. & Maehama, T. Regulation of PTEN phosphorylation and stability by a tumor suppressor candidate protein. J. Biol. Chem. 279, 45300–45303 (2004).

    Article  CAS  PubMed  Google Scholar 

  49. Li, Z. et al. Regulation of PTEN by Rho small GTPases. Nat. Cell Biol. 7, 399–404 (2005).

    Article  CAS  PubMed  Google Scholar 

  50. Yim, E. K. et al. Rak functions as a tumor suppressor by regulating PTEN protein stability and function. Cancer Cell 15, 304–314 (2009).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  51. Bagchi, A. et al. CHD5 is a tumor suppressor at human 1p36. Cell 128, 459–475 (2007).

    Article  CAS  PubMed  Google Scholar 

  52. Munirajan, A. K. et al. KIF1Bβ functions as a haploinsufficient tumor suppressor gene mapped to chromosome 1p36.2 by inducing apoptotic cell death. J. Biol. Chem. 283, 24426–24434 (2008).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  53. Climent, J. et al. Deletion of the PER3 gene on chromosome 1p36 in recurrent ER-positive breast cancer. J. Clin. Oncol. 28, 3770–3778 (2010).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

Download references

Acknowledgements

We thank Q. Ye (Beijing Institute of Biotechnology, Beijing, China), Z. Liu (Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China) and S. Maddika (Centre for DNA Fingerprinting and Diagnostics, Nampally, Hyderabad, India) for kindly providing materials and H. Li, P. Xie, Y. Chen, S. Wang, J. Feng, M. Lai and D. Zhao for technical assistance. This work was supported by Chinese National Basic Research Programs (2012CB910702, 2011CB910602, 2012CB910304), the Program of International S&T Cooperation (2014DFB30020), Chinese National Natural Science Foundation Projects (31330021, 31125010, 81221004) and Beijing Natural Science Foundation Project (5142020).

Author information

Authors and Affiliations

Authors

Contributions

The project was conceived by L.Z. The experiments were designed by L.Z., L.Y., Y.Lv, H.L., Y.Y. and F.H. Most of the experiments were performed by L.Y., Y.Lv and H.L. The establishment and phenotype analysis of transgenic mice were contributed by H.L. The protein ubiquitylation assays, the protein interaction assays and animal experiments were contributed by S.S., Y.Z., G.X., X.K. and L.W. The breast cancer sample collection and immunohistochemical analysis were contributed by H.G., Y.Li, T.Z. and D.G. The data were analysed by L.Z., L.Y., Y.Lv, H.L., Z.-X.X., Y.Y., W.W. and F.H. The manuscript was written by L.Z., L.Y. and Y.Lv.

Corresponding author

Correspondence to Lingqiang Zhang.

Ethics declarations

Competing interests

The authors declare no competing financial interests.

Integrated supplementary information

Supplementary Figure 4 OTUD3 stabilizes PTEN protein.

a, Protein level analysis of PTEN in the presence or absence of overexpressed DUBs. HCT116 colon cancer cells were overexpressed with each of the Flag-tagged DUBs. b, Quantification of the PTEN protein levels relative to β-actin for Fig. 1b. n = 3 independent experiments (**, P < 0.01, Student’s t-test). c, Increasing amounts of plasmids encoding other members of OTU sub-family of DUBs were each transfected into HCT116 cells. The endogenous PTEN protein levels were examined by western blot. d, Knockdown of OTUD3 by pool siRNAs (left) or individual siRNAs (middle) or overexpression of OTUD3 (right) had no significant effect on PTEN mRNA level in MCF7 cells. n = 3 independent experiments (NS: no significance, Student’s t-test). e, MCF7 cells stably expressing the indicated shRNA were treated with cycloheximide (10 μg ml−1), and harvested at the indicated times. The left panels show immunoblots of PTEN and OTUD7A. Right panel: quantification of the PTEN protein levels relative to β-actin. n = 3 independent experiments (NS: no significance, two-way ANOVA test). f, Domain structure of OTUD3 and OTUD7A. g, Co-immunoprecipitation assays in four types of cancer cell lines to detect the endogenous interactions. The total cell lysates were immunoprecipitated with anti-OTUD3, anti-PTEN or control IgG as indicated. Both the lysates and the immunoprecipitates were subjected to western blot analysis. h, GST pull-down assays were performed to map the domain of OTUD3 required for interaction with PTEN. i, GST pull-down assays were performed to map the domain of PTEN required for interaction with OTUD3. All panels are representative results of three or more independent experiments. Statistics source data can be found in Supplementary Table 4. Uncropped images of blots are shown in Supplementary Fig. 9.

Supplementary Figure 5 OTUD3 is a more potent regulator of PTEN than USP13, and HAUSP has no significant effect on PTEN protein level.

a, Increasing amounts of Flag-USP13 were transfected into MCF7 (left) and HEK293T (right) cells. After 48 h, PTEN protein levels were detected. b, Co-immunoprecipitation of endogenous PTEN, USP13 and OTUD3 in MCF7 cells. Protein extracts were immunoprecipitated with antibody against PTEN, USP13 or OTUD3, followed by immunoblotting with antibodies indicated. c, Increasing amounts of shRNA-USP13 were transfected into MCF7 cells. After 72 h, PTEN protein levels were detected. d, Protein level analysis of PTEN in the presence of overexpressed USP13 or OTUD3. MCF7 cells were overexpressed with Myc-USP13 or Flag-OTUD3. e, Protein level analysis of PTEN in MCF7 cells stably expressing the indicated shRNA. f, MCF7 cells were transfected with a constant amount of shRNA-USP13 and increasing amounts of Myc-OTUD3 (left), or a constant amount of shRNA-OTUD3 and increasing amounts of Myc-USP13 (right). After 48 h, PTEN protein levels were detected. g, HCT116 cells transfected with the indicated constructs were treated with MG132 for 8 h before harvest. PTEN was immunoprecipitated with anti-PTEN and immunoblotted with anti-HA. h, siRNA-resistant OTUD3 was introduced into the HCT116 cells together with the OTUD3 siRNA. PTEN ubiquitination was measured. i, The PTEN ubiquitination linkage was analysed in HCT116 cells transfected with OTUD3and the indicated ubiquitin WT or K48-only plasmids. j, Increasing amounts of HAUSP plasmids were transfected into the MCF7 cells and the lysates were subjected to western blot analysis. k, Knockdown of HAUSP by individual siRNAs had no significant effect on PTEN protein level but downregulated MDM2 protein level (positive) in MCF7 cells. l, Effectiveness of HAUSP knockdown by individual siRNAs was measured by quantitative RT-PCR. Data are shown as mean ± s.d. n = 3 independent experiments. (**, P < 0.01, Student’s t-test). All panels are representative results of three or more independent experiments. Statistics source data can be found in Supplementary Table 4. Uncropped images of blots are shown in Supplementary Fig. 9.

Supplementary Figure 6 OTUD3 negatively regulates Akt signaling in a PTEN-dependent manner.

a,b, Quantitative RT-PCR analysis of indicated gene mRNA level in MCF7 (PTEN wild-type) (a) or BT549 (PTEN-null) (b) cells stably expressing the shRNA-Control or shOTUD3#1. Data were analysed using two-tailed unpaired Student’s t-test. Data are shown as mean ± s.d. n = 3 independent experiments. *: P < 0.05,**: P < 0.01, NS: no significance. c, Quantitative RT-PCR analysis of VEGF gene mRNA level in MCF7 and BT549 cells stably expressing shRNA-Control or shOTUD3 #1. Data were analysed using two-tailed unpaired Student’s t-test. Data are shown as mean ± s.d. n = 3 independent experiments. d,e, OTUD3 shRNA-transduced MCF7 or BT549 cells were serum-starved and treated with EGF for various times. Quantification of the phosphorylation levels of Akt, p38 and Erk protein relative to total Akt, p38 and Erk protein levels for Fig. 3f and g. Data are shown as mean ± s.d. n = 3 independent experiments (two-way ANOVA test). Statistics source data can be found in Supplementary Table 4.

Supplementary Figure 7 OTUD3 inhibits cell proliferation, promotes cell apoptosis, and suppresses cell migration.

a, Anchorage-independent growth of MCF7 cells stably expressing the indicated shRNA on soft agar. Viable colonies after 3 weeks were counted and the data from n = 3 independent experiments were presented (mean ± s.d.). **: P < 0.01, Student’s t-test. b, MCF7 cells stably expressing control shRNA, OTUD3 shRNA or PTEN shRNA were transfected with either OTUD3 or PTEN as indicated. Cells were tested for growth in colony assay. Colonies after one week were counted and the data (mean ± s.d.) from n = 3 independent experiments were presented (**: P < 0.01, NS: no significance, Student’s t-test, compared with cells expressing control shRNA;). c, MCF7 cells stably expressing control shRNA, OTUD3 shRNA or PTEN shRNA were seeded and cell proliferation was measured by MTS assay for 5 days. Data are shown as mean ± s.d. n = 3 independent experiments (**: P < 0.01, two-way ANOVA test). d, The indicated HCT116 cells were treated with cisplatin or DMSO to induce apoptosis. The percentage of apoptotic cells was measured by Annexin-V staining. Data are shown as mean ± s.d. n = 3 independent experiments (*: P < 0.05; **: P < 0.01, Student’s t-test). e, The cellular morphology was analysed in MCF7 cells stably expressing the indicated shRNA and/or transfected with the OTUD3 or PTEN plasmids. Cells were fixed and stained for F-actin (red). Nuclei were stained with DAPI (blue). Scale bar, 20 μm. f, Box plots showed the distribution of the size (largest in each section) of metastasis per section. Data were analysed using Mann–Whitney test (n = 50 fields from 10 mice, p = 0.0025). g, Immunoblotting of OTUD3, PTEN, p-Akt, Akt, F-actin and β-actin in lysates of primary tumors and metastatic livers from mice injected with the stable shRNA-control or shRNA-OTUD3 MCF7 cells. h, Pie charts for mice died by liver metastasis. Panel g is representative results of 3 independent experiments. Statistics source data can be found in Supplementary Table 4. Uncropped images of blots are shown in Supplementary Fig. 9.

Supplementary Figure 8 The possible synergy between OTUD3 and USP13, and the phenotype of OTUD3 transgenic mice.

a, Akt phosphorylation was analysed in MCF7 cells stably expressing the indicated shRNA. bd, The MCF7 cells expressing OTUD3 WT or mutants were measured for cell proliferation (b, two-way ANOVA test), anchorage-independent growth in soft agar (c, Student’s t-test) and transwell migration assay (d, Student’s t-test). Data are shown as mean ± s.d. n = 3 independent experiments. **: P < 0.01, *: P < 0.05. e, Representative bright-field imaging of the tumors. MCF7cells stably expressing indicated shRNA were implanted into a subcutaneous site in the skin on one flank of athymic nude mice (n = 8). On 4 weeks, mice receiving transplants of indicated cells were sacrificed. Scale bar, 1 cm. f, Whisker plots showed the distribution of the tumor weights (n = 8 tumors from 8 mice). Data were shown as mean ± s.d. and analysed using Kruskal-Wallis test. g, WT and TG MEFs were serum-starved and treated with EGF for various times. Quantification of the phosphorylation levels of Akt, p38 and Erk protein relative to total Akt, p38 and Erk protein levels for Fig. 5i. Data are shown as mean ± s.d. n = 3 independent experiments (two-way ANOVA test). h, The mating strategy of OTUD3 TG mice crossing with MMTV-PyMT mice. Panels a,g are representative results of 3 or more independent experiments. Statistics source data can be found in Supplementary Table 4. Uncropped images of blots are shown in Supplementary Fig. 9.

Supplementary Figure 9 OTUD3 E86K mutation in OTU domain leads to PTEN protein destabilization and increased tumorigenesis.

a, Sequencing the region encoding OTUD3 OTU domain from 50 cases of Chinese breast cancer patients. Breast cancer tissues (n = 50) and the corresponding adjacent tissues (n = 50) were collected and subjected to RNA extraction and reverse transcription. The cDNAs were then sequenced. If the original sequencing results displayed heterozygous peaks, the corresponding cDNAs were ligated with pGEM-T vector (Promega) and transformed into the competent bacteria. Then the colonies were sent out for sequencing. b, The mutation sites of OTUD3 in domain structure. Amino acid sequence alignment spanning OTUD3 R79 and E86 across species. c, Half-life of PTEN in the presence or absence of ectopic OTUD3 mutants. Quantification of the PTEN protein levels relative to β-actin for Fig. 6d (**: P < 0.01, two-way ANOVA test). Data are depicted as bar graphs with mean ± s.d. n = 3 independent experiments. Statistics source data can be found in Supplementary Table 4.

Supplementary Figure 10 OTUD3 expression is reduced in human breast cancer, which has no correlation with estrogen receptor, progesterone receptor or HER-2 expression.

a, Relative OTUD3 mRNA level in breast cancer tissues and matched normal control from 30 subjects. Data were analysed using Chi-square test. bd, Whisker plots show the OTUD3 mRNA level in the cancer tissues classified into negative and positive groups based on estrogen receptor (b), progesterone receptor (c) or HER-2 (d) expression from 50 subjects. Data were analysed using Mann–Whitney test. e, Representative images from immunohistochemical staining of OTUD3, PTEN and p-Akt(S473) in three serial sections of the same tumor and matched adjacent tissue. The boxed areas in the left images were magnified on the right. A, adjacent tissue; C, carcinoma. Scale bar, 50 μm.

Supplementary Figure 11 OTUD3 and USP13 exhibit a synergy effect on PTEN expression in breast cancer samples.

a,b, Potential synergy between OTUD3 and USP13 for PTEN protein levels. Serial sections of tissue arrays 68 patient breast specimens were subjected to immunohistochemistry with anti-OTUD3, anti-USP13 and anti-PTEN antibodies, respectively, and visualized by the DAB staining before imaging. Scale bar represents 50 μm. Representative images were shown in (a), and the summary of the IHC results was listed in (b). P < 0.001, calculated by both Chi-square and χ2 test. Scale bar, 50 μm. c, Regression analysis comparing HAUSP and PTEN expression in breast cancer tissues. n = 37. d, Proposed working model of OTUD3 on PTEN stability control. PTEN is a phosphatase that catalyses the conversion of the lipid second messenger PtdIns(3,4,5)P3 to PtdIns(4,5)P2 and inhibits PI3K-Akt signaling. PTEN protein is relatively stable. This study identifies the deubiquitinase OTUD3 catalyses the removal of poly-ubiquitin chain of PTEN in the cytoplasm. OTUD3 reverses the poly-ubiquitination of PTEN by E3 ubiquitin ligases (such as Nedd4-1, WWP2, XIAP and CHIP) and prevents PTEN degradation by 26S proteasome. On the other hand, a previous study (ref. 22) identified that the deubiquitinase HAUSP is mainly localized in the nucleus and catalyses the removal of mono-ubiquitination of PTEN. HAUSP regulates the PTEN cytoplasm-nucleus shuttling but has no effects on PTEN stability.

Supplementary Table 1 OTUD3 mutations in human cancer.
Supplementary Table 2 Primers for OTUD3 mutation selection in human breast cancer.
Supplementary Table 3 Primers for qPCR analysis.

Supplementary information

Supplementary Information

Supplementary Information (PDF 5384 kb)

Supplementary Table 4

Supplementary Information (XLSX 57 kb)

Rights and permissions

Reprints and permissions

About this article

Check for updates. Verify currency and authenticity via CrossMark

Cite this article

Yuan, L., Lv, Y., Li, H. et al. Deubiquitylase OTUD3 regulates PTEN stability and suppresses tumorigenesis. Nat Cell Biol 17, 1169–1181 (2015). https://doi.org/10.1038/ncb3218

Download citation

  • Received:

  • Accepted:

  • Published:

  • Issue Date:

  • DOI: https://doi.org/10.1038/ncb3218

This article is cited by

Search

Quick links

Nature Briefing

Sign up for the Nature Briefing newsletter — what matters in science, free to your inbox daily.

Get the most important science stories of the day, free in your inbox. Sign up for Nature Briefing