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:

A single-copy Sleeping Beauty transposon mutagenesis screen identifies new PTEN-cooperating tumor suppressor genes

Abstract

The overwhelming number of genetic alterations identified through cancer genome sequencing requires complementary approaches to interpret their significance and interactions. Here we developed a novel whole-body insertional mutagenesis screen in mice, which was designed for the discovery of Pten-cooperating tumor suppressors. Toward this aim, we coupled mobilization of a single-copy inactivating Sleeping Beauty transposon to Pten disruption within the same genome. The analysis of 278 transposition-induced prostate, breast and skin tumors detected tissue-specific and shared data sets of known and candidate genes involved in cancer. We validated ZBTB20, CELF2, PARD3, AKAP13 and WAC, which were identified by our screens in multiple cancer types, as new tumor suppressor genes in prostate cancer. We demonstrated their synergy with PTEN in preventing invasion in vitro and confirmed their clinical relevance. Further characterization of Wac in vivo showed obligate haploinsufficiency for this gene (which encodes an autophagy-regulating factor) in a Pten-deficient context. Our study identified complex PTEN-cooperating tumor suppressor networks in different cancer types, with potential clinical implications.

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: A Sleeping Beauty–dependent inactivatable Pten allele for coupled whole-body mutagenesis and tumor suppressor discovery in mice.
Figure 2: Characterization of Pten-inactivated prostate cancer and identification of genes that potentially drive its progression.
Figure 3: Perturbed biological pathways, cellular processes and previously uncharacterized tumor suppressor candidates enriched among the PSB∩PISB CIS genes.
Figure 4: Genetic inhibition of tumor suppressor genes drives prostate cancer progression through canonical signaling pathways.
Figure 5: Clinical significance of validated genes in human prostate cancer.
Figure 6: In vivo validation of Wac as a new obligate haploinsufficient gene in prostate cancer.

Similar content being viewed by others

Accession codes

Primary accessions

ArrayExpress

References

  1. Alexandrov, L.B. et al. Signatures of mutational processes in human cancer. Nature 500, 415–421 (2013).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  2. Davoli, T. et al. Cumulative haploinsufficiency and triplosensitivity drive aneuploidy patterns and shape the cancer genome. Cell 155, 948–962 (2013).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  3. Takeda, H. et al. Transposon mutagenesis identifies genes and evolutionary forces driving gastrointestinal tract tumor progression. Nat. Genet. 47, 142–150 (2015).

    Article  CAS  PubMed  Google Scholar 

  4. Rad, R. et al. A conditional piggyBac transposition system for genetic screening in mice identifies oncogenic networks in pancreatic cancer. Nat. Genet. 47, 47–56 (2015).

    Article  CAS  PubMed  Google Scholar 

  5. Moriarity, B.S. & Largaespada, D.A. Sleeping Beauty transposon insertional mutagenesis–based mouse models for cancer gene discovery. Curr. Opin. Genet. Dev. 30, 66–72 (2015).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  6. Mann, M.B. et al. Transposon mutagenesis identifies genetic drivers of BrafV600E melanoma. Nat. Genet. 47, 486–495 (2015).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  7. Jones, K.B. Transposon mutagenesis disentangles osteosarcoma genetic drivers. Nat. Genet. 47, 564–565 (2015).

    Article  CAS  PubMed  Google Scholar 

  8. Vassiliou, G.S. et al. Mutant nucleophosmin and cooperating pathways drive leukemia initiation and progression in mice. Nat. Genet. 43, 470–475 (2011).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  9. Rad, R. et al. PiggyBac transposon mutagenesis: a tool for cancer gene discovery in mice. Science 330, 1104–1107 (2010).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  10. Bard-Chapeau, E.A. et al. Transposon mutagenesis identifies genes driving hepatocellular carcinoma in a chronic hepatitis B mouse model. Nat. Genet. 46, 24–32 (2014).

    Article  CAS  PubMed  Google Scholar 

  11. Pérez-Mancera, P.A. et al. The deubiquitinase USP9X suppresses pancreatic ductal adenocarcinoma. Nature 486, 266–270 (2012).

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  12. Rangel, R. et al. Transposon mutagenesis identifies genes that cooperate with mutant Pten in breast cancer progression. Proc. Natl. Acad. Sci. USA 113, E7749–E7758 (2016).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  13. Ahmad, I. et al. Sleeping Beauty screen reveals Pparg activation in metastatic prostate cancer. Proc. Natl. Acad. Sci. USA 113, 8290–8295 (2016).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  14. Dupuy, A.J., Akagi, K., Largaespada, D.A., Copeland, N.G. & Jenkins, N.A. Mammalian mutagenesis using a highly mobile somatic Sleeping Beauty transposon system. Nature 436, 221–226 (2005).

    Article  CAS  PubMed  Google Scholar 

  15. Luo, G. et al. Cancer predisposition caused by elevated mitotic recombination in Bloom mice. Nat. Genet. 26, 424–429 (2000).

    Article  CAS  PubMed  Google Scholar 

  16. Hollander, M.C., Blumenthal, G.M. & Dennis, P.A. PTEN loss in the continuum of common cancers, rare syndromes and mouse models. Nat. Rev. Cancer 11, 289–301 (2011).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  17. Friedrich, M.J. et al. Genome-wide transposon screening and quantitative insertion site sequencing for cancer gene discovery in mice. Nat. Protoc. 12, 289–309 (2017).

    Article  CAS  PubMed  Google Scholar 

  18. de Ridder, J., Uren, A., Kool, J., Reinders, M. & Wessels, L. Detecting statistically significant common insertion sites in retroviral insertional mutagenesis screens. PLOS Comput. Biol. 2, e166 (2006).

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  19. Collier, L.S., Carlson, C.M., Ravimohan, S., Dupuy, A.J. & Largaespada, D.A. Cancer gene discovery in solid tumors using transposon-based somatic mutagenesis in the mouse. Nature 436, 272–276 (2005).

    Article  CAS  PubMed  Google Scholar 

  20. Karreth, F.A. et al. In vivo identification of tumor-suppressive PTEN ceRNAs in an oncogenic BRAF-induced mouse model of melanoma. Cell 147, 382–395 (2011).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  21. Keng, V.W. et al. PTEN and NF1 inactivation in Schwann cells produces a severe phenotype in the peripheral nervous system that promotes the development and malignant progression of peripheral nerve sheath tumors. Cancer Res. 72, 3405–3413 (2012).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  22. Dorr, C. et al. Transposon mutagenesis screen identifies potential lung cancer drivers and CUL3 as a tumor suppressor. Mol. Cancer Res. 13, 1238–1247 (2015).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  23. 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 

  24. Trotman, L.C. et al. Pten dose dictates cancer progression in the prostate. PLoS Biol. 1, E59 (2003).

    Article  PubMed  PubMed Central  Google Scholar 

  25. Ai, J. et al. Concomitant loss of Eaf2 (U19) and Pten synergistically promotes prostate carcinogenesis in the mouse model. Oncogene 33, 2286–2294 (2014).

    Article  CAS  PubMed  Google Scholar 

  26. Patel, R. et al. Sprouty2, PTEN and PP2A interact to regulate prostate cancer progression. J. Clin. Invest. 123, 1157–1175 (2013).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  27. Fernández-Marcos, P.J. et al. Simultaneous inactivation of Par-4 and Pten in vivo leads to synergistic NF-κB activation and invasive prostate carcinoma. Proc. Natl. Acad. Sci. USA 106, 12962–12967 (2009).

    Article  PubMed  PubMed Central  Google Scholar 

  28. Carver, B.S. et al. Aberrant ERG expression cooperates with loss of PTEN to promote cancer progression in the prostate. Nat. Genet. 41, 619–624 (2009).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  29. Abate-Shen, C. et al. Nkx3.1;Pten mutant mice develop invasive prostate adenocarcinoma and lymph node metastases. Cancer Res. 63, 3886–3890 (2003).

    CAS  PubMed  Google Scholar 

  30. Di Cristofano, A., De Acetis, M., Koff, A., Cordon-Cardo, C. & Pandolfi, P.P. Pten and p27KIP1 cooperate in prostate cancer tumor suppression in the mouse. Nat. Genet. 27, 222–224 (2001).

    Article  CAS  PubMed  Google Scholar 

  31. An, O. et al. NCG 4.0: the network of cancer genes in the era of massive mutational screenings of cancer genomes. Database 7, bau015 (2014).

    Google Scholar 

  32. Gundem, G. et al. The evolutionary history of lethal metastatic prostate cancer. Nature 520, 353–357 (2015).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  33. Cooper, C.S. et al. Analysis of the genetic phylogeny of multifocal prostate cancer identifies multiple independent clonal expansions in neoplastic and morphologically normal prostate tissue. Nat. Genet. 47, 367–372 (2015).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  34. Boutros, P.C. et al. Spatial genomic heterogeneity within localized, multifocal prostate cancer. Nat. Genet. 47, 736–745 (2015).

    Article  CAS  PubMed  Google Scholar 

  35. Taylor, B.S. et al. Integrative genomic profiling of human prostate cancer. Cancer Cell 18, 11–22 (2010).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  36. Barbieri, C.E. et al. Exome sequencing identifies recurrent SPOP, FOXA1 and MED12 mutations in prostate cancer. Nat. Genet. 44, 685–689 (2012).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  37. Huang, W., Sherman, B.T. & Lempicki, R.A. Systematic and integrative analysis of large gene lists using DAVID bioinformatics resources. Nat. Protoc. 4, 44–57 (2009).

    Article  CAS  Google Scholar 

  38. Grasso, C.S. et al. The mutational landscape of lethal castration-resistant prostate cancer. Nature 487, 239–243 (2012).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  39. Ding, L. et al. CBP loss cooperates with PTEN haploinsufficiency to drive prostate cancer: implications for epigenetic therapy. Cancer Res. 74, 2050–2061 (2014).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  40. Chen, J.L. et al. Deregulation of a Hox protein regulatory network spanning prostate cancer initiation and progression. Clin. Cancer Res. 18, 4291–4302 (2012).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  41. Koon, H.B., Ippolito, G.C., Banham, A.H. & Tucker, P.W. FOXP1: a potential therapeutic target in cancer. Expert Opin. Ther. Targets 11, 955–965 (2007).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  42. Takayama, K. et al. Integrative analysis of FOXP1 function reveals a tumor-suppressive effect in prostate cancer. Mol. Endocrinol. 28, 2012–2024 (2014).

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  43. Demichelis, F. et al. Distinct genomic aberrations associated with ERG-rearranged prostate cancer. Genes Chromosom. Cancer 48, 366–380 (2009).

    Article  CAS  PubMed  Google Scholar 

  44. Naguib, A. et al. PTEN functions by recruitment to cytoplasmic vesicles. Mol. Cell 58, 255–268 (2015).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  45. Leslie, N.R., Batty, I.H., Maccario, H., Davidson, L. & Downes, C.P. Understanding PTEN regulation: PIP2, polarity and protein stability. Oncogene 27, 5464–5476 (2008).

    Article  CAS  PubMed  Google Scholar 

  46. Tay, Y. et al. Coding-independent regulation of the tumor suppressor PTEN by competing endogenous mRNAs. Cell 147, 344–357 (2011).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  47. Sarver, A.L. & Subramanian, S. Competing endogenous RNA database. Bioinformation 8, 731–733 (2012).

    Article  PubMed  PubMed Central  Google Scholar 

  48. Carver, B.S. et al. Reciprocal feedback regulation of PI3K and androgen receptor signaling in PTEN-deficient prostate cancer. Cancer Cell 19, 575–586 (2011).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  49. White, E. Deconvoluting the context-dependent role for autophagy in cancer. Nat. Rev. Cancer 12, 401–410 (2012).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  50. McKnight, N.C. et al. Genome-wide siRNA screen reveals amino acid starvation–induced autophagy requires SCOC and WAC. EMBO J. 31, 1931–1946 (2012).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  51. Joachim, J. et al. Activation of ULK kinase and autophagy by GABARAP trafficking from the centrosome is regulated by WAC and GM130. Mol. Cell 60, 899–913 (2015).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  52. Fernández, A.F. & López-Otín, C. The functional and pathologic relevance of autophagy proteases. J. Clin. Invest. 125, 33–41 (2015).

    Article  PubMed  PubMed Central  Google Scholar 

  53. Yin, Y. & Shen, W.H. PTEN: a new guardian of the genome. Oncogene 27, 5443–5453 (2008).

    Article  CAS  PubMed  Google Scholar 

  54. Milella, M. et al. PTEN: multiple functions in human malignant tumors. Front. Oncol. 5, 24 (2015).

    Article  PubMed  PubMed Central  Google Scholar 

  55. Qu, X. et al. Promotion of tumorigenesis by heterozygous disruption of the beclin 1 autophagy gene. J. Clin. Invest. 112, 1809–1820 (2003).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  56. Santanam, U. et al. Atg7 cooperates with Pten loss to drive prostate cancer tumor growth. Genes Dev. 30, 399–407 (2016).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  57. Skarnes, W.C. et al. A conditional-knockout resource for the genome-wide study of mouse gene function. Nature 474, 337–342 (2011).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  58. Izsvák, Z. et al. Involvement of a bifunctional, paired-like DNA-binding domain and a transpositional enhancer in Sleeping Beauty transposition. J. Biol. Chem. 277, 34581–34588 (2002).

    Article  PubMed  Google Scholar 

  59. Lee, E.C. et al. A highly efficient Escherichia coli–based chromosome engineering system adapted for recombinogenic targeting and subcloning of BAC DNA. Genomics 73, 56–65 (2001).

    Article  CAS  PubMed  Google Scholar 

  60. Kim, D. et al. TopHat2: accurate alignment of transcriptomes in the presence of insertions, deletions and gene fusions. Genome Biol. 14, R36 (2013).

    Article  PubMed  PubMed Central  Google Scholar 

  61. Anders, S., Pyl, P.T. & Huber, W. HTSeq—a Python framework to work with high-throughput sequencing data. Bioinformatics 31, 166–169 (2015).

    Article  CAS  PubMed  Google Scholar 

  62. Love, M.I., Huber, W. & Anders, S. Moderated estimation of fold change and dispersion for RNA-seq data with DESeq2. Genome Biol. 15, 550 (2014).

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  63. Temiz, N.A. et al. RNA sequencing of Sleeping Beauty transposon-induced tumors detects transposon–RNA fusions in forward genetic cancer screens. Genome Res. 26, 119–129 (2016).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  64. Wu, T.D. & Nacu, S. Fast and SNP-tolerant detection of complex variants and splicing in short reads. Bioinformatics 26, 873–881 (2010).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  65. Subramanian, A. et al. Gene set enrichment analysis: a knowledge-based approach for interpreting genome-wide expression profiles. Proc. Natl. Acad. Sci. USA 102, 15545–15550 (2005).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  66. Merico, D., Isserlin, R., Stueker, O., Emili, A. & Bader, G.D. Enrichment map: a network-based method for gene set enrichment visualization and interpretation. PLoS One 5, e13984 (2010).

    Article  PubMed  PubMed Central  CAS  Google Scholar 

Download references

Acknowledgements

We thank the staff members of the Research Support Facility at the Wellcome Trust Sanger Institute, the Laboratory of Molecular Medicine at IMOMA, and those at the Transgenic Animal Unit, the Molecular Histopathology Unit, the Department of Biochemistry and Molecular Biology and the Biobank of the Principality of Asturias at IUOPA, for excellent technical assistance. This work was supported by grants from the Wellcome Trust (grant no. 098051; A.B.), the Ministerio de Economía y Competitividad–Spain (grant no. SAF2014-52413; C.L.-O.) and the German Research Society (grant no. SFB1243; R.R.), as well as by funding from the Fundación María Cristina Masaveu Peterson (J.C.), the Fundación Centro Médico de Asturias (J.C.), the Fundación Bancaria Caja de Ahorros de Asturias/Liberbank (J.C. and A.A.), FEBS (J.d.L.R. and J.C.), CIBERONC (Plan Feder) (C.L.-O.), the Progeria Research Foundation (C.L.-O.), the EDP Foundation (C.L.-O.) and the German Cancer Consortium (R.R.). G.S.V. is funded by a Wellcome Trust Senior Fellowship in Clinical Science (WT095663MA). J.d.l.R. is a recipient of a FEBS Long-Term Fellowship and was a recipient of a fellowship from the Fundación María Cristina Masaveu Peterson during part of this work. J.C. was a recipient of a FEBS Long-Term Fellowship in the initial phases of this work.

Author information

Authors and Affiliations

Authors

Contributions

J.d.l.R., R.R., C.L.-O., A.B. and J.C. designed the study; J.d.l.R., J.W., R.R. and J.C. generated mouse alleles and cohorts, and performed experiments; J.d.l.R., J.W., L.R., Q.L., M.A.L., G.S.V., R.R. and J.C. performed mouse necropsies; A.A., M.S.F.-G., M.T.F.-G. and G.J.H. performed histopathological analysis; J.d.l.R., M.J.F., Y.L and H.P. did bioinformatics analyses; J.d.l.R., C.L.-O. and J.C. interpreted results; M.J.F., S.B.d.Q., I.N., E.M., A.S. and R.F. contributed to some of the experiments; R.R., C.L.-O., A.B. and J.C supervised the study; and J.d.l.R. and J.C. wrote the manuscript. All authors discussed the results and commented on the manuscript.

Corresponding author

Correspondence to Juan Cadiñanos.

Ethics declarations

Competing interests

The authors declare no competing financial interests.

Supplementary information

Supplementary Text and Figures

Supplementary Figures 1–13 (PDF 10687 kb)

Supplementary Tables

Supplementary Tables 1–36 (XLSX 18564 kb)

Rights and permissions

Reprints and permissions

About this article

Check for updates. Verify currency and authenticity via CrossMark

Cite this article

de la Rosa, J., Weber, J., Friedrich, M. et al. A single-copy Sleeping Beauty transposon mutagenesis screen identifies new PTEN-cooperating tumor suppressor genes. Nat Genet 49, 730–741 (2017). https://doi.org/10.1038/ng.3817

Download citation

  • Received:

  • Accepted:

  • Published:

  • Issue Date:

  • DOI: https://doi.org/10.1038/ng.3817

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