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.

Rescue of Hippo coactivator YAP1 triggers DNA damage–induced apoptosis in hematological cancers

Subjects

Abstract

Oncogene-induced DNA damage elicits genomic instability in epithelial cancer cells, but apoptosis is blocked through inactivation of the tumor suppressor p53. In hematological cancers, the relevance of ongoing DNA damage and the mechanisms by which apoptosis is suppressed are largely unknown. We found pervasive DNA damage in hematologic malignancies, including multiple myeloma, lymphoma and leukemia, which leads to activation of a p53-independent, proapoptotic network centered on nuclear relocalization of ABL1 kinase. Although nuclear ABL1 triggers cell death through its interaction with the Hippo pathway coactivator YAP1 in normal cells, we show that low YAP1 levels prevent nuclear ABL1-induced apoptosis in these hematologic malignancies. YAP1 is under the control of a serine-threonine kinase, STK4. Notably, genetic inactivation of STK4 restores YAP1 levels, triggering cell death in vitro and in vivo. Our data therefore identify a new synthetic-lethal strategy to selectively target cancer cells presenting with endogenous DNA damage and low YAP1 levels.

This is a preview of subscription content, access via your institution

Access options

Rent or buy this article

Prices vary by article type

from$1.95

to$39.95

Prices may be subject to local taxes which are calculated during checkout

Figure 1: MM cells present ongoing DNA damage, driving ABL1 inside the nucleus.
Figure 2: pATM and pJNK modulate ABL1 nuclear relocalization.
Figure 3: YAP1 deletions and expression in MM cell lines and samples from subjects with MM.
Figure 4: YAP1 re-expression leads to ABL1-dependent proliferation reduction and cell death.
Figure 5: STK4 knockdown triggers YAP1 re-expression, reducing MM cell proliferation and increasing apoptosis in vitro and in vivo.
Figure 6: Lymphoma, leukemia and Waldenström macroglobulinemia cells present ongoing DNA damage and ABL1 nuclear localization and undergo apoptosis after STK4-mediated increases in YAP1 levels.

Similar content being viewed by others

Accession codes

Accessions

Gene Expression Omnibus

References

  1. Halazonetis, T.D., Gorgoulis, V.G. & Bartek, J. An oncogene-induced DNA damage model for cancer development. Science 319, 1352–1355 (2008).

    Article  CAS  Google Scholar 

  2. Boehrer, S. et al. Suppression of the DNA damage response in acute myeloid leukemia versus myelodysplastic syndrome. Oncogene 28, 2205–2218 (2009).

    Article  CAS  Google Scholar 

  3. Walters, D.K. et al. Evidence for ongoing DNA damage in multiple myeloma cells as revealed by constitutive phosphorylation of H2AX. Leukemia 25, 1344–1353 (2011).

    Article  CAS  Google Scholar 

  4. Xu-Monette, Z.Y. et al. Dysfunction of the TP53 tumor suppressor gene in lymphoid malignancies. Blood 119, 3668–3683 (2012).

    Article  CAS  Google Scholar 

  5. Chapman, M.A. et al. Initial genome sequencing and analysis of multiple myeloma. Nature 471, 467–472 (2011).

    Article  CAS  Google Scholar 

  6. Baskaran, R. et al. Ataxia telangiectasia mutant protein activates c-Abl tyrosine kinase in response to ionizing radiation. Nature 387, 516–519 (1997).

    Article  CAS  Google Scholar 

  7. Kharbanda, S. et al. Activation of the c-Abl tyrosine kinase in the stress response to DNA-damaging agents. Nature 376, 785–788 (1995).

    Article  CAS  Google Scholar 

  8. Shafman, T. et al. Interaction between ATM protein and c-Abl in response to DNA damage. Nature 387, 520–523 (1997).

    Article  CAS  Google Scholar 

  9. Yuan, Z.M. et al. Regulation of DNA damage–induced apoptosis by the c-Abl tyrosine kinase. Proc. Natl. Acad. Sci. USA 94, 1437–1440 (1997).

    Article  CAS  Google Scholar 

  10. Yuan, Z.M. et al. Role for c-Abl tyrosine kinase in growth arrest response to DNA damage. Nature 382, 272–274 (1996).

    Article  CAS  Google Scholar 

  11. Gorgoulis, V.G. et al. Activation of the DNA damage checkpoint and genomic instability in human precancerous lesions. Nature 434, 907–913 (2005).

    Article  CAS  Google Scholar 

  12. Bartkova, J. et al. DNA damage response as a candidate anti-cancer barrier in early human tumorigenesis. Nature 434, 864–870 (2005).

    Article  CAS  Google Scholar 

  13. Brown, L. & McCarthy, N. DNA repair. A sense-abl response? Nature 387, 450–451 (1997).

    Article  CAS  Google Scholar 

  14. Yoshida, K., Yamaguchi, T., Natsume, T., Kufe, D. & Miki, Y. JNK phosphorylation of 14-3-3 proteins regulates nuclear targeting of c-Abl in the apoptotic response to DNA damage. Nat. Cell Biol. 7, 278–285 (2005).

    Article  CAS  Google Scholar 

  15. Hickson, I. et al. Identification and characterization of a novel and specific inhibitor of the ataxia-telangiectasia mutated kinase ATM. Cancer Res. 64, 9152–9159 (2004).

    Article  CAS  Google Scholar 

  16. Bennett, B.L. et al. SP600125, an anthrapyrazolone inhibitor of Jun N-terminal kinase. Proc. Natl. Acad. Sci. USA 98, 13681–13686 (2001).

    Article  CAS  Google Scholar 

  17. White, E. & Prives, C. DNA damage enables p73. Nature 399, 734–735, 737 (1999).

    Article  CAS  Google Scholar 

  18. Sudol, M. Yes-associated protein (YAP65) is a proline-rich phosphoprotein that binds to the SH3 domain of the Yes proto-oncogene product. Oncogene 9, 2145–2152 (1994).

    CAS  PubMed  Google Scholar 

  19. Levy, D., Adamovich, Y., Reuven, N. & Shaul, Y. Yap1 phosphorylation by c-Abl is a critical step in selective activation of proapoptotic genes in response to DNA damage. Mol. Cell 29, 350–361 (2008).

    Article  CAS  Google Scholar 

  20. Levy, D., Adamovich, Y., Reuven, N. & Shaul, Y. The Yes-associated protein 1 stabilizes p73 by preventing Itch-mediated ubiquitination of p73. Cell Death Differ. 14, 743–751 (2007).

    Article  CAS  Google Scholar 

  21. Keats, J.J. et al. Promiscuous mutations activate the noncanonical NF-κB pathway in multiple myeloma. Cancer Cell 12, 131–144 (2007).

    Article  CAS  Google Scholar 

  22. Annunziata, C.M. et al. Frequent engagement of the classical and alternative NF-κB pathways by diverse genetic abnormalities in multiple myeloma. Cancer Cell 12, 115–130 (2007).

    Article  CAS  Google Scholar 

  23. Carrasco, D.R. et al. High-resolution genomic profiles define distinct clinico-pathogenetic subgroups of multiple myeloma patients. Cancer Cell 9, 313–325 (2006).

    Article  CAS  Google Scholar 

  24. Walker, B.A. et al. A compendium of myeloma-associated chromosomal copy number abnormalities and their prognostic value. Blood 116, e56–e65 (2010).

    Article  CAS  Google Scholar 

  25. Basu, S., Totty, N.F., Irwin, M.S., Sudol, M. & Downward, J. Akt phosphorylates the Yes-associated protein, YAP, to induce interaction with 14-3-3 and attenuation of p73-mediated apoptosis. Mol. Cell 11, 11–23 (2003).

    Article  CAS  Google Scholar 

  26. Strano, S. et al. Physical interaction with Yes-associated protein enhances p73 transcriptional activity. J. Biol. Chem. 276, 15164–15173 (2001).

    Article  CAS  Google Scholar 

  27. Strano, S. et al. The transcriptional coactivator Yes-associated protein drives p73 gene-target specificity in response to DNA Damage. Mol. Cell 18, 447–459 (2005).

    Article  CAS  Google Scholar 

  28. Lapi, E. et al. PML, YAP, and p73 are components of a proapoptotic autoregulatory feedback loop. Mol. Cell 32, 803–814 (2008).

    Article  CAS  Google Scholar 

  29. Zhou, D. et al. Mst1 and Mst2 maintain hepatocyte quiescence and suppress hepatocellular carcinoma development through inactivation of the Yap1 oncogene. Cancer Cell 16, 425–438 (2009).

    Article  CAS  Google Scholar 

  30. Zhou, D. et al. Mst1 and Mst2 protein kinases restrain intestinal stem cell proliferation and colonic tumorigenesis by inhibition of Yes-associated protein (Yap) overabundance. Proc. Natl. Acad. Sci. USA 108, E1312–E1320 (2011).

    Article  CAS  Google Scholar 

  31. Creasy, C.L., Ambrose, D.M. & Chernoff, J. The Ste20-like protein kinase, Mst1, dimerizes and contains an inhibitory domain. J. Biol. Chem. 271, 21049–21053 (1996).

    Article  CAS  Google Scholar 

  32. Kaelin, W.G. Jr. The concept of synthetic lethality in the context of anticancer therapy. Nat. Rev. Cancer 5, 689–698 (2005).

    Article  CAS  Google Scholar 

  33. Pan, D. The Hippo signaling pathway in development and cancer. Dev. Cell 19, 491–505 (2010).

    Article  CAS  Google Scholar 

  34. Bertini, E., Oka, T., Sudol, M., Strano, S. & Blandino, G. YAP: at the crossroad between transformation and tumor suppression. Cell Cycle 8, 49–57 (2009).

    Article  CAS  Google Scholar 

  35. Harvey, K.F., Zhang, X. & Thomas, D.M. The Hippo pathway and human cancer. Nat. Rev. Cancer 13, 246–257 (2013).

    Article  CAS  Google Scholar 

  36. Reuven, N., Adler, J., Meltser, V. & Shaul, Y. The Hippo pathway kinase Lats2 prevents DNA damage–induced apoptosis through inhibition of the tyrosine kinase c-Abl. Cell Death Differ. 20, 1330–1340 (2013).

    Article  CAS  Google Scholar 

  37. Tschop, K. et al. A kinase shRNA screen links LATS2 and the pRB tumor suppressor. Genes Dev. 25, 814–830 (2011).

    Article  CAS  Google Scholar 

  38. Piccolo, S., Cordenonsi, M. & Dupont, S. Molecular pathways: YAP and TAZ take center stage in organ growth and tumorigenesis. Clin. Cancer Res. 19, 4925–4930 (2013).

    Article  CAS  Google Scholar 

Download references

Acknowledgements

We thank M. Sudol (Mount Sinai School of Medicine) for the eGFP-YAP1 construct, W. Hahn (Dana-Farber Cancer Institute) for pLKO.1 shRNA lentiviral vectors and S. Rosen (Northwestern University) and R. Burger (University of Kiel) for MM.1R and INA-6 MM cells. We also thank E. Di Cairano and L. Spagnuolo for immunohistochemical stains, F. Ghini and A.M. Gasparri for technical help, the Dana-Farber Cancer Institute Flow cytometry facility, D. Kufe and F. Bernassola for insightful suggestions, C. Brennan for the bioinformatics analysis and members of the Anderson and Tonon lab for sharing reagents and critical reading of the manuscript. W.M.K. is supported by the Intramural Research Program of the US National Institutes of Health (NIH), National Cancer Institute, Center for Cancer Research. This work was supported by Fondazione CARIPLO, a Marie Curie International Reintegration Grant and the Associazione Italiana per la Ricerca sul Cancro (AIRC; AIRC Special Program Molecular Clinical Oncology, 5 per mille no. 9980 to A.N. and Investigator Grants and Special Program Molecular Clinical Oncology, 5 per mille no. 9965 to G.T.) (G.T.). K.C.A. is an American Cancer Society Clinical Research Professor and is supported by NIH grants NIH SPORE P50 100707, PO-1 78378 and RO-1 50947.

Author information

Authors and Affiliations

Authors

Contributions

F.C. designed research, performed experiments, analyzed data and wrote the manuscript. T.H. designed research and analyzed data. C.X., E.t.H., E.A. and M.T.B. performed mouse experiments. M.S., A.C.K., K.-K.W., G.B. and F.C.-C. contributed to designing experiments. M.D., L.A. and A.N. analyzed genomic and microarray data. M.P. and R.C. analyzed immunohistochemistry staining. M.M. and P.G.R. provided patient samples. W.M.K. analyzed data and wrote the manuscript. K.C.A. analyzed data and wrote the manuscript. G.T. designed research, analyzed data and wrote the manuscript.

Corresponding authors

Correspondence to Kenneth C Anderson or Giovanni Tonon.

Ethics declarations

Competing interests

The authors declare no competing financial interests.

Supplementary information

Supplementary Text and Figures

Supplementary Figures 1–15 (PDF 5371 kb)

Rights and permissions

Reprints and permissions

About this article

Check for updates. Verify currency and authenticity via CrossMark

Cite this article

Cottini, F., Hideshima, T., Xu, C. et al. Rescue of Hippo coactivator YAP1 triggers DNA damage–induced apoptosis in hematological cancers. Nat Med 20, 599–606 (2014). https://doi.org/10.1038/nm.3562

Download citation

  • Received:

  • Accepted:

  • Published:

  • Issue Date:

  • DOI: https://doi.org/10.1038/nm.3562

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