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.

  • Letter
  • Published:

Regulation of Hippo pathway transcription factor TEAD by p38 MAPK-induced cytoplasmic translocation

An Author Correction to this article was published on 17 July 2018

Abstract

The Hippo pathway controls organ size and tissue homeostasis, with deregulation leading to cancer. The core Hippo components in mammals are composed of the upstream serine/threonine kinases Mst1/2, MAPK4Ks and Lats1/2. Inactivation of these upstream kinases leads to dephosphorylation, stabilization, nuclear translocation and thus activation of the major functional transducers of the Hippo pathway, YAP and its paralogue TAZ1,2. YAP/TAZ are transcription co-activators that regulate gene expression primarily through interaction with the TEA domain DNA-binding family of transcription factors (TEAD)3. The current paradigm for regulation of this pathway centres on phosphorylation-dependent nucleocytoplasmic shuttling of YAP/TAZ through a complex network of upstream components2. However, unlike other transcription factors, such as SMAD, NF-κB, NFAT and STAT, the regulation of TEAD nucleocytoplasmic shuttling has been largely overlooked. In the present study, we show that environmental stress promotes TEAD cytoplasmic translocation via p38 MAPK in a Hippo-independent manner. Importantly, stress-induced TEAD inhibition predominates YAP-activating signals and selectively suppresses YAP-driven cancer cell growth. Our data reveal a mechanism governing TEAD nucleocytoplasmic shuttling and show that TEAD localization is a critical determinant of Hippo signalling output.

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: p38 mediates stress-induced TEAD cytoplasmic translocation.
Figure 2: p38 mediates stress-induced TEAD cytoplasmic translocation via protein–protein interaction.
Figure 3: TEAD cytoplasmic translocation prevents YAP activation.
Figure 4: TEAD inhibition restricts YAP-driven cancer cell growth.

Similar content being viewed by others

References

  1. Johnson, R. & Halder, G. The two faces of Hippo: targeting the Hippo pathway for regenerative medicine and cancer treatment. Nat. Rev. Drug Discov. 13, 63–79 (2014).

    Article  CAS  Google Scholar 

  2. Yu, F. X., Zhao, B. & Guan, K. L. Hippo pathway in organ size control, tissue homeostasis, and cancer. Cell 163, 811–828 (2015).

    Article  CAS  Google Scholar 

  3. Zhao, B. et al. TEAD mediates YAP-dependent gene induction and growth control. Genes Dev. 22, 1962–1971 (2008).

    Article  CAS  Google Scholar 

  4. Yu, F. X. et al. Regulation of the Hippo-YAP pathway by G-protein-coupled receptor signaling. Cell 150, 780–791 (2012).

    Article  CAS  Google Scholar 

  5. Mo, J. S. et al. Cellular energy stress induces AMPK-mediated regulation of YAP and the Hippo pathway. Nat. Cell Biol. 17, 500–510 (2015).

    Article  CAS  Google Scholar 

  6. Wang, W. et al. AMPK modulates Hippo pathway activity to regulate energy homeostasis. Nat. Cell Biol. 17, 490–499 (2015).

    Article  CAS  Google Scholar 

  7. Dupont, S. et al. Role of YAP/TAZ in mechanotransduction. Nature 474, 179–183 (2011).

    Article  CAS  Google Scholar 

  8. Zhao, B. et al. Cell detachment activates the Hippo pathway via cytoskeleton reorganization to induce anoikis. Genes Dev. 26, 54–68 (2012).

    Article  Google Scholar 

  9. Kim, N. G. & Gumbiner, B. M. Adhesion to fibronectin regulates Hippo signaling via the FAK-Src-PI3K pathway. J. Cell Biol. 210, 503–515 (2015).

    Article  CAS  Google Scholar 

  10. Sorrentino, G. et al. Metabolic control of YAP and TAZ by the mevalonate pathway. Nat. Cell Biol. 16, 357–366 (2014).

    Article  CAS  Google Scholar 

  11. de Nadal, E., Ammerer, G. & Posas, F. Controlling gene expression in response to stress. Nat. Rev. Genet. 12, 833–845 (2011).

    Article  CAS  Google Scholar 

  12. Cargnello, M. & Roux, P. P. Activation and function of the MAPKs and their substrates, the MAPK-activated protein kinases. Microbiol. Mol. Biol. Rev. 75, 50–83 (2011).

    Article  CAS  Google Scholar 

  13. Tanoue, T., Adachi, M., Moriguchi, T. & Nishida, E. A conserved docking motif in MAP kinases common to substrates, activators and regulators. Nat. Cell Biol. 2, 110–116 (2000).

    Article  CAS  Google Scholar 

  14. Tanoue, T., Maeda, R., Adachi, M. & Nishida, E. Identification of a docking groove on ERK and p38 MAP kinases that regulates the specificity of docking interactions. EMBO J. 20, 466–479 (2001).

    Article  CAS  Google Scholar 

  15. Estrada-Gelonch, A., Aramburu, J. & Lopez-Rodriguez, C. Exclusion of NFAT5 from mitotic chromatin resets its nucleo-cytoplasmic distribution in interphase. PLoS ONE 4, e7036 (2009).

    Article  Google Scholar 

  16. Moroishi, T., Hansen, C. G. & Guan, K. L. The emerging roles of YAP and TAZ in cancer. Nat. Rev. Cancer 15, 73–79 (2015).

    Article  CAS  Google Scholar 

  17. Yu, F. X. et al. Mutant Gq/11 promote uveal melanoma tumorigenesis by activating YAP. Cancer Cell 25, 822–830 (2014).

    Article  CAS  Google Scholar 

  18. Murakami, H. et al. LATS2 is a tumor suppressor gene of malignant mesothelioma. Cancer Res. 71, 873–883 (2011).

    Article  CAS  Google Scholar 

  19. Hong, A. W. et al. Osmotic stress-induced phosphorylation by NLK at Ser128 activates YAP. EMBO Rep. 18, 72–86 (2017).

    Article  CAS  Google Scholar 

  20. Moon, S. et al. Phosphorylation by NLK inhibits YAP-14-3-3-interactions and induces its nuclear localization. EMBO Rep. 18, 61–71 (2017).

    Article  CAS  Google Scholar 

  21. Home, P. et al. Altered subcellular localization of transcription factor TEAD4 regulates first mammalian cell lineage commitment. Proc. Natl Acad. Sci. USA 109, 7362–7367 (2012).

    Article  CAS  Google Scholar 

  22. Cagliero, J., Forget, A., Daldello, E., Silber, J. & Zider, A. The Hippo kinase promotes Scalloped cytoplasmic localization independently of Warts in a CRM1/Exportin1-dependent manner in Drosophila. FASEB J. 27, 1330–1341 (2013).

    Article  CAS  Google Scholar 

  23. Ran, F. A. et al. Genome engineering using the CRISPR-Cas9 system. Nat. Protoc. 8, 2281–2308 (2013).

    Article  CAS  Google Scholar 

  24. Park, H. W. et al. Alternative Wnt signaling activates YAP/TAZ. Cell 162, 780–794 (2015).

    Article  CAS  Google Scholar 

  25. Meng, Z. et al. MAP4K family kinases act in parallel to MST1/2 to activate LATS1/2 in the Hippo pathway. Nat. Commun. 6, 8357 (2015).

    Article  CAS  Google Scholar 

Download references

Acknowledgements

This work was supported by grants from the National Institutes of Health (CA196878, DE15964 and GM51586) to K.-L.G., the National Research Foundation of Korea (NRF) grant funded by the Korea government (MSIP) (2014R1A5A2009936) to H.-S.J. and by a grant of the Korea Health Technology R&D Project through the Korea Health Industry Development Institute (KHIDI), funded by the Ministry of Health & Welfare, Republic of Korea (grant number: HI17C1560), and the National Research Foundation of Korea (NRF) grant funded by the Korea government (MOE) (2017R1D1A1B03034797) and (MSIP) (2017R1A4A1015328) to H.W.P. K.C.L. and S.W.P. were supported in part by the University of California, San Diego (UCSD) Graduate Training Program in Cellular and Molecular Pharmacology (T32 GM007752).

Author information

Authors and Affiliations

Authors

Contributions

K.C.L. and H.W.P. designed experiments, performed research, analysed data and wrote the manuscript. T.M., Z.M., H.-S.J. and S.W.P. designed experiments, performed research, analysed data and reviewed the manuscript. Y.S. and J.H. provided reagents and reviewed the manuscript. K.-L.G. designed experiments, analysed data and wrote the manuscript.

Corresponding authors

Correspondence to Hyun Woo Park or Kun-Liang Guan.

Ethics declarations

Competing interests

K.-L.G. is a co-founder of and has an equity interest in Vivace Therapeutics. The terms of this arrangement have been reviewed and approved by the University of California, San Diego in accordance with its conflict of interest policies.

Integrated supplementary information

Supplementary Figure 1 Osmotic stress induces TEAD cytoplasmic translocation.

(a) Time course of osmotic stress-induced TEAD cytoplasmic translocation. HEK293A cells were treated with NaCl for 0, 1, 3, or 6 h and stained for immunofluorescence. (b) Dose response for NaCl-induced TEAD cytoplasmic translocation. HEK293A cells were stimulated with different concentrations of NaCl for 6 h and stained for immunofluorescence. TEAD cytoplasmic translocation occurs from 100 mM NaCl. (c) High cell density-induced TEAD cytoplasmic localization is p38 independent. Immunofluorescence shows inhibition or KO of p38 upon high cell density has no effect on TEAD cytoplasmic translocation. (d) Cells ectopically expressing MKK3/p38 were stained for immunofluorescence. MKK3/p38 promotes TEAD4 cytoplasmic translocation. (e) Dose response for p38 inhibitor treatment. HEK293A cells were pre-treated with different doses of p38 inhibitors as indicated, followed by NaCl stimulation and stained for immunofluorescence. (f) Western blot of p38α/β knockout cells show upregulation of p38δ/γ isoforms. (g) p38 2KO cells were stained for immunofluorescence. Deletion of p38α/β isoforms is not sufficient to inhibit TEAD cytoplasmic translocation. (h) Immunoblotting for p-p38 in p38 4KO cells shows impaired p38 activity under various p38 activating stimuli (200 mM NaCl, 500 μM sorbitol, 500 μM arsenite). (i,j) p38 4KO shows no effect on TEAD localization (i) or target gene expression, as measured by qRT-PCR (j) under basal conditions. Scale bars in ae, g, and i are 20 μm. Data are presented as mean ± s.e.m. from n = 3 independent experiments. Statistics source data are shown in Supplementary Table 1. Unprocessed scans of blots are shown in Supplementary Fig. 5.

Supplementary Figure 2 Phosphorylation of TEAD by p38 is not required for cytoplasmic translocation.

(a) Sequence of TEAD4-4SP construct harboring mutations in putative p38 phosphorylation sites. (bd) TEAD is a poor substrate for p38 phosphorylation. In vitro kinase assay for p38α (b), p38γ (c), and p38δ (d) using TEAD as a substrate. No phosphorylation or weak phosphorylation was detected in TEAD4 WT, which was further ablated in TEAD4-4SP, whereas ATF2 was effectively phosphorylated by p38. (e) Detection of TEAD4-4SP cytoplasmic translocation by p38 using immunofluorescence. (f) Sequence of putative TEAD nuclear export signal. (g) TEAD cytoplasmic translocation requires a nuclear export signal. Immunofluorescence shows truncation of TEAD disrupting a putative nuclear export signal in TEAD (1-382) mutant inhibits CRM1-dependent TEAD nuclear export compared to full length TEAD. (h) TEAD cytoplasmic translocation is a CRM1-dependent process. HEK293A cells were pretreated with LMB for the indicated times followed by NaCl stimulation and staining for immunofluorescence. Pretreatment with LMB inhibits TEAD cytoplasmic translocation. (i) Immunofluorescence of NFAT5 nuclear translocation upon NaCl stimulation. Scale bars in e and gi are 20 μm. Unprocessed scans of blots are shown in Supplementary Fig. 5.

Supplementary Figure 3 Stress-induced TEAD cytoplasmic translocation is independent of Hippo pathway.

(a,b) Stress-induced TEAD cytoplasmic translocation is independent of Lats1/2. WT and Lats KO cells were stimulated with NaCl and stained with anti-TEAD1 (a) or anti-TEAD4 (b) antibody for immunofluorescence. (c,d,e) Stress-induced TEAD cytoplasmic translocation is independent of Hippo core kinases and MAP4K. Lats KO (c), Mst KO (d), and MAP4K KO (e) cells were treated with NaCl and stained for immunofluorescence. (f) Western blot showing MAP4K and p38 are independent branches of the MAPK pathway. (g) Western blot showing stress-activated p38 does not affect YAP phosphorylation status. Scale bars in ae are 20 μm. Unprocessed scans of blots are shown in Supplementary Fig. 5.

Supplementary Figure 4 Stress-induced TEAD inhibition uncouples YAP localization and dephosphorylation in YAP-driven cancer cells.

(a,b) Stress-induced TEAD and YAP/TAZ cytoplasmic translocation in H2373 mesothelioma cells, which have homozygous deletion of NF2. Immunofluorescence showing NaCl stimulation induces TEAD and subsequent YAP/TAZ cytoplasmic sequestration (a), despite constitutive dephosphorylation of YAP as shown by western blot (b). NC, normal condition. (c) Nuclear localization of TEAD-VP16 in the presence of osmotic stress. MSTO-211H cells stably expressing TEAD1/4-VP16 construct were treated with NaCl and stained for immunofluorescence. (d,e) Stress promotes TEAD and YAP cytoplasmic sequestration in YAP-driven uveal melanoma cells. 92.1 cells were treated with YAP-inhibiting stimuli as in Fig. 1a, b. NaCl treatment elicits TEAD and YAP cytoplasmic translocation shown by immunofluorescence (d), despite constitutive dephosphorylation of YAP shown by western blot (e). (f,g) p38 mediates stress-induced TEAD cytoplasmic translocation in UM cell lines. Immunofluorescence shows treatment with SB203580 blocks NaCl-induced cytoplasmic translocation of TEAD in 92.1 (f) and OCM1 (g). (h) p38 expression inhibits colony formation of GNAQ-mutant 92.1 cells but not BRAF-mutant OCM1 cells. (i) Colony growth assay showing osmotic stress inhibits anchorage independent growth of YAP-5SA transformed MCF10A. (j) Expression of p38 reduces target gene expression induced by hyperactive YAP as measured by qRT-PCR. Target gene expression is rescued by constitutively active TEAD. Data are presented as mean from n = 2 independent experiments. (km) Immunohistochemistry staining of TEAD. Negative control staining for pan-TEAD antibody (left) and normal kidney tissue staining with pan-TEAD (right) (k). Nuclear staining of TEAD detected in mouse spleen and lung tissues (l). Cytoplasmic staining of TEAD is detected in tubule cells of normal kidney while nuclear staining is detected in renal clear cell carcinomas derived from transformed tubule cells (m). Scale bars in a, c,d, and f,g are 20 μm. Scale bars in km are 50 μm. Statistics source data are shown in Supplementary Table 1. Unprocessed scans of blots are shown in Supplementary Fig. 5.

Supplementary information

Supplementary Information

Supplementary Information (PDF 5690 kb)

Life Sciences Reporting Summary (PDF 71 kb)

Supplementary Table 1

Supplementary Information (XLSX 31 kb)

Rights and permissions

Reprints and permissions

About this article

Check for updates. Verify currency and authenticity via CrossMark

Cite this article

Lin, K., Moroishi, T., Meng, Z. et al. Regulation of Hippo pathway transcription factor TEAD by p38 MAPK-induced cytoplasmic translocation. Nat Cell Biol 19, 996–1002 (2017). https://doi.org/10.1038/ncb3581

Download citation

  • Received:

  • Accepted:

  • Published:

  • Issue Date:

  • DOI: https://doi.org/10.1038/ncb3581

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