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.

  • Matters Arising
  • Published:

Hippo signalling maintains ER expression and ER+ breast cancer growth

Matters Arising to this article was published on 03 March 2021

The Original Article was published on 09 January 2017

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: LATS is essential to maintaining ER expression.
Fig. 2: LATS knockout inhibits the growth of ER+ breast cancer cells by abolishing expression of ERα.

Data availability

The RNA sequencing data are available in Gene Expression Omnibus database, with the accession number GSE134615. All other data supporting the findings of this study are available from the corresponding author upon request. Source data are provided with this paper.

References

  1. 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 

  2. Zheng, Y. & Pan, D. The Hippo signaling pathway in development and disease. Dev. Cell 50, 264–282 (2019).

    Article  CAS  Google Scholar 

  3. Britschgi, A. et al. The Hippo kinases LATS1 and 2 control human breast cell fate via crosstalk with ERα. Nature 541, 541–545 (2017).

    Article  ADS  CAS  Google Scholar 

  4. Jardé, T. et al. Wnt and Neuregulin1/ErbB signalling extends 3D culture of hormone responsive mammary organoids. Nat. Commun. 7, 13207 (2016).

    Article  ADS  Google Scholar 

  5. Kessler, M. et al. The Notch and Wnt pathways regulate stemness and differentiation in human fallopian tube organoids. Nat. Commun. 6, 8989 (2015).

    Article  ADS  CAS  Google Scholar 

  6. Turco, M. Y. et al. Long-term, hormone-responsive organoid cultures of human endometrium in a chemically defined medium. Nat. Cell Biol. 19, 568–577 (2017).

    Article  CAS  Google Scholar 

  7. Zhao, B. et al. Inactivation of YAP oncoprotein by the Hippo pathway is involved in cell contact inhibition and tissue growth control. Genes Dev. 21, 2747–2761 (2007).

    Article  CAS  Google Scholar 

  8. Dong, J. et al. Elucidation of a universal size-control mechanism in Drosophila and mammals. Cell 130, 1120–1133 (2007).

    Article  CAS  Google Scholar 

  9. Zhao, B., Kim, J., Ye, X., Lai, Z. C. & Guan, K. L. Both TEAD-binding and WW domains are required for the growth stimulation and oncogenic transformation activity of yes-associated protein. Cancer Res. 69, 1089–1098 (2009).

    Article  CAS  Google Scholar 

  10. Plouffe, S. W. et al. Characterization of Hippo pathway components by gene inactivation. Mol. Cell 64, 993–1008 (2016).

    Article  CAS  Google Scholar 

  11. Yin, F. et al. Spatial organization of Hippo signaling at the plasma membrane mediated by the tumor suppressor Merlin/NF2. Cell 154, 1342–1355 (2013).

    Article  CAS  Google Scholar 

  12. 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 

  13. Li, Q. et al. Lats1/2 sustain intestinal stem cells and Wnt activation through TEAD-dependent and independent transcription. Cell Stem Cell 26, 675–692.e8 (2020).

    Article  CAS  Google Scholar 

  14. Cheung, P. et al. Regenerative reprogramming of the intestinal stem cell state via Hippo signaling suppresses metastatic colorectal cancer. Cell Stem Cell 27, 590–604.e9 (2020).

    Article  CAS  Google Scholar 

Download references

Acknowledgements

We thank members of the laboratory of K.-L.G. for discussions. This work was supported by National Cancer Institute grant CA207504 (to J.Z.) and National Institutes of Health grants R01CA217642 and GM51586 (to K.-L.G.).

Author information

Authors and Affiliations

Authors

Contributions

S.M. and K.-L.G. conceived the study with advice from J.Z., R.L.J. and M.G.R. S.M. performed the majority of the experiments and data analyses with help from Z.W. and F.Y. The manuscript was written by S.M. and K.-L.G., with input from all of the authors.

Corresponding author

Correspondence to Kun-Liang Guan.

Ethics declarations

Competing interests

K.-L.G. is a cofounder of and has equity interest in Vivace Therapeutics. The other authors declare no competing interests.

Extended data figures and tables

Extended Data Fig. 1 LATS1/2 are required to maintain ERα and target-gene expression.

a, Impaired YAP phosphorylation in LATS1/2 double-knockout cells. Wild-type MCF-7 cells, and two MCF-7 LATS1/2 double-knockout clones, were serum-starved or treated with 1 μM cerivastatin (for 1 h), and subjected to immunoblot analysis. b, Increased YAP and TAZ transcriptional activity in LATS1/2 double-knockout cells. qPCR of the YAP and TAZ target-genes CTGF and CYR61 (also known as CCN1). c, LATS1/2 deficiency inhibits growth of MCF-7 cells. d, YAP signature and oestrogen receptor signature are among the top upregulated and downregulated gene sets, respectively, in LATS1/2 double-knockout cells. Gene enrichment analysis of LATS1/2-deficient (n = 3) and wild-type (n = 3) MCF-7 cells. Circle size represents the relative gene numbers in each set. Red, enriched in LATS1/2-deficient cells; blue, enriched in wild-type cells. e, Opposite effects of LATS1/2 deletion on the YAP signature genes (red) and oestrogen response signature genes (blue) in MCF-7 cells. Significance, false-discovery rate-adjusted P value. Magnitude of difference, fold change. n = 3 independent samples. f, YAP, TAZ and ERα localization and intensity in wild-type or LATS1/2 double-knockout MCF-7 cells. Scale bar, 20 μm. g, h, Redundancy of LATS1/2 in regulating YAP activity and ERα expression. MCF-7 clones with different sgRNA or shRNA targeting LATS1, LATS2 or both were subjected to qPCR analysis for CTGF and ESR1 (g) or immunoblot (h). ik, Deletion of Lats1/2 reduces ERα in mouse endometrial organoids. Organoids derived from the endometrial tissues of Lats1+/+Lats2+/+ and Lats1fl/flLats2fl/fl mice were infected with Cre-encoding adenovirus and subjected to immunohistochemistry (i), immunoblot (j) and qPCR analysis (k). Scale bar, 100 μm for bright field, 30 μm for immunohistochemistry. ln, Deletion of Lats1/2 reduces ERα in mouse Fallopian tube organoids. Organoids derived from the Fallopian tube tissues of Lats1+/+Lats2+/+ and Lats1fl/f/Lats2fl/fl mice were infected with Cre-encoding adenovirus and subjected to immunohistochemistry (l), immunoblot (m) and qPCR analysis (n). Scale bar, 200 μm for bright field, 25 μm for immunohistochemistry staining. ***P < 0.001; mean + s.d.. Gel source data are provided in Supplementary Fig 1.

Source data

Extended Data Fig. 2 YAP and TAZ mediate Hippo signalling to repress expression of ESR1.

a, Repression of levels of ERα by TAZ. MCF-7 cells transduced with a control vector, or cDNA encoding Flag–TAZ(4SA) or Flag–TAZ(4SA/S51A), were subjected to immunoblot. TAZ(4SA) is a constitutively active mutant with mutation of the four LATS1/2 phosphorylation sites, and TAZ(4SA/S51A) is defective in TEAD binding. b, c, YAP reduces levels of ERα in additional ER+ breast cancer cell lines. T47D (b) and ZR-75-1 (c) cells transduced with a control vector, Flag–YAP(5SA) or Flag–YAP(5SA/S94A) were analysed by immunoblot. d, The TEAD-binding, but not WW, domain of YAP is essential for CTGF induction. e, f, YAP and TAZ have redundant roles in repressing ERα expression. Wild-type, YAP knockout, TAZ knockout or YAP and TAZ double-knockout MCF-7 cells were subjected to immunoblot (e) or qPCR for ESR1 (f). g, YAP and TAZ double knockout increases expression of the ERα target-genes TFF1 and GREB1. h, i, NF2 deficiency decreases ERα expression, YAP phosphorylation and LATS1 phosphorylation, without affecting LATS1 protein. Wild-type, or three independent clones of NF2-null MCF-7 cells were subjected to immunoblot (h) or qPCR (i). ***P < 0.001; mean + s.d., n.s., not significant. Gel source data are provided in Supplementary Fig 1.

Source data

Extended Data Fig. 3 Deletion of LATS1/2 inhibits growth of ER+ breast cancer cells.

a, Image representative of six biologically independent xenografts for Fig. 2h. Scale bar, 10 mm. bd, LATS1/2 deficiency inhibits tumour cell proliferation and promotes apoptosis in vivo. Representative images (b) and analysis (c, d) of ERα, phosphorylated histone H3 (S10) and cleaved caspase 3 immunostaining in LATS1/2-deficient and wild-type MCF-7 xenografts. Scale bar, 100 μm. Box plots indicate median and interquartile range.

Source data

Supplementary information

Supplementary Figure 1

This file contains the uncropped western blot source images.

Reporting Summary

Source data

Rights and permissions

Reprints and permissions

About this article

Check for updates. Verify currency and authenticity via CrossMark

Cite this article

Ma, S., Wu, Z., Yang, F. et al. Hippo signalling maintains ER expression and ER+ breast cancer growth. Nature 591, E1–E10 (2021). https://doi.org/10.1038/s41586-020-03131-5

Download citation

  • Received:

  • Accepted:

  • Published:

  • Issue Date:

  • DOI: https://doi.org/10.1038/s41586-020-03131-5

This article is cited by

Comments

By submitting a comment you agree to abide by our Terms and Community Guidelines. If you find something abusive or that does not comply with our terms or guidelines please flag it as inappropriate.

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