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:

YAP induces an oncogenic transcriptional program through TET1-mediated epigenetic remodeling in liver growth and tumorigenesis

Abstract

Epigenetic remodeling is essential for oncogene-induced cellular transformation and malignancy. In contrast to histone post-translational modifications, how DNA methylation is remodeled by oncogenic signaling remains poorly understood. The oncoprotein YAP, a coactivator of the TEAD transcription factors mediating Hippo signaling, is widely activated in human cancers. Here, we identify the 5-methylcytosine dioxygenase TET1 as a direct YAP target and a master regulator that coordinates the genome-wide epigenetic and transcriptional reprogramming of YAP target genes in the liver. YAP activation induces the expression of TET1, which physically interacts with TEAD to cause regional DNA demethylation, histone H3K27 acetylation and chromatin opening in YAP target genes to facilitate transcriptional activation. Loss of TET1 not only reverses YAP-induced epigenetic and transcriptional changes but also suppresses YAP-induced hepatomegaly and tumorigenesis. These findings exemplify how oncogenic signaling regulates the site specificity of DNA demethylation to promote tumorigenesis and implicate TET1 as a potential target for modulating YAP signaling in physiology and disease.

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

Fig. 1: Tet1 is a direct target of the YAP–TEAD complex.
Fig. 2: YAP activation causes regional DNA demethylation in enhancers.
Fig. 3: TET1 interacts with transcription factor TEAD4 to cause regional histone acetylation and chromatin opening.
Fig. 4: Loss of Tet1 suppresses YAP-induced hepatomegaly and transcriptional activation.
Fig. 5: Tet1 deletion suppresses YAP-induced hepatomegaly and tumorigenesis in diverse genetic models of YAP activation.
Fig. 6: YAP activation drives TEAD4 expression through TET1-dependent epigenetic remodeling.
Fig. 7: Drug-induced DNA demethylation partially compensates for TET1 function in YAPTg Tet1 -/- livers.

Similar content being viewed by others

Data availability

RNA-seq, MBD–seq and ChIP–seq data that support the findings of this study have been deposited in the Gene Expression Omnibus (GEO) under accession number GSE178227. Source data are provided with this paper. All other data and reagents supporting the findings of this study are available from the corresponding author on reasonable request.

References

  1. Baylin, S. B. & Jones, P. A. Epigenetic determinants of cancer. Cold Spring Harb. Perspect. Biol. 8, a019505 (2016).

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  2. Suva, M. L., Riggi, N. & Bernstein, B. E. Epigenetic reprogramming in cancer. Science 339, 1567–1570 (2013).

    Article  CAS  PubMed  Google Scholar 

  3. Allis, C. D. & Jenuwein, T. The molecular hallmarks of epigenetic control. Nat. Rev. Genet. 17, 487–500 (2016).

    Article  CAS  PubMed  Google Scholar 

  4. Suganuma, T. & Workman, J. L. Signals and combinatorial functions of histone modifications. Annu. Rev. Biochem. 80, 473–499 (2011).

    Article  CAS  PubMed  Google Scholar 

  5. Audia, J. E. & Campbell, R. M. Histone modifications and cancer. Cold Spring Harb. Perspect. Biol. 8, a019521 (2016).

    Article  PubMed  PubMed Central  Google Scholar 

  6. Rasmussen, K. D. & Helin, K. Role of TET enzymes in DNA methylation, development, and cancer. Genes Dev. 30, 733–750 (2016).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  7. Wu, X. & Zhang, Y. TET-mediated active DNA demethylation: mechanism, function and beyond. Nat. Rev. Genet. 18, 517–534 (2017).

    Article  CAS  PubMed  Google Scholar 

  8. Jones, P. A. & Baylin, S. B. The fundamental role of epigenetic events in cancer. Nat. Rev. Genet. 3, 415–428 (2002).

    Article  CAS  PubMed  Google Scholar 

  9. Jones, P. A. & Baylin, S. B. The epigenomics of cancer. Cell 128, 683–692 (2007).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  10. Robertson, K. D. DNA methylation and human disease. Nat. Rev. Genet. 6, 597–610 (2005).

    Article  CAS  PubMed  Google Scholar 

  11. Issa, J. P. CpG island methylator phenotype in cancer. Nat. Rev. Cancer 4, 988–993 (2004).

    Article  CAS  PubMed  Google Scholar 

  12. Belinsky, S. A. Gene-promoter hypermethylation as a biomarker in lung cancer. Nat. Rev. Cancer 4, 707–717 (2004).

    Article  CAS  PubMed  Google Scholar 

  13. Greenberg, M. V. C. & Bourc’his, D. The diverse roles of DNA methylation in mammalian development and disease. Nat. Rev. Mol. Cell Biol. 20, 590–607 (2019).

    Article  CAS  PubMed  Google Scholar 

  14. Bestor, T. H. The DNA methyltransferases of mammals. Hum. Mol. Genet. 9, 2395–2402 (2000).

    Article  CAS  PubMed  Google Scholar 

  15. Pastor, W. A., Aravind, L. & Rao, A. TETonic shift: biological roles of TET proteins in DNA demethylation and transcription. Nat. Rev. Mol. Cell Biol. 14, 341–356 (2013).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  16. Kohli, R. M. & Zhang, Y. TET enzymes, TDG and the dynamics of DNA demethylation. Nature 502, 472–479 (2013).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  17. Tahiliani, M. et al. Conversion of 5-methylcytosine to 5-hydroxymethylcytosine in mammalian DNA by MLL partner TET1. Science 324, 930–935 (2009).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  18. Ito, S. et al. Role of Tet proteins in 5mC to 5hmC conversion, ES-cell self-renewal and inner cell mass specification. Nature 466, 1129–1133 (2010).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  19. Ito, S. et al. Tet proteins can convert 5-methylcytosine to 5-formylcytosine and 5-carboxylcytosine. Science 333, 1300–1303 (2011).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  20. Dawlaty, M. M. et al. Tet1 is dispensable for maintaining pluripotency and its loss is compatible with embryonic and postnatal development. Cell Stem Cell 9, 166–175 (2011).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  21. Ficz, G. et al. Dynamic regulation of 5-hydroxymethylcytosine in mouse ES cells and during differentiation. Nature 473, 398–402 (2011).

    Article  CAS  PubMed  Google Scholar 

  22. Koh, K. P. et al. Tet1 and Tet2 regulate 5-hydroxymethylcytosine production and cell lineage specification in mouse embryonic stem cells. Cell Stem Cell 8, 200–213 (2011).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  23. Zhang, R. R. et al. Tet1 regulates adult hippocampal neurogenesis and cognition. Cell Stem Cell 13, 237–245 (2013).

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  24. Watanabe, K. et al. Emergence of the dedifferentiated phenotype in hepatocyte-derived tumors in mice: roles of oncogene-induced epigenetic alterations. Hepatol. Commun. 3, 697–715 (2019).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  25. Shirai, K. et al. TET1 upregulation drives cancer cell growth through aberrant enhancer hydroxymethylation of HMGA2 in hepatocellular carcinoma. Cancer Sci. 112, 2855–2869 (2021).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  26. Bai, X. et al. Ten-eleven translocation 1 promotes malignant progression of cholangiocarcinoma with wild-type isocitrate dehydrogenase 1. Hepatology 73, 1747–1763 (2021).

    Article  CAS  PubMed  Google Scholar 

  27. Good, C. R. et al. TET1-mediated hypomethylation activates oncogenic signaling in triple-negative breast cancer. Cancer Res. 78, 4126–4137 (2018).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  28. Filipczak, P. T. et al. p53-suppressed oncogene TET1 prevents cellular aging in lung cancer. Cancer Res. 79, 1758–1768 (2019).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  29. Takai, H. et al. 5-Hydroxymethylcytosine plays a critical role in glioblastomagenesis by recruiting the CHTOP-methylosome complex. Cell Rep. 9, 48–60 (2014).

    Article  CAS  PubMed  Google Scholar 

  30. Huang, H. et al. TET1 plays an essential oncogenic role in MLL-rearranged leukemia. Proc. Natl Acad. Sci. USA 110, 11994–11999 (2013).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  31. Halder, G. & Johnson, R. L. Hippo signaling: growth control and beyond. Development 138, 9–22 (2011).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  32. 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  PubMed  PubMed Central  Google Scholar 

  33. Yimlamai, D., Fowl, B. H. & Camargo, F. D. Emerging evidence on the role of the Hippo/YAP pathway in liver physiology and cancer. J. Hepatol. 63, 1491–1501 (2015).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  34. Zanconato, F., Cordenonsi, M. & Piccolo, S. YAP/TAZ at the roots of cancer. Cancer Cell 29, 783–803 (2016).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

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

    Article  CAS  PubMed  PubMed Central  Google Scholar 

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

    Article  CAS  PubMed  Google Scholar 

  37. Moya, I. M. & Halder, G. Hippo-YAP/TAZ signalling in organ regeneration and regenerative medicine. Nat. Rev. Mol. Cell Biol. 20, 211–226 (2019).

    Article  CAS  PubMed  Google Scholar 

  38. Patel, S. H., Camargo, F. D. & Yimlamai, D. Hippo signaling in the liver regulates organ size, cell fate, and carcinogenesis. Gastroenterology 152, 533–545 (2017).

    Article  CAS  PubMed  Google Scholar 

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

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  40. Sanchez-Vega, F. et al. Oncogenic signaling pathways in the cancer genome atlas. Cell 173, 321–337.e10 (2018).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  41. Prior, N. et al. Lgr5+ stem and progenitor cells reside at the apex of a heterogeneous embryonic hepatoblast pool. Development 146, dev174557 (2019).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  42. 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  PubMed  PubMed Central  Google Scholar 

  43. Yimlamai, D. et al. Hippo pathway activity influences liver cell fate. Cell 157, 1324–1338 (2014).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  44. Zhang, N. et al. The Merlin/NF2 tumor suppressor functions through the YAP oncoprotein to regulate tissue homeostasis in mammals. Dev. Cell 19, 27–38 (2010).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  45. Baskin-Bey, E. S. et al. Constitutive androstane receptor (CAR) ligand, TCPOBOP, attenuates Fas-induced murine liver injury by altering Bcl-2 proteins. Hepatology 44, 252–262 (2006).

    Article  CAS  PubMed  Google Scholar 

  46. Huang, Y. et al. The behaviour of 5-hydroxymethylcytosine in bisulfite sequencing. PLoS ONE 5, e8888 (2010).

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  47. Yu, M. et al. Base-resolution analysis of 5-hydroxymethylcytosine in the mammalian genome. Cell 149, 1368–1380 (2012).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  48. Davis, C. A. et al. The Encyclopedia of DNA Elements (ENCODE): data portal update. Nucleic Acids Res. 46, D794–D801 (2018).

    Article  CAS  PubMed  Google Scholar 

  49. Zanconato, F. et al. Genome-wide association between YAP/TAZ/TEAD and AP-1 at enhancers drives oncogenic growth. Nat. Cell Biol. 17, 1218–1227 (2015).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  50. Yagi, R., Chen, L. F., Shigesada, K., Murakami, Y. & Ito, Y. A WW domain-containing yes-associated protein (YAP) is a novel transcriptional co-activator. EMBO J. 18, 2551–2562 (1999).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  51. Stein, C. et al. YAP1 exerts its transcriptional control via TEAD-mediated activation of enhancers. PLoS Genet. 11, e1005465 (2015).

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  52. Vassilev, A., Kaneko, K. J., Shu, H., Zhao, Y. & DePamphilis, M. L. TEAD/TEF transcription factors utilize the activation domain of YAP65, a Src/Yes-associated protein localized in the cytoplasm. Genes Dev. 15, 1229–1241 (2001).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  53. Suzuki, T. et al. RUNX1 regulates site specificity of DNA demethylation by recruitment of DNA demethylation machineries in hematopoietic cells. Blood Adv. 1, 1699–1711 (2017).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

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

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  55. Zhang, W. et al. Isoform switch of TET1 regulates DNA demethylation and mouse development. Mol. Cell 64, 1062–1073 (2016).

    Article  CAS  PubMed  Google Scholar 

  56. Yuan, W. C. et al. NUAK2 is a critical YAP target in liver cancer. Nat. Commun. 9, 4834 (2018).

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  57. Simon, J. M., Giresi, P. G., Davis, I. J. & Lieb, J. D. Using formaldehyde-assisted isolation of regulatory elements (FAIRE) to isolate active regulatory DNA. Nat. Protoc. 7, 256–267 (2012).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  58. Tsai, H. C. et al. Transient low doses of DNA-demethylating agents exert durable antitumor effects on hematological and epithelial tumor cells. Cancer Cell 21, 430–446 (2012).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  59. Cheng, Y. et al. Targeting epigenetic regulators for cancer therapy: mechanisms and advances in clinical trials. Signal Transduct. Target. Ther. 4, 62 (2019).

    Article  PubMed  PubMed Central  Google Scholar 

  60. Reilly, B. M. et al. 5-Azacytidine transiently restores dysregulated erythroid differentiation gene expression in TET2-deficient erythroleukemia cells. Mol. Cancer Res. 19, 451–464 (2021).

    Article  CAS  PubMed  Google Scholar 

  61. Oh, H. et al. Yorkie promotes transcription by recruiting a histone methyltransferase complex. Cell Rep. 8, 449–459 (2014).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  62. Qing, Y. et al. The Hippo effector Yorkie activates transcription by interacting with a histone methyltransferase complex through Ncoa6. eLife 3, e02564 (2014).

    Article  PubMed Central  Google Scholar 

  63. Chang, L. et al. The SWI/SNF complex is a mechanoregulated inhibitor of YAP and TAZ. Nature 563, 265–269 (2018).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  64. Skibinski, A. et al. The Hippo transducer TAZ interacts with the SWI/SNF complex to regulate breast epithelial lineage commitment. Cell Rep. 6, 1059–1072 (2014).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  65. Oh, H. et al. Genome-wide association of Yorkie with chromatin and chromatin-remodeling complexes. Cell Rep. 3, 309–318 (2013).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  66. Zanconato, F. et al. Transcriptional addiction in cancer cells is mediated by YAP/TAZ through BRD4. Nat. Med. 24, 1599–1610 (2018).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  67. Joshi, S. et al. TEAD transcription factors are required for normal primary myoblast differentiation in vitro and muscle regeneration in vivo. PLoS Genet. 13, e1006600 (2017).

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  68. Liu, X. et al. Tead and AP1 coordinate transcription and motility. Cell Rep. 14, 1169–1180 (2016).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  69. Chen, Q. et al. Homeostatic control of Hippo signaling activity revealed by an endogenous activating mutation in YAP. Genes Dev. 29, 1285–1297 (2015).

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  70. Hamaratoglu, F. et al. The tumour-suppressor genes NF2/Merlin and Expanded act through Hippo signalling to regulate cell proliferation and apoptosis. Nat. Cell Biol. 8, 27–36 (2006).

    Article  CAS  PubMed  Google Scholar 

  71. Genevet, A., Wehr, M. C., Brain, R., Thompson, B. J. & Tapon, N. Kibra is a regulator of the Salvador/Warts/Hippo signaling network. Dev. Cell 18, 300–308 (2010).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  72. Aloia, L. et al. Epigenetic remodelling licences adult cholangiocytes for organoid formation and liver regeneration. Nat. Cell Biol. 21, 1321–1333 (2019).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  73. Driskill, J. H. et al. WWTR1(TAZ)-CAMTA1 reprograms endothelial cells to drive epithelioid hemangioendothelioma. Genes Dev. 35, 495–511 (2021).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  74. Sohni, A. et al. Dynamic switching of active promoter and enhancer domains regulates Tet1 and Tet2 expression during cell state transitions between pluripotency and differentiation. Mol. Cell. Biol. 35, 1026–1042 (2015).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  75. Chandru, A., Bate, N., Vuister, G. W. & Cowley, S. M. Sin3A recruits Tet1 to the PAH1 domain via a highly conserved Sin3-interaction domain. Sci. Rep. 8, 14689 (2018).

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  76. Jin, C. et al. TET1 is a maintenance DNA demethylase that prevents methylation spreading in differentiated cells. Nucleic Acids Res. 42, 6956–6971 (2014).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  77. Ko, M. et al. Impaired hydroxylation of 5-methylcytosine in myeloid cancers with mutant TET2. Nature 468, 839–843 (2010).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  78. Ko, M. et al. Modulation of TET2 expression and 5-methylcytosine oxidation by the CXXC domain protein IDAX. Nature 497, 122–126 (2013).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  79. Li, Z. et al. Structural insights into the YAP and TEAD complex. Genes Dev. 24, 235–240 (2010).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  80. Zuber, J. et al. Toolkit for evaluating genes required for proliferation and survival using tetracycline-regulated RNAi. Nat. Biotechnol. 29, 79–83 (2011).

    Article  CAS  PubMed  Google Scholar 

  81. Ge, S. X., Son, E. W. & Yao, R. iDEP: an integrated web application for differential expression and pathway analysis of RNA-Seq data. BMC Bioinformatics 19, 534 (2018).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  82. Tang, Z., Kang, B., Li, C., Chen, T. & Zhang, Z. GEPIA2: an enhanced web server for large-scale expression profiling and interactive analysis. Nucleic Acids Res. 47, W556–W560 (2019).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  83. Tang, Z. et al. GEPIA: a web server for cancer and normal gene expression profiling and interactive analyses. Nucleic Acids Res. 45, W98–W102 (2017).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  84. McLean, C. Y. et al. GREAT improves functional interpretation of cis-regulatory regions. Nat. Biotechnol. 28, 495–501 (2010).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  85. Yu, G., Wang, L. G. & He, Q. Y. ChIPseeker: an R/Bioconductor package for ChIP peak annotation, comparison and visualization. Bioinformatics 31, 2382–2383 (2015).

    Article  CAS  PubMed  Google Scholar 

  86. Thomas-Chollier, M. et al. A complete workflow for the analysis of full-size ChIP-seq (and similar) data sets using peak-motifs. Nat. Protoc. 7, 1551–1568 (2012).

    Article  CAS  PubMed  Google Scholar 

  87. Leporcq, C. et al. TFmotifView: a webserver for the visualization of transcription factor motifs in genomic regions. Nucleic Acids Res. 48, W208–W217 (2020).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  88. 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  PubMed  PubMed Central  Google Scholar 

Download references

Acknowledgements

We thank the McDermott Sequencing Core, the Next Generation Sequencing Core, the Bioinformatics Lab, the Animal Resource Center and the Transgenic Core Facility at University of Texas Southwestern Medical Center for assistance with this work. This work was supported in part by grants from the National Institutes of Health (EY015708 to D.P. and R35GM136316 to E.H.C.) and Department of Defense (PR190360 to D.P.). D.P. is an investigator of the Howard Hughes Medical Institute.

Author information

Authors and Affiliations

Authors

Contributions

B.W. and D.P. conceived the study. B.W. performed experiments. B.W., S.M. and E.H.C. contributed to the bioinformatic data analysis. B.W., Y.Z. and D.P. wrote the paper.

Corresponding author

Correspondence to Duojia Pan.

Ethics declarations

Competing interests

The authors declare no competing interests.

Peer review

Peer review information

Nature Genetics thanks Yi Zhang, Georg Halder and the other, anonymous, reviewer(s) for their contribution to the peer review of this work.

Additional information

Publisher’s note Springer Nature remains neutral with regard to jurisdictional claims in published maps and institutional affiliations.

Extended data

Extended Data Fig. 1 YAP activation induces TET1 expression.

a, RNA-seq analysis showing the expression of canonical YAP target genes, YAP upstream tumor suppressor genes and DNA methylation-related genes in induced YAPTg compared to control livers. Mice were treated with 50 mg/l Dox for 10 days starting at 1 month of age. b, Representative immunostaining of livers from induced YAPTg and Mst1/2 mutant mice at 1 month of age (n = 4), showing the induction of the hepatoblast marker AFP, but not the bile duct marker CK19, in YAP-activated hepatocytes. Also note the normal expression of the hepatocyte markers ALB and HNF4A in the proliferating hepatocytes (PH3). White scale bar, 100 μm. c, Heatmap of representative gene expression, derived from the published microarray data from YAPS127A transgenic livers after 7 days Dox induction. Note the induction of Tet1 and hepatoblast marker (Afp), but not bile duct markers (Krt7 and Krt19), in YAPS127A transgenic livers. d-e, RT-qPCR of Tet1 mRNA level in Yap mutant livers at neonatal P0.5 (n = 6) (d) and active mutant TAZ4SA transgenic (TAZ4SATg) livers (CTL n = 3; TAZ4SATg n = 4) which were treated with 50 mg/l Dox for 3 days starting at 1 month of age (e). Tet1 mRNA level was normalized to control livers. f, Western blot of TET1 protein in Mst1/2 mutant livers. Arrowhead marks the specific TET1 protein band. Images are representative of three independent experiments. g, Genomic tracks displaying MBD-seq, TET1 ChIP-seq, TEAD4 ChIP-seq and H3K27ac ChIP-seq reads at TET1 promoter. h, Genomic tracks displaying TEAD4 ChIP-seq reads at TET1 promoter in human embryonic stem cells (H1-hESC, green) and in mouse trophoblast stem cells (mTS, red). i, ChIP-qPCR at Tet1 promoter in Mst1/2 mutant livers (n = 3). Values represent mean ± s.e.m. (d, e, i). P values are calculated with unpaired two-tailed Student’s t-test (d, e, i).

Source data

Extended Data Fig. 2 YAP and TET1 are dispensable for TCP-induced hepatomegaly.

a, Representative gross image of livers (top panel) and quantification of liver-to-body weight ratio (bottom panel) from vehicle-treated (n = 8) and TCP-treated (n = 7) mice. 1-month-old mice were injected intraperitoneally with a single dose of 3 mg/kg TCP. Mice were sacrificed 7 days post injection. b, c, RT-qPCR showing mRNA levels of the YAP downstream target genes (b) or upstream suppressor genes (c) in TCP-treated livers. mRNA levels were normalized to vehicle-treated control livers (n = 4). d, Representative gross image of livers (top panel) and quantification of liver-to-body weight ratio (bottom panel) from 1-month-old Yap mutant mice (CTL n = 12; Yap/ n = 13) subjected to TCP-induced hepatomegaly. e, MBD-qPCR of methylation levels at the indicated rDMRs in TCP-treated mouse livers. Methylation levels were normalized to vehicle-treated control livers (n = 3). f, Representative gross image of livers (top panel) and quantification of liver-to-body weight ratio (bottom panel) from 1-month-old Tet1 mutant mice (CTL n = 10; Tet1/ n = 15) subjected to TCP-induced hepatomegaly. Values represent mean ± s.e.m. (a, b, c, d, e, f). P values are calculated with unpaired two-tailed Student’s t-test (a, b, c, d, e, f).

Source data

Extended Data Fig. 3 Elevated YAP/TAZ and TET1 abundance in foregut cancers shows a significant and positive correlation.

a, Elevated expression of YAP and TAZ in five human cancers (LIHC, CHOL, ESCA, PAAD and STAD) obtained from the TCGA database. The center lines show the medians, the box limits mark the 25th to the 75th percentiles and the whiskers indicate 1.5× the interquartile range. b, c, Spearman’s correlation coefficient analysis between the expression of YAP/TAZ and TET1 in LIHC, CHOL, ESCA, PAAD, STAD (b), LGG and GBM (c). d, Elevated expression of TEAD1 and TEAD4 in human foregut cancers (LIHC, CHOL, ESCA, PAAD and STAD) obtained from the TCGA database. e, Spearman’s correlation coefficient analysis between the expression of YAP/TAZ and TEAD1 (left panel) or TEAD4 (right panel) in human foregut cancers (LIHC, CHOL, ESCA, PAAD and STAD) obtained from the TCGA database. f, Kaplan-Meier analysis of overall survival in patients with human foregut cancers (LIHC, CHOL, ESCA, PAAD and STAD) according to high or low mRNA expression of YAP/TAZ, TET1, TEAD1 or TEAD4. P values are calculated with two-tailed Spearman’s correlation test (b, c, e) or two-sided log-rank test (f).

Extended Data Fig. 4 YAP activation causes regional DNA demethylation in canonical YAP target genes.

a, DNA dot blot of genomic 5mC and 5hmC levels in 1-month-old Mst1/2 mutant livers (top panels). The blot was then stained with methylene blue as loading control (bottom panels). Images are representative of two independent experiments. b, 5mC and 5hmC validation at the indicated rDMRs in Mst1/2 mutant livers. Methylation (top panel) and hydroxymethylation (bottom panel) levels were determined by MBD-qPCR and hMeDIP-qPCR, respectively (n = 4). Values represent mean ± s.e.m. P values are calculated with unpaired two-tailed Student’s t-test. c, Genomic tracks displaying MBD-seq and H3K4me1 ChIP-seq reads at the rDMR loci of the indicated YAP target genes. Grey columns represent YAP-induced rDMRs. The cloning-based locus-specific bisulfite sequencing sites are also marked. Note the robust enhancer mark H3K4me1 occupancy at the YAP-induced rDMRs. d, Cloning-based traditional bisulfite sequencing (left panels) and TAB-seq (right panels) of CpG sites at the rDMR loci of YAP target genes. All sequencing results include 10 independent clones.

Source data

Extended Data Fig. 5 YAP activation causes regional DNA demethylation in enhancers.

a, Pie charts showing the distribution of methylation peaks and YAP-induced rDMRs to mm10 mouse genomic features. b, Average peak profile of methylation peaks and YAP-induced rDMRs mapped to nearest TSS. Note the disassociation of YAP-induced rDMRs from TSS. c, The de novo motif analysis of YAP-induced rDMRs. Both RUNX and TEAD recognition motifs were identified in the top 10 enriched motifs. d, e, Enrichment profiles representing the TEAD motif centered at YAP-induced rDMRs (d) and TET1 peaks (e). f, Enrichment profiles representing the TET1, TEAD4 and H3K27ac ChIP-seq reads centered at YAP-induced rDMRs.

Extended Data Fig. 6 TEAD4 interacts with TET1 through its YAP binding domain (YBD).

a, Co-IP assay showing physical interactions between TET1 and RUNX1 or TEAD1/TEAD4. The indicated constructs were transfected into HEK293T cells, lysed and subjected to immunoprecipitation assays. Normal IgG immunoprecipitates serve as negative control. b, Co-IP assay showing no interactions between TET1 and YAP. Co-IP assay was conducted under same condition as in (a). c, Co-IP assay showing physical interactions between TET1 and RUNX1 or TEAD1/TEAD4 in the presence or absence of DNA/RNA nuclease. The indicated constructs were transfected into HEK293T cells, lysed with or without benzonase treatment before immunoprecipitation assays. Note the co-IP between TET1 and RUNX1 or TEAD1/TEAD4, regardless of nuclease treatment. d, Co-IP assay showing physical interactions between TET1 and TEAD1/TEAD4. Note that the TET1-TEAD1/TEAD4 interaction were abolished by deletion of the CXXC domain in TET1 protein. e, Co-IP assay showing physical interactions between TEAD4 and TET1. The schematic diagram indicates the TEAD4 mutants used: TEAD4ΔΤΕΑ is defective in DNA-binding and TEAD4-Y429H is defective in YAP-binding. Both TEAD4 mutants interacted with TET1. f, Co-IP assay showing physical interactions between TEAD4 and TET1. Note that TEAD4 interacts with TET1 through its YAP binding domain (YBD). g, Co-IP assay showing physical interactions between TEAD4 and TET1/2/3. Note the co-IP between TEAD4 and TET1, not TET2 or TET3. h, Co-IP assay showing physical interactions between endogenous TET1 and TEAD1 in YAPTg livers. i, Genome track showing Tet1 RNA-seq in YAPTg livers. j, TET1 exon usage profile in LIHC samples from the TCGA database. Exon usage profile was visualized with TSVdb web tool. Patients are represents in x-axis and exon usage of each exon are showed in y-axis. Images are representative of two (a, b, c, g) or three (d, e, f, h) independent experiments.

Source data

Extended Data Fig. 7 TET1 interacts with TEAD4 transcription factor to cause regional histone acetylation and chromatin opening at the rDMRs of canonical YAP target genes.

a, Validation of TEAD4 and TET1 antibody specificity. Western blot of TEAD4 or TET1 protein in HEK293T cells with non-targeting control (siCon) or siRNA targeting TEAD4 (siTEAD4) or siRNA targeting TET1 (siTET1) treatment. Images are representative of two independent experiments. b, Enrichment profiles representing the TET1 ChIP-seq reads from control and YAPTg livers centered at TET1 peaks (left panel) and the TEAD4 ChIP-seq reads from control and YAPTg livers centered at TEAD4 peaks (right panel). c, Spearman’s correlation coefficient analysis using the signals of peaks between H3K27ac and TET1 or TEAD4 from the identified 8,022 TET1/TEAD4/H3K27ac overlapped peaks. d, Genomic tracks displaying MBD-seq and ChIP-seq reads at the rDMR loci of Cyr61 and Ctgf. Grey columns represent YAP-induced rDMR regions. e, ChIP-qPCR at rDMRs in YAPTg livers (n = 3). f, g, ChIP-qPCR (n = 3) (f) and FAIRE-qPCR (n = 4) (g) at rDMRs in Mst1/2 mutant livers. h, Luciferase reporter assay showing enhancer activity of unmethylated and methylated Myc and Cyr61 rDMRs of three independent experiments. Reporter construct containing respective rDMRs with or without in vitro methylation was co-transfected with YAP-expressing vector into HEK293T cells. Luciferase activity was normalized to unmethylated rDMR. i, Luciferase reporter assay showing enhancer activity of methylated Myc or Cyr61 rDMR with non-targeting control (siCon) or siRNA targeting TET1 (siTET1) of three independent experiments. Reporter construct containing methylated rDMR was co-transfected with YAP-expressing vector together with either non-targeting control or siRNA targeting TET1 into HEK293T cells. Luciferase activity was normalized to non-targeting control. Values represent mean ± s.e.m. (e, f, g, h, i). P values are calculated with unpaired two-tailed Student’s t-test (e, f, g, h, i) or two-tailed Spearman’s correlation test (c).

Source data

Extended Data Fig. 8 Loss of Tet1 suppresses YAP-induced hepatomegaly and tumorigenesis.

a, Normal liver function in Tet1 mutant mice, as indicated by similar serum levels of ALB, AST and ALT compared to control mice (n = 5). b, c, Representative H&E and Ki-67 staining of liver sections (b) and quantification of Ki-67 positive cells (c) from the indicated mice (n = 4) treated with Dox for 10 days starting at 1 month of age. d, Quantification of Ki-67 positive cells in YAPTg livers treated with Tet1 shRNA lentivirus (n = 4). e, Principal components analysis (PCA) for transcriptome profiles from the indicated mice (n = 3). f, A volcano plot showing differential expression of 2,963 YAP-upregulated genes in YAPTg Tet1/ compared to YAPTg livers. Red dots represent remarkable (fold change ≥ 2) and significant (P < 0.00001) genes. g, i, Representative gross image and H&E staining of livers (g) and quantification of liver-to-body weight ratio (i) from CTL (n = 7), Tet1−/− (n = 9), YAPTg (n = 13) and YAPTg Tet1−/− (n = 18) mice treated with Dox starting at birth for ~2 months. h, Kaplan-Meier survival curve showing the survival of the indicated mice treated with Dox starting at birth, monitored for 120 days. j, Quantification of LIHC incidence under same condition as in (g). k, l, Representative gross image and H&E staining of livers (k) and quantification of liver-to-body weight ratio (l) from CTL (n = 6), Tet1−/− (n = 6), YAPTg (n = 22) and YAPTg Tet1−/− (n = 14) mice at ~14 months of age without Dox treatment. m, Quantification of LIHC incidence under same condition as in (k). Values represent mean ± s.e.m. (a, c, d, i, l). P values are calculated with unpaired two-tailed Student’s t-test (a) or one-way ANOVA with Tukey’s test (c, d, i, l).

Source data

Extended Data Fig. 9 TET1-mediated DNA methylation is required for transcriptional induction of TEAD1/4.

a, Clustered heatmaps of MBD-seq data (top panel) and RNA-seq data (bottom panel) displaying sample-to-sample distance between the indicated genetic background. b, Methylation profile representing the MBD-seq signals from the indicated mutant livers centered at TET1 peaks. c, Absolute distance of methylation peaks, YAP-induced rDMRs and TET1-dependent rDMRs relative to nearest TSS. d, Genomic distribution of methylation peaks, YAP-induced rDMRs and TET1-dependent rDMRs. e, Average peak profile of methylation peaks, YAP-induced rDMRs and TET1-dependent rDMRs mapped to nearest TSS. f, Genomic tracks displaying MBD-seq, TET1 ChIP-seq, TEAD4 ChIP-seq, H3K4me1 ChIP-seq and H3K27ac ChIP-seq reads at the Tead1 rDMR locus. Grey column represents a YAP-induced rDMR region. g, Cloning-based traditional bisulfite sequencing (left panels) and TAB-seq (right panels) of 16 CpG sites at the Tead1 rDMR locus. All sequencing results include 10 independent clones. h, 5mC and 5hmC validation at the Tead1 and Tead4 rDMRs in the indicated mutant livers. Methylation (top panel) and hydroxymethylation (bottom panel) levels were determined by MBD-qPCR and hMeDIP-qPCR, respectively (n = 4). i, RT-qPCR of Tead1 and Tead4 in the indicated mutant livers. mRNA level was normalized to control livers (n = 6). j, RT-qPCR of Tead1 and Tead4 in the indicated 1-month-old mutant livers (CTL n = 5, 5, 6 and KO n = 7, 6, 6 from left to right). mRNA level was normalized to control livers. k, Western blot of TEAD1 and TEAD4 protein in Mst1/2 mutant livers. Images are representative of three independent experiments. l, Genomic tracks displaying TEAD4 ChIP-seq reads at TEAD1 and TEAD4 genes in human embryonic stem cells (H1-hESC).Values represent mean ± s.e.m. (h, i, j). P values are calculated with unpaired two-tailed Student’s t-test (j) or one-way ANOVA with Tukey’s test (h, i).

Source data

Extended Data Fig. 10 YAP directly induces the transcription of Tead1 and Tead4 in a TET1-dependent manner.

a, b, ChIP-qPCR (n = 3) (a) and FAIRE-qPCR (n = 3) (b) at rDMRs in the indicated mutant livers (also see Fig. 3l and Extended Data Fig. 7e). c, d, ChIP-qPCR (n = 3) (c) and FAIRE-qPCR (n =4) (d) at Tead1 and Tead4 rDMRs in Mst1/2 mutant livers. e, A volcano plot for differential DNA methylation of 1,376 rDMRs sites (associated with 1,003 TET1-dependent YAP target genes) in DAC-treated compared to untreated YAPTg Tet1/ livers. Red dots represent remarkable (fold change ≥ 2) and significant (P < 0.1) genes. Note that 27.4% of rDMRs (377 out of 1,376) were DNA demethylation by DAC treatment. f, Heatmaps of RNA-seq (left panel) and MBD-seq (right panel) data displaying changes of 1,003 TET1-dependent YAP target genes between the indicated mice, ranked based on P values (between YAPTg Tet1/ and YAPTg Tet1/ /DAC) with the most significant on top. g, Schematic model showing a YAP-TET1-TEAD positive feedforward loop sustains YAP signaling. In control livers, the YAP-TEAD complex has limited access to the promoter and enhancer of YAP target genes due to closed chromatin. In YAP-activated livers, YAP-induced expression of TET1 protein causes regional DNA demethylation and local chromatin opening to facilitate YAP-TEAD binding. As a transcriptional coactivator, YAP then recruits histone-modifying enzymes such as NCOA6 and P300, as well as chromatin remodelers such as SWI/SNF complex, to further increase chromatin opening. This further facilitates the association of the YAP-TEAD complex to YAP target genes to activate their transcription. Induction of Tead1 and Tead4 by the YAP-TEAD complex constitutes a feedforward loop to amplify YAP signaling. Values represent mean ± s.e.m. (a, b, c, d). P values are calculated with unpaired two-tailed Student’s t-test (c, d) or one-way ANOVA with Tukey’s test (a, b).

Source data

Supplementary information

Source data

Source Data Fig. 1

Statistical source data.

Source Data Fig. 1

Unprocessed western blots.

Source Data Fig. 2

Statistical source data.

Source Data Fig. 2

Unprocessed western blots.

Source Data Fig. 3

Statistical source data.

Source Data Fig. 3

Unprocessed western blots.

Source Data Fig. 4

Statistical source data.

Source Data Fig. 5

Statistical source data.

Source Data Fig. 6

Unprocessed western blots.

Source Data Fig. 6

Statistical source data.

Source Data Fig. 7

Statistical Source Data.

Source Data Extended Data Fig. 1

Statistical source data.

Source Data Extended Data Fig. 1

Unprocessed western blots.

Source Data Extended Data Fig. 2

Statistical source data.

Source Data Extended Data Fig. 4

Statistical source data.

Source Data Extended Data Fig. 4

Unprocessed western blots.

Source Data Extended Data Fig. 6

Unprocessed western blots.

Source Data Extended Data Fig. 7

Statistical source data.

Source Data Extended Data Fig. 7

Unprocessed western blots.

Source Data Extended Data Fig. 8

Statistical source data.

Source Data Extended Data Fig. 9

Statistical source data.

Source Data Extended Data Fig. 9

Unprocessed western blots.

Source Data Extended Data Fig. 10

Statistical source data.

Rights and permissions

Reprints and permissions

About this article

Check for updates. Verify currency and authenticity via CrossMark

Cite this article

Wu, BK., Mei, SC., Chen, E.H. et al. YAP induces an oncogenic transcriptional program through TET1-mediated epigenetic remodeling in liver growth and tumorigenesis. Nat Genet 54, 1202–1213 (2022). https://doi.org/10.1038/s41588-022-01119-7

Download citation

  • Received:

  • Accepted:

  • Published:

  • Issue Date:

  • DOI: https://doi.org/10.1038/s41588-022-01119-7

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