Skip to main content

This is a preprint.

It has not yet been peer reviewed by a journal.

The National Library of Medicine is running a pilot to include preprints that result from research funded by NIH in PMC and PubMed.

bioRxiv logoLink to bioRxiv
[Preprint]. 2024 Oct 3:2024.10.03.615022. [Version 1] doi: 10.1101/2024.10.03.615022

An improved TEAD dominant-negative protein inhibitor to study Hippo YAP1/TAZ-dependent transcription

Briana Branch 1,2, Yao Yuan 1, Mariastella Cascone 3, Francesco Raimondi 3, Ramiro Iglesias-Bartolome 1,*
PMCID: PMC11537315  PMID: 39502361

Abstract

Hippo signaling is one of the top pathways altered in human cancer, and intensive focus has been devoted to developing therapies targeting Hippo-dependent transcription mediated by YAP1 and TAZ interaction with TEAD proteins. However, a significant challenge in evaluating the efficacy of these approaches is the lack of models that can precisely characterize the consequences of TEAD inhibition. To address this gap, our laboratory developed a strategy that utilizes a fluorescently traceable, dominant-negative protein named TEADi. TEADi specifically blocks the nuclear interactions of TEAD with YAP1 and TAZ, enabling precise dissection of Hippo TEAD-dependent and independent effects on cell fate. In this study, we aimed to enhance TEADi effectiveness by altering post-transcriptional modification sites within its TEAD-binding domains (TBDs). We demonstrate that a D93E mutation in the YAP1 TBD significantly increases TEADi inhibitory capacity. Additionally, we find that TBDs derived from VGLL4 and YAP1 are insufficient to block TAZ-induced TEAD activity, revealing crucial differences in YAP1 and TAZ displacement mechanisms by dominant-negative TBDs. Structural differences in YAP1 and TAZ TBDs were also identified, which may contribute to the distinct binding of these proteins to TEAD. Our findings expand our understanding of TEAD regulation and highlight the potential of an optimized TEADi as a more potent, specific, and versatile tool for studying TEAD-transcriptional activity.


Hippo signaling is downstream from mechanosensing and receptor-mediated pathways that regulate cell fate, helping cells balance cell growth and differentiation13. At its core, Hippo signaling is a kinase cascade that results in the cytoplasmic retention and inactivation of YAP1 and its paralog TAZ (also known as WWTR1). YAP1 and TAZ interact with numerous effectors, although their regulatory and oncogenic functions are mainly mediated by binding the family of TEAD transcription factors (TEAD1 to 4)46.

The recognized oncogenic properties of YAP1 and TAZ have solidified them as primary targets for developing inhibitors to treat cancer and other hyperproliferative diseases, mainly focused on blocking YAP1 and TAZ interaction with TEAD. However, the lack of models to precisely characterize the consequences of TEAD inhibition is a significant challenge in studying the efficacy of this approach. In addition, YAP1 and TAZ interact with numerous other proteins in the cytoplasm and the nucleus, making it difficult to discern TEAD-dependent and independent events.

Several tools are available to characterize the disruption of YAP1/TAZ-TEAD binding and its use for cancer treatment, including dominant-negative proteins, peptides that block YAP1-TEAD interactions, and drugs that inhibit TEAD activity. One of the first strategies used was to block TEAD function by overexpressing truncated TEAD proteins lacking the DNA-binding domain, acting as dominant-negative forms5,7. A similar approach has been to utilize fusion proteins of the TEAD4 transcription factor with the repression domain of Drosophila Engrailed (Tead4EnR) or the transcriptional activation domain of herpes simplex virus VP16 (Tead4VP16)8. Small peptide inhibitors, such as the Super-TDU peptide9, have been derived from the VGLL4 and YAP1 TEAD binding domains (TBDs) to block YAP1-TEAD interaction. Verteporfin, the first drug characterized to inhibit TEAD, works by binding YAP1 and blocking its interaction with TEAD7. IAG933 is another drug that directly binds TEAD and prevents its interaction with YAP and TAZ10. Recent studies have established that TEAD proteins require palmitoylation for activation11,12, leading to the development of small molecule inhibitors that target the TEAD lipid site1315. While each of these tools offers unique advantages, they lack one or more essential elements, including traceability, efficient inhibition of both YAP1 and TAZ or specific blockage of TEAD nuclear events without affecting YAP1 and TAZ interaction with cytoplasmic effectors.

To address these limitations, our laboratory developed a complementary strategy that utilizes a fluorescently traceable, dominant-negative protein named TEADi that specifically blocks the nuclear interaction of TEAD with YAP1 and TAZ16 (Fig. 1A). TEADi offers several advantages for studying YAP1/TAZ-TEAD dependent transcription, including easy visualization, rapid and simple inhibition of TEAD, and specific blockage of nuclear events mediated by both YAP1 and TAZ without affecting their structural or cytoplasmic functions16. TEADi has been used to elucidate Hippo signaling across diverse contexts1728. Despite TEADi advantages, a key challenge of dominant-negative proteins is the need for high expression levels and strong binding to effectively outcompete and displace endogenous interacting partners of the target protein. Hence, improving TEADi stability and affinity for TEAD could improve its inhibitory capacity, allowing for effective TEAD inhibition at lower expression levels.

Figure 1: Analysis of post-transcriptional modifications in TEAD binding domains and development of a next-generation TEAD inhibitor peptide.

Figure 1:

A. Model showing Hippo signaling and how TEADi disrupts YAP1/TAZ binding to TEAD transcription factors, specifically in the nucleus. The building blocks of TEADi are shown. TBD= TEAD-binding domain, NLS= nuclear localization signal. B. Detail of the TBDs that compose the TEAD inhibitor. Colors indicate the stretch of the amino acid sequence from each protein found in TEADi. Reported post-transcriptional modifications in TBDs are indicated (www.phosphosite.org29). p= phosphorylation, ub= ubiquitination, sm= SUMOylation, ac=acetylation. The homologous site in YAP1 and TAZ that binds TEAD is underlined. C. Detail of the homologous site in YAP1 and TAZ that binds TEAD and mutations introduced in TEADiv1 and TEADiv2. D. Depiction of the building blocks for constructions used in this study (not at scale).

TEADi is built upon the TEAD binding domains (TBDs) of VGLL4, YAP1, and TAZ16 (Fig. 1A). Since YAP1 and TAZ nuclear localization and interaction with TEAD are tightly regulated by phosphorylation and ubiquitination2, we hypothesized that altering post-transcriptional modification sites in the TEADi TBDs could enhance its stability and inhibitory capacity. Analysis of post-transcriptional modifications in TEADi TBDs indicated the presence of phosphorylation, ubiquitination, and SUMOylation sites, particularly in the YAP1 TBD (www.phosphosite.org29) (Fig. 1B). Phosphorylation S94 in YAP1 is known to disrupt its binding to TEAD transcription factors30,31 and it is in a region essential for YAP1-TEAD interaction, indicating that preventing S94 phosphorylation could improve binding between these proteins. However, it has been shown that mutating the S94 residue leads to a loss of YAP1-induced TEAD activity and TEAD binding5,30. A different way to prevent S94 phosphorylation would be to mutate surrounding residues to avoid kinase binding. Since the homologous site in TAZ (S51) is not phosphorylated (Fig. 1B, underlined) and this region has a high affinity for TEAD32, we changed aspartic acid 93 in the YAP1 TBD to glutamic acid (D93E), mimicking the TAZ sequence (Fig. 1C). The original TEADi had already a P98E substitution in the YAP1 TBD that increases its affinity to TEAD16,32 (Fig. 1C). We named the new version TEADiv2 and the original as TEADiv1 (Fig. 1C and D). We also constructed a version of TEADi without the TAZ TBD that resembles the Super-TDU inhibitor peptide9 (minimal TEADi or TEADimin) and a TEADi version with an HA tag instead of GFP to characterize potential effects from the fluorescent tag (Fig. 1D).

Transduction of cells with plasmids corresponding to each construct indicates that they are well expressed at the predicted molecular weight (Fig. 2A). All TEADi constructs reduce the expression of the YAP1/TAZ-TEAD target CYR61, while, in general, they do not affect the expression of TEAD, YAP1 or TAZ (Fig. 2A). In agreement with the effect of TEADiv116, coimmunoprecipitation experiments confirmed that TEADiv2 blocks the interaction of YAP1 and TAZ with TEAD transcription factors (Fig. 2B). To compare the efficacy of TEADi versions we measured TEAD-induced transcriptional activity by co-transfecting cells with different doses of DNA for each dominant-negative protein in the presence of YAP1 or TAZ and a luciferase reporter composed of eight repeats of the TEAD binding site (8x-TEAD-Luc33). TEADiv2 showed increased inhibition of YAP1-induced TEAD activity at lower doses than TEADiv2 (Fig. 2C). In contrast, both proteins showed a similar profile of inhibition towards TAZ (Fig. 2D). These results are in agreement with the modifications of TEADiv2 being in the YAP1 TBD (Fig. 1C). We also observed that TEADi without a TAZ TBD (TEADimin) showed a decrease inhibition towards TAZ (Fig. 2E), indicating that the presence of the TAZ TBD enhances the activity of TEADi towards this protein and pointing towards potential differences in YAP1 and TAZ displacement from TEAD by the dominant negative TBDs.

Figure 2: TEADiv2 shows enhanced TEAD inhibition towards YAP1-TEAD activity.

Figure 2:

A. Western blot showing the expression of TEAD inhibitors in HEK293 cells 24hs after transfection. Unless otherwise indicated, TEADiv1 and TEADiv2 refer to the GFP-tagged version. B. Western blot of coimmunoprecipitation experiments showing that TEADiv2 reduces TEAD interaction with YAP1 and TAZ. C-D. Dose curve comparing TEADiv1 and TEADiv2 TEAD inhibition with 8x-TEAD-Luc reporter in HEK293 cells overexpressing YAP1 (C) or TAZ (D). n=3 independent experiments with technical duplicates. ns=p>0.05, **=p<0.005, paired t test. Mean ± SEM is shown. E. Dose curve comparing TEADimin TEAD inhibition with an 8x-TEAD-Luc reporter in HEK293 cells overexpressing YAP1 or TAZ. n=3 independent experiments with technical duplicates. Mean ± SEM is shown.

To measure the effectivity of TEADiv2 against each member of the TEAD family, we utilized TEAD transcription factors fused to a Gal4 DNA-binding domain5. As shown in Fig. 3A, TEADiv2 efficiently blocked YAP1 and TAZ-induced activity for all TEAD family members. Finally, we measured TEAD inhibition by a GFP or HA-tagged TEADiv2 protein using the 8x-TEAD-Luc reporter. Our results show that YAP1 and TAZ-induced TEAD activity was similarly blocked by both tagged versions of TEADiv2 (Fig. 3C), indicating that the efficiency of the dominant-negative protein is independent of the tag.

Figure 3: TEADiv2 inhibits YAP1 and TAZ TEAD activity for all TEAD family members, and its activity is independent of the tag.

Figure 3:

A-B. Graphs showing TEAD activity inhibition by TEADiv2. Transcriptional activity of TEAD was measured by luciferase assay in HEK293 cells transduced with TEADiv2 or GFP, each corresponding TEAD fused to the Gal4 DNA binding domain, UAS-luciferase, and either YAP1 (A) or TAZ (B). TEAD activity is reported as the fold decrease in TEADi compared with GFP control (dotted line indicates activity in GFP condition). n=3 independent experiments with technical duplicates. **=p<0.005, ***=p<0.001, ****=p<0.0001, t test comparing GFP vs TEADi. Mean ± SEM is shown. C. Comparison of GFP-tagged and HA-tagged TEADiv2 with 8x-TEAD-Luc reporter in HEK293 cells overexpressing YAP1 or TAZ. TEAD activity is reported as percentage inhibition over the activity of cells transfected with GFP instead of TEADi. n=3 independent experiments with technical duplicates. ns=p>0.05, t test. Mean ± SEM is shown.

Next, we performed structural predictions with AlphaFold-Multimer34,35 to understand the potential differences between TEADiv1 and TEADiv2. We observed that, in isolation, TEADiv2 adopts an alternative conformation with respect to TEADiv1 (Fig. 4A). We then used RosettaRelax36 to refine and score predicted structures, with lower energy values translating to a more favorable conformation37. TEADiv2 is predicted to be more energetically stable, since its structural models (considering the top10 predictions) are associated with more negative energies compared to TEADiv1 (Fig. 4B). This also correlates in part with a local structuring of TEADiv2 N- and C-terminals, predicted to have higher α-helix content (Figure 4A). Prediction of the complex between TEADi and TEAD4 suggested different 3D conformations for the TEADiv2 complex (Fig. 4C), correlated with overall lower energies of the complexes (Fig. 4D). The predicted structural differences between TEADis are interesting considering that the only difference between TEADiv1 and TEADiv2 is the change of aspartic acid 93 in the YAP1 TBD to glutamic acid (D93E). Our modeling indicates that this D93E substitution does not affect the local structure in the mutation site but translates into a different predicted peptide conformation. Despite the similarities between aspartates and glutamates, it has been proposed that for intrinsically disordered regions aspartates support extended structures and helical caps, while glutamates support increased helicity, which is necessary for folding and binding38. These differences in the properties of aspartates and glutamates to drive structure suggest that the increased activity of TEADiv2 could be partly due to a more stable conformation of the inhibitor resulting from the D93E change in the YAP1 TBD.

Figure 4: TEADiv2 is structurally more stable and has increased affinity for TEAD.

Figure 4:

A. Predicted 3D conformations of isolated TEADiv1 and TEADiv2 via AlphaFold-Multimer v2.3.2. B. Violin plot depicting the energy distribution -in Rosetta energy units (REU)- of the top 10 predicted models of TEADiv1 and TEADiv2. Violin plots show density of data and lines indicate the corresponding average. C. Predicted 3D modeling of TEADiv1 and TEADiv2 interaction with TEAD4 (purple). D. Violin plot depicting the Rosetta energy units (REU) of the top 10 predicted models of TEADiv1 and TEADiv2 in complex with TEAD. Violin plots show density of data and lines indicate the corresponding average.

Overall, our study reveals new insights into the regulation of TEAD activity by YAP1 and TAZ and highlights the potential for improving dominant-negative TEAD inhibitors. We demonstrate that altering post-transcriptional modification sites can enhance the activity of peptides based on TBDs, making them more effective in blocking TEAD-mediated transcriptional activity. We also find that the VGLL4-YAP1 TBDs are insufficient to inhibit TAZ-induced TEAD activity, indicating differences in how YAP1 and TAZ are displaced from TEAD by dominant-negative TBDs. Our results further demonstrate that TEADi effectively inhibits the activity of all four TEAD transcription factors driven by YAP1 and TAZ. Finally, we also identify structural differences in the TBDs of YAP1 and TAZ that may underlie their differential binding and affinity for TEAD. Our work highlights the potential of TEADiv2 as a more effective, specific, and versatile tool for studying TEAD transcriptional activity.

MATERIALS & METHODS

DNA constructs and cell culture:

TEADiv1 construct has been described before16. TEADiv2 and TEADimin were constructed using gBlocks Gene Fragments (Integrated DNA Technologies) downstream of GFP or 2 sequences of the hemagglutinin A tag (2xHA) into a pCEFL vector. Human YAP1 plasmid was constructed in gBlocks Gene Fragments (Integrated DNA Technologies) using sequence optimized YAP1 (NM_001282101) with an N-terminal 3xHA tag followed by a 3xGS linker and cloned into pCEFL. pCEFL vector has an EF-1α promoter and was a gift from Silvio Gutkind. 8x-TEAD-Luc (pGL3b 8xGTIIC-luciferase) was a gift from Stefano Piccolo33 (Addgene plasmid #34615). pcDNA3-HA-TAZ39 and pCMX-GAL4-TEAD1 to 45 constructs were a gift from Kunliang Guan (Addgene plasmids 32839, 33108, 33107, 33106, and 33105 respectively). pGL4.31[luc2P/Gal4UAS/Hygro] was purchased from Promega (catalog #C9351). HEK293 cells were cultured at 37 °C in the presence of 5% CO2. These cells were obtained from AddexBio and cultured in DMEM (Sigma-Aldrich Inc) supplemented with 10% fetal bovine serum (FBS) (Sigma-Aldrich Inc) and antibiotic/antimycotic solution (Sigma-Aldrich Inc).

Luciferase assays:

Luciferase assays were performed in HEK293 cells. To measure pan-TEAD activity, cells were plated in poly-d-lysine coated 24-well plates and transfected 24 h after plating. Cells were co-transfected overnight with 8x-TEAD-Luc (0.3 μg cm−2) plus the indicated DNA constructs. Unless stated otherwise, the DNA amount used was: GFP (0.3 μg cm−2), TEADis (0.3 μg cm−2), YAP1 (0.1 μg cm−2), TAZ (0.1 μg cm−2). The following day, cells were harvested, and luciferase activity was measured using a Dual-Glo Luciferase Assay Kit (Promega) and a Microtiter plate luminometer (SpectrMax iD3, Molecular Devices LLC). To measure individual TEAD family member activity, cells were co-transfected with pCMX-GAL4-TEAD1 to 4 constructs (0.3 μg cm−2), pGL4.31[luc2P/Gal4UAS/Hygro] Vector (0.2 μg cm−2) plus the indicated DNA constructs at 0.3 μg cm−2 and processed as described above. Luciferase normalization was performed in every case by co-transfecting a Renilla luciferase vector (0.01 μg cm−2).

Immunoprecipitation:

For immunoprecipitation (IP), the anti-HA tag antibody was used (Covance; clone no. 16B12; catalog no. MMS-101R; 1:100). Proteins were extracted in IP lysis buffer (50 mM Tris-Cl pH 7.5, 150 mM NaCl, 10% glycerol, 2 mM MgCl2, and 1% NP40) supplemented with a complete protease inhibitor cocktail (Roche, #6538304001). Protein extracts were subjected to centrifugation at max speed for 10 min, and then supernatant protein was quantified. Equal amounts of protein were incubated in a rotator at 4 °C overnight with primary antibody. Then, protein extracts were incubated in a rotator for two hours at 4 °C with Dynabeads Protein G (Invitrogen, #10004D). Finally, beads were washed 4 times with IP lysis buffer, and proteins were eluted with denaturing loading buffer for Western blot analysis.

Immunoblot analysis:

Cells were lysed by sonication at 4 °C in lysis buffer (50 mM Tris-HCl, 150 mM NaCl, 1 mM EDTA, 1% Nonidet P-40, 0.2% sodium deoxycholate, 0.1% SDS, 1 mM DTT) supplemented with complete protease inhibitor cocktail (Roche, #6538304001) and phosphatase inhibitors (PhosSTOP, Sigma-Aldrich, #4906837001). Equal amounts of total cell lysate proteins were subjected to SDS-polyacrylamide gel electrophoresis and transferred to PVDF membranes. Protein visualization was performed using the following antibodies: anti-GAPDH (Cell Signaling; clone no. 14C10; catalogue no. 2118; 1:2000), anti-GFP (Cell Signaling; clone no. D5.1; catalog no. 2956; 1:2000), anti-HA tag antibody (Cell Signaling; clone no. C29F4; catalog no. 3724; 1:1000), YAP1 (Cell Signaling; clone no. D8H1X; catalog no. 14074; 1:1000), TAZ (Cell Signaling; clone no. V386; catalog no. 4883; 1:1000), CYR61 (Cell Signaling; clone no. D4H5D; catalog no. 14479; 1:1000), and Pan-TEAD (Cell Signaling; clone no. D3F7L; catalog no. 13295; 1:1000). Secondary HRP-conjugated antibodies used were: Pierce peroxidase goat antimouse IGG (H + L) (ThermoFisher, catalog no. 31432; 1:2500), and Multi-rAb HRP-Goat Anti Rabbit Recombinant Secondary Antibody (H + L) (Proteintech, catalog no. RGAR001; 1:30000). Secondary antibody was incubated at room temperature for 2 h. Bands were detected using a ChemiDoc Imaging System (Bio-Rad) with Clarity Western ECL Blotting Substrates (Bio-Rad) according to the manufacturer’s instructions.

Structural predictions:

we employed AlphaFold-Multimer v2.3.234,35 with jackhmmer to produce 25 structures of each complex (TEAD-TEADiv1 and TEAD-TEADiv2) and 25 of each inhibitor in its isolated form (TEADiv1 and TEADiv2). We inputted the following sequence stretches for TEADiv1 and TEADiv2, encompassing the VGLL4, YAP1 and TAZ domains:TEADiv1:SVDDHFAKALGDTWLQIGDPPVATNPKTANVPQTVPMRLRKLPDSFFKEPEG DPPVATNPKPSSWRKKILPESFFKEP;TEADiv2:SVDDHFAKALGDTWLQIGDPPVATNPKTANVP QTVPMRLRKLPESFFKEPEGDPPVATNPKPSSWRKKILPESFFKEP. For TEAD4, we inputted the human canonical sequence from Uniprot (Uniprot Accession: Q15561). For each modeling run, the top10 scoring structures were then relaxed using Rosetta Relax version 2021.16.61629 (fast relax protocol)36, employing backbone restraints (flag: “constrain_relax_to_start_coords”). From the outputs of the relax, we then extracted the total energies (in Rosetta Energy Units - REU) and structures. We generated violin plots using the “violinplot” function of the seaborn (v0.13.2) python library. 3D representation was generated using VMD (v1.9.4)40.

Statistics:

All analyses were performed in triplicate or greater and the means obtained were used for t-tests. Statistical analyses, variation estimation and validation of test assumptions were carried out using the Prism 9 statistical analysis program (GraphPad).

ACKNOWLEDGMENTS

This research was supported by the Intramural Research Program of the National Institutes of Health, National Cancer Institute, Center for Cancer Research (ZIA BC 011763).

Footnotes

CONFLICT OF INTEREST

Ramiro Iglesias-Bartolome holds an NCI Employee Invention Report (EIR) for commercial licensing of Peptide inhibitors of YAP1/TAZ-TEAD, E-108–2019-0. The other authors do not have any conflict of interest to declare.

REFERENCES

  • 1.Misra J.R., and Irvine K.D. (2018). The Hippo Signaling Network and Its Biological Functions. Annu Rev Genet 52, 65–87. 10.1146/annurev-genet-120417-031621. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 2.Ma S., Meng Z., Chen R., and Guan K.L. (2019). The Hippo Pathway: Biology and Pathophysiology. Annu Rev Biochem 88, 577–604. 10.1146/annurev-biochem-013118-111829. [DOI] [PubMed] [Google Scholar]
  • 3.Driskill J.H., and Pan D. (2023). Control of stem cell renewal and fate by YAP and TAZ. Nature reviews. Molecular cell biology 24, 895–911. 10.1038/s41580-023-00644-5. [DOI] [PubMed] [Google Scholar]
  • 4.Zhang H., Liu C.Y., Zha Z.Y., Zhao B., Yao J., Zhao S., Xiong Y., Lei Q.Y., and Guan K.L. (2009). TEAD transcription factors mediate the function of TAZ in cell growth and epithelial-mesenchymal transition. The Journal of biological chemistry 284, 13355–13362. 10.1074/jbc.M900843200. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 5.Zhao B., Ye X., Yu J., Li L., Li W., Li S., Yu J., Lin J.D., Wang C.Y., Chinnaiyan A.M., Lai Z.C., and Guan K.L. (2008). TEAD mediates YAP-dependent gene induction and growth control. Gene Dev 22, 1962–1971. 10.1101/gad.1664408. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 6.Lin K.C., Park H.W., and Guan K.L. (2017). Regulation of the Hippo Pathway Transcription Factor TEAD. Trends Biochem Sci 42, 862–872. 10.1016/j.tibs.2017.09.003. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 7.Liu-Chittenden Y., Huang B., Shim J.S., Chen Q., Lee S.J., Anders R.A., Liu J.O., and Pan D. (2012). Genetic and pharmacological disruption of the TEAD-YAP complex suppresses the oncogenic activity of YAP. Gene Dev 26, 1300–1305. 10.1101/gad.192856.112. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 8.Nishioka N., Inoue K., Adachi K., Kiyonari H., Ota M., Ralston A., Yabuta N., Hirahara S., Stephenson R.O., Ogonuki N., et al. (2009). The Hippo signaling pathway components Lats and Yap pattern Tead4 activity to distinguish mouse trophectoderm from inner cell mass. Developmental cell 16, 398–410. 10.1016/j.devcel.2009.02.003. [DOI] [PubMed] [Google Scholar]
  • 9.Jiao S., Wang H., Shi Z., Dong A., Zhang W., Song X., He F., Wang Y., Zhang Z., Wang W., et al. (2014). A peptide mimicking VGLL4 function acts as a YAP antagonist therapy against gastric cancer. Cancer Cell 25, 166–180. 10.1016/j.ccr.2014.01.010. [DOI] [PubMed] [Google Scholar]
  • 10.Chapeau E.A., Sansregret L., Galli G.G., Chene P., Wartmann M., Mourikis T.P., Jaaks P., Baltschukat S., Barbosa I.A.M., Bauer D., et al. (2024). Direct and selective pharmacological disruption of the YAP-TEAD interface by IAG933 inhibits Hippo-dependent and RAS-MAPK-altered cancers. Nat Cancer 5, 1102–1120. 10.1038/s43018-024-00754-9. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 11.Chan P., Han X., Zheng B., DeRan M., Yu J., Jarugumilli G.K., Deng H., Pan D., Luo X., and Wu X. (2016). Autopalmitoylation of TEAD proteins regulates transcriptional output of the Hippo pathway. Nat Chem Biol 12, 282–289. 10.1038/nchembio.2036. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 12.Noland C.L., Gierke S., Schnier P.D., Murray J., Sandoval W.N., Sagolla M., Dey A., Hannoush R.N., Fairbrother W.J., and Cunningham C.N. (2016). Palmitoylation of TEAD Transcription Factors Is Required for Their Stability and Function in Hippo Pathway Signaling. Structure 24, 179–186. 10.1016/j.str.2015.11.005. [DOI] [PubMed] [Google Scholar]
  • 13.Bum-Erdene K., Zhou D., Gonzalez-Gutierrez G., Ghozayel M.K., Si Y., Xu D., Shannon H.E., Bailey B.J., Corson T.W., Pollok K.E., Wells C.D., and Meroueh S.O. (2019). Small-Molecule Covalent Modification of Conserved Cysteine Leads to Allosteric Inhibition of the TEAD·Yap Protein-Protein Interaction. Cell Chem Biol 26, 378–389 e313. 10.1016/j.chembiol.2018.11.010. [DOI] [PubMed] [Google Scholar]
  • 14.Hagenbeek T.J., Zbieg J.R., Hafner M., Mroue R., Lacap J.A., Sodir N.M., Noland C.L., Afghani S., Kishore A., Bhat K.P., et al. (2023). An allosteric pan-TEAD inhibitor blocks oncogenic YAP/TAZ signaling and overcomes KRAS G12C inhibitor resistance. Nat Cancer 4, 812–828. 10.1038/s43018-023-00577-0. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 15.Tang T.T., Konradi A.W., Feng Y., Peng X., Ma M., Li J., Yu F.X., Guan K.L., and Post L. (2021). Small Molecule Inhibitors of TEAD Auto-palmitoylation Selectively Inhibit Proliferation and Tumor Growth of NF2-deficient Mesothelioma. Mol Cancer Ther 20, 986–998. 10.1158/1535-7163.MCT-20-0717. [DOI] [PubMed] [Google Scholar]
  • 16.Yuan Y., Park J., Feng A., Awasthi P., Wang Z., Chen Q., and Iglesias-Bartolome R. (2020). YAP1/TAZ-TEAD transcriptional networks maintain skin homeostasis by regulating cell proliferation and limiting KLF4 activity. Nat Commun 11, 1472. 10.1038/s41467-020-15301-0. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 17.McGinn J., Hallou A., Han S., Krizic K., Ulyanchenko S., Iglesias-Bartolome R., England F.J., Verstreken C., Chalut K.J., Jensen K.B., Simons B.D., and Alcolea M.P. (2021). A biomechanical switch regulates the transition towards homeostasis in oesophageal epithelium. Nature cell biology 23, 511–525. 10.1038/s41556-021-00679-w. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 18.Yuan Y., Salinas Parra N., Chen Q., and Iglesias-Bartolome R. (2022). Oncogenic Hedgehog-Smoothened Signaling Depends on YAP1TAZ/TEAD Transcription to Restrain Differentiation in Basal Cell Carcinoma. The Journal of investigative dermatology 142, 65–76 e67. 10.1016/j.jid.2021.06.020. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 19.Hile G.A., Coit P., Xu B., Victory A.M., Gharaee-Kermani M., Estadt S.N., Maz M.P., Martens J.W.S., Wasikowski R., Dobry C., et al. (2023). Regulation of Photosensitivity by the Hippo Pathway in Lupus Skin. Arthritis Rheumatol 75, 1216–1228. 10.1002/art.42460. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 20.Canellas-Socias A., Cortina C., Hernando-Momblona X., Palomo-Ponce S., Mulholland E.J., Turon G., Mateo L., Conti S., Roman O., Sevillano M., et al. (2022). Metastatic recurrence in colorectal cancer arises from residual EMP1(+) cells. Nature 611, 603–613. 10.1038/s41586-022-05402-9. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 21.Hatterschide J., Castagnino P., Kim H.W., Sperry S.M., Montone K.T., Basu D., and White E.A. (2022). YAP1 activation by human papillomavirus E7 promotes basal cell identity in squamous epithelia. Elife 11. 10.7554/eLife.75466. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 22.Claude-Taupin A., Isnard P., Bagattin A., Kuperwasser N., Roccio F., Ruscica B., Goudin N., Garfa-Traore M., Regnier A., Turinsky L., et al. (2023). The AMPK-Sirtuin 1-YAP axis is regulated by fluid flow intensity and controls autophagy flux in kidney epithelial cells. Nat Commun 14, 8056. 10.1038/s41467-023-43775-1. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 23.Rogg M., Maier J.I., Helmstadter M., Sammarco A., Kliewe F., Kretz O., Weisser L., Van Wymersch C., Findeisen K., Koessinger A.L., et al. (2023). A YAP/TAZ-ARHGAP29-RhoA Signaling Axis Regulates Podocyte Protrusions and Integrin Adhesions. Cells 12. 10.3390/cells12131795. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 24.El Yousfi Y., Mora-Molina R., Lopez-Rivas A., and Yerbes R. (2023). Role of the YAP/TAZ-TEAD Transcriptional Complex in the Metabolic Control of TRAIL Sensitivity by the Mevalonate Pathway in Cancer Cells. Cells 12. 10.3390/cells12192370. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 25.Liu Y., Xu C., Li J., Zhang Y., Wang X., Wang Y., Qin J., Zheng Z., and Xia Y. (2023). YAP promotes AP-1 expression in tubular epithelial cells in the kidney. American journal of physiology. Renal physiology 324, F581–F589. 10.1152/ajprenal.00246.2022. [DOI] [PubMed] [Google Scholar]
  • 26.Mukhopadhyay S., Huang H.Y., Lin Z., Ranieri M., Li S., Sahu S., Liu Y., Ban Y., Guidry K., Hu H., et al. (2023). Genome-Wide CRISPR Screens Identify Multiple Synthetic Lethal Targets That Enhance KRASG12C Inhibitor Efficacy. Cancer research. 10.1158/0008-5472.CAN-23-2729. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 27.Edwards A.C., Stalnecker C.A., Jean Morales A., Taylor K.E., Klomp J.E., Klomp J.A., Waters A.M., Sudhakar N., Hallin J., Tang T.T., et al. (2023). TEAD Inhibition Overcomes YAP1/TAZ-Driven Primary and Acquired Resistance to KRASG12C Inhibitors. Cancer research 83, 4112–4129. 10.1158/0008-5472.CAN-23-2994. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 28.Liu Y., Wang Y., Xu C., Zhang Y., Wang Y., Qin J., Lan H.Y., Wang L., Huang Y., Mak K.K., Zheng Z., and Xia Y. (2024). Activation of the YAP/KLF5 transcriptional cascade in renal tubular cells aggravates kidney injury. Mol Ther 32, 1526–1539. 10.1016/j.ymthe.2024.02.031. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 29.Hornbeck P.V., Zhang B., Murray B., Kornhauser J.M., Latham V., and Skrzypek E. (2015). PhosphoSitePlus, 2014: mutations, PTMs and recalibrations. Nucleic Acids Res 43, D512–520. 10.1093/nar/gku1267. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 30.Mo J.S., Meng Z., Kim Y.C., Park H.W., Hansen C.G., Kim S., Lim D.S., and Guan K.L. (2015). Cellular energy stress induces AMPK-mediated regulation of YAP and the Hippo pathway. Nature cell biology 17, 500–510. 10.1038/ncb3111. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 31.Wang W., Xiao Z.D., Li X., Aziz K.E., Gan B., Johnson R.L., and Chen J. (2015). AMPK modulates Hippo pathway activity to regulate energy homeostasis. Nature cell biology 17, 490–499. 10.1038/ncb3113. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 32.Hau J.C., Erdmann D., Mesrouze Y., Furet P., Fontana P., Zimmermann C., Schmelzle T., Hofmann F., and Chene P. (2013). The TEAD4-YAP/TAZ protein-protein interaction: expected similarities and unexpected differences. Chembiochem 14, 1218–1225. 10.1002/cbic.201300163. [DOI] [PubMed] [Google Scholar]
  • 33.Dupont S., Morsut L., Aragona M., Enzo E., Giulitti S., Cordenonsi M., Zanconato F., Le Digabel J., Forcato M., Bicciato S., Elvassore N., and Piccolo S. (2011). Role of YAP/TAZ in mechanotransduction. Nature 474, 179–183. 10.1038/nature10137. [DOI] [PubMed] [Google Scholar]
  • 34.Jumper J., Evans R., Pritzel A., Green T., Figurnov M., Ronneberger O., Tunyasuvunakool K., Bates R., Zidek A., Potapenko A., et al. (2021). Highly accurate protein structure prediction with AlphaFold. Nature 596, 583–589. 10.1038/s41586-021-03819-2. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 35.Evans R., O’Neill M., Pritzel A., Antropova N., Senior A., Green T., Žídek A., Bates R., Blackwell S., Yim J., et al. (2022). Protein complex prediction with AlphaFold-Multimer. bioRxiv, 2021.2010.2004.463034. 10.1101/2021.10.04.463034. [DOI] [Google Scholar]
  • 36.Nivon L.G., Moretti R., and Baker D. (2013). A Pareto-optimal refinement method for protein design scaffolds. PloS one 8, e59004. 10.1371/journal.pone.0059004. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 37.Alford R.F., Leaver-Fay A., Jeliazkov J.R., O’Meara M.J., DiMaio F.P., Park H., Shapovalov M.V., Renfrew P.D., Mulligan V.K., Kappel K., et al. (2017). The Rosetta All-Atom Energy Function for Macromolecular Modeling and Design. J Chem Theory Comput 13, 3031–3048. 10.1021/acs.jctc.7b00125. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 38.Roesgaard M.A., Lundsgaard J.E., Newcombe E.A., Jacobsen N.L., Pesce F., Tranchant E.E., Lindemose S., Prestel A., Hartmann-Petersen R., Lindorff-Larsen K., and Kragelund B.B. (2022). Deciphering the Alphabet of Disorder-Glu and Asp Act Differently on Local but Not Global Properties. Biomolecules 12. 10.3390/biom12101426. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 39.Lei Q.Y., Zhang H., Zhao B., Zha Z.Y., Bai F., Pei X.H., Zhao S., Xiong Y., and Guan K.L. (2008). TAZ promotes cell proliferation and epithelial-mesenchymal transition and is inhibited by the hippo pathway. Mol Cell Biol 28, 2426–2436. 10.1128/MCB.01874-07. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 40.Humphrey W., Dalke A., and Schulten K. (1996). VMD: visual molecular dynamics. J Mol Graph 14, 33–38, 27–38. 10.1016/0263-7855(96)00018-5. [DOI] [PubMed] [Google Scholar]

Articles from bioRxiv are provided here courtesy of Cold Spring Harbor Laboratory Preprints

RESOURCES