Abstract Transcriptional coactivators Yes-associated protein (YAP) and transcriptional coactivator with PDZ-binding motif (TAZ) play key roles in cancers through transcriptional outputs. However, their transactivation mechanisms remain unclear, and effective targeting strategies are lacking. Here, we show that YAP/TAZ possess a hydrophobic transactivation domain (TAD). TAD knockout prevents tumor establishment due to growth defects and enhances immune attack. Mechanistically, TADs facilitate preinitiation complex (PIC) assembly by recruiting the TATA-binding protein-associated factor 4 (TAF4)-dependent TFIID complex and enhance RNA polymerase II (Pol II) elongation through mediator complex subunit 15 (MED15)-dependent mediator recruitment for the expressions of oncogenic/immune-suppressive programs. The synthesized peptide TJ-M11 selectively disrupts TAD interactions with MED15 and TAF4, suppressing tumor growth and sensitizing tumors to immunotherapy. Our findings demonstrate that YAP/TAZ TADs exhibit dual functions in PIC assembly and Pol II elongation via hydrophobic interactions, which represent actionable targets for cancer therapy and combination immunotherapy. Subject terms: Oncogenes, Transcription __________________________________________________________________ The mechanism underlying YAP/TAZ transactivation remains to be explored. Here the authors report that the hydrophobic transcriptional activation domains of YAP/TAZ exert dual functions on transcriptional initiation and elongation through interactions with TAF4 and MED15, which could be targeted for anti-cancer therapy. Introduction YAP/TAZ, a pair of paralogs of potent transcriptional coactivators, function downstream of the Hippo pathway in cancer initiation, progression, and immunotherapy resistance by promoting the transcription of target genes transcription^[44]1–[45]7. Compared to their functions in covalent and noncovalent epigenetic regulation^[46]8–[47]11, the mechanism and in vivo function of YAP/TAZ to initiate transcription is not well known. Like other transcription events, the specific binding of YAP/TAZ to DNA through TEA domain (TEAD) family transcription factors (TFs) has been well documented, whereas how their intrinsically disordered regions (IDRs) of TAD promoting transactivation has been unresolved questions^[48]12. There are over 2000 TFs with hundreds of TADs in human genome^[49]13,[50]14 and these TADs exhibited obvious primary sequences variation and functional conservation through interacting with only a few cofactors for driving transcription. The mechanism underlying TAD and cofactors interactions driving transcriptions needs to be addressed^[51]15. Thus, dissecting the structure and function of YAP/TAZ-TADs will facilitate the understanding of the transcription output of YAP/TAZ as well as provide potential targeting strategies. YAP/TAZ are upregulated in numerous human cancers and exhibit strong multipotency in autonomous tumor cell growth, non-autonomous immune suppression, microenvironment regulation, and immunotherapy resistance^[52]1,[53]6,[54]16. Because of the key function of YAP/TAZ in solid tumors, targeting them for clinical applications is striking^[55]17. Previous studies have focused mainly on the interaction between TEAD and YAP^[56]18. Several strategies have been developed to identify small molecules and peptides that block TEAD-YAP interactions^[57]19,[58]20. However, to date, no drugs that inhibit YAP have been clinically used because of their limited activity or cytotoxicity. The prevention of transcriptional activation by small peptides or chemicals has recently shown good efficacy against some key onco-transcription factors^[59]21,[60]22. Thus, targeting the interactions between intrinsically disordered TFs and cofactors is a potential field for drug development. However, challenges remain due to the dynamic interface of these interactions^[61]23. Here, we show the structure and function of YAP and TAZ transactivation domains. They exhibit dual functions during transcriptional activation including initiation and elongation. TADs assemble the pre-initiation complex for Pol II loading by interacting with TAF4 and promote Pol II efficient elongations via recruiting the mediator complex. Both TFIID and the Mediator complex bind to transcriptional activation domains (TADs) through hydrophobic interactions involving an α-helix and the linear FLTWL motif. This interaction promotes the folding of the IDR, facilitating the formation of transcriptional condensates via co-condensation. Knockout of TADs of YAP/TAZ leads to tumor growth defects and enhances immune response due to reducing oncogenic and immunosuppressive transcriptional program expression. Dual targeting of the interaction between the TADs of YAP/TAZ and the cofactors TAF4 and MED15 by small peptides mimicking FLTWL inhibits their transactivation activity. This suppresses tumor growth and enhances the efficacy of immunotherapy, supporting the strategy of targeting the transactivation domains of YAP/TAZ for cancer therapy. Results Structural characterization of TADs of YAP and TAZ Our previous studies identified a 55-amino acid TAD at the C-terminus of YAP^[62]24. To determine whether TAZ has a similar TAD, we performed protein sequence alignments and evaluated their activities using Gal4-UAS-luciferase reporter assays (Fig. [63]1a). Similarly, the final 35 residues (366-400) at the C-terminus of TAZ could initiate reporter gene transcription, although with only one-third of the activity seen in YAP-TAD (Fig. [64]1b). Notably, both TADs were composed of over 50% hydrophobic and ~20% acidic amino acids (Fig. [65]1c). Interestingly, at the termini of these TADs is a five-residue linear hydrophobic motif FLTWL, previously identified as the PDZ binding domain^[66]25. Hydrophilic replacement with four serine residues (S) disrupted the ability to drive the transcription of cellular target genes (Fig. [67]1d). Fig. 1. Structural characterization of TADs of YAP and TAZ. [68]Fig. 1 [69]Open in a new tab a Diagram of the conserved domains of YAP/TAZ and the Gal4-UAS-Luciferase reporter system for the TAD activity test. (Pro: Proline, CC: Coiled coil domain, WW: WW domain, sIDR: small Intrinsic Disorder Region. TAD: Transactivation Domain, 40×UAS: 40 repeats of Upstream Activating Sequence). b Luciferase activity of HEK293T cells co-transfected with the indicated plasmids and 40× UAS-luciferase reporters (n = 3 biological replicates). c Sequence alignment of YAP and TAZ TADs. Acidic amino acids are shown in red, and hydrophobic amino acids are shown in green. d RT-qPCR shows YAP target genes in HEK293T cell transfected with YAP, YAPΔTAD, or F500L501W503L504 mutating to S (n = 3 biological replicates). e Assignments of backbone amide groups of YAP-TAD and NMR structure ensemble of YAP-TAD consisting of the 20 lowest-energy models. The α-helix is shown in pink and the intrinsically disordered region in gray. f Assignments of backbone amide groups of TAZ-TAD and NMR structure ensemble of TAZ-TAD consisting of the 20 lowest-energy models. g Predicted structural model of the α-helix of YAP-TAD by Alphafold2. Hydrophilic (blue) and hydrophobic (red) residues are indicated. h RT-qPCR analysis of YAP target genes in HEK293T cells transfected with YAP, YAPΔTAD, L483M486L490 to S, I482M486V489 to S, or N484S488 to L (n = 3 biological replicates). Data are presented as mean ± standard error of the mean (SEM). Statistical analyses were performed using one-way ANOVA (d, h). To dissect the structures of both TADs, we performed nuclear magnetic resonance (NMR) analysis (Fig. [70]1e, [71]f) on highly purified protein samples expressed and purified from E. coli (Supplementary Fig. [72]1a, [73]b). The solution structure of YAP-TAD has been resolved and submitted to Protein Data Bank (PDB) under accession number 8WRG. Our results revealed that the TADs were predominantly composed of IDRs (Fig. [74]1e, f and Supplementary Fig. [75]1c–f). Compared to the TAD of TAZ, the TAD of YAP exhibited a short amphipathic α-helix spanning seven amino acids, with Alphafold2 predictions suggesting a potential extension of up to 17 residues (Fig. [76]1g). Interestingly, substituting residues asparagine (N) and serine (S) on the hydrophilic surface with hydrophobic residue leucine (L) enhanced transcriptional activation. Conversely, replacing hydrophobic residues on the hydrophobic surface with serine (S) reduced transcriptional activity (Fig. [77]1h and Supplementary Fig. [78]1g). These results indicate that YAP possesses a hydrophobic TAD, and the interactions between the amphipathic helix and the FLTWL motif with TFs are crucial for YAP over-expression induced gene activation. The TAD of YAP initiates immunosuppressive transcriptional program Next, we evaluated the functional significance of TADs of YAP/TAZ in transcriptional promotion and tumor growth using a Kras^LSLG12D;Trp53^f/f mouse lung tumor-derived cell line (TDCL)^[79]26 (Supplementary Fig. [80]2a). To this end, we knocked out the TAD of YAP in TAZ ^-/- background by generating a frameshift near the start residue of the TAD of YAP using the clustered regularly interspaced short palindromic repeats (CRISPR-Cas9) assay (Supplementary Fig. [81]2b). The loss of TAD or FLTWL in YAP did not affect its stability or nuclear localization (Supplementary Fig. [82]2c, d). To test whether TADs are essential for tumor cell growth, we examined the growth of TDCL with TADs depletion using plate cloning and allograft assays in C57BL/6J mice. The results showed that knockout of TADs partially impaired tumor cell growth in vitro (Supplementary Fig. [83]2e). Strikingly, tumors with TAD depletion could not be established in immune-competent animals (Supplementary Fig. [84]2f), suggesting a strong immune attack from the hosts when TADs are depleted. To evaluate the contribution of TADs of YAP/TAZ in immunosuppression, we transplanted tumors with TADs depletion in immunodeficient NCG (NOD/ShiLtJGpt-Prkdc^em26Cd52Il2rg^em26Cd22/Gpt) mice and compared their growth in C57BL/6 J mice. Obviously, the tumors in immunodeficient mice can grow bigger than those in C57BL/6J mice (Fig. [85]2a). To assess tumor-specific immune responses, we examined CD8^+ T cells specific to ovalbumin (OVA) using H-2K(b) OVA tetramer antibodies. Initially, we transfected wild-type or TAD-deficient TDCL cells with a lentiviral vector expressing OVA fused with nano-luciferase (nLuc). Comparable intracellular luciferase activity confirmed similar OVA expression levels between the two cell lines (Supplementary Fig. [86]2g). Subsequently, we subcutaneously implanted these cells into C57BL/6 J mice. Our results demonstrated that tumors lacking TAD exhibited increased infiltration of OVA-tetramer-positive CD8^+ T cells (Fig. [87]2b, c). Moreover, infiltrating CD8^+ T cells from TAD-depleted tumors displayed higher fraction and enhanced cytotoxic activity (Fig. [88]2d, e and Supplementary Fig. [89]2h), suggesting a role for YAP-TAD in mediating immunosuppression within the tumor microenvironment. Importantly, depletion of CD8^+ T cells using anti-CD8 antibody in C57BL/6 mice restored tumor growth in TAD-depleted tumors (Supplementary Fig. [90]2i, j). To exclude potential effects of Cas9 neoantigens, we restored YAP expression in TAD-deficient cells. Our results showed that restorage of YAP expression reversed both the tumor growth defects and target gene downregulations due to TAD deficiency (Supplementary Fig. [91]2j, k). Collectively, these findings highlight the critical role of CD8^+ T cell-mediated antitumor immunity in the context of TAD deficiency. Fig. 2. The TAD of YAP initiates immunosuppressive transcriptional program. [92]Fig. 2 [93]Open in a new tab a Tumor growth of TDCL WT, YapΔTAD Taz^-/-#1, and YapΔTAD Taz^-/-#2 cells injected subcutaneously to C57BL/6J mice or NCG mice (n = 5 mice per group). b Tetramer experimental design of tumor-specific T cells against OVA^+ cancer cells. c Flow cytometry analysis of OVA-tetramer^+ in CD8 T cells of TDCL WT-OVA, YapΔTAD Taz^-/- OVA tumors in C57BL/6J mice (n = 5 mice per group). d Flow cytometry analysis of CD8^+ T cells in CD45^+ cells, GZMB^+ cells, and IFN-γ^+ cells of TDCL WT, YapΔTAD Taz^-/- tumors in C57BL/6J mice (n = 6 mice per group). e Immunofluorescence images of TDCL WT, YapΔTAD Taz^-/- tumors were stained for CD8 (red), and macrophage markers CD206 (red), CD163 (red) and F4/80 (green). The value was calculated by counting total CD8^+, CD206^+, or CD163^+ cells / tumor area (n = 5 per group). f GSEA analysis of YAP signature genes from RNA-seq data of TDCL YapΔTAD Taz^-/- and WT Cells. YAP signature genes set was obtained from the Molecular Signatures Database (MsigDB). g Heatmap showed the YAP target genes and immune suppressive genes from RNA-seq. h RT-qPCR analysis of gene expression in indicated genotypes of TDCL cells (n = 3 biological replicates). Data are presented as mean ± SEM. Statistical analysis was performed using two-way ANOVA (a), 2-tailed Student’s t test (c–e), one-way ANOVA (h). To identify YAP-TAD target genes involved in immunosuppression, we performed RNA sequencing followed by Gene Set Enrichment Analysis (GSEA). Our results showed significant downregulation of classic YAP/TAZ target genes, highlighting the essential role of YAP-TAD in transcriptional regulation (Fig. [94]2f). Additionally, we identified reduced expression of both known and novel immunosuppressive genes, including Tgfb2, Il33, and Itgb2, which play key roles in immunosuppression (Fig. [95]2g, Supplementary Fig. [96]2l). To validate these genes as direct YAP-TAD targets, we conducted RT-qPCR in TAD-depleted and WT cells, confirming their reduced expression upon TAD depletion (Fig. [97]2h, Supplementary Fig. [98]2m). Notably, Csf1 and Il33 are known to promote macrophage recruitment and M2 polarization, contributing to tumor immunosuppression^[99]27–[100]29. Immunofluorescence (IF) staining for CD206 and CD163 further revealed that YAP-TAD depletion significantly decreased M2 macrophage infiltration, which in turn enhanced CD8^+ T cell infiltration and cytotoxicity (Fig. [101]2e). In summary, our findings demonstrate that the TADs of YAP/TAZ are essential for driving oncogenic and immunosuppressive transcriptional programs in tumorigenesis. TADs of YAP/TAZ initiate PIC assembly and Pol II loading through recruiting TAF4 To explore the detail molecular mechanism underlying transcriptional activation by the TADs of YAP/TAZ, we first performed the spatial interactome of YAP TAD using a Proximity-dependent Biotin Identification assay (BioID) (Supplementary Fig. [102]3a). Enrichment analysis of the mass spectrometry profile identified several transcriptional components, including TFIID, the Mediator complex, and transcription coactivators (Fig. [103]3a and Supplementary Fig. [104]3b), suggesting the involvement of TADs in transcription initiation via interactions with these transcriptional cofactors. Fig. 3. TADs of YAP/TAZ initiate PIC assembly and Pol II loading through recruiting TAF4. [105]Fig. 3 [106]Open in a new tab a BirA^*-Gal4 and BirA^*-Gal4-YAP-TAD were expressed with 40×UAS in HEK293T cells, and the cell lysates were subjected to BioID-MS. b HA-YAP or YAPΔTAD was co-expressed with Flag-TAF4-v and subjected to coimmunoprecipitation assay (n = 3 independent experiments) (TAF4-v: TAF4 splice variant). c Flag-YAP was co-expressed with HA-TAF4 M[373-837] or HA-TAF4 C[837-1086] and subjected to coimmunoprecipitation assay (n = 3 independent experiments). d Analysis for the CSP in ^1H-^15N-HSQC of ^15N-TAFH upon titration with YAP-TAD. For (d, f), CSP represents the difference in chemical shift of the peak position before and after titration. The detailed calculation method can be found in the Methods section. e The residues with significant CSP in (d) are mapped on the AlphaFold-predicted TAFH structure. f Analysis for CSP in ^1H-^15N-HSQC of ^15N-labeled YAP-TAD upon titrations with TAFH. g The residues with significant CSP in (f) are mapped on the YAP-TAD structure (red). h The CSP analysis for NMR titration of ^15N-TAFH with the pentapeptide FLTWL. i The residues with significant CSP in (h) are mapped on the Alphafold-predicted TAFH structure (blue). j A heatmap of YAP target genes and immunosuppressive genes in NCI-H1299 siNC or siTAF4 cells, derived from RNA-seq. k Upper panel: YAP/TAZ siRNA CUT&Tag analysis Pol II occupancy of YAP target genes (n = 269) in NCI-H1299 cells transfected with siNC or siYAP/TAZ (siYT). Lower panel: Metagene analysis showing total Pol II occupancy of YAP target genes (n = 242) in NCI-H1299 PLKO.1 or shTAF4 cells (TSS transcription start sites, TES transcription end sites). l Representative genome browser tracks of Pol II CUT&Tag in NCI-H1299 siYT or shTAF4 cells for YAP target genes. The x-axis indicates the chromosome position, and the y-axis represents normalized read density in reads per million. To further confirm these interactions, we initially performed co-immunoprecipitation (Co-IP) assays to investigate the interactions between YAP and members of the TFIID family, which scored highly in the BioID assay and are key regulators of promoter occupancy. Our results showed that YAP physically interacts with TAF4 but not with TAF6, TAF7, or TAF9a (Supplementary Fig. [107]3c, d). Notably, depletion of the TAD of YAP or FLTWL motif significantly weakened these interactions (Fig. [108]3b and Supplementary Fig. [109]3e). Similarly, depletion the TAD of TAZ reduced its interaction with TAF4 (Supplementary Fig. [110]3f), aligning with previous transcriptional activity analysis (Fig. [111]1d). Further Co-IP mapping revealed that the middle portion of TAF4 is crucial for interacting with YAP (Fig. [112]3c). Prompted by the direct interactions observed in cells between the TAD of YAP and the middle fragment of TAF4, we employed NMR techniques to gain a more comprehensive and refined understanding of the binding interface between the two proteins. By dissecting the middle part of TAF4 into M1 (residues 373-582), M2 (residues 679-837), and the highly conserved TAF4 homology (TAFH) domain (residues 582−678)^[113]30 (Fig. [114]3c), we expressed and purified the recombinant TAF4 M1, TAFH, M2, and TAF4[582-837] (i.e., TAFH + M2) proteins (Supplementary Fig. [115]3g–j) for biophysical characterization. Follow-up NMR titration assays revealed that TAF4 M1 and M2 were not observed to significantly interact with YAP-TAD (Supplementary Fig. [116]4a, c), but the binding affinity between TAF4[582-837] and YAP-TAD is similar to that of TAFH and YAP-TAD (Supplementary Fig. [117]4b and Supplementary Fig. [118]5a), which narrowed the binding site of TAF4 to the TAFH domain. We subsequently obtained most of the backbone resonance assignments for TAFH, enabling further biophysical characterization. The ^1H-^15N-HSQC spectrum for TAFH exhibits heterogeneously distributed peak intensities, resulting in the inability to assign the backbone resonances for certain residues in α1 (Supplementary Fig. [119]5b). These observations, possibly caused by severe peak broadening, reflect the masked conformational dynamics of the putative hydrophobic pocket enclosed by these residues. Based on the backbone assignments for TAFH, we investigated the binding interface of TAFH involved in the YAP-TAD and TAFH interactions using NMR titration experiments. Observations from the overlaid ^1H-^15N-HSQC spectra of ^15N-TAFH in the presence or absence of YAP-TAD showed that peaks of certain TAFH residues experienced chemical shift perturbations (CSPs) upon YAP-TAD binding (Fig. [120]3d, e, and Supplementary Fig. [121]6a), indicating near μM-level binding affinity. These residues especially hydrophobic residues like L603, L653, L657, Y642, L606, and F599, were primarily scattered around the α1/α3/α4 helices, enclosing a hydrophobic pocket (Fig. [122]3e). The structural model of TAFH was predicted by AlphaFold2. In a reverse series of NMR titration assays, in which ^15N-labeled YAP-TAD was titrated with TAFH, both the C-terminal FLTWL motif and part of the helix of YAP-TAD displayed major chemical shift perturbations, indicating their deep involvement in TAD-TAFH interactions (Fig. [123]3f, g). Notably, the results of the two titrations indicated the involvement of the hydrophobic regions in both YAP-TAD and TAFH. Considering the similarity in hydrophobicity between the helix and the FLTWL of YAP-TAD, we conducted two additional NMR titration experiments to examine the binding properties of the standalone peptide fragments of these two regions toward TAFH. Interestingly, we observed that both fragments bound to almost the same hydrophobic pocket of TAFH as full-length YAP-TAD (Fig. [124]3h, i and Supplementary Fig. [125]6b, c). Collectively, NMR analysis revealed that the YAP-TAF4 interaction was heavily dependent on hydrophobic interactions, specifically involving a hydrophobic pocket on TAFH domain of TAF4 and both the α-helical region and FLTWL motif on YAP-TAD. To further investigate the functional implications of the protein–protein interaction (PPI) between TAF4 and YAP-TAD in YAP-driven transcription, we measured the mRNA levels of target genes by RT-qPCR following TAF4 knockdown, both with and without YAP overexpression. TAF4 knockdown consistently reduced the classic target genes (CTGF, CYR61, and ANKRD1) expression in both conditions (Supplementary Fig. [126]7a, b). Moreover, overexpression of YAP did not reverse the reduction in YAP target gene expression by TAF4 knockdown (Supplementary Fig. [127]7c). To assess the broader impact of TAF4 on YAP/TAZ genome-wide transcriptional activity, we conducted transcriptome profiling and observed the downregulation of YAP typical target genes ANKRD1, CYR61, MYC and the immunosuppressive genes CSF1, IL33, and ITGB2. (Fig. [128]3j and Supplementary Fig. [129]7d). To determine whether the downregulation of target gene expression by TAF4 knockdown was associated with reduced Pol II loading on the transcription start site (TSS) regions of YAP/TAZ target genes, we performed cleavage under targets and tagmentation (CUT&Tag) analyses of Pol II occupancy in TAF4-or YAP/TAZ-knockdown cells. Indeed, as expected, TAF4 depletion led to a genome-wide reduction in Pol II enrichment at both the TSS and gene body, supporting the role of TAF4 as a broadly acting transcription initiation factor belonging to the TFIID family (Fig. [130]3k down, and Supplementary Fig. [131]7e, f). In contrast, siYAP/TAZ had a smaller impact on Pol II, with the most significant changes observed in Pol II occupancy at YAP target genes, indicating that YAP primarily regulates its specific target genes by interaction with TAF4 (Fig. [132]3k up and Supplementary Fig. [133]7g–j). For instance, Pol II occupancy at the typical target loci—CYR61, CTGF, MYC, AMOTL2, and CD155—was reduced in TAF4- or YAP/TAZ-knockdown cells (Fig. [134]3l and Supplementary Fig. [135]7k). In summary, the TADs of YAP/TAZ initiate Pol II loading for target gene transcription through TAF4 dependent Pol II loading, where the TAD-TAF4 interaction is heavily reliant on hydrophobic interactions, specifically involving a hydrophobic pocket on TAF4 and the helix, and FLTWL on YAP-TAD. TADs of YAP/TAZ recruit MED15 for Pol II efficient elongations In addition to TAF4, our interactome results showed potential interactions of mediator components, including MED1, MED15, and MED23, associated with the TAD of YAP (Fig. [136]3a). Previous studies have suggested a role for the mediator complex in facilitating Pol II release from promoters at YAP target loci^[137]31. This prompted us to explore whether the TAD of YAP directly recruits the mediator. Through an immune co-precipitation assay, we observed that YAP interacted with MED15, but not with MED23 or MED24 (Fig. [138]4a, b). Further examination of the interaction regions of MED15 and YAP revealed that the deletion of TAD completely abolished their interactions (Supplementary Fig. [139]8a). To explore the function of MED15 in YAP transcriptional activation, we performed a BioID assay to determine its spatial interactome (Supplementary Fig. [140]8b). As expected, we identified more than ten mediator components and numerous Pol II elongation factors (Supplementary Fig. [141]8c, d), suggesting that the mediator complex recruited elongation factors for YAP target gene elongation. Co-IP assays demonstrated that MED15 interacts with elongation factors such as PAF1, SPT5, and CDK9 (Fig. [142]4c). These interactions prompted us to examine the function of MED15 in Pol II elongation at the YAP target loci using the CUT&Tag assay. Knockdown of MED15 resulted in the downregulation of target genes (Supplementary Fig. [143]8e), and reduced Ki67 staining due to impaired growth (Supplementary Fig. [144]8f, g). Moreover, reconstitution of YAP/TAZ did not restore the YAP target gene reduction by MED15 depletion, suggesting that MED15 functions downstream of YAP/TAZ (Supplementary Fig. [145]8h). CUT&Tag analysis revealed that MED15 knockdown increased both the TSS and gene body occupancy of Pol II in several YAP/TAZ target genes, including CTGF, CYR61, AMOTL2, and FOSL1 (Fig. [146]4d, e). However, increased Pol II loading at target loci led to decreased target gene expression, reminiscent of previous findings, wherein the loss of PAF1 and SPT5 resulted in inefficient Pol II elongation^[147]32. To explore whether MED15 depletion leads to inefficient elongation, we examined premature termination events in the classic target gene CTGF by comparing the levels of nascent RNA and mRNA using RT-qPCR. The results revealed an increased ratio of nascent RNA to mRNA, indicating the inefficient elongation of Pol II on the target gene bodies (Fig. [148]4f). The above results implicate an MED15-involving YAP transcriptional activation process, necessitating a deeper understanding of the underlying mechanisms. Fig. 4. TADs of YAP/TAZ recruit MED15 for Pol II efficient elongations. [149]Fig. 4 [150]Open in a new tab a HA-YAP was co-expressed with Flag-MED15/MED23/MED24 and subjected to coimmunoprecipitation assay (n = 3 independent experiments). b Endogenous Co-IP of MED15 and YAP in NCI-H1299 cells (n = 3 independent experiments). c Endogenous Co-IP of PAF1, SPT5, CDK9 and MED15 in NCI-H1299 cells (n = 3 independent experiments). d Metagene profile of Pol II CUT&Tag reads of YAP/TAZ target genes (n = 150). e Representative genome browser tracks of Pol II CUT&Tag in NCI-H1299 PLKO.1 or shMED15 cells. f RT-qPCR was used to detect both mature mRNA and nascent RNA levels of CTGF in NCI-H1299 PLKO.1 or shMED15 cells (n = 3 biological replicates). For nascent RNA detection, the reverse transcription primer was designed to target the first intron of CTGF, 87 nucleotides downstream of the transcription start site (TSS). Expression levels were normalized to GAPDH. g Heatmap shows YAP target genes of NCI-H1299 MED15 KO or WT cells. h GSEA analysis of YAP signature genes from RNA-seq data of NCI-H1299 WT and MED15 knockout cells. i Tumor growth of NCI-H1299 WT or MED15 KO cells in xenograft models (n = 5 mice). j Model depicting YAP/TAZ-TAD promotion of PIC assembly through TAF4 and Pol II elongation via MED15. Data are presented as mean ± SEM. Statistical analysis was performed using 2-tailed Student’s t test (f), and two-way ANOVA (i). To evaluate the function of MED15 in YAP-associated transcription, tumor cell growth, and tumorigenesis in vivo, we established MED15 knockout cell lines in NCI-H1299 cells in a CRISPR-Cas9 dependent manner (Supplementary Fig. [151]8i). Initially, we compared the transcriptomes of MED15 KO and WT cells. The profiles revealed significant downregulation of several YAP target genes associated with cancer initiation and the progression of immunosuppressive genes (Fig. [152]4g). Moreover, GSEA analysis revealed that MED15 depletion downregulated YAP signature genes (Fig. [153]4h). To evaluate the function of MED15 in cell growth in vivo, we performed xenograft assays and observed that the knockout of MED15 robustly reduced the growth rate of tumor cells (Fig. [154]4i and Supplementary Fig. [155]8j, k). Additionally, restoring MED15 expression in NCI-H1299 MED15^-/- cells, rescued tumor growth defects (Supplementary Fig. [156]8l, m). Collectively, these findings indicate that MED15 is indispensable for the oncogenic transcriptional program regulated by YAP and for tumor cell growth, likely due to its role in transcriptional elongation. Taken together, TADs of YAP/TAZ have dual functions in initiation and elongation by recruiting TAF4 and Mediator 15 (Fig. [157]4j). The hydrophobic interaction of YAP-TAD and MED15 boosts transcriptional hub formation by co-condensations Multiple reports support the notion that YAP/TAZ functions as a transcriptional hub through coiled-coil (CC) domain-mediated phase separation facilitated by hydrophobic interactions. Key transcription regulators are recruited to the condensates for efficient transcription in a spatially partitioned manner^[158]24,[159]33–[160]35. This prompted us to test whether MED15 was recruited to YAP condensates. Interestingly, in contrast to other mediator components, MED15 was recruited as well as boosting YAP condensation, including increased condensate size and number when co-expressed with YAP (Fig. [161]5a and Supplementary Fig. [162]9a, b). To examine whether co-condensation and enhancement occurred in vitro, we synthesized and purified YAP and MED15 (Supplementary Fig. [163]9c). Upon mixing them in a physiological concentration of NaCl, we observed similar co-condensation and enhancement of YAP droplet formation (Fig. [164]5b, c). We further identified the region involved in the co-condensation of YAP and MED15 by co-expressing different regions of MED15 and YAP, which revealed that the middle fragment of MED15 is required for co-condensation (Fig. [165]5d). Previous studies in yeast have identified several activation-binding domains (ABDs) in the middle region of MED15^[166]36,[167]37. Multiple sequence alignment revealed three relatively conserved ABDs (ABD1-3) in human MED15. Individual expression of ABDs with YAP demonstrated obvious co-condensation in the nucleus, with ABD3 showing particularly pronounced effects (Fig. [168]5e). These results suggested that the hydrophobic interactions of MED15-YAP acted as another driving forces for YAP condensation paralleled to CC mediated hydrophobic interactions. To test this hypothesis, we examined whether the condensation defect of YAP^S127A△CC could be restored by co-expression with MED15. Surprisingly, MED15 co-expression with YAP^S127A△CC restored its phase separation, subsequently rescuing its transcriptional activity defects (Fig. [169]5f). To test whether co-condensation drives transcription in the droplets, we fused the TAD of YAP/TAZ with rTetR, co-expressed it with PP7 coat protein (PCP) -3×mCherry and TetO-PP7, and visualized and monitored the levels of nascent RNA by microscopy^[170]38. We observed increased transcription in the condensates after doxycycline treatment (Fig. [171]5g). Fig. 5. The hydrophobic interaction of YAP-TAD and MED15 boosts transcriptional hub formation by co-condensations. [172]Fig. 5 [173]Open in a new tab a Confocal sections of YAP^S127A -mEGFP expressed with or without Flag-MED15, and scatter plot showed the puncta diameter (n = 24 cells per group). b Fluorescence images of in vitro His-YAP-mEGFP (green) and His-MED15-CY3 (red) were mixed with the indicated module concentration in 10% PEG solution. c Plot shows mean droplet size. DIC images of His-YAP-mEGFP and His-MED15-CY3 mixture (1:1 at 5 µM, 10% PEG) shows numerous droplets at room temperature in a coverslip chamber. d mEGFP-YAP^S127A was expressed with Flag-MED15-N, Flag-MED15-M, and Flag-MED15-C. e Schematic representation of ABDs of MED15. mEGFP-YAP^S127A was expressed with MED15-ABD1, ABD2, Linker, and ABD3. f Images of Flag-MED15 co-expressed with mEGFP- YAP^S127A or mEGFP YAP^S127AΔCC, histogram shows the ratio of cells with puncta (n = 100 cells per group). RT-qPCR analysis of YAP target genes in cells transfected with Flag-MED15 (1 μg), mEGFP- YAP^S127A (200 ng) or mEGFP- YAP^S127AΔCC (200 ng) (n = 3 biological replicates). g Schematic of visualizing transcriptional activation in YAP/TAZ-TAD and MED15 condensates. Arrows indicate the nascent RNA signals (bound by PCP-3×mCherry) colocalizing with TAD/MED15 condensates (rTetR-BFP-YAP/TAZ-TAD and mEGFP-MED15) after addition of doxycycline (0.01 mg/ml). h Schematic of the hydrophobic interaction of YAP and MED15 forming a transcriptional hub to regulate the transcription initiation and elongation. Data are presented as mean ± SEM. Statistical analysis was performed using 2-tailed Student’s t test (a), one-way ANOVA (c, f). Scale bars: 10 μm (a, f), 5 μm (b, c, d, e, g). These results indicated that the hydrophobic interactions of TAD-ABD3 enhanced YAP condensation parallel to CC-mediated hydrophobic interactions (Fig. [174]5h). The mechanism of interaction between TADs and MED15 To further understand elongation, we investigated the molecular mechanisms underlying the interactions between TADs and MED15. By performing multiple sequence alignments, we identified three relatively conserved ABDs (ABD1-3) in human MED15 (Fig. [175]5e). This result prompted us to examine whether YAP-TAD binds to MED15-ABDs in humans similarly to the fuzzy interactions observed in yeast. We prepared protein samples from MED15 and its ABDs (Supplementary Fig. [176]10a–e) and performed hydrogen-deuterium exchange (HDX) detected by mass spectrometry (MS) (HDX-MS) assays to analyze the binding sites on YAP and MED15. Unlike the fuzzy interactions in yeast, the results indicated that the TAD domain of YAP and the ABD3 domain of MED15 were the primary binding sites responsible for the YAP-MED15 interaction (Fig. [177]6a; Supplementary Fig. [178]10f–h, Supplementary Fig. [179]11a, b; Supplementary Fig. [180]12a, b; Supplementary Fig. [181]13a). Follow-up NMR titration assays confirmed that ABD1 and ABD2 did not bind YAP-TAD (Supplementary Fig. [182]13b, c). Consistent with our expectations, surface plasmon resonance (SPR) characterization revealed a substantial interaction between YAP-TAD and MED15 (particularly its ABD3 domain), evidenced by comparable dissociation constants (Kd = 27.5 ± 14.8 nM for MED15 and YAP, Kd = 160.2 ± 54.3 nM for YAP-TAD and ABD3; Supplementary Fig. [183]14a–c). These in vitro biophysical assays aligned with findings from co-condensation experiments for individual ABDs, prompting us to focus on ABD3-TAD interactions in subsequent studies. Fig. 6. The mechanism of interaction between TADs and MED15. [184]Fig. 6 [185]Open in a new tab a HDX-MS characterization for identification of YAP-MED15 binding sites. Deuterium uptake plots of MED15 peptides (Region I: AA 492-511, +4 charge; Region II: AA 531-551, +5 charge) and YAP peptide (AA 491-502, +3) were measured in the presence and absence of their binding partners. Data are plotted as percent deuterium uptake versus time (logarithmic scale). Red and green plots represent the unbound and bound states, respectively (n = 3 technical replicates). b NMR structure ensemble of MED15-ABD3 consisting of 20 lowest-energy models. c Analysis for the CSP of ^15N-YAP-TAD upon titration with ABD3. d The residues with significant CSP during NMR titration in (c) are mapped on the YAP-TAD structure in red. e Predicted helix formation propensity for YAP-TAD with or without ABD3 using TALOS-N webserver. f Analysis for the change in CSP of ^15N-ABD3 upon titration with YAP-TAD. g The residues with significant CSP change during NMR titration in (f) are mapped on the MED15-ABD3 structure in blue. h Analysis for the change in CSP of ^15N-ABD3 upon titration with the pentapeptide FLTWL. i The residues with significant CSP change in (h) during NMR titration experiment are mapped on the MED15-ABD3 structure in blue. Data are presented as mean ± SD. To acquire detailed properties of the TAD-MED15 interaction, we assigned most of the backbone and side-chain chemical shifts of a standalone truncation of the ABD3 domain of MED15 (residues 453-561 of MED15 isoform 2, named MED15-ABD3) using NMR spectroscopy. We next determined the solution structure of MED15-ABD3 under PDB accession number 8J9A, which consists of a long N-terminal loop and three α-helices (designated as α1, α2, and α3) (Fig. [186]6b and Supplementary Fig. [187]15a, b). Subsequently, we conducted 2D HSQC spectrum-based NMR titration experiments, using the CSPs observed during the titration of MED15-ABD3 into ^15N-YAP-TAD to identify the binding sites on YAP-TAD for the YAP-ABD3 interaction (Fig. [188]6c and Supplementary Fig. [189]16a). Notably, certain hydrophobic residues in the helix and C-terminal FLTWL of YAP-TAD exhibited significant chemical shift perturbations, indicating their involvement in the binding of MED15-ABD3 (Fig. [190]6c, d). Interestingly, ABD3 binding induced the elongation of the α-helix of YAP-TAD (Fig. [191]6e). The extension of the secondary structural elements of the initially disordered region of YAP-TAD depicts the flexible and binding-ready nature of this dual-targeting transcriptional coactivator, reflecting its versatile use in various situations regarding the transcription processes. Backbone assignments and the MED15-ABD3 structure enabled us to perform reverse NMR titrations where serial concentrations of YAP-TAD were titrated into ^15N-MED15-ABD3. These experiments revealed that residues involved in binding were scattered mainly on α2 and α3 helices, with the majority located at the helix-helix interfaces, forming a clustering pattern of a line-like continuum (Fig. [192]6f, g and Supplementary Fig. [193]16b). Similarly, titration involving ^15N-TAZ-TAD with MED15-ABD3, as well as its reverse titration, showed a similar binding pattern to interactions between YAP-TAD and MED15-ABD3 (Supplementary Fig. [194]16c–f). Based on these findings on the TAD-ABD3 interaction and TAD-TAF4 binding sites, we investigated the roles played by the helix and FLTWL motif of YAP-TAD in the TAD-ABD3 interaction by titrating peptides containing each specific fragment into ^15N-MED15-ABD3. The FLTWL peptide had a binding interface analogous to YAP-TAD on MED15-ABD3, with most binding residues being hydrophobic and positioned at the aforementioned helix-helix junction interface (Fig. [195]6h, i and Supplementary Fig. [196]17a). Contrary to the ^15N-YAP-TAD titration results (Fig. [197]6c, d), the α-helix-based peptide showed reduced affinity for binding ABD3 in a standalone state, characterized by hardly observed CSPs or peak intensity changes (Supplementary Fig. [198]17b). This observation suggests a weaker α-helix-ABD3 interaction compared to α-helix-TAF4 binding (Supplementary Fig. [199]6c and Supplementary Fig. [200]17b), potentially reflecting differentiated binding mechanisms and preferences for the two target proteins. In summary, the