INTRODUCTION
Transcription factors are the gatekeepers of gene expression. Aberrant expression or deregulation of transcription factors contributes to human diseases including cancer [
1Targeting transcription factors in cancer - from undruggable to reality.
]. The transcriptional enhanced associate domain family members 1-4 (TEADs) are a set of four transcription factor paralogs that are highly conserved and ubiquitously expressed in higher order organisms. The domain architecture of TEADs is comprised of a DNA binding domain (TEA domain) at the N-terminus and a cofactor-binding domain at the C-terminus. The N-terminal domain directly interacts with the major and minor grooves of DNA [
2- Shi Z.
- He F.
- Chen M.
- Hua L.
- Wang W.
- Jiao S.
- Zhou Z.
DNA-binding mechanism of the Hippo pathway transcription factor TEAD4.
] and contains a nuclear localization sequence (NLS) [
3Identification of a classical bipartite nuclear localization signal in the Drosophila TEA/ATTS protein scalloped.
]. The C-terminal domain mediates interaction with co-activating proteins such as Yes-associated protein (YAP), and its paralog, transcriptional co-activator with PDZ-binding motif (TAZ) [
4- Kaneko K.J.
- DePamphilis M.L.
(1998) Regulation of gene expression at the beginning of mammalian development and the TEAD family of transcription factors.
,
5- 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.
]. Importantly, protein interactions are central to the cellular functions of TEADs.
TEADs function as the transcriptional effectors of the Hippo pathway [
6- Zhou Y.
- Huang T.
- Cheng A.S.
- Yu J.
- Kang W.
- To K.F.
The TEAD Family and Its Oncogenic Role in Promoting Tumorigenesis.
] which is a highly conserved kinase cascade that maintains organ growth and tissue homeostasis [
7- Ma S.
- Meng Z.
- Chen R.
- Guan K.L.
The Hippo Pathway: Biology and Pathophysiology.
]. Activation of this pathway is dependent on two serine/threonine kinases, mammalian STE20-like 1/2 (MST1/2) and large tumor suppressor 1/2 (LATS1/2), that phosphorylate and negatively regulate YAP and TAZ. In the cytoplasm, phosphorylated YAP and TAZ are sequestered by binding to 14-3-3 and subsequently degraded by the proteasome [
8- Harvey K.F.
- Zhang X.
- Thomas D.M.
The Hippo pathway and human cancer.
]. When the upstream kinases of the Hippo pathway are inactive, YAP and TAZ remain unphosphorylated which allows them to translocate to the nucleus where they directly interact with TEADs to mediate the transcription of genes involved in cellular proliferation, homeostasis, polarity, survival, and cell fate [
8- Harvey K.F.
- Zhang X.
- Thomas D.M.
The Hippo pathway and human cancer.
]. In addition to YAP and TAZ, TEADs are known to engage with other protein binding partners in the nucleus. TEADs interact with the VGLL (VGLL1-4) family of transcriptional coactivators, and these interactions can be transcriptionally activating or repressive depending on the context [
9- Simon E.
- Faucheux C.
- Zider A.
- Theze N.
- Thiebaud P.
From vestigial to vestigial-like: the Drosophila gene that has taken wing.
,
10- Hori N.
- Okada K.
- Takakura Y.
- Takano H.
- Yamaguchi N.
- Yamaguchi N.
Vestigial-like family member 3 (VGLL3), a cofactor for TEAD transcription factors, promotes cancer cell proliferation by activating the Hippo pathway.
,
11- Jiao S.
- Wang H.
- Shi Z.
- Dong A.
- Zhang W.
- Song X.
- He F.
- Wang Y.
- Zhang Z.
- Wang W.
- Wang X.
- Guo T.
- Li P.
- Zhao Y.
- Ji H.
- Zhang L.
- Zhou Z.
A peptide mimicking VGLL4 function acts as a YAP antagonist therapy against gastric cancer.
]. TEADs also interact with AP-1 transcription factors [
12- Zanconato F.
- Forcato M.
- Battilana G.
- Azzolin L.
- Quaranta E.
- Bodega B.
- Rosato A.
- Bicciato S.
- Cordenonsi M.
- Piccolo S.
Genome-wide association between YAP/TAZ/TEAD and AP-1 at enhancers drives oncogenic growth.
,
13- Liu X.
- Li H.
- Rajurkar M.
- Li Q.
- Cotton J.L.
- Ou J.
- Zhu L.J.
- Goel H.L.
- Mercurio A.M.
- Park J.S.
- Davis R.J.
- Mao J.
Tead and AP1 Coordinate Transcription and Motility.
] and with chromatin remodeling complexes [
14- Chang C.Y.
- Shipony Z.
- Lin S.G.
- Kuo A.
- Xiong X.
- Loh K.M.
- Greenleaf W.J.
- Crabtree G.R.
Increased ACTL6A occupancy within mSWI/SNF chromatin remodelers drives human squamous cell carcinoma.
]. Clearly the repertoire of TEAD interactions is heavily networked and influences a various of cellular processes, the full extent to which remains unclear.
The Hippo pathway repeatedly emerges as relevant during cancer development, progression, and therapy resistance [
8- Harvey K.F.
- Zhang X.
- Thomas D.M.
The Hippo pathway and human cancer.
,
15- Wang Y.
- Xu X.
- Maglic D.
- Dill M.T.
- Mojumdar K.
- Ng P.K.
- Jeong K.J.
- Tsang Y.H.
- Moreno D.
- Bhavana V.H.
- Peng X.
- Ge Z.
- Chen H.
- Li J.
- Chen Z.
- Zhang H.
- Han L.
- Du D.
- Creighton C.J.
- Mills G.B.
- Cancer Genome Atlas Research N.
- Camargo F.
- Liang H.
Comprehensive Molecular Characterization of the Hippo Signaling Pathway in Cancer.
,
16The Hippo Signaling Pathway in Drug Resistance in Cancer.
]. A prominent example is the frequent amplification of the effector proteins, YAP, TAZ, and TEADs in squamous, head and neck, and gastrointestinal malignancies [
6- Zhou Y.
- Huang T.
- Cheng A.S.
- Yu J.
- Kang W.
- To K.F.
The TEAD Family and Its Oncogenic Role in Promoting Tumorigenesis.
,
15- Wang Y.
- Xu X.
- Maglic D.
- Dill M.T.
- Mojumdar K.
- Ng P.K.
- Jeong K.J.
- Tsang Y.H.
- Moreno D.
- Bhavana V.H.
- Peng X.
- Ge Z.
- Chen H.
- Li J.
- Chen Z.
- Zhang H.
- Han L.
- Du D.
- Creighton C.J.
- Mills G.B.
- Cancer Genome Atlas Research N.
- Camargo F.
- Liang H.
Comprehensive Molecular Characterization of the Hippo Signaling Pathway in Cancer.
,
17- Sanchez-Vega F.
- Mina M.
- Armenia J.
- Chatila W.K.
- Luna A.
- La K.C.
- Dimitriadoy S.
- Liu D.L.
- Kantheti H.S.
- Saghafinia S.
- Chakravarty D.
- Daian F.
- Gao Q.
- Bailey M.H.
- Liang W.W.
- Foltz S.M.
- Shmulevich I.
- Ding L.
- Heins Z.
- Ochoa A.
- Gross B.
- Gao J.
- Zhang H.
- Kundra R.
- Kandoth C.
- Bahceci I.
- Dervishi L.
- Dogrusoz U.
- Zhou W.
- Shen H.
- Laird P.W.
- Way G.P.
- Greene C.S.
- Liang H.
- Xiao Y.
- Wang C.
- Iavarone A.
- Berger A.H.
- Bivona T.G.
- Lazar A.J.
- Hammer G.D.
- Giordano T.
- Kwong L.N.
- McArthur G.
- Huang C.
- Tward A.D.
- Frederick M.J.
- McCormick F.
- Meyerson M.
- Cancer Genome Atlas Research N.
- Van Allen E.M.
- Cherniack A.D.
- Ciriello G.
- Sander C.
- Schultz N.
Oncogenic Signaling Pathways in The Cancer Genome Atlas.
]. Additionally, inducing YAP expression transforms normal epithelial cells to metastatic cells [
18- Overholtzer M.
- Zhang J.
- Smolen G.A.
- Muir B.
- Li W.
- Sgroi D.C.
- Deng C.X.
- Brugge J.S.
- Haber D.A.
Transforming properties of YAP, a candidate oncogene on the chromosome 11q22 amplicon.
], and plays an important role in bypassing RAS signaling in pancreatic and colon cancer progression [
19- Calses P.C.
- Crawford J.J.
- Lill J.R.
- Dey A.
Hippo Pathway in Cancer: Aberrant Regulation and Therapeutic Opportunities.
,
20- Kapoor A.
- Yao W.
- Ying H.
- Hua S.
- Liewen A.
- Wang Q.
- Zhong Y.
- Wu C.J.
- Sadanandam A.
- Hu B.
- Chang Q.
- Chu G.C.
- Al-Khalil R.
- Jiang S.
- Xia H.
- Fletcher-Sananikone E.
- Lim C.
- Horwitz G.I.
- Viale A.
- Pettazzoni P.
- Sanchez N.
- Wang H.
- Protopopov A.
- Zhang J.
- Heffernan T.
- Johnson R.L.
- Chin L.
- Wang Y.A.
- Draetta G.
- DePinho R.A.
Yap1 activation enables bypass of oncogenic Kras addiction in pancreatic cancer.
,
21- Shao D.D.
- Xue W.
- Krall E.B.
- Bhutkar A.
- Piccioni F.
- Wang X.
- Schinzel A.C.
- Sood S.
- Rosenbluh J.
- Kim J.W.
- Zwang Y.
- Roberts T.M.
- Root D.E.
- Jacks T.
- Hahn W.C.
KRAS and YAP1 converge to regulate EMT and tumor survival.
]. Although significant progress is being made in elucidating how other signaling pathways can regulate YAP and TAZ in cancer, less is understood about how TEADs are regulated. Moreover, efforts to target the Hippo pathway therapeutically are well underway, in particular, the development of TEAD-targeting small molecules aimed at disrupting the protein interactions that are necessary for TEADs transcriptional function [
22- Holden J.K.
- Cunningham C.N.
Targeting the Hippo Pathway and Cancer through the TEAD Family of Transcription Factors.
,
23- Zagiel B.
- Melnyk P.
- Cotelle P.
Progress with YAP/TAZ-TEAD inhibitors: a patent review (2018-present).
]. Therefore, understanding the full repertoire of the cellular functions of TEADs will be critical in anticipating the efficacy and potential toxicity of these approaches.
Here, we describe a connection between TEADs and the DNA damage response (DDR) pathway. We performed protein-protein interaction studies using AP-MS and found that TEADs interact with several factors known to be involved in DNA double-stranded break repair. We found that TEADs colocalize with nuclear DNA damage foci, and that RNAi knockdown of TEADs decreased cellular repair of double-stranded breaks and promoted genomic instability. The proteomic and cellular studies presented here provide a rich interaction dataset and describe a connection between the TEAD transcription factors and the crucial cellular response to genomic insults.
EXPERIMENTAL PROCEDURES
Experimental Design and Statistical Rationale
The control, WT TEAD3-Myc/Flag, and NLS mutant TEAD3-Myc/Flag (one control, two baits) immunoprecipitations were performed using isotype IgG and anti-Myc Ab respectively in HEK293 cell lysates as described in “Cellular fractionation, Western blotting, Immunoprecipitation'' section below. We carried out the analysis in three biological replicates where each set of control and bait IPs were collected simultaneously to minimize the difference in background proteins between the control and bait IPs. Each biological replicate was done on a different batch of cells. The IP and subsequent mass spectrometric workflow were performed on different days. Similarly, we applied the same strategy for the experiments in Detroit 562 cell lysates. For this analysis we used the GeLC-MS approach to maximize the chance of detecting the low abundant interactors. Therefore, for practical reasons, we did not perform technical replicates. In addition, our lab has witnessed a high level of reproducibility within technical replicates.
Software
Figure schemes were created using
BioRender.com. Graphs were created using R or using Graphpad Prism. Flow cytometry data were analyzed using FlowJo.
Cell lines
HEK293, Detroit 562 (pharyngeal carcinoma cells), MDA-MB-231 and SK-N-F1 (neuroblastoma cells) were cultured in DMEM or RPMI supplemented with 2 mM glutamine, 50 U/mL penicillin, 50 μg/mL streptomycin, 10% heat-inactivated fetal calf serum at 37 °C and 5% CO2. All cell lines described in this study were sourced from ATCC. To generate stable lines used in our experiments, MYC-FLAG tagged-WT TEAD3 cDNA (OHu08942, GeneScript) was first subcloned into PiggyBac (pBind, Genentech) plasmid containing a puromycin resistance and doxycycline inducible cassette by LakePharma. The WT-TEAD3 plasmid was subsequently mutated for seven point mutations to generate the NLS mutant TEAD3 (R87N, R88N, R99N, R100N, K101N, V102N, and R103N) (LakePharma). Stable cell lines were generated by co-transfection of these plasmids with the transposase vector (pBO, Transposagen Biopharmaceuticals) using TransIT-LT1 (Mirus, MIR2300). Transfected cells were then selected with Puromycin (P9620, Millipore-Sigma) at 2 μg/mL. DNA damage in cells was induced by exposing the cells to 6 Gy γ-irradiation (γIR) or treatment with the indicated DNA damaging agents (ex. etoposide, bleomycin).
Antibodies
Antibodies used for western blotting: YAP (D8H1X (#14074) or 1A12 (#12395)), TAZ (D3I6D (#70148) or E5P2N (#71192)), Myc-tag (9B11 (#2276) or 71D10 (#2278)), panTEAD (D3F7L (#13295)), RIF1 (D2F2M (#95558)) GAPDH (D16H11 (#5174)), α-Tubulin (DM1A (#3873)), pS139 H2A.X (20E3 (#9718)), Ku70 (D10A7 (#4588)), and Ku80 (#2753) from Cell Signaling Technology. MAX ((H2) sc-8011) from Santa Cruz Biotechnology. Secondary antibodies: Goat anti-mouse (926-3220, Li-cor), Goat-anti-rabbit (926-68071, Li-Cor).
Antibody used for immunoprecipitation: Myc-tag (9B11 (#2276) or 71D10 (#2278), Cell Signaling Technology), Mouse (G3A1) mAb IgG1 Isotype Control (#5415, Cell Signaling Technology).
Antibodies used for immunofluorescence: panTEAD (D3F7L (#13295), Cell Signaling Technologies), RIF1 (NBP2-26219, Novus biologicals), pS139 H2A.X (05-636, Millipore-Sigma).
siRNAs and shRNAs
siRNAs for TEAD-1 (L-012603-00), TEAD-2 (L-012611-01), TEAD3 (L-012604), TEAD4 (L-019570-00), DNA ligase 4 (L-004254-00), 53BP1 (L-003548-00), and BRCA2 (L-003462-00) are from Horizon Discovery. Non-targeting control (NTC) siRNA is from Qiagen (1027310).
MDA-MB-231 shTEAD_1 target sequences:
TEAD1: 5’-GCTCAAACACTTACCAGAGAA-3’
TEAD2: 5’-ATGACCTGTGAGATCACAAAG-3’
TEAD3: 5’-CCTGGAGTATTCAGCCTTCAT-3’
TEAD4: 5’-GAGACAGAGTATGCTCGCTAT-3’
MDA-MD-231 shTEAD_2 target sequences are the same as the target sequences for shTEAD_1 except for TEAD2 sequence: 5’-GCCTGAGCGATACATGATGAA-3’.
Detroit 562 shRNA target sequences for TEAD are as follows:
shTEAD1/3/4 #1 sequence 5’-ATGATCAACTTCATCCACAAT-3’
shTEAD1/3/4 #2 sequence 5’-GATCAACTTCATCCACAAGCT-3’
These sequences were cloned into PiggyBac (pBind, Genentech), similar to as described in [
24- Watanabe C.
- Cuellar T.L.
- Haley B.
Quantitative evaluation of first, second, and third generation hairpin systems reveals the limit of mammalian vector-based RNAi.
].
Cellular fractionation, Western blotting, and Immunoprecipitation
Cellular fractionation between the cytoplasm, nucleus and chromatin fraction was performed using a modified protocol established by Mendez
et al. [
25Chromatin association of human origin recognition complex, cdc6, and minichromosome maintenance proteins during the cell cycle: assembly of prereplication complexes in late mitosis.
] using fresh cell pellets resuspended in buffer A (10 mM HEPES, 10mM KCl, 1.5 mM MgCl
2, 340 mM Sucrose, 10% Glycerol, 1 mM DTT, 0.1% Triton-X, 1X Halt™ Protease and Phosphatase Inhibitor Cocktail (Thermo Fisher Scientific)). The lysates were incubated on ice for 5 min. Then, the nuclear fraction was isolated by centrifugation (1300 x g) for 4 min at 4 °C. The supernatant (cytoplasm) was collected, and the nuclear pellet was washed several times with buffer A. The nuclei were lysed using buffer B (3 mM EDTA, 0.2 M EGTA, 1 mM DTT, 1X Halt™ Protease and Phosphatase Inhibitor Cocktail). The supernatant (nucleoplasm) was collected following centrifugation (1700 x g) for 4 min at 4 °C. Then, the cell pellet was washed several times with buffer B. The cell pellet (chromatin) was lysed with RIPA buffer (Thermo Fisher Scientific) according to manufacturer’s instructions, supplemented with 2 mM MgCl
2, universal nuclease (Thermo Fisher Scientific), and 1X Halt™ Protease and Phosphatase Inhibitor Cocktail (Thermo Fisher Scientific) for 20 min on ice. Western blotting for proteins of interest were lysed using RIPA buffer according to manufacturer’s instructions supplemented with 2 mM MgCl
2, universal nuclease, and 1X Halt™ Protease and Phosphatase Inhibitor Cocktail for 20 min on ice. Protein concentration was measured by Bradford reagent (5000205, BioRad) using SpectraMax M5 (Molecular devices) at 595 nm. Cell lysates were run on a 4-12% Bis-Tris Plus protein gel (Thermo Fisher Scientific), and subsequently transferred onto a nitrocellulose membrane using Transblot Turbo transfer system (BioRad). The membrane was blocked, incubated with primary antibodies in 5% milk for 24 h at 4 °C. After several washes with TBST, membranes were incubated with secondary antibodies: Goat anti-mouse (926-3220, Li-cor), Goat-anti-rabbit (926-68071, Li-Cor). Images were acquired using the Odyssey Clx (Li-Cor) or by SuperSignal West Pico ECL (1856136, Thermo Fisher Scientific)
For immunoprecipitation experiments, expression was induced by doxycycline treatment of MYC/FLAG WT or NLS mutant TEAD3 HEK293 or Detroit 562 cells for 48 h. Cells were harvested, washed with ice-cold PBS, and lysed using 20 mM Tris-HCl pH 7.4, 150 mM NaCl, 0.5% NP-40, 2 mM MgCl2, universal nuclease (Thermo Fisher Scientific), and 1X Halt™ Protease and Phosphatase Inhibitor Cocktail (Thermo Fisher Scientific) for 20 min on ice. 20 mg/mL of whole cell lysates were clarified by centrifugation at 4000 x g 30 min at 4 °C. Immunoprecipitation was carried out on cell lysates using Myc-tag antibody with protein A and G Dynabeads magnetic beads (Thermo Fisher Scientific) according to manufacturer’s instructions and Mouse IgG1 Isotype Control was used as control to preclear the protein lysates and used for IP. Immunoprecipitated proteins were eluted from the magnetic beads at 95 °C for 10 min in 2X NuPAGE® LDS Sample Buffer (Thermo Fisher Scientific) without any reducing agent.
In-gel reduction/alkylation and tryptic digestion
In preparation for mass spectrometry analysis, samples were separated on 4-12% Bis-Tris gel (NW04120, Thermo Fisher Scientific) under non-reduced conditions to separate any antibody species from where TEAD proteins run ∼55 kDa. Proteins were stained with SimplyBlue stain (Invitrogen) and de-stained in water. The gel was excised from top to bottom into 14 bands per lane. Gel pieces were further de-stained in 50 mM ammonium bicarbonate (NH4HCO3)/30% acetonitrile (ACN) and dehydrated in 100% ACN. In-gel reduction was performed in 50 mM NH4HCO3 containing 50 mM of dithiothreitol (DTT) at 37 °C for 1 h, followed by a quick wash with 50 mM NH4HCO3 buffer. Alkylation was done using 50 mM iodoacetamide (IAA) at room temperature for 20 min in the dark. Excess amount of IAA was removed by washing the gel pieces with 50 mM NH4HCO3 /30% ACN followed by dehydration in 100% ACN. In-gel tryptic digestion was performed by hydrating the gel pieces in 10 ng/μL trypsin solution in 25 mM NH4HCO3 and chilled on ice for 1 h. Excess trypsin solution was removed and digestion was performed overnight in 25 mM NH4HCO3 at 37 °C. Peptides were extracted with 0.1% trifluoroacetic acid (TFA) in ACN. Peptides were dried to completion and re-suspended in 2% ACN/0.1% formic acid (FA)/water.
LC-MS/MS analysis
Samples were reconstituted in solvent A (2% ACN/0.1% FA/water) and injected via an auto-sampler for separation by reverse phase chromatography on a NanoAcquity UPLC system (Waters, Dublin, CA). Peptides were loaded onto the Symmetry ® C18 column (1.7 mm BEH-130, 0.1 x 100 mm, Waters, Dublin, CA) with a flow rate of 1 μL/min. A gradient of 2% solvent B to 25% solvent B (solvent A is 0.1% FA/2% ACN/water and solvent B is 0.1% FA/2% water/ACN) was applied over 35 min with a total analysis time of 60 min. Peptides were eluted directly into an Advance CaptiveSpray ionization source (Michrom BioResources/Bruker, Auburn, CA) with a spray voltage of 1.3 kV, and were analyzed using an LTQ Orbitrap Elite mass spectrometer (Thermo Fisher Scientific). Precursor ions were acquired in the FTMS at 60,000 resolution; MS/MS was performed in the LTQ using resonance excitation, with the instrument operated in data-dependent mode, whereby the 15 most abundant ions were selected for fragmentation in each duty cycle.
Mass spectrometry data analysis and statistical analysis
For peptide identification, MS/MS spectra were searched using the search algorithm Mascot version 2.4.1 (Matrix Sciences, London, UK) against the concatenated target-decoy database comprised of UniProt human protein sequences (UniProt, version 2016_06 that includes 42,279 Swissprot sequences of canonical and protein isoforms and 134,256 unreviewed TREMBL sequences), one custom NLS mutant TEAD sequence, known contaminants, and the reversed versions of each sequence. A 50 ppm precursor ion mass tolerance and 0.8 Da fragment ion tolerant were selected with tryptic specificity with up to 3 miscleavages. Variable modifications were permitted for methionine oxidation (+15.9949 Da), IAA adduct for cysteine residues (+57.0215 Da), phosphorylation on serine, threonine, and tyrosine (+79.9663 Da), and ubiquitination on lysine residues (+114.0429 Da)
. Peptide assignments were first filtered to a 1% false discovery rate (FDR) at the peptide level and subsequently at 2% FDR at protein level. We used SAINTexpress (Significance Analysis of INTeractome), which is a statistical method for probabilistically scoring protein-protein interaction data from AP-MS experiment (SAINTExpress-spc v.3.6.1) (Teo et al., 2014). We compared spectral counts for each bait IP sample (WT-TEAD3 or NLS-mutant TEAD3) and to control IP samples and to assign confidence scores to observed protein-protein interactions. Protein spectral counts for each sample were calculated at the sum from the peptide spectral counts (all modified and shared peptides) for one protein in a given sample, across all fractions from the GeLC-MS experiment. No filtering for protein spectral counts was performed. A single peptide is also used for protein quantification. The default setting for SAINTexpress was used (low Mod=0, minFold=1, normalization=1). We didn’t compress control or bait samples. Interactions with average sum PSMs > 10, a SAINT score > 0.99, and a Bayesian False Discovery Rate (BFDR) < 0.001 were marked as significant. Significant interactions for the WT and NLS mutant TEAD3 in each cell line were submitted for ‘Compute overlaps’ tool against the Hallmark gene sets from MsigDB (v2022.1.Hs updated August 2022) [
26- Subramanian A.
- Tamayo P.
- Mootha V.K.
- Mukherjee S.
- Ebert B.L.
- Gillette M.A.
- Paulovich A.
- Pomeroy S.L.
- Golub T.R.
- Lander E.S.
- Mesirov J.P.
Gene set enrichment analysis: a knowledge-based approach for interpreting genome-wide expression profiles.
] using the Broad institute’s online tool [
26- Subramanian A.
- Tamayo P.
- Mootha V.K.
- Mukherjee S.
- Ebert B.L.
- Gillette M.A.
- Paulovich A.
- Pomeroy S.L.
- Golub T.R.
- Lander E.S.
- Mesirov J.P.
Gene set enrichment analysis: a knowledge-based approach for interpreting genome-wide expression profiles.
,
27- Liberzon A.
- Birger C.
- Thorvaldsdottir H.
- Ghandi M.
- Mesirov J.P.
- Tamayo P.
The Molecular Signatures Database (MSigDB) hallmark gene set collection.
]. Hallmark gene sets with a q-value < 0.05 were marked as significant.
Immunofluorescence
Cells were either cultured on 1.5 glass coverslips (cat. 64-0714, Warner instruments) or 96 well plates, fixed with 4% paraformaldehyde for 10 min at room temperature, permeabilized with Triton buffer (0.5% Triton X-100, 20 mM HEPES, pH 7.9, 50 mM NaCl, 3 mM MgCl2, and 300 mM sucrose). Then, cells were washed with PBS (2x) followed by blocking using 2% BSA for 1 h at room temperature. Primary antibodies were incubated in 0.1% Tween-20 in 2% BSA overnight, followed by 3 washes with 0.1% Tween-20/PBS. Secondary antibodies were incubated for 1 h in 0.1% Tween-20 in 2% BSA. Cells were counterstained with DAPI for the last 10 min of secondary incubation, followed by 3 quick washes with 0.1% Tween-20/PBS. Coverslips were mounted onto slides using VectaMount (H-5000, Vector Labs.)
TEAD transcriptional reporter assay
Detroit 562 and MDA-MB-231 were transduced with a lentiviral vector expressing secreted NanoLuc luciferase under the control of multimerized 8xTEAD consensus element (5′-CACATTCCA-3′). Within the same construct, Firefly Luciferase is constitutively expressed from a PGK Promoter and serves as an internal control (schematic in
Figure S2A). 10,000 cells were seeded per well in a 96 well-plate and incubated for 24 h. Cells were then treated with 500 nM dasatinib, exposed to 6 Gy of γIR, or left untreated. At the indicated time points post treatment (4, 6, 24, 30, 48 h), TEAD reporter activity was assayed by performing Nano-Glo Dual-Luciferase Reporter Assay System (Promega N1610) following the manufacturer’s instructions. Plates were read on an Envision multimode microplate reader. TEAD reporter activity was calculated by normalizing the NanoLuc luminescence signal to the Firefly Luciferase signal, where the Luciferase signal represents the overall cell viability.
DNA repair reporter assay
To test homologous recombination (HR) efficiency, we used pDR-GFP in U-2 OS cells, originally developed in the Jasin lab [
28- Pierce A.J.
- Johnson R.D.
- Thompson L.H.
- Jasin M.
XRCC3 promotes homology-directed repair of DNA damage in mammalian cells.
]. siRNAs for various targets were transfected using Lipofectamine RNAiMAX (Thermo Fisher Scientific) according to the manufacturer’s instructions. After 24 h, transfection of the plasmid encoding I-SceΙ was performed with Lipofectamine 2000 (Thermo Fisher Scientific) according to the manufacturer’s instructions. Cells were allowed to repair the DSBs for at least 48 h before GFP analysis by FACS using Attune NxT Flow Cytometer. Recombination efficiencies were calculated as % GFP positive cells relative to siRNA control: (% GFP positive siRNA X) / (% GFP positive siRNA control) *100.
To test non-homologous end joining repair (NHEJ) efficiency, we used iHN HeLa-Tet cell line (DR5001-iHNHeLa-Tet, TopoGEN) according to manufacturer’s instructions. After siRNA transfection as above, cells were treated with 2 μg/mL doxycycline to induce I-Sce1 mediated DSBs for 24 h. Cells were then allowed to repair DSBs for at least 48 h before GFP analysis by FACS using BDCelesta analyzer (BD Biosciences). The % repair efficiency is the fraction of GFP positive cells in siRNA treated samples normalized to the fraction of GFP positive cells in control siRNA treated cells. Data were quantified using FlowJo software. Recombination efficiencies were calculated as % GFP positive cells relative to siRNA control: (% GFP positive siRNA X) / (% GFP positive siRNA control) *100.
Comet Assay
The Comet assay was performed using OxiSelect comet assay kit (STA-351, Cell Biolabs, Inc.) following manufacturer’s instructions. MDA-MB-231 cells expressing shNTC, shTEAD_1 or shTEAD_2 were treated with DMSO or bleomycin for 2 h. Cells were allowed to recover in fresh media for 24 h. Then, cells were gently trypsinized and pelleted by centrifugation. To form a base layer, 75 μL of molten LMAgarose was pipetted onto the comet slide and allowed to solidify at 4 °C for 15 min. Cells were resuspended at 1 × 105 cells/mL in cold PBS, followed by combining cells with molten LMAgarose at a ratio of 1:10 (v/v), and quickly pipetting 75 μL onto a comet slide, without disrupting the base gel layer. The slide was placed at 4 °C in the dark for 15 min, then immersed in pre-cooled lysis buffer and kept at 4 °C in the dark for 45 min. The comet slide was immersed in pre-chilled alkaline solution for 30 min. Then, the comet slide was transferred to a horizontal electrophoresis apparatus to perform alkaline electrophoresis at 20 V for 25 min. Samples were then rinsed three times with pre-chilled PBS for 5 min. Samples were dried at room temperature in the dark, then stained with 100 μL of diluted SYBR Green I Nucleic Acid Gel Stain (S7567, Thermo Fisher Scientific Scientific) for 30 min. Images were captured by a confocal microscope (SP5, Leica microsystems) from four different fields and data were analyzed by CometScore.
Survival assay
Fifty thousand cells were initially seeded in 6-well dishes before being exposed to 6 Gy γIR or being treated with the indicated concentrations of bleomycin or etoposide. At 24 h post γIR exposure or treatment, cells were incubated with fresh media and incubated for 5-6 days. Thereafter, cells were fixed with 10% glacial acetic acid and 10% methanol for 10 min and stained with 0.5% crystal violet. After several rinses with ddH2O and allowing the plates to dry, the crystal violet was solubilized with 0.01% SDS in methanol. The absorbance was measured using SpectraMax M5 (Molecular devices) at 595 nm.
Cell Cycle Analysis
HeLa and U-2 OS cells were transfected with siControl or siTEAD using Lipofectamine RNAiMAX (Thermo Fisher Scientific) according to the manufacturer’s instructions. After 72 h, cells were treated with 10 μM EdU for 15 min at 37 °C, fixed with 4% PFA at room temperature, and permeabilized and blocked in PBS containing 10% FBS, 1% BSA, 0.1% TX-100, and 0.01% NaN
3 for 1 h at room temperature. Click reaction was then performed following manufacturer's instructions (Thermo Fisher plus EdU cell proliferation kit). Cells were washed with PBS, re-blocked for 30 min, and treated with Hoechst 33342 1:10000 in PBS for 10 min. After washing out Hoechst, cells were imaged on OperaPhenix (PerkinElmer). Images were segmented and quantified following the protocol outlined in [
29- Chung M.
- Liu C.
- Yang H.W.
- Koberlin M.S.
- Cappell S.D.
- Meyer T.
Transient Hysteresis in CDK4/6 Activity Underlies Passage of the Restriction Point in G1.
].
DISCUSSION
DNA damage response is a network of processes that safeguard genomes from deleterious endogenous and exogenous DNA damage. If left unrepaired, DNA damage can produce irreversible mutations that may lead to various deleterious effects, including cancer. Here, we identified binding partners for TEADs by performing semi-quantitative mass spectrometry analysis using cell lines overexpressing either WT-TEAD3 or NLS-mutant TEAD3. We found that PARP1, RIF1, and XRCC5 and XRCC6 were enriched in the WT-TEAD3 samples but were absent or significantly decreased in the NLS-mutant TEAD3 samples. In contrast, the canonical binding partners of TEADs, YAP, TAZ, and VGLL4, displayed no change in interaction between mutant and wildtype conditions. Because the NLS region of TEADs is highly conserved, we anticipate that the results we observe with TEAD3 are likely apply to all four TEAD paralogs, although future experimental testing is required to validate such interactions with TEAD1, TEAD2 and TEAD4. Notably, previous studies have identified XRCC5 and XRCC6 as interacting partners for TEAD2 and TEAD4 [
15- Wang Y.
- Xu X.
- Maglic D.
- Dill M.T.
- Mojumdar K.
- Ng P.K.
- Jeong K.J.
- Tsang Y.H.
- Moreno D.
- Bhavana V.H.
- Peng X.
- Ge Z.
- Chen H.
- Li J.
- Chen Z.
- Zhang H.
- Han L.
- Du D.
- Creighton C.J.
- Mills G.B.
- Cancer Genome Atlas Research N.
- Camargo F.
- Liang H.
Comprehensive Molecular Characterization of the Hippo Signaling Pathway in Cancer.
].
To evaluate the role of TEADs associating with these DNA damage markers, we used RNAi-based depletion systems targeting all four TEADs as well as agents that induce DNA damage. Upon the introduction of DNA damage in cells, we observed that TEADs are recruited to sites of DSBs, co-localizing with DNA damage markers γH2AX and RIF1. We also observed that interaction of TEADs with XRCC5 and XRCC6 is increased after DNA damage. Additionally, depletion of TEADs by RNAi reduces the efficient maintenance of genome stability, sensitizes cells to agents that cause double-stranded breaks, and reduces DNA repair in both HR and NHEJ GFP reporter assays. These cellular findings indicate that TEADs play a role in DNA damage response, however, the RNAi-centered approach for TEADs depletion we used in this study is an important limitation due to potential off-target effects that are inherent to RNAi. Follow up experiments that disrupt TEADs in other ways, such as with CRISPR approaches or with TEAD inhibitors, would provide valuable insights in evaluating the functional significance of TEADs in maintaining genomic stability.
Although TEADs function as transcription factors, perhaps they have an additional role in regulating DNA repair after DNA damage to restore cellular homeostasis. Several studies have shown that other transcription factors play an important role in DNA damage response; for instance, several transcription factors localize to DNA damage sites, some of which are PARP1-dependent [
40- Izhar L.
- Adamson B.
- Ciccia A.
- Lewis J.
- Pontano-Vaites L.
- Leng Y.
- Liang A.C.
- Westbrook T.F.
- Harper J.W.
- Elledge S.J.
A Systematic Analysis of Factors Localized to Damaged Chromatin Reveals PARP-Dependent Recruitment of Transcription Factors.
]. However, the exact physiological role of these transcription factors at the DNA damage sites remains unknown. Several independent studies have begun to unravel the role of transcription factors in DNA repair. Most notably, E2F transcription factor 2 (E2F2) functions to regulate genes involved in proliferation and apoptosis, and has been implicated to directly participate in DNA repair by forming DNA damage induced foci mediated by ATM and ATR after ultraviolet (UV)-induced DNA damage [
41- Biswas A.K.
- Mitchell D.L.
- Johnson D.G.
E2F1 responds to ultraviolet radiation by directly stimulating DNA repair and suppressing carcinogenesis.
,
42- Guo R.
- Chen J.
- Zhu F.
- Biswas A.K.
- Berton T.R.
- Mitchell D.L.
- Johnson D.G.
E2F1 localizes to sites of UV-induced DNA damage to enhance nucleotide excision repair.
]. ATF2 is another transcription factor belonging to the AP1 family of stress responsive transcriptional activators and has been shown to localize at sites of DSBs by ATM-induced phosphorylation [
43- Bhoumik A.
- Takahashi S.
- Breitweiser W.
- Shiloh Y.
- Jones N.
- Ronai Z.
ATM-dependent phosphorylation of ATF2 is required for the DNA damage response.
]. POU domain transcription factor, BRN2 has been shown to bind to and be recruited by PARP1 along with XRCC5 and XRCC6 to sites of ultraviolet B-induced DNA damage to suppress apoptosis [
44- Herbert K.
- Binet R.
- Lambert J.P.
- Louphrasitthiphol P.
- Kalkavan H.
- Sesma-Sanz L.
- Robles-Espinoza C.D.
- Sarkar S.
- Suer E.
- Andrews S.
- Chauhan J.
- Roberts N.D.
- Middleton M.R.
- Gingras A.C.
- Masson J.Y.
- Larue L.
- Falletta P.
- Goding C.R.
BRN2 suppresses apoptosis, reprograms DNA damage repair, and is associated with a high somatic mutation burden in melanoma.
]. These studies suggest that transcription factors play an important role in DNA damage response and repair; the details of how TEADs may function in DNA damage is only beginning to come to light.
The Hippo pathway is tightly networked into a variety of cellular functions, including DNA damage. For example, YAP is known to respond to DNA damage signals. Levy
et al. have demonstrated that upon DNA damage, ataxia telangiectasia mutated (ATM) phosphorylates c-Abl tyrosine kinase, and activated c-Abl in turn phosphorylates YAP [
45- Levy D.
- Adamovich Y.
- Reuven N.
- Shaul Y.
Yap1 phosphorylation by c-Abl is a critical step in selective activation of proapoptotic genes in response to DNA damage.
]. This phosphorylation stabilizes YAP and promotes interaction with p73, leading to transcriptional activation of proapoptotic genes, including promyelocytic leukemia tumor suppressor (PML). PML is then able to bind to YAP and regulates YAP stability [
45- Levy D.
- Adamovich Y.
- Reuven N.
- Shaul Y.
Yap1 phosphorylation by c-Abl is a critical step in selective activation of proapoptotic genes in response to DNA damage.
,
46- Lapi E.
- Di Agostino S.
- Donzelli S.
- Gal H.
- Domany E.
- Rechavi G.
- Pandolfi P.P.
- Givol D.
- Strano S.
- Lu X.
- Blandino G.
PML, YAP, and p73 are components of a proapoptotic autoregulatory feedback loop.
]. In a parallel DNA damage response pathway, ATM activation by DNA damage phosphorylates the Hippo pathway activator, RASSF1A, and stimulates the activity of MST1/2 and LATS1/2 to promote YAP and p73 binding [
47Hippo pathway and protection of genome stability in response to DNA damage.
,
48- Reuven N.
- Adler J.
- Meltser V.
- Shaul Y.
The Hippo pathway kinase Lats2 prevents DNA damage-induced apoptosis through inhibition of the tyrosine kinase c-Abl.
]. These findings outline a tumor suppressive mechanism by which YAP-mediated protein interactions become induced by DNA damage to promote apoptosis [
49- Basu S.
- Totty N.F.
- Irwin M.S.
- Sudol M.
- Downward J.
Akt phosphorylates the Yes-associated protein, YAP, to induce interaction with 14-3-3 and attenuation of p73-mediated apoptosis.
]. Here, we describe our observations that TEADs may also respond to DNA damage. We found that TEADs localize to DNA damage-induced nuclear foci even in cells that do not express YAP and TAZ, indicating that the function of TEADs in response to DNA damage is separate from interaction with YAP and TAZ. The details of what mechanistic role TEADs play in mediating DNA damage repair is still to be determined.
Over the past decade the Hippo pathway proteins YAP, TAZ, and TEADs have been shown to be active players in tumorigenesis, metastasis, resistance to chemotherapies, and targeted immunotherapies [
8- Harvey K.F.
- Zhang X.
- Thomas D.M.
The Hippo pathway and human cancer.
,
19- Calses P.C.
- Crawford J.J.
- Lill J.R.
- Dey A.
Hippo Pathway in Cancer: Aberrant Regulation and Therapeutic Opportunities.
,
50- Moon S.
- Yeon Park S.
- Woo Park H.
Regulation of the Hippo pathway in cancer biology.
]. Inhibitors to TEAD, aimed at disrupting protein-protein interactions, are in development and advancing in the clinic. Thus, deepening our understanding of the protein interactions involving TEADs are crucial for effectively evaluating the therapeutic potential and impact of TEAD inhibitors. Our study suggests that knockdown of TEADs slows cellular proliferation, and that treatment with γ-irradiation or chemotherapeutic agents can further slow cell growth and reduce DNA repair, leading to accumulation of DNA lesions. A combination of a TEAD inhibitor and chemotherapy may have a potential clinical benefit. Clearly, the TEADs-DNA damage connection deserves further scrutiny moving forward.
AUTHOR CONTRIBUTIONS
Conceptualization: P.C.C, J.R.L., A.D.; Methodology: P.C.C., V.C.P.; Formal analysis: P.C.C., V.C.P., E.V., S.T.B., C.L., M.T.C., T.H., C.B., M.C.; Investigation: P.C.C., V.C.P., S.T.B., C.L.; Resources: B.H., N.K., J.Z., C.Y., X.S., X.L., R.R., A.A.; Data Curation: V.C.P., A.D.G., T.H., C.B., M.C., T.P.; Writing – Original Draft: P.C.C, J.R.L., A.D.; Writing - Review & Editing: P.C.C, V.C.P., A.D.G., J.R.L., A.D., T.P.; Visualization: P.C.C., V.C.P., A.D.G., E.V., C.B., M.C.; Supervision: J.R.L., A.D.; Project administration: J.R.L., A.D.; Funding acquisition: A.A., J.R.L., A.D.
Article info
Publication history
Accepted:
January 4,
2023
Received in revised form:
December 15,
2022
Received:
September 23,
2022
Publication stage
In Press Accepted ManuscriptFootnotes
This manuscript describes an affinity purification mass spectrometry approach to evaluate the protein interactions of TEAD transcription factors and identifies DNA damage-associated proteins as nuclear interacting partners for TEADs. Follow up validation experiments with biochemical and cellular assays finds that TEADs colocalize with DNA damage-induced foci and that depletion of TEADs in cells decreases DNA repair efficiencies and promotes genomic instability. These experiments indicate a connection between TEAD transcription factors and DNA repair processes.
Alan Ashworth is an employee of UCSF. Other authors are affiliated with Genentech Inc.
For transparency, authors are required to submit an author statement file outlining their individual contributions to the paper using the relevant CRediT roles: Conceptualization; Data curation; Formal analysis; Funding acquisition; Investigation; Methodology; Project administration; Resources; Software; Supervision; Validation; Visualization; Roles/Writing - original draft; Writing - review & editing.
Philamer C. Calses1,2*, Conceptualization; Data curation; Formal analysis
Victoria C. Pham2*Alissa D. Guarnaccia1,2* Roles/Writing - original draft; Writing - review & editing
Meena Choi2, Data curation; Formal analysis
Erik Verschueren2, Data curation; Formal analysis
Sietske T. Bakker3, Data curation; Formal analysis
Trang H. Pham1, Data curation; Formal analysis
Trent Hinkle2, Data curation; Formal analysis
Chad Liu1, Data curation; Formal analysis
Matthew T. Chang4, Data curation; Formal analysis
Noelyn Kljavin5, Data curation; Formal analysis
Corey Bakalarski2, Data curation; Formal analysis
Benjamin Haley1, Data curation; Formal analysis
Jianing Zou6, Data curation; Formal analysis
Cuicui Yan6, Data curation; Formal analysis
Xia Song6, Data curation; Formal analysis
Xiaoyan Lin6, Data curation; Formal analysis
Rebecca Rowntree5, Data curation; Formal analysis
Alan Ashworth3, Data curation; Formal analysis
Anwesha Dey1#, Conceptualization; Data curation; Formal analysis Roles/Writing - original draft; Writing - review & editing.
Jennie R. Lill2# Conceptualization; Data curation; Formal analysis Roles/Writing - original draft; Writing - review & editing.
Copyright
© 2023 THE AUTHORS. Published by Elsevier Inc on behalf of American Society for Biochemistry and Molecular Biology.