1. Introduction
Despite decades of research and significant advances in our understanding of the molecular and cellular events that drive cancer development and progression, cancer remains a leading cause of death worldwide. Most cancer deaths result from metastatic disease, which requires systemic therapies because it cannot be effectively treated surgically. In melanoma specifically, cytotoxic chemotherapies are largely ineffective, so most patients are treated with immune therapies and targeted therapies. Immune therapies have proven successful, but not all patients have durable responses, and some patients are contraindicated due to other health issues. Targeted therapies, such as BRAF and MEK inhibitors, can also be effective, but patients typically develop resistance. Therefore, there is an urgent need for new targeted therapies and a way to predict which patients will benefit from them.
Yes-associated protein (YAP) and transcriptional co-activator with PDZ-binding motif (TAZ) are inappropriately active in many cancer types [
4,
5,
6,
7], including melanoma [
1,
8,
9,
10]. Hundreds of studies collectively demonstrate that increased YAP or TAZ activity can enhance tumor formation and growth, and also promote tumor progression and metastasis [
4,
6,
7,
11,
12]. YAP and TAZ are negatively regulated by the Hippo Pathway, a serine/threonine kinase cascade that, when active, results in LATS-mediated phosphorylation of multiple serine residues in YAP and TAZ, promoting either cytoplasmic sequestration or proteasomal degradation [
13,
14,
15,
16]. In addition, numerous other cellular pathways can influence YAP and TAZ function either by regulating the Hippo Pathway or through Hippo Pathway-independent mechanisms [
17,
18,
19]. As transcriptional co-activators, YAP and TAZ regulate gene expression programs to mediate their effects on cells. However, both lack DNA binding domains, so they must partner with other transcription factors to regulate target gene expression. Although YAP and TAZ can interact with numerous transcription factors, the TEAD family members play critical roles in YAP/TAZ-dependent gene expression [
20,
21].
Given the established roles for YAP, TAZ, and TEADs in cancer and other diseases, there has been substantial interest in developing compounds to target these proteins. Although YAP and TAZ have proven difficult to target directly, compounds that target the TEADs, and thus prevent YAP/TAZ-TEAD-mediated transcription, have great promise [
21,
22]. Indeed, numerous compounds that target YAP/TAZ-TEAD have been described (reviewed in [
23]) and 3 TEAD inhibitors (VT3989, IK-930, IAG933) have recently entered early phase clinical trials in cancer patients (NCT04665206, NCT05228015, and NCT04857372). An antisense oligonucleotide inhibitor of YAP1 that proved effective in pre-clinical models has also entered clinical trials (NCT04659096). However, as is the case for most targeted therapies, the success of these treatments will depend upon whether tumors that are dependent upon YAP/TAZ-TEAD can be distinguished from those that are not. This will be straightforward for cancers driven by mutations known to cause YAP or TAZ activation, such as NF2-mutant mesothelioma [
24], GNAQ mutant uveal melanoma [
25], Epithelioid Hemangioendothelioma (EHE), which is driven by the oncogenic TAZ-CAMTA1 fusion protein [
26,
27,
28], and other cancers driven by YAP fusions [
29]. However, in most of the cancers where YAP and TAZ play causal roles, mutations or alterations in the Hippo Pathway or YAP and TAZ themselves are rare, so biomarkers that can predict sensitivity to YAP/TAZ-TEAD inhibition will be essential. Given the complexities of how YAP and TAZ are regulated, increased protein expression or nuclear localization do not necessarily indicate increased YAP/TAZ-TEAD activity. The expression of YAP/TAZ-TEAD target genes could provide a more direct readout for YAP/TAZ activation, but whether they can predict sensitivity to YAP/TAZ-TEAD inhibition remains unclear.
Here, we describe the development of a YAP/TAZ gene signature from metastatic human melanoma cells, which is also highly enriched in cell lines dependent upon YAP, TAZ, or TEADs. Despite being developed from melanoma cell lines, we show that the genes in this signature are YAP/TAZ-dependent in other cancer types and that this signature is predictive of cancer cell dependence upon YAP, TAZ, and TEAD. This work suggests that this YAP/TAZ-TEAD gene signature, and others like it, could have diagnostic value for identifying cancer patients that will benefit from TEAD inhibitors.
2. Materials and Methods
Cell lines, vectors, and cloning: Human melanoma cell lines (A375, A375-MA2, A2058) were cultured in growth media (DMEM+ 10% fetal bovine serum, 2 mM L-glutamine) at 37° C and 5% CO2 and maintained at low passage number. Cell lines were routinely tested for mycoplasma and other bacterial contaminants. A375 and A2058 cells were obtained from ATCC. A375-MA2 cells were derived from A375 by Richard Hynes [
30]. All vectors used in this work are listed in
Table S1. If vectors were previously described, received as gifts, or purchased from commercial vendors, the source of the vector is listed. New vectors were generated using standard cloning procedures, and the source constructs used for each insert and vector backbone are indicated.
Generation of retrovirus and lentivirus: Retrovirus and lentivirus were packaged as described previously [
31]. Briefly, 293FT cells were plated on 6-well plates at roughly 50% confluence in growth media. After 16-24 hours, cells were transfected (according to the manufacturer’s protocol) with a transfection mixture containing 1 μg of viral vector, 0.5 μg of packaging vector (gag/pol), 0.5 μg of coat protein (VSVG), 5 μL of X-tremeGENE™ 9 (Sigma-Aldrich, Cat#6365779001), and 95 μL of Opti-MEM™ (Thermo Fisher Scientific, Cat#31985062). The transfection mixture was added to the cells for 24 hours, after which, the mixture was removed, and the cells were fed with fresh growth media. Culture supernatant was collected and filtered through a 0.45 μm filter 24 hours later. For stable transduction, cells at roughly 60-80% confluence were incubated with viral supernatant diluted 1:1 with fresh growth media and Polybrene (Sigma-Aldrich, Cat#45-H9268) (final concentration 8 µg/mL) for 24 hours and then viral supernatants were removed, and cells were fed with fresh growth media and stably selected with the appropriate antibiotic.
RNAi: siRNA experiments used Horizon Discovery SMARTPools and included a non-targeting control siRNA SMARTPool (Horizon Discovery ON-TARGETplus Non-targeting siRNA #1, Cat#D-001810-01-05) or SMARTPools targeting human YAP (Horizon Discovery ON-TARGETplus Human YAP1 (10413) siRNA, Cat#L-012200-00-0010) and human TAZ (Horizon Discovery ON-TARGETplus Human WWTR1 (25937) siRNA, Cat#L-016083-00-0010). Cells were plated at 4x105 cells per 6 cm well and cultured in growth media for 24 hours. A transfection mixture containing 9 μL (90 pmol) of siRNA, 27 μL of Lipofectamine™ RNAiMAX (Thermo Fisher Scientific Cat#13778075) and 900 μL Opti-MEMTM was setup according to the manufacturer’s protocol. For the combined knockdown of both YAP and TAZ, 4.5 μL (45pmol) of each siRNA SMARTPool was used so that the total volume of siRNA remained 9 μL (90 pmol). Twenty-four hours after the transfection, the cells were trypsinized and plated for Western blots or qPCR and cultured for an additional 48 hours in growth media before lysing as described below.
YAP/TAZ transcriptional activity: The YAP/TAZ-TEAD transcriptional reporter assays utilized a TEAD reporter construct (pGL3-5xMCAT(SV)-49 [
31,
32] that consists of 5 repeats of a TEAD binding element upstream of a minimal SV40 promoter that drives expression of the gene the encodes Firefly Luciferase. A control vector encoding a constitutively expressed
Renilla luciferase (PRL-TK (Promega, Cat#E2231)) is co-transfected and used for normalization. Cells were plated on a 12-well in duplicate in 1 mL of growth media. Aer 24 hours cells were co-transfected with a transfection mixture containing 400 ng of a 20:1 mixture of pGL3-5xMCAT(SV)-49 and PRL-TK, 4 μL/well of Lipofectamine 3000, 2 μL/well of the P3000 reagent (Invitrogen, Cat#L3000001) and 94 μL of Opti-MEM. After 24 hours, luciferase activity was assayed using the Dual-Luciferase Reporter Assay System (Promega, Cat#E1910) and normalized luciferase levels were calculated as described previously [
31]. For some experiments, cells were transfected with siRNAs or infected with viral constructs prior to assaying luciferase activity.
Western blotting and qPCR: For Western blots, cells were lysed in Cell Lysis Buffer (Cell Signaling Technology, Cat#9803) containing Pierce™ Protease Inhibitor Mini Tablets (Thermo Fisher Scientific, Cat#88665) and Pierce™ Phosphatase Inhibitor Mini Tablets (Thermo Fisher Scientific, Cat#88667). Protein concentration was determined by Pierce™ BCA protein assay kit (Thermo Fisher Scientific, Cat#23225) and equal protein (20-30 μg) was subjected to 10% SDS-PAGE, transferred to nitrocellulose membranes, and assayed by Western blot. The following primary antibodies were used at 1:1000 dilutions: total YAP (D8H1X) XP (Cell Signaling Technology, Cat#14074); total YAP (Cell Signaling Technology, Cat#4912,); total TAZ (V386) (Cell Signaling Technology, Cat#4883); and GAPDH Cell Signaling Technology, Cat#2118). The following horseradish-peroxidase-conjugated secondary antibodies were used at 1:5000 dilutions: goat anti-rabbit IgG (Thermo Fisher Scientific, Cat#31460); and goat anti-mouse IgG (Thermo Fisher Scientific, Cat#32430). Primary and secondary antibodies were diluted in 5% BSA. Western blotting images were captured with a Fujifilm LAS-3000 gel imager. For qPCR, cells were lysed with TRIzol (Thermo Fisher Scientific, Cat#15596018) and RNA was isolated following the manufacturer’s protocol. cDNA was made from 200 ng of the total RNA using qScript cDNA SuperMix at 42 °C (QuantaBio, Cat#95048) following the manufacturer’s protocol. qPCR reactions were carried out on 2 μL of cDNA, using 2 pmol of each primer (Table S1) and 10 µL of iTaq Universal SYBR Green (Bio-Rad, Cat#1725120). The reaction mixture was brought to a total of 20 µL with nuclease-free water. qPCR reactions were run using MyiQTM real-time PCR detection system according to the manufacturer’s instructions (Bio-Rad, Cat#1855201). PCR conditions were 95°C for 30 seconds, followed by 40 cycles of 95°C for 10 seconds, 60°C for 30 seconds, followed by a melt temperature analysis. For data processing, the Bio-Rad CFX Maestro software was used to calculate the fold change in mRNA for each indicated gene for each sample relative to a pre-determined control sample using the ΔΔCt method and GAPDH as a reference gene.
In vivo metastasis assays: The Albany Medical College Institutional Animal Care and Use Committee approved all mouse studies. Mice were housed in specific pathogen-free conditions in the Albany Medical College Animal Resources Facility, which is licensed by both the USDA and the NYS Department of Health, Division of Laboratories and Research, and accredited by the AAALAC. These studies used immunocompromised NOD/Scid mice (NOD/MrkBomTac-Prkdcscid, Taconic). To assay metastatic colonization, fluorescently labeled A375 cells expressing the indicated constructs were injected into the lateral tail veins of mice at 1x106 cells per mouse in 100 μL of PBS, and after 6 weeks, mice were euthanized, and lung metastases were counted using a fluorescent stereomicroscope.
RNA-seq: A375 cells stably expressing the indicated constructs were cultured in growth media, lysed with TRIzol (Thermo Fisher Scientific, Cat#15596018), and then RNA was isolated following the manufacturer’s protocol. cDNA was prepared with Illumina TruSeq chemistry and libraries were prepared using SPRIworks (Beckman Coulter) and sequenced using TruSeq SBS Kit v3 on the Illumina HiSeq2000. Sequence reads were aligned to the UCSC known genes version 2012 hg19 human assembly with using bowtie2 version 2.0.0-beta6 [
33] and tophat version 2.0.4 [
34]. Transcript assembly, gene summary, and differential expression was performed using cufflinks version 2.0.2 [
35]. RNA-seq data is available in the NCBI Gene Expression Omnibus (GSE234083).
Bioinformatic Analyses: Publicly available gene expression datasets downloaded from the NCBI Gene Expression Omnibus (
https://www.ncbi.nlm.nih.gov/geo/) are listed in
Table S1, Tab 3. Analysis of datasets downloaded from The Cancer Genome Atlas (TCGA) (
https://www.cancer.gov/tcga), The Broad Institute Cancer Dependency Map (DepMap) Portal (
https://depmap.org/portal/), or The ENCODE Project Database (
https://www.encodeproject.org/) [
36] are described below. Metascape [
2] was accessed via this link
https://metascape.org. For GSEA, downloaded NCBI-GEO, TCGA, or DepMap datasets were used to generate .gct files, and .cls phenotype files were generated for the indicated comparisons. GSEA [
37,
38] was run to test for enrichment of genesets described in
Table S1, Tab 4 using the appropriate CHIP platform and default settings. Probe/transcript Ids were collapsed to gene symbols, 1000 permutations were run, and the permutation type used was “phenotype” if the number of samples in each group was 8 or more, or “geneset” if the number of samples in each group was less than 8. Rank-ordered lists were generated by running GSEAPreranked with the same settings described above. Heatmaps were generated using MORPHEUS (
https://software.broadinstitute.org/morpheus/). To calculate Z-Scored expression, TPM values were converted to log2(1+x) and Z-Scored in Morpheus. Similarity matrices were generated from the Z-Scored expression values by first calculating the Spearman’s Rank Correlation of each gene with either
CTGF or
CYR61 using the nearest neighbor function and 1000 permutations. Genes were then sorted by Spearman’s Rank Correlation values (highest to lowest) and the Similarity Matrix function was used to generate the Spearman’s Rank Correlation for each pairwise comparison of genes in the dataset.
ENCODE: Conservative IDR Threshold Peaks for the indicated ChIP-Seq datasets were downloaded from the ENCODE Portal [
36] (
https://www.encodeproject.org/) (
Table S1, Tab 5). Using the ChIPseeker package in R [
39], the datasets were annotated based on the GRCh38 genome assembly, with the transcriptional start sites being defined as +/- 1kb. ENTREZ IDs for annotated peaks were converted to gene symbols using the ENSEMBL Database Homo Sapiens V86 as a reference.
DepMap: The RNA-seq data file “OmicsExpressionProteinCodingGenesTPMLogp1.csv” was downloaded from “DepMap Public 23Q2 Primary Files” on The Broad Institute Cancer Dependency Map (DepMap) Portal (
https://depmap.org/portal/). Dependency scores for
TEADs 1-4,
YAP1, and
WWTR1 (TAZ) were downloaded from DepMap. The RNA-seq data from the 1019 cell lines that have reported
YAP1,
WWTR1, and
TEADs dependency scores were analyzed for enrichment of genesets using GSEA analysis as described above. To generate ROC curves, the GSVA package in R [
40] was used to calculate enrichment scores for the indicated genesets in the indicated cell lines, and the resulting GSVA enrichment scores were used to perform ROC curve analyses using MedCalc for Windows, version 22.013 (MedCalc Software, Ostend, Belgium). Cell lines were considered dependent upon a gene if the Chronos Dependency Score was
< -0.65.
The Cancer Genome Atlas (TCGA): RNA-seq data from 303 human melanoma samples in the TCGA-Skin Cutaneous Melanoma (SKCM) project was downloaded from the NCI Genomic Data Commons Data Portal. Only datasets with an RNA integrity number between 7-10 were used. The transcripts per million (TPM) data for each sample was compiled and used for GSEA analysis, to generate heatmaps, and for similarity matrix calculations.
Statistical Analyses: Statistical analyses were performed in GraphPad Prism. The statistical test used to determine significance and the number of n’s is indicated in the legends. All scatter plots show mean + S.D unless noted otherwise in the legend.
Author Contributions
Conceptualization, J.M.L., R.O.H., E.N., R.K., P.S.; methodology, J.M.L., R.O.H., E.N., R.K., P.S.; validation, J.M.L., E.N., R.K., P.S.; formal analysis J.M.L., E.N., R.K., P.S.; investigation, J.M.L., E.N., R.K., P.S.; resources, J.M.L., R.O.H.; data curation, J.M.L., E.N., R.K., P.S.; writing J.M.L., E.N., R.K.; writing—review and editing, J.M.L., R.O.H., E.N., R.K., P.S.; visualization, J.M.L., E.N., R.K.; supervision, J.M.L., R.O.H.; project administration, J.M.L., R.O.H.; funding acquisition, J.M.L., R.O.H.; All authors have read and agreed to the published version of the manuscript.