Following chemotherapy and relapse, high-risk neuroblastoma tumors harbor more genomic alterations than at diagnosis, including increased transcriptional activity of the Yes-associated protein (YAP), a key downstream component of the Hippo signaling network. Although YAP has been implicated in many cancer types, its functional role in the aggressive pediatric cancer neuroblastoma is not well-characterized. In this study, we performed genetic manipulation of YAP in human-derived neuroblastoma cell lines to investigate YAP function in key aspects of the malignant phenotype, including mesenchymal properties, tumor growth, chemotherapy response, and MEK inhibitor response. Standard cytotoxic therapy induced YAP expression and transcriptional activity in patient-derived xenografts treated in vivo, which may contribute to neuroblastoma recurrence. Moreover, YAP promoted a mesenchymal phenotype in high-risk neuroblastoma that modulated tumor growth and therapy resistance in vivo. Finally, the BH3-only protein, Harakiri (HRK), was identified as a novel target inhibited by YAP, which, when suppressed, prevented apoptosis in response to nutrient deprivation in vitro and promoted tumor aggression, chemotherapy resistance, and MEK inhibitor resistance in vivo. Collectively, these findings suggest that YAP inhibition may improve chemotherapy response in patients with neuroblastoma via its regulation of HRK, thus providing a critical strategic complement to MEK inhibitor therapy.

Significance:

This study identifies HRK as a novel tumor suppressor in neuroblastoma and suggests dual MEK and YAP inhibition as a potential therapeutic strategy in RAS-hyperactivated neuroblastomas.

Despite intense multimodal therapy, more than 50% of patients with high-risk neuroblastoma relapse with aggressive chemotherapy-resistant disease, stressing an exigent need for therapies targeting pathways that drive recurrence (1). Following treatment and relapse, high-risk neuroblastoma tumors display significantly more RAS/RAF/MAPK genetic alterations than at diagnosis (2, 3). Gene set enrichment analyses (GSEA) also show increased transcriptional activity of the Yes-associated protein (YAP) in posttherapy recurrent neuroblastoma tumors (4). YAP is a transcriptional coactivator that binds with TEA domain (TEAD) family transcription factors (TF) to regulate genes promoting organ growth, cell self-renewal, and survival (5–8). YAP, and its paralog TAZ, are negatively regulated by proteins of the Hippo pathway that phosphorylate YAP/TAZ, leading to their cytoplasmic retention and inactivation. When YAP is dephosphorylated, it translocates into the nucleus to bind TEAD and mediate transcription. YAP's aberrant transcription functionally contributes to many aspects of cancer formation and survival (8–12). YAP's transcriptional regulation of downstream target genes such as EGFR, SOX2, and OCT4 has been shown to promote the mesenchymal phenotype and resistance to therapy (8–13). In addition, YAP regulates genes associated with epithelial-to-mesenchymal transition, thus promoting metastasis in many cancers (14). In an in vivo metastatic mouse model of neuroblastoma, neuroblastoma tumors that had metastasized to the brain displayed increased levels of YAP, suggesting a potential microenvironmental and metastatic role for YAP (15). While YAP “genetic signatures” are associated with tumor recurrence and independently predict poor neuroblastoma patient outcome, the functional role for YAP itself in neuroblastoma has not been characterized (3, 15). Furthermore, given the lack of YAP mutations or upstream Hippo pathway mutations in relapsed neuroblastoma tumors, it is unclear how YAP transcriptional activity increases at or before tumor recurrence and whether it contributes to neuroblastoma relapse. We now show that standard cytotoxic chemotherapy induces both YAP expression and transcriptional activity in murine models of neuroblastoma. We also demonstrate that YAP promotes a mesenchymal phenotype to affect in vivo neuroblastoma tumor growth and therapy response, and further inhibits apoptosis in response to nutrient deprivation through a novel regulation of the proapoptotic protein, Harakiri (HRK).

Cell lines and patient-derived xenograft models

Human-derived neuroblastoma cell lines were obtained from the Children's Oncology Group Childhood Cancer Repository and the ATCC. Neuroblastoma cell lines were cultured in RPMI1640 (Sigma) with 10% FBS (Gemini) and 1% penicillin–streptomycin (Gemini) and incubated at 37°C with 5% CO2 (16). All cells used were maintained at low passage, which did not exceed 15 passages. Cell lines and transduced cell models underwent short tandem repeat–based genotyping (Texas Tech Cancer Cell Repository) and identities were verified using the Children's Oncology Group cell line genotype database (www.cccells.org). Cells were routinely tested for Mycoplasma with MycoAlert Mycoplasma Detection Kit (Lonza). Patient-derived xenografts (PDX) were created from fresh deidentified human tumor tissue obtained from patients at diagnosis or at relapse under a Children's Healthcare of Atlanta Biorepository (Atlanta, GA) protocol approved by the institutional review board (IRB00034535). Written informed consent was obtained from patients/families for tumor tissue procurement, including cell line and PDX derivation, and deidentification for research investigations. Studies were conducted in accordance with recognized ethical guidelines (Declaration of Helsinki).

qRT-PCR

RNA was purified from neuroblastoma cell lines and snap-frozen xenograft tumors using the RNeasy Mini Kit (Qiagen) and TRIzol (Ambion) and Chloroform (Sigma), respectively. cDNA was created using the high-capacity cDNA Reverse Transcription Kit (Applied Biosystems). Power SYBR Green PCR Master Mix (Applied Biosystems) and the Bio-Rad System were utilized for quantitative PCR, with samples run in triplicate. Relative gene expression was normalized to GAPDH and HPRT using the CFX Manager Software (Bio-Rad). Primer sequences are detailed in Supplementary Table S1.

Western blot analysis

Snap-frozen xenograft tumors were dissociated with mortar and pestle. Neuroblastoma cell lines were consistently harvested at 80% confluency, as YAP expression can be influenced by cell contact (17). Neuroblastoma cells were lysed with CHAPS buffer (10 mmol/L HEPES, 150 mmol/L NaCl, and 2% CHAPS) with protease inhibitors (Roche Protease Inhibitor Cocktail 1×, 2 mmol/L sodium orthovanadate, and 2 mmol/L PMSF), run on Nu-PAGE 4%–12% Bis-Tris Gels (Invitrogen), transferred to polyvinylidene difluoride Membranes (Invitrogen), and detected for proteins by chemiluminescence as described previously (18).

Antibodies

Antibodies from Cell Signaling Technology used include: YAP (#4912, RRID:AB_2218911), phospho-YAP (Ser127) (#4911, RRID:AB_2218913), TAZ (V386) (#4883, RRID:AB_1904158), pan-TEAD (D3F7L) (#13295, RRID:AB_2687902), EGFR (C74B9) (#2646, RRID:AB_2230881), phospho-EGFR (Tyr1068) (#3777, RRID:AB_2096270), CYR61 (E5W3H) (#39382, RRID:AB_2799154), GAPDH (14C10) (#2118, RRID:AB_561053), BCL-XL (#2762, RRID:AB_10694844), BIM (#2933, RRID:AB_1030947), and cytochrome c (#11940, RRID:AB_2637071). Other antibodies used were: β-Tubulin (Santa Cruz Biotechnology, #sc-53140, RRID:AB_793543), BCL2 (Agilent, t# M0887, RRID:AB_2064429), and MCL1 (Enzo Life Sciences, #ADI-AAP-240, RRID:AB_10997659).

Stable transduction of YAP genetic inhibition and expression

YAP gene expression was stably knocked down in SK-N-AS (RRID:CVCL_1700) and NLF (RRID:CVCL_E217) using MISSION YAP shRNA Lentiviral Particles (Sigma, SHCLNV-NM_006106). Cells were transduced with shYAP or control vector (CV) containing lentiviral particles with 8 μg/mL polybrene for 48 hours, and then, transfectants were selected for and maintained in puromycin. For YAP expression in IMR5 (RRID:CVCL_1306) and NGP (RRID:CVCL_2141), pGAMA-empty (Addgene, plasmid #74755; RRID: Addgene_74755) and pGAMA-YAP (Addgene, plasmid #74942; RRID:Addgene_74942) were gifts from Miguel Ramalho-Santos (University of Toronto, Toronto, Canada). HEK293T (RRID:CVCL_0063) cells were transfected with 3 μg of plasmid DNA, pMD2.G (gift from Didier Trono, EPFL, Lausanne, Switzerland; Addgene, plasmid #12259; RRID:Addgene_12259) and psPAX2 (gift from Didier Trono; Addgene, plasmid # 12260; RRID:Addgene_12260) using Fugene 6 (Promega) transfection reagent protocol at ratio of 1:3 (plasmid DNA:Fugene 6). Plasmid containing viral particles were collected from the supernatant and used to transduce neuroblastoma cells.

Cell proliferation assays

Cells were plated in triplicate at 10,000 cells/well and CellTiter-Glo (Promega) was used to quantify live cells at different timepoints.

Colony formation assays

Cells were plated at 2,000 cells/well and cultured for 7 days. Cells were fixed with formaldehyde (1%) and stained with crystal violet (0.1%, w/v) in 20% methanol. Wells were imaged with a Lionheart FX Microscope (BioTek) and quantification was performed with ImageJ (NIH; RRID:SCR_003070).

Neurosphere assays

Cells were plated at 15,000 cells/mL in neurosphere media (50/50 F12/DMEM, Thermo Fisher Scientific; 1× B27, Thermo Fisher Scientific; 1× N2, Thermo Fisher Scientific; 0.1 mmol/L BME, Sigma; 2 μg/mL heparin, Stemcell Technologies; 1% penicillin–streptomycin, Gemini; 20 ng/mL EGF, Corning; and 40 ng/mL FGF, Corning) and cultured in Ultra-Low Attachment Plates (Corning). Neurospheres were harvested after 7 days for qRT-PCR. For quantification assays, cells were plated in triplicate at a density of 2,000 cells/100 μL in each well and grown for 7 days, and then imaged.

In vitro drug studies

Cells were plated in triplicate at 10,000 cells/well. After 24 hours, cells were treated with vehicle (DMSO), etoposide (Sigma), irinotecan (Sigma), topotecan (Sagent Pharmaceuticals), and melphalan (Sigma) for 48 hours, or trametinib (Selleck Chemicals) for 72 hours and evaluated by CellTiter-Glo (Promega).

Cytosolic/mitochondrial fractionation

Harvested cells were suspended in subcellular fractionation buffer (250 mmol/L sucrose, 20 mmol/L HEPES, 10 mmol/L KCl, 1.5 mmol/L MgCl2, 1 mmol/L EDTA, and 1 mmol/L EGTA) with protease inhibitors (Roche Protease Inhibitor Cocktail 1×, 2 mmol/L sodium orthovanadate, and 2 mmol/L PMSF) and lysed. Lysate was incubated on ice for 20 minutes, and then centrifuged at 720 × g for 5 minutes. The resulting supernatant was centrifuged at 10,000 × g for 5 minutes to produce cytosolic (supernatant) and mitochondrial (pellet) fractions. The mitochondrial fraction was washed and resuspended in subcellular fractionation buffer supplemented with 10% glycerol and 0.1% SDS. The resulting fractions were immunoblotted for cytochrome c.

Apoptosis assays

For serum starvation experiments, cells were grown in RPMI with 10% FBS for 24 hours, media were replaced with RPMI containing 10% FBS or 0.1% FBS, and cells were incubated for 48 hours. For etoposide (Sigma) treatments, cells were exposed to either vehicle (DMSO) or etoposide (5 μmol/L) for 48 hours. BD ViaProbe/7-AAD (Thermo Fisher Scientific) and APC Annexin V (BioLegend) were added and detected by Flow Cytometry (Cytek Aurora). Analysis was performed using FlowJo software (v10, RRID:SCR_008520). Caspase Glo 9 (Promega) was used per protocol to quantify caspase-9 activity.

Ethics statement

All animal studies were conducted in accordance with policies set forth by the Emory University (Atlanta, GA) Institutional Animal Care and Use Committee (IACUC). Our protocol was approved by the Emory IACUC (PROTO201700089). Euthanasia was performed by CO2 and cervical dislocation.

Mouse xenograft in vivo studies

Mice were approximately 6 weeks of age at time of injection and male/female mice were distributed equally among experimental arms. For all tumor growth and drug treatment studies, mice were randomized on the basis of tumor size and gender into the different treatment arms to maintain equivalent median tumor sizes per arm of each study. We measured binary endpoints (i.e., tumor growth or tumor regression), 4–6 mice per group were used to detect 50% differences in group means, assuming sample SDs < 50% of the mean (α = 0.05 and power = 0.8). For PDX studies, low passage (≤3rd) PDX tumors were removed from one NOD scid gamma (NSG; The Jackson Laboratory; IMSR catalog no., ARC:NSG, RRID:IMSR_ARC:NSG) mouse, dissociated, mixed 1:1 in Matrigel (Corning) as a single-cell suspension with 4 × 106 cells/injection, and reinjected into the subcutaneous flank of additional NSG mice. Tumor volumes were measured with calipers and calculated using formula L × W × H × π/6. When tumors reached approximately 150 mm3, mice were treated with vehicle control (PBS) or 0.05 mg/kg topotecan (Sagent Pharmaceuticals) and 20 mg/kg cyclophosphamide (Jiangsu Hengrui Medicine) via intraperitoneal injections once daily for 5 days. Mice were sacrificed on day 6 and tumor was harvested and evaluated. For tumor formation studies, SK-N-AS CV, shYAP#1, and shYAP#2 cells (4 × 106 cells/injection) were injected into the subcutaneous flanks of NSG mice (n = 4 per arm). Mice were sacrificed when tumor volumes reached 1,500 mm3. For cyclophosphamide treatment studies, SK-N-AS xenografts were established as above in nu/nu athymic mice (The Jackson Laboratory; MGI #3848172, RRID:MGI:3848172). Mice (n = 5 per arm) were treated with vehicle (PBS) or 75 mg/kg cyclophosphamide by intraperitoneal injection twice weekly for four doses and sacrificed when tumor volumes reached 1,500 mm3. For trametinib studies, SK-N-AS xenografts established subcutaneously in NSG mice (n = 6 per arm) were treated with vehicle (35% Kollisolv PEG E 400, 60% Phosal, and 5% DMSO) or 3 mg/kg Trametinib (Selleck Chemicals) for 14 days once daily via oral gavage and mice were sacrificed when tumor volumes reached 1,500 mm3.

RNA-sequencing

SK-N-AS CV, shYAP#1, and shYAP#2 pellets in triplicates were harvested, RNA was isolated, and quality checked at the Emory Integrated Genomics Core. RNA-sequencing (RNA-seq; poly-A selection, 20 M paired-end 150 bp reads on the Illumina NovaSeq Platform) was performed at Novogene, with data analyzed by the Emory Integrated Computational Core. Sequencing data were first checked by FastQC (RRID:SCR_014583) for quality control, and then aligned to the human “indexed” reference genome (hg38) using STAR (RRID:SCR_015899; ref. 19). Gene quantification was performed using HTSeq-count (RRID:SCR_011867; ref. 20) and gene count data were filtered before analysis to remove lowly expressed genes. Counts per million (CPM) data were generated after normalization using trimmed method of M-values, as implemented in the Bioconductor package (RRID:SCR_006442), EdgeR (RRID:SCR_012802; ref. 21). To determine genes important for a case–control, a t test followed by FDR (Benjamini–Hochberg method) on normalized log2-transformed CPM was performed. A gene was defined as differentially expressed gene (DEG) if its FDR ≤ 0.05. To characterize expressed genes, a preranked permutation-based GSEA was performed with weighted Kolmogorov–Smirnov statistical test (22). A gene set from the Molecular Signatures Database (v7.0) was used as input data for this analysis. The identified DEGs were used in pathway analysis.

Statistical analysis

A Student t test or a one-way ANOVA was used to assess statistical significance. A log-rank (Mantel–Cox) test was performed on the Kaplan–Meier survival curves for xenograft studies. Statistical significance was defined as P < 0.05. GraphPad Prism (v8.0; RRID:SCR_002798) was used for all analyses.

YAP tumor expression and transcriptional activity increase in response to in vivo standard chemotherapy treatment of neuroblastoma tumors

Given previous reports demonstrating increased YAP transcription in primary recurrent neuroblastomas, we first sought to determine YAP expression in neuroblastoma PDXs from the same patient's prechemotherapy diagnostic (NBX-4) and post-chemotherapy recurrent (NBX-4R) tumor. Results demonstrate that YAP expression and transcriptional activity increased in the relapsed PDX compared with the pretherapy PDX from the same patient (Fig. 1A). In keeping with this finding, paired isogenic cell lines derived from the same patient at diagnosis prechemotherapy (SK-N-BE1) and at relapse following chemotherapy (SK-N-BE2) showed increased YAP expression in the relapsed SK-N-BE2 (Supplementary Fig. S1). To gain a sense for the timing of YAP increase during or after therapy, we established in vivo tumors using chemotherapy-naïve NBX-4 cells and subsequently treated mice with 5 days of vehicle or topotecan and cyclophosphamide at doses and regimen comparable with a standard cycle of chemotherapy used for neuroblastoma patient induction (23). The tumors grew through chemotherapy or vehicle treatment and were harvested on day 6. Notably, YAP gene and protein expression increased in the chemotherapy-treated tumors and remained absent in the vehicle-treated control and expression of YAP transcriptional targets, CTGF and EGFR, increased in the chemotherapy-treated tumor, but not in the vehicle control (Fig. 1B and C). We treated a second neuroblastoma PDX derived from another patient's diagnostic chemotherapy-naïve tumor (NBX-1a) with topotecan and cyclophosphamide. Tumors grew through the topotecan and cyclophosphamide treatment, and again we observed that YAP protein expression increased by day 6 in the chemotherapy-treated PDX, but not in the vehicle control (Fig. 1D). We then evaluated a panel of human-derived neuroblastoma cell lines and found YAP protein to be heterogeneously expressed (Fig. 1E). Phosphorylated and inactive YAP (pYAP) was present in some cell lines. TAZ was neither always coexpressed with YAP nor was it increased in YAP-null cells. TEAD protein was present throughout all cell lines. Expression of known downstream targets of YAP-mediated transcription, EGFR and CYR61 (24, 25), was increased in neuroblastomas with YAP expression, suggesting that YAP transcription may be active despite the concurrent presence of pYAP in certain cells. We also found that YAP expression does not correlate with MYCN amplification status and YAP was commonly seen in neuroblastoma cell lines harboring RAS/MAPK mutations, but not ALK mutations (Fig. 1E; Supplementary Table S2).

Figure 1.

YAP transcriptional activity and protein expression increase after standard chemotherapy treatment in neuroblastoma tumors. A,YAP and downstream target gene expression in NBX-4 and NBX-4R PDX tumors. Data represent mean ± SEM (n = 3); *, P < 0.05; **, P < 0.01. B,YAP and downstream target gene expression in NBX-4 vehicle-treated and NBX-4 topotecan/cyclophosphamide (topo/cy)-treated PDX tumors. Data represent mean ± SEM (n = 3); **, P < 0.01; ****, P < 0.0001. C, YAP protein expression in the topotecan and cyclophosphamide-treated NBX-4 tumor. D, YAP protein expression in a different PDX tumor (NBX-1a) derived at diagnosis, pretreatment. E, Human-derived neuroblastoma cell line panel showing YAP and related protein expression. See Supplementary Table S2 for more detailed mutational profile for each neuroblastoma cell line. n.s. not significant.

Figure 1.

YAP transcriptional activity and protein expression increase after standard chemotherapy treatment in neuroblastoma tumors. A,YAP and downstream target gene expression in NBX-4 and NBX-4R PDX tumors. Data represent mean ± SEM (n = 3); *, P < 0.05; **, P < 0.01. B,YAP and downstream target gene expression in NBX-4 vehicle-treated and NBX-4 topotecan/cyclophosphamide (topo/cy)-treated PDX tumors. Data represent mean ± SEM (n = 3); **, P < 0.01; ****, P < 0.0001. C, YAP protein expression in the topotecan and cyclophosphamide-treated NBX-4 tumor. D, YAP protein expression in a different PDX tumor (NBX-1a) derived at diagnosis, pretreatment. E, Human-derived neuroblastoma cell line panel showing YAP and related protein expression. See Supplementary Table S2 for more detailed mutational profile for each neuroblastoma cell line. n.s. not significant.

Close modal

YAP genetic inhibition suppresses neuroblastoma tumor growth in vivo

A prime role for YAP in both normal development and in cancer is to promote growth through transcriptional as well as posttranslational regulation of genes involved in cell proliferation (26, 27). To test the effects of YAP on neuroblastoma growth, we used both gain- and loss-of-function approaches to perturb YAP expression. Western blot analysis of these cell lines confirmed YAP knockdown (Fig. 2A) and overexpression (Fig. 2B; Supplementary Fig. S2A). TAZ and TEAD expression were not altered by YAP knockdown (Fig. 2A). While EGFR and phosphorylated EGFR (pEGFR) did not significantly change, YAP targets, CTGF and CYR61, decreased with YAP knockdown and increased with YAP overexpression, suggesting these models efficiently modulate YAP transcriptional activity (Fig. 2A and B). In vitro, YAP perturbation does not significantly alter cell proliferation rates (Fig. 2C; Supplementary Fig. S2B). Yet, in the in vivo setting, time to sacrifice occurred later in the SK-N-AS shYAP xenografts compared with the SK-N-AS CV tumors (Fig. 2D). Importantly, YAP remained knocked down at time of sacrifice (Fig. 2D). These results suggest that YAP affects neuroblastoma proliferation within the heterotypic tumor microenvironment (TME) and may promote the initiation of neuroblastoma growth.

Figure 2.

YAP affects neuroblastoma tumor growth in vivo. A and B, YAP protein and related protein in stably expressing shYAP models of cell lines SK-N-AS (top left) and NLF (top right) with YAP downstream target gene expression (bottom; A) and in YAP stable overexpression model (pGAMA) of cell line IMR5 (top) with YAP downstream target gene expression (bottom; B). Data represent mean ± SEM (n = 3). C, Cell survival measured using CellTiter-Glo at 0, 24, 48, and 72 hours after SK-N-AS shYAP model (left), NLF shYAP model (middle), and IMR5 pGAMA-YAP model (right) cells were plated. Data represent mean ± SEM (n = 3). D, Kaplan–Meier curve (top left) showing survival for mice injected with SK-N-AS CV (n = 4) or shYAP#1 (n = 4) and shYAP#2 (n = 4) cells. There was a significant (***, P < 0.001) delay in time to reach tumor volume of 1,500 mm3 with YAP knockdown as shown in average tumor growth curve (right). This experiment was repeated three separate times. Western blot analysis (bottom left) showing YAP protein expression of SK-N-AS CV, shYAP#1, and shYAP#2 xenografts, with two representative tumors from each group obtained at time of sacrifice. n.s., not significant; *, P < 0.05; **, P < 0.01; ***, P < 0.001; ****, P < 0.0001. WT, wild-type.

Figure 2.

YAP affects neuroblastoma tumor growth in vivo. A and B, YAP protein and related protein in stably expressing shYAP models of cell lines SK-N-AS (top left) and NLF (top right) with YAP downstream target gene expression (bottom; A) and in YAP stable overexpression model (pGAMA) of cell line IMR5 (top) with YAP downstream target gene expression (bottom; B). Data represent mean ± SEM (n = 3). C, Cell survival measured using CellTiter-Glo at 0, 24, 48, and 72 hours after SK-N-AS shYAP model (left), NLF shYAP model (middle), and IMR5 pGAMA-YAP model (right) cells were plated. Data represent mean ± SEM (n = 3). D, Kaplan–Meier curve (top left) showing survival for mice injected with SK-N-AS CV (n = 4) or shYAP#1 (n = 4) and shYAP#2 (n = 4) cells. There was a significant (***, P < 0.001) delay in time to reach tumor volume of 1,500 mm3 with YAP knockdown as shown in average tumor growth curve (right). This experiment was repeated three separate times. Western blot analysis (bottom left) showing YAP protein expression of SK-N-AS CV, shYAP#1, and shYAP#2 xenografts, with two representative tumors from each group obtained at time of sacrifice. n.s., not significant; *, P < 0.05; **, P < 0.01; ***, P < 0.001; ****, P < 0.0001. WT, wild-type.

Close modal

YAP is increased by stem-like conditions and regulates genes promoting a mesenchymal phenotype

In other tumor models, YAP regulates tumor cell growth by mediating contact-independent cell proliferation and expression of stem-like genes (9, 10, 28). We, therefore, evaluated the colony formation potential of NLF shYAP cells and show a significant decrease in the number of colonies formed compared with NLF CV cells (Fig. 3A). To further investigate for mesenchymal properties of YAP, we grew human-derived neuroblastoma cell lines as neurospheres in neurobasal media, thus mirroring the stem-like environment. YAP gene expression was significantly increased upon neurosphere formation (Fig. 3B). OCT4 and SOX2, known mesenchymal genes regulated by YAP (28, 29), increased under neurosphere conditions, compared with regular culture conditions (Fig. 3B). We evaluated for changes in mesenchymal gene expression in the NLF shYAP model versus NLF CV cells grown in neurobasal media and noted that YAP knockdown leads to decreased levels of these genes (Fig. 3C). Furthermore, RNA-seq of SK-N-AS shYAP models compared with SK-N-AS CV shows genes associated with a mesenchymal and stem-like state in other tumor types, such as JAK1, IL6ST, TBX3, and PPP2R2B (30, 31), decreased when YAP was knocked down in neuroblastoma (Supplementary Fig. S3). We also noted fewer neurospheres in the NLF shYAP cells, and an increased number of neurospheres in the YAP-overexpressed cells (Fig. 3D). Together, these results suggest that YAP promotes a dedifferentiated mesenchymal phenotype.

Figure 3.

YAP regulates genes promoting a mesenchymal phenotype. A, Colony formation assay quantified using crystal violet staining in NLF shYAP model. Left, quantified number of colonies shown in box plots. Data represent mean ± range (n = 2); *, P < 0.05; **, P < 0.01. Right, representative images of staining from wells. B,YAP and downstream target gene (OCT4 and SOX2) expression in IMR5 cells (top) and NLF cells (bottom) grown as a monolayer in RPMI and as neurospheres (NS) in neurobasal media. Data represent mean ± SEM (n = 3); *, P < 0.05; **, P < 0.01; ****, P < 0.0001. C,YAP and downstream target gene (OCT4 and SOX2) expression in NLF CV and NLF shYAP cells grown in stem-like conditions as neurospheres. Data represent mean ± SEM (n = 3); **, P < 0.01. D, Neurosphere count after 7 days in neurobasal media of NLF shYAP model (top) and IMR5 pGAMA-YAP model (bottom). Data represent mean ± SEM (n = 3). **, P < 0.01; ***, P < 0.001; n.s., not significant.

Figure 3.

YAP regulates genes promoting a mesenchymal phenotype. A, Colony formation assay quantified using crystal violet staining in NLF shYAP model. Left, quantified number of colonies shown in box plots. Data represent mean ± range (n = 2); *, P < 0.05; **, P < 0.01. Right, representative images of staining from wells. B,YAP and downstream target gene (OCT4 and SOX2) expression in IMR5 cells (top) and NLF cells (bottom) grown as a monolayer in RPMI and as neurospheres (NS) in neurobasal media. Data represent mean ± SEM (n = 3); *, P < 0.05; **, P < 0.01; ****, P < 0.0001. C,YAP and downstream target gene (OCT4 and SOX2) expression in NLF CV and NLF shYAP cells grown in stem-like conditions as neurospheres. Data represent mean ± SEM (n = 3); **, P < 0.01. D, Neurosphere count after 7 days in neurobasal media of NLF shYAP model (top) and IMR5 pGAMA-YAP model (bottom). Data represent mean ± SEM (n = 3). **, P < 0.01; ***, P < 0.001; n.s., not significant.

Close modal

YAP does not influence chemotherapy-induced apoptosis in neuroblastoma in vitro

Neuroblastoma cells with a mesenchymal phenotype represent the most chemotherapy-resistant population (29, 32–34). Given YAP is increased at relapse and under stem-like media conditions, we evaluated whether YAP expression and transcriptional activity affected neuroblastoma response to chemotherapy. We treated neuroblastoma cells harboring YAP knockdown or CV in vitro with cytotoxic agents used in high-risk neuroblastoma first-line therapy or in relapsed salvage therapy, melphalan, irinotecan, topotecan, and etoposide, and evaluated for cell death. YAP genetic knockdown or YAP overexpression neither sensitizes nor promotes resistance to cytotoxic therapy, respectively, in vitro (Fig. 4A; Supplementary Fig. S4A–S4D). Short hairpin RNA (shRNA) inhibition of YAP also fails to augment apoptosis in NLF shYAP cells treated with etoposide in vitro (Supplementary Fig. S5). YAP has been shown to transcriptionally upregulate prosurvival multidomain BCL2 family members (BCL2 and BCL-XL) in other cancers (35, 36), however, YAP knockdown does not affect baseline BCL2 family prosurvival protein expression in neuroblastoma (Supplementary Fig. S6A). Although YAP knockdown leads to an increase in proapoptotic BIM protein expression, it does not alter BIM binding to prosurvival BCL2 members that are known to prevent apoptosis (Supplementary Fig. S6B; ref. 37).

Figure 4.

YAP regulates cytotoxic therapy and MEK inhibitor response in neuroblastoma in vivo. A, Cell survival measured by CellTiter-Glo after 48 hours of melphalan treatment of NLF shYAP (left) and SK-N-AS shYAP (right) cells. Data represent mean ± SEM (n = 3). B, Average tumor volume curve (left) and Kaplan–Meier curve (right) showing survival for mice injected with SK-N-AS CV or shYAP#1 cells and then treated with vehicle or cyclophosphamide twice weekly for four treatments (indicated by downward arrows on left figure). N = 5 for each arm. Tumor growth curve shows significant delay in tumor growth in SK-N-AS shYAP#1 cyclophosphamide-treated xenografts. **, P < 0.01; ***, P < 0.001; ****, P < 0.0001. There was a significant delay in time to sacrifice for the shYAP#1 cyclophosphamide-treated group. **, P < 0.01. C, Cell survival measured by CellTiter-Glo after 72 hours of trametinib treatment of NLF shYAP (top) and SK-N-AS shYAP (bottom) model cells. Data represent mean ± SEM (n = 3). D, Average tumor growth curve of SK-N-AS shYAP model xenograft tumors treated with vehicle or trametinib (n = 6 in each arm) showing significant delay to reach tumor volume in shYAP model xenograft tumors treated with trametinib. ***, P < 0.001; ****, P < 0.0001. Arrow, treatment duration. This experiment was repeated two separate times.

Figure 4.

YAP regulates cytotoxic therapy and MEK inhibitor response in neuroblastoma in vivo. A, Cell survival measured by CellTiter-Glo after 48 hours of melphalan treatment of NLF shYAP (left) and SK-N-AS shYAP (right) cells. Data represent mean ± SEM (n = 3). B, Average tumor volume curve (left) and Kaplan–Meier curve (right) showing survival for mice injected with SK-N-AS CV or shYAP#1 cells and then treated with vehicle or cyclophosphamide twice weekly for four treatments (indicated by downward arrows on left figure). N = 5 for each arm. Tumor growth curve shows significant delay in tumor growth in SK-N-AS shYAP#1 cyclophosphamide-treated xenografts. **, P < 0.01; ***, P < 0.001; ****, P < 0.0001. There was a significant delay in time to sacrifice for the shYAP#1 cyclophosphamide-treated group. **, P < 0.01. C, Cell survival measured by CellTiter-Glo after 72 hours of trametinib treatment of NLF shYAP (top) and SK-N-AS shYAP (bottom) model cells. Data represent mean ± SEM (n = 3). D, Average tumor growth curve of SK-N-AS shYAP model xenograft tumors treated with vehicle or trametinib (n = 6 in each arm) showing significant delay to reach tumor volume in shYAP model xenograft tumors treated with trametinib. ***, P < 0.001; ****, P < 0.0001. Arrow, treatment duration. This experiment was repeated two separate times.

Close modal

YAP genetic inhibition sensitizes neuroblastoma xenografts to cytotoxic and MEK inhibitor therapy in vivo

Given the heterogeneity of solid tumors due to their TME (38), we ascertained whether YAP affects cytotoxic response in vivo. We treated SK-N-AS CV and shYAP xenografts with cyclophosphamide and monitored tumor growth. SK-N-AS shYAP xenografts were significantly more sensitive to cyclophosphamide in vivo compared with control xenografts (Fig. 4B). Because of strong evidence in other cancers for an interaction between YAP and oncogenic RAS (39), we queried whether YAP expression also affects MEK inhibitor therapy response. We treated the NRAS-mutated SK-N-AS shYAP and CV cells with the MEK inhibitor, trametinib, both in vitro and in vivo. Similar to chemotherapy responses, there was no change in trametinib-induced cell death in vitro with YAP genetic inhibition in neuroblastoma cell lines harboring NRAS (SK-N-AS) and NF1 (NLF) mutations (Fig. 4C). In contrast, SK-N-AS shYAP xenografts were significantly more sensitive to trametinib in vivo with more tumor regression in YAP shRNA–transduced cell line xenografts compared with SK-N-AS CV xenografts (Fig. 4D). Importantly, for both the cytotoxic and trametinib xenograft studies, the decrease in tumor growth in response to therapy in the YAP-knockdown models remained significant after considering the delay in normal tumor growth imparted by YAP knockdown alone. Therefore, YAP knockdown has an increased effect on cytotoxic and trametinib responses in situ, supporting a potential role for YAP-influencing responses to stresses imparted by the 3-dimensional microenvironment in neuroblastoma.

YAP regulates BH3 genes involved in intrinsic apoptosis in neuroblastoma

To understand genes regulated by YAP that may be contributing to the more profound effects seen on tumor growth and treatment response in vivo, we performed RNA-seq on SK-N-AS shYAP#1 and #2 models and SK-N-AS CV cells. Hierarchical clustering analysis revealed that the gene expression profiles of the SK-N-AS shYAP#1 and SK-NAS shYAP#2 were more similar than SK-N-AS CV cells (Fig. 5A). While the direction of gene expression (increased or decreased) in the two YAP-knockdown models was not identical, this was likely due to the higher degree of YAP genetic inhibition in shYAP#1 compared with shYAP#2 (Fig. 5B). We, therefore, focused on the shYAP#1-knockdown model and compared it with SK-N-AS CV RNA-seq results to prioritize our evaluation of genes affected by changes in YAP expression, and then confirmed those genes were expressed similarly in SK-N-AS shYAP#2. As expected, YAP was one of the downregulated genes in the Hippo signaling pathway analysis, confirming effective knockdown in this model (Fig. 5C). Hippo pathway genes that are YAP transcriptional targets, such as SMAD1, and the stemness gene, PPP2R2B, were downregulated with YAP knockdown, again supporting inhibition of YAP transcription (Fig. 5C). We evaluated for changes in apoptosis genes and confirmed that BCL2 antiapoptotic members were not altered upon YAP knockdown and that proapoptotic BIM (BCL2L11) expression increased, as was seen at the protein level (Fig. 5D; Supplementary Fig. S6A).

Figure 5.

RNA-seq of SK-N-AS CV and shYAP cells shows that YAP regulates genes involved in intrinsic apoptosis in neuroblastoma. A, Hierarchical clustering analysis of the gene expression profiles of SK-N-AS CV (CV), shYAP#1, and shYAP#2 cells shows similar profiles between shYAP cells compared with CV cells. B,YAP gene expression in CV and shYAP cells plotted on a log2 scale ± ranges shows decreased YAP expression in shYAP (#1 > #2) cells compared with CV cells. C, GSEA of Hippo signaling pathway genes. D, GSEA of apoptosis pathway genes. E,HRK gene expression in CV and shYAP cells plotted on a log2 scale ± ranges shows increased HRK expression in shYAP (#1 > #2) cells compared with CV cells. F,HRK and YAP gene signatures of Maris cohort were assessed from R2 database (http://r2.amc.nl/) and Kaplan–Meier survival curves were generated.

Figure 5.

RNA-seq of SK-N-AS CV and shYAP cells shows that YAP regulates genes involved in intrinsic apoptosis in neuroblastoma. A, Hierarchical clustering analysis of the gene expression profiles of SK-N-AS CV (CV), shYAP#1, and shYAP#2 cells shows similar profiles between shYAP cells compared with CV cells. B,YAP gene expression in CV and shYAP cells plotted on a log2 scale ± ranges shows decreased YAP expression in shYAP (#1 > #2) cells compared with CV cells. C, GSEA of Hippo signaling pathway genes. D, GSEA of apoptosis pathway genes. E,HRK gene expression in CV and shYAP cells plotted on a log2 scale ± ranges shows increased HRK expression in shYAP (#1 > #2) cells compared with CV cells. F,HRK and YAP gene signatures of Maris cohort were assessed from R2 database (http://r2.amc.nl/) and Kaplan–Meier survival curves were generated.

Close modal

Given that tumor environmental factors, such as hypoxia and nutrient deprivation, can contribute to solid tumor therapy resistance in situ, and that increased BIM in shYAP neuroblastoma cells failed to enhance in vitro apoptosis from chemotherapy, we honed in on other YAP-regulated genes that mediate apoptosis in response to TME stress. Notably, HRK, a BH3-only prodeath protein that activates mitochondrial apoptosis in response to cytokine deprivation (40, 41), had one of the highest fold increases in expression of all genes altered between the YAP shRNA neuroblastoma models compared with CV (Fig. 5D; Supplementary Tables S3 and S4). We confirmed upregulated HRK in both shYAP#1 and #2 models compared with CV (Fig. 5E). We queried a high-risk neuroblastoma primary tumor gene expression dataset (R2 database: http://r2.amc.nl) and found that patients with high-risk neuroblastoma with decreased tumor expression of HRK had significantly worse overall survival, supporting a potential tumor suppressive role for HRK in neuroblastoma. In that same dataset, high YAP expression portended a worse prognosis, supporting the reciprocal relationship (Fig. 5F). HRK tumor expression did not correlate with MYCN status, but was noted to have lower expression in higher staged tumors (Supplementary Fig. S7).

YAP suppresses HRK to inhibit apoptotic responses to serum deprivation

To confirm RNA-seq data, we evaluated SK-N-AS and NLF shYAP models for HRK expression and found that HRK gene expression was indeed upregulated when YAP was inhibited genetically. We complemented this analysis by showing HRK gene expression decreases significantly in the IMR5 cell line with YAP overexpression (Fig. 6A). HRK is a BH3-only prodeath protein that is activated by environmental stresses such as cytokine depletion and severe hypoxia in neurons, conditions that are often found in aggressive solid tumors such as neuroblastoma (40, 41). We, therefore, evaluated neuroblastoma cells with and without YAP knockdown for their survival in both hypoxic and normoxic conditions and found no difference in apoptosis (Supplementary Fig. S8). Given the role of metabolic stress in the tumor environment, we evaluated neuroblastoma cells with and without YAP knockdown for their responses to serum starvation. Upon 48 hours of serum starvation, NLF shYAP showed increased apoptosis in low serum (0.1% FBS) conditions compared with YAP-expressing NLF CV cells in low serum conditions (Fig. 6B). Importantly, under serum starvation, HRK gene expression increased significantly in both NLF shYAP and SK-N-AS shYAP cells compared with their control counterparts (Fig. 6C). Furthermore, cytochrome c release and caspase-9 activation were increased in serum-starved shYAP neuroblastoma cells, confirming that apoptosis proceeds through the intrinsic apoptosis pathway, where HRK-mediated apoptosis occurs (Fig. 6D; Supplementary Fig. S9; ref. 42). Most importantly, apoptosis in response to etoposide treatment in vitro was significantly increased in NLF shYAP and SK-N-AS shYAP cells, but not in control cells treated with etoposide under serum-starved conditions (Fig. 6E).

Figure 6.

YAP suppresses HRK expression to inhibit apoptotic responses to serum deprivation. A,HRK gene expression in SK-N-AS shYAP model (left), NLF shYAP model (middle), and IMR5 pGAMA-YAP model (right) by qRT-PCR. Data represent mean ± SEM (n = 3); **, P < 0.01; ***, P < 0.001. B, Apoptosis quantified via flow for Annexin V in NLF CV and shYAP#1 cells in 10% serum or 0.1% serum for 48 hours. Data represent mean ± SEM (n = 3); **, P < 0.01; ***, P < 0.001; NLF shYAP#1 in 10% serum versus 0.1% serum (P = 0.063). C,HRK gene expression in NLF shYAP model (left) and SK-N-AS shYAP model (right) in standard 10% serum- or 0.1% serum-deprived conditions for 48 hours. Data represent mean ± SEM (n = 3); ***, P < 0.001; ****, P < 0.0001. D, Western blot analysis of cytochrome c (Cyto. C) measured in the cytosolic fraction of NLF CV (CV) and NLF shYAP (shYAP #1) cells grown under normal serum (10% FBS) and serum-starved (0.1% FBS) conditions. E, Caspase-9 activity (normalized to 0 hour activity) measured in NLF shYAP model (left) and SK-N-AS shYAP model (right) cells grown in normal 10% serum or 0.1% serum-deprived conditions for 24 hours and treated with etoposide for 48 hours. Data represent mean ± SEM (n = 3); *, P < 0.05; ****, P < 0.0001. F,HRK gene expression in NBX-4 and NBX-4R PDX tumors (left) and NBX-4 vehicle-treated and NBX-4 topotecan/cyclophosphamide (topo/cy)-treated PDX tumors (right). Data represent mean ± SEM (n = 3). **, P < 0.01; ***, P < 0.001.

Figure 6.

YAP suppresses HRK expression to inhibit apoptotic responses to serum deprivation. A,HRK gene expression in SK-N-AS shYAP model (left), NLF shYAP model (middle), and IMR5 pGAMA-YAP model (right) by qRT-PCR. Data represent mean ± SEM (n = 3); **, P < 0.01; ***, P < 0.001. B, Apoptosis quantified via flow for Annexin V in NLF CV and shYAP#1 cells in 10% serum or 0.1% serum for 48 hours. Data represent mean ± SEM (n = 3); **, P < 0.01; ***, P < 0.001; NLF shYAP#1 in 10% serum versus 0.1% serum (P = 0.063). C,HRK gene expression in NLF shYAP model (left) and SK-N-AS shYAP model (right) in standard 10% serum- or 0.1% serum-deprived conditions for 48 hours. Data represent mean ± SEM (n = 3); ***, P < 0.001; ****, P < 0.0001. D, Western blot analysis of cytochrome c (Cyto. C) measured in the cytosolic fraction of NLF CV (CV) and NLF shYAP (shYAP #1) cells grown under normal serum (10% FBS) and serum-starved (0.1% FBS) conditions. E, Caspase-9 activity (normalized to 0 hour activity) measured in NLF shYAP model (left) and SK-N-AS shYAP model (right) cells grown in normal 10% serum or 0.1% serum-deprived conditions for 24 hours and treated with etoposide for 48 hours. Data represent mean ± SEM (n = 3); *, P < 0.05; ****, P < 0.0001. F,HRK gene expression in NBX-4 and NBX-4R PDX tumors (left) and NBX-4 vehicle-treated and NBX-4 topotecan/cyclophosphamide (topo/cy)-treated PDX tumors (right). Data represent mean ± SEM (n = 3). **, P < 0.01; ***, P < 0.001.

Close modal

We initially showed that YAP expression increases in relapsed PDXs compared with matched diagnostic tumors and YAP also increases in response to a single cycle of chemotherapy in vivo (Fig. 1A and B). We, therefore, evaluated for HRK expression in these same models. The inverse correlation for YAP and HRK holds true in vivo, as HRK expression was significantly decreased in NBX-4R PDX compared with the NBX-4 PDX (Fig. 6F). Furthermore, following topotecan and cyclophosphamide treatment in vivo, not only did YAP expression increase, but HRK gene expression concurrently decreased in the chemotherapy-treated NBX-4 PDX (Fig. 1B and F). Thus, YAP may be driving the aggressive nature of recurrent neuroblastoma through modulation of in vivo stress conditions, promoting tumor growth and therapy resistance via inhibition of HRK.

Despite the finding of increased YAP transcriptional activity (4), the role of YAP in relapsed neuroblastoma has not been well-characterized. Here, we describe a role for YAP in regulating neuroblastoma tumor growth and therapy response in vivo that may, in part, be due to YAP repression of the proapoptotic protein, HRK. We have confirmed that YAP transcriptional activity increases in relapsed neuroblastoma models, and now show that YAP expression and transcriptional activity immediately increase in response to standard high-risk neuroblastoma chemotherapy in vivo. Given the brief period of drug exposure and the continued growth of tumors throughout chemotherapy or vehicle treatment, our findings suggest that increased YAP expression in the chemotherapy-treated tumors may represent a cell intrinsic response to treatment rather than a therapy-induced cell death and selection of a YAP-expressing drug-resistant clone.

YAP is necessary for organ growth during the embryonic stages of development (43) and is involved in normal neural crest migration and fate (44). YAP also contributes to the mesenchymal state in pediatric solid tumors like rhabdomyosarcoma (39). Our results concur with such findings in neuroblastoma, with both neurosphere studies and RNA-seq showing the transcription of stem-like genes to be dependent on YAP expression in neuroblastoma. Importantly, we have uniquely shown that mesenchymal properties such as colony formation, tumor growth, and stem cell gene expression are negatively affected upon YAP genetic knockdown, supporting a potential tumor-initiating role for YAP in neuroblastoma.

A hallmark of the mesenchymal phenotype in neuroblastoma and other cancers is extreme therapy resistance (29, 45, 46). This may be attributed to YAP's transcriptional regulation of mesenchymal genes that are also known to impart therapy resistance, such as EGFR. For example, YAP transcriptionally upregulates EGFR in esophageal cancer to mediate resistance to chemotherapy (13). Our results also show that EGFR is overexpressed in YAP-expressing neuroblastoma cells and upregulated in PDXs in response to chemotherapy treatment. Yet, EGFR expression does not change in response to YAP knockdown or overexpression, perhaps because EGFR can be influenced by many additional pathways (47). Despite the persistence of EGFR in SK-N-AS shYAP xenografts, the shYAP tumors regress more in response to trametinib treatment compared with control, suggesting that alternative YAP-regulated genes are responsible for in vivo therapy resistance, such as the suppression of HRK. YAP has also been shown to promote therapy resistance through inhibition of mitochondrial apoptosis through upregulation of prosurvival BCL2 family genes (35, 36). In neuroblastoma, however, we show that YAP knockdown does not affect prosurvival BCL2 family gene or protein expression and it does not restore apoptosis in response to multiple cytotoxic agents in vitro. Despite the increased expression of BIM with YAP knockdown, BIM remains sequestered to and inactivated by MCL1, which may explain the lack of in vitro chemosensitization. BIM overexpression has also been a marker of trametinib sensitivity in other cell models (48, 49), yet the lack of in vitro sensitization to trametinib with YAP knockdown again negates BIM as the etiology for in vivo trametinib sensitization by YAP genetic inhibition.

YAP regulates MEK inhibitor resistance in multiple adult cancers (50, 51). Coggins and colleagues recently demonstrated that YAP transcription is activated in response to trametinib therapy in RAS-mutated neuroblastoma cell lines in vitro and YAP reciprocally induces MEK inhibitor resistance through transcriptional activation of MYCN and E2F in an NF1-mutated MYCN-amplified neuroblastoma cell treated with trametinib (52). We extend this to show that in neuroblastoma PDXs without RAS-hyperactivating mutations, YAP expression and transcriptional activity also increase in response to cytotoxic chemotherapy. Coggins and colleagues also show that complete knockout of YAP in NLF sensitized the cells to trametinib in vitro (52). While complete knockdown of YAP in NLF was not achieved by us, we provide data showing that complete knockdown of YAP in NRAS-mutated SK-N-AS and overexpression of YAP in NF1-mutated IMR5 and NGP did not significantly affect cell proliferation, chemotherapy response, or trametinib response in vitro. Importantly, we show enhanced tumor regression to trametinib in MYCN-nonamplified NRAS-mutated SK-N-AS xenografts when YAP was genetically inhibited in vivo. Our results, therefore, both build upon others' findings and add new dimensions by confirming that this combination holds true in vivo and highlighting the therapeutic potential of YAP and MEK coinhibition in all RAS/RAF/MAPK-mutated neuroblastomas, regardless of MYCN status.

The differences in our cell culture and xenograft responses to YAP knockdown lead us to conclude that YAP's influence on cytotoxic and targeted therapy resistance in vivo may be due to its effect on tumor environmental factors that notably contribute to solid tumor therapy resistance in situ (38). Apoptosis pathway analyses showed that gene expression of HRK significantly increased upon YAP knockdown. Rather than being activated in response to chemotherapy or radiation, HRK instead is activated and promotes apoptosis in response to environmental stresses such as cytokine withdrawal, nutrient deprivation, and hypoxia, tumor properties that have all been shown to promote therapy resistance in vivo (38, 40, 41). Therefore, we focused on HRK and its potential effects on neuroblastoma therapy response. Under serum deprivation conditions, we demonstrate that HRK expression increases, mitochondrial apoptosis ensues, and cells are more sensitive to chemotherapy in vitro, but only in neuroblastoma cells with YAP knocked down. These environmental stresses may explain the more profound differences in tumor growth and xenograft response to therapy seen in vivo, but not in vitro (in normal media conditions) when YAP is genetically inhibited. Importantly, the reciprocal relationship of HRK inhibition in the setting of YAP expression was maintained in vivo. When YAP expression was increased, HRK was downregulated in the relapsed compared with diagnostic PDX paired tumors. HRK also decreases in response to chemotherapy treatment of a chemotherapy-naïve PDX when YAP increases.

While HRK has not been extensively investigated in neuroblastoma or other cancers, primary tumor gene expression data confirm a significantly negative impact on high-risk neuroblastoma patient survival when HRK is low, legitimizing HRK as a potential tumor suppressor in neuroblastoma. HRK is inactivated in other cancers via epigenetic silencing through promoter methylation (53, 54). Preliminary methylation analysis of the HRK promoter region following PCR amplification reveals that this region is not methylated at baseline in YAP-expressing neuroblastoma cells (Supplementary Fig. S10). We have also interrogated TF binding site databases (JASPAR, TFBIND, and ALGGEN), which do not show putative TEAD binding sites (CATTCCA/T-3′) on the HRK promoter. YAP and TEAD1–4 family members have been described to bind to other transcriptional cofactors (such as MEF2, AP-1, and SRF) that can bind to the promoter and enhancer regions of target genes and recruit YAP/TEAD to regulate these genes (55). While TEAD might not bind to the HRK promoter directly, there may be a potential for other transcriptional cofactors to directly interact with HRK and recruit YAP/TEAD to suppress HRK transcription (55, 56). Therefore, investigations are ongoing to deduce the direct versus indirect regulation of HRK by YAP/TEAD and other potential coregulators.

Overall, these data demonstrate a novel role for YAP in suppressing HRK to restrict stress-induced apoptosis in vitro and inhibit therapy response and promote tumor growth in vivo. This strongly supports YAP as a logical therapeutic target to combine with MEK inhibitors or chemotherapy, and HRK as a novel tumor suppressor worthy of further study in neuroblastoma and perhaps other aggressive solid malignancies.

No potential conflicts of interest were disclosed.

J. Shim: Conceptualization, resources, data curation, software, formal analysis, supervision, funding acquisition, validation, investigation, visualization, methodology, writing-original draft, project administration, writing-review and editing. J.Y. Lee: Data curation, formal analysis, methodology. H.C. Jonus: Data curation, formal analysis, methodology. A. Arnold: Data curation. R.W. Schnepp: Conceptualization, writing-review and editing. K.M. Janssen: Data curation. V. Maximov: Data curation. K.C. Goldsmith: Conceptualization, resources, supervision, funding acquisition, validation, visualization, methodology, project administration, writing-review and editing.

This work was supported by CURE Childhood Cancer Foundation and Hyundai Hope on Wheels (to K.C. Goldsmith) and Hyundai Hope on Wheels and Emory Pediatrics Fellow Research Fund (to J. Shim). We are grateful for the thoughtful advice and reviews on our work by Drs. Lawrence Boise and Anna Kenney.

The costs of publication of this article were defrayed in part by the payment of page charges. This article must therefore be hereby marked advertisement in accordance with 18 U.S.C. Section 1734 solely to indicate this fact.

1.
Maris
JM
,
Hogarty
MD
,
Bagatell
R
,
Cohn
SL
. 
Neuroblastoma
.
Lancet
2007
;
369
:
2106
20
.
2.
Pugh
TJ
,
Morozova
O
,
Attiyeh
EF
,
Asgharzadeh
S
,
Wei
JS
,
Auclair
D
, et al
The genetic landscape of high-risk neuroblastoma
.
Nat Genet
2013
;
45
:
279
84
.
3.
Eleveld
TF
,
Oldridge
DA
,
Bernard
V
,
Koster
J
,
Colmet Daage
L
,
Diskin
SJ
, et al
Relapsed neuroblastomas show frequent RAS-MAPK pathway mutations
.
Nat Genet
2015
;
47
:
864
71
.
4.
Schramm
A
,
Koster
J
,
Assenov
Y
,
Althoff
K
,
Peifer
M
,
Mahlow
E
, et al
Mutational dynamics between primary and relapse neuroblastomas
.
Nat Genet
2015
;
47
:
872
7
.
5.
Moroishi
T
,
Hansen
CG
,
Guan
KL
. 
The emerging roles of YAP and TAZ in cancer
.
Nat Rev Cancer
2015
;
15
:
73
9
.
6.
Zhao
B
,
Li
L
,
Lei
Q
,
Guan
KL
. 
The Hippo-YAP pathway in organ size control and tumorigenesis: an updated version
.
Genes Dev
2010
;
24
:
862
74
.
7.
Lian
I
,
Kim
J
,
Okazawa
H
,
Zhao
J
,
Zhao
B
,
Yu
J
, et al
The role of YAP transcription coactivator in regulating stem cell self-renewal and differentiation
.
Genes Dev
2010
;
24
:
1106
18
.
8.
Camargo
FD
,
Gokhale
S
,
Johnnidis
JB
,
Fu
D
,
Bell
GW
,
Jaenisch
R
, et al
YAP1 increases organ size and expands undifferentiated progenitor cells
.
Curr Biol
2007
;
17
:
2054
60
.
9.
Wang
L
,
Shi
S
,
Guo
Z
,
Zhang
X
,
Han
S
,
Yang
A
, et al
Overexpression of YAP and TAZ is an independent predictor of prognosis in colorectal cancer and related to the proliferation and metastasis of colon cancer cells
.
PLoS One
2013
;
8
:
e65539
.
10.
Wang
X
,
Wu
B
,
Zhong
Z
. 
Downregulation of YAP inhibits proliferation, invasion and increases cisplatin sensitivity in human hepatocellular carcinoma cells
.
Oncol Lett
2018
;
16
:
585
93
.
11.
Zanconato
F
,
Cordenonsi
M
,
Piccolo
S
. 
YAP/TAZ at the roots of cancer
.
Cancer Cell
2016
;
29
:
783
803
.
12.
Ahmed
AA
,
Mohamed
AD
,
Gener
M
,
Li
W
,
Taboada
E
. 
YAP and the Hippo pathway in pediatric cancer
.
Mol Cell Oncol
2017
;
4
:
e1295127
.
13.
Song
S
,
Honjo
S
,
Jin
J
,
Chang
SS
,
Scott
AW
,
Chen
Q
, et al
The Hippo coactivator YAP1 mediates EGFR overexpression and confers chemoresistance in esophageal cancer
.
Clin Cancer Res
2015
;
21
:
2580
90
.
14.
Warren
JSA
,
Xiao
Y
,
Lamar
JM
. 
YAP/TAZ activation as a target for treating metastatic cancer
.
Cancers
2018
;
10
:
115
.
15.
Seong
BK
,
Fathers
KE
,
Hallett
R
,
Yung
CK
,
Stein
LD
,
Mouaaz
S
, et al
A metastatic mouse model identifies genes that regulate neuroblastoma metastasis
.
Cancer Res
2017
;
77
:
696
706
.
16.
Tumilowicz
JJ
,
Nichols
WW
,
Cholon
JJ
,
Greene
AE
. 
Definition of a continuous human cell line derived from neuroblastoma
.
Cancer Res
1970
;
30
:
2110
8
.
17.
Zhao
B
,
Wei
X
,
Li
W
,
Udan
RS
,
Yang
Q
,
Kim
J
, et al
Inactivation of YAP oncoprotein by the Hippo pathway is involved in cell contact inhibition and tissue growth control
.
Genes Dev
2007
;
21
:
2747
61
.
18.
Lestini
BJ
,
Goldsmith
KC
,
Fluchel
MN
,
Liu
X
,
Chen
NL
,
Goyal
B
, et al
Mcl1 downregulation sensitizes neuroblastoma to cytotoxic chemotherapy and small molecule Bcl2-family antagonists
.
Cancer Biol Ther
2009
;
8
:
1587
95
.
19.
Dobin
A
,
Davis
CA
,
Schlesinger
F
,
Drenkow
J
,
Zaleski
C
,
Jha
S
, et al
STAR: ultrafast universal RNA-seq aligner
.
Bioinformatics
2013
;
29
:
15
21
.
20.
Anders
S
,
Pyl
PT
,
Huber
W
. 
HTSeq–a Python framework to work with high-throughput sequencing data
.
Bioinformatics
2015
;
31
:
166
9
.
21.
Robinson
MD
,
Smyth
GK
. 
Moderated statistical tests for assessing differences in tag abundance
.
Bioinformatics
2007
;
23
:
2881
7
.
22.
Subramanian
A
,
Tamayo
P
,
Mootha
VK
,
Mukherjee
S
,
Ebert
BL
,
Gillette
MA
, et al
Gene set enrichment analysis: a knowledge-based approach for interpreting genome-wide expression profiles
.
Proc Natl Acad Sci U S A
2005
;
102
:
15545
50
.
23.
Krytska
K
,
Ryles
HT
,
Sano
R
,
Raman
P
,
Infarinato
NR
,
Hansel
TD
, et al
Crizotinib synergizes with chemotherapy in preclinical models of neuroblastoma
.
Clin Cancer Res
2016
;
22
:
948
60
.
24.
Moroishi
T
,
Park
HW
,
Qin
B
,
Chen
Q
,
Meng
Z
,
Plouffe
SW
, et al
A YAP/TAZ-induced feedback mechanism regulates Hippo pathway homeostasis
.
Genes Dev
2015
;
29
:
1271
84
.
25.
Wang
Y
,
Xu
X
,
Maglic
D
,
Dill
MT
,
Mojumdar
K
,
Ng
PK
, et al
Comprehensive molecular characterization of the Hippo signaling pathway in cancer
.
Cell Rep
2018
;
25
:
1304
17
.
26.
Lange
AW
,
Sridharan
A
,
Xu
Y
,
Stripp
BR
,
Perl
AK
,
Whitsett
JA
. 
Hippo/Yap signaling controls epithelial progenitor cell proliferation and differentiation in the embryonic and adult lung
.
J Mol Cell Biol
2015
;
7
:
35
47
.
27.
Di Agostino
S
,
Sorrentino
G
,
Ingallina
E
,
Valenti
F
,
Ferraiuolo
M
,
Bicciato
S
, et al
YAP enhances the pro-proliferative transcriptional activity of mutant p53 proteins
.
EMBO Rep
2016
;
17
:
188
201
.
28.
Slemmons
KK
,
Crose
LES
,
Riedel
S
,
Sushnitha
M
,
Belyea
B
,
Linardic
CM
. 
A novel Notch-YAP circuit drives stemness and tumorigenesis in embryonal rhabdomyosarcoma
.
Mol Cancer Res
2017
;
15
:
1777
91
.
29.
van Groningen
T
,
Koster
J
,
Valentijn
LJ
,
Zwijnenburg
DA
,
Akogul
N
,
Hasselt
NE
, et al
Neuroblastoma is composed of two super-enhancer-associated differentiation states
.
Nat Genet
2017
;
49
:
1261
6
.
30.
Dong
L
,
Lyu
X
,
Faleti
OD
,
He
ML
. 
The special stemness functions of Tbx3 in stem cells and cancer development
.
Semin Cancer Biol
2019
;
57
:
105
10
.
31.
Taniguchi
K
,
Moroishi
T
,
de Jong
PR
,
Krawczyk
M
,
Grebbin
BM
,
Luo
H
, et al
YAP-IL-6ST autoregulatory loop activated on APC loss controls colonic tumorigenesis
.
Proc Natl Acad Sci U S A
2017
;
114
:
1643
8
.
32.
Chen
J
,
Li
Y
,
Yu
TS
,
McKay
RM
,
Burns
DK
,
Kernie
SG
, et al
A restricted cell population propagates glioblastoma growth after chemotherapy
.
Nature
2012
;
488
:
522
6
.
33.
Huang
R
,
Rofstad
EK
. 
Cancer stem cells (CSCs), cervical CSCs and targeted therapies
.
Oncotarget
2017
;
8
:
35351
67
.
34.
Veschi
V
,
Verona
F
,
Thiele
CJ
. 
Cancer stem cells and neuroblastoma: characteristics and therapeutic targeting options
.
Front Endocrinol
2019
;
10
:
782
.
35.
Chen
X
,
Gu
W
,
Wang
Q
,
Fu
X
,
Wang
Y
,
Xu
X
, et al
C-MYC and BCL-2 mediate YAP-regulated tumorigenesis in OSCC
.
Oncotarget
2018
;
9
:
668
79
.
36.
Li
H
,
He
F
,
Zhao
X
,
Zhang
Y
,
Chu
X
,
Hua
C
, et al
YAP inhibits the apoptosis and migration of human rectal cancer cells via suppression of JNK-Drp1-mitochondrial fission-HtrA2/Omi pathways
.
Cell Physiol Biochem
2017
;
44
:
2073
89
.
37.
Goldsmith
KC
,
Gross
M
,
Peirce
S
,
Luyindula
D
,
Liu
X
,
Vu
A
, et al
Mitochondrial Bcl-2 family dynamics define therapy response and resistance in neuroblastoma
.
Cancer Res
2012
;
72
:
2565
77
.
38.
Gkretsi
V
,
Stylianou
A
,
Papageorgis
P
,
Polydorou
C
,
Stylianopoulos
T
. 
Remodeling components of the tumor microenvironment to enhance cancer therapy
.
Front Oncol
2015
;
5
:
214
.
39.
Slemmons
KK
,
Crose
LE
,
Rudzinski
E
,
Bentley
RC
,
Linardic
CM
. 
Role of the YAP oncoprotein in priming Ras-driven rhabdomyosarcoma
.
PLoS One
2015
;
10
:
e0140781
.
40.
Nakamura
M
,
Shimada
K
,
Konishi
N
. 
The role of HRK gene in human cancer
.
Oncogene
2008
;
27
:
S105
13
.
41.
Pike
LR
,
Phadwal
K
,
Simon
AK
,
Harris
AL
. 
ATF4 orchestrates a program of BH3-only protein expression in severe hypoxia
.
Mol Biol Rep
2012
;
39
:
10811
22
.
42.
Kaya-Aksoy
E
,
Cingoz
A
,
Senbabaoglu
F
,
Seker
F
,
Sur-Erdem
I
,
Kayabolen
A
, et al
The pro-apoptotic Bcl-2 family member Harakiri (HRK) induces cell death in glioblastoma multiforme
.
Cell Death Discov
2019
;
5
:
64
.
43.
Varelas
X
. 
The Hippo pathway effectors TAZ and YAP in development, homeostasis and disease
.
Development
2014
;
141
:
1614
26
.
44.
Hindley
CJ
,
Condurat
AL
,
Menon
V
,
Thomas
R
,
Azmitia
LM
,
Davis
JA
, et al
The Hippo pathway member YAP enhances human neural crest cell fate and migration
.
Sci Rep
2016
;
6
:
23208
.
45.
Singh
A
,
Settleman
J
. 
EMT, cancer stem cells and drug resistance: an emerging axis of evil in the war on cancer
.
Oncogene
2010
;
29
:
4741
51
.
46.
Santamaria
PG
,
Moreno-Bueno
G
,
Cano
A
. 
Contribution of epithelial plasticity to therapy resistance
.
J Clin Med
2019
;
8
:
676
.
47.
Avraham
R
,
Yarden
Y
. 
Feedback regulation of EGFR signalling: decision making by early and delayed loops
.
Nat Rev Mol Cell Biol
2011
;
12
:
104
17
.
48.
Joshi
M
,
Rice
SJ
,
Liu
X
,
Miller
B
,
Belani
CP
. 
Trametinib with or without vemurafenib in BRAF mutated non-small cell lung cancer
.
PLoS One
2015
;
10
:
e0118210
.
49.
Florent
R
,
Weiswald
LB
,
Lambert
B
,
Brotin
E
,
Abeilard
E
,
Louis
MH
, et al
Bim
, 
Puma and Noxa upregulation by naftopidil sensitizes ovarian cancer to the BH3-mimetic ABT-737 and the MEK inhibitor trametinib
.
Cell Death Dis
2020
;
11
:
380
.
50.
Kim
MH
,
Kim
J
,
Hong
H
,
Lee
SH
,
Lee
JK
,
Jung
E
, et al
Actin remodeling confers BRAF inhibitor resistance to melanoma cells through YAP/TAZ activation
.
EMBO J
2016
;
35
:
462
78
.
51.
Lin
L
,
Sabnis
AJ
,
Chan
E
,
Olivas
V
,
Cade
L
,
Pazarentzos
E
, et al
The Hippo effector YAP promotes resistance to RAF- and MEK-targeted cancer therapies
.
Nat Genet
2015
;
47
:
250
6
.
52.
Coggins
GE
,
Farrel
A
,
Rathi
KS
,
Hayes
CM
,
Scolaro
L
,
Rokita
JL
, et al
YAP1 mediates resistance to MEK1/2 inhibition in neuroblastomas with hyperactivated RAS signalling
.
Cancer Res
2019
;
79
:
6204
14
.
53.
Obata
T
,
Toyota
M
,
Satoh
A
,
Sasaki
Y
,
Ogi
K
,
Akino
K
, et al
Identification of HRK as a target of epigenetic inactivation in colorectal and gastric cancer
.
Clin Cancer Res
2003
;
9
:
6410
8
.
54.
Higuchi
T
,
Nakamura
M
,
Shimada
K
,
Ishida
E
,
Hirao
K
,
Konishi
N
. 
HRK inactivation associated with promoter methylation and LOH in prostate cancer
.
Prostate
2008
;
68
:
105
13
.
55.
Landin-Malt
A
,
Benhaddou
A
,
Zider
A
,
Flagiello
D
. 
An evolutionary, structural and functional overview of the mammalian TEAD1 and TEAD2 transcription factors
.
Gene
2016
;
591
:
292
303
.
56.
Pobbati
AV
,
Hong
W
. 
Emerging roles of TEAD transcription factors and its coactivators in cancers
.
Cancer Biol Ther
2013
;
14
:
390
8
.