Open Access

Cucurbitacin E inhibits the Yes‑associated protein signaling pathway and suppresses brain metastasis of human non‑small cell lung cancer in a murine model

  • Authors:
    • Ping‑Chih Hsu
    • Bo Tian
    • Yi‑Lin Yang
    • Yu‑Cheng Wang
    • Shu Liu
    • Anatoly Urisman
    • Cheng‑Ta Yang
    • Zhidong Xu
    • David M. Jablons
    • Liang You
  • View Affiliations

  • Published online on: June 20, 2019     https://doi.org/10.3892/or.2019.7207
  • Pages: 697-707
  • Copyright: © Hsu et al. This is an open access article distributed under the terms of Creative Commons Attribution License.

Metrics: Total Views: 0 (Spandidos Publications: | PMC Statistics: )
Total PDF Downloads: 0 (Spandidos Publications: | PMC Statistics: )


Abstract

Human non‑small cell lung cancer (NSCLC) is associated with an extremely poor prognosis especially for the 40% of patients who develop brain metastasis, and few treatment strategies exist. Cucurbitacin E (CuE), an oxygenated tetracyclic triterpenoid isolated from plants particularly of the family Cucurbitaceae, has shown anti‑tumorigenic properties in several types of cancer, yet the mechanism remains unclear. Yes‑associated protein (YAP), a main mediator of the Hippo signaling pathway, promotes tumorigenesis, drug resistance and metastasis in human NSCLC. The present study was designed to ascertain whether CuE inhibits YAP and its downstream gene expression in the human NSCLC cell lines H2030‑BrM3 (K‑rasG12C mutation) and PC9‑BrM3 (EGFRΔexon19 mutation), which have high potential for brain metastasis. The efficacy of CuE in suppressing brain metastasis of H2030‑BrM3 cells in a murine model was also investigated. It was found that after CuE treatment in H2030‑BrM3 and PC9‑BrM3 cells, YAP protein expression was decreased, and YAP signaling GTIIC reporter activity and expression of the downstream genes CTGF and CYR61 were significantly (P<0.01) decreased. CuE treatment also reduced the migration and invasion abilities of the H2030‑BrM3 and PC9‑BrM3 cells. Finally, our in vivo study showed that CuE treatment (0.2 mg/kg) suppressed H2030‑BrM3 cell brain metastasis and that mice treated with CuE survived longer than the control mice treated with 10% DMSO (P=0.02). The present study is the first to demonstrate that CuE treatment inhibits YAP and the signaling downstream gene expression in human NSCLC in vitro, and suppresses brain metastasis of NSCLC in a murine model. More studies to verify the promising efficacy of CuE in inhibiting brain metastasis of NSCLC and various other cancers may be warranted.

Introduction

Brain metastasis affects approximately 40% of patients with non-small cell lung cancer (NSCLC) (1,2). The prognosis for these patients is extremely poor; median survival is less than three months without treatment (3,4). Treatment options for those patients whose tumors lack targetable mutations remain quite limited. For example, erlotinib, an epidermal growth factor receptor (EGFR)-tyrosine kinase inhibitor (TKI), is an effective target therapy for brain metastatic lung adenocarcinoma harboring EGFR exon 19 deletion or exon 21 L858R mutations (58). Anaplastic lymphoma kinase (ALK) inhibitors including ceritinib and alectinib have better response rates than conventional chemotherapy for brain metastatic NSCLC patients with ALK-rearrangement mutation (9,10). Although the K-ras mutation is common in NSCLC patients (15–30%), there is still no approved target therapy drug for K-ras mutant metastatic NSCLC (11,12). There is therefore an important need to develop novel drugs for the treatment of brain metastatic NSCLC patients without targetable driven mutations.

Cucurbitacin E (CuE, a-elaterin) (Fig. 1A) is a tetracyclic triterpene and a biochemical natural compound from the family of cucurbitacins. CuE can be isolated from natural plants such as those of the family Cucurbitaceae (gourd, cucumber, pumpkins) (13). Previous studies have shown that CuE inhibits the proliferation, invasion and migration abilities of various types of cancers, including lung, colorectal and breast, yet the mechanism by which it performs these functions and the signaling pathway remain unclear (1319).

Yes-associated protein (YAP) is a key mediator protein in the Hippo (also known as the Salvador-Warts-Hippo) signaling pathway, and is reportedly an important oncogenic protein in various types of cancer (20,21). Recently, YAP was found to promote tumorigenesis, invasion, drug resistance, brain metastasis and immune evasion in human NSCLC (2228). Thus, YAP has been suggested as a therapeutic target for metastatic NSCLC. However, to date, no molecular inhibitor targeting YAP has been developed for the treatment of NSCLC. Therefore, identification of a natural biochemical compound that can inhibit the YAP signaling pathway is an alternative strategy.

In the present study, we sought to determine whether CuE inhibits YAP expression in the NSCLC cell lines H2030-BrM3 (K-rasG12C mutation) and PC9-BrM3 (EGFRΔexon19 mutation), which have high potential for brain metastasis. We also investigated the efficacy of CuE in suppressing human NSCLC brain metastasis in a murine model.

Materials and methods

Cell culture

Human metastatic NSCLC cell lines H2030-BrM3 (K-rasG12C mutation) (P9) and PC9-BrM3 (EGFRΔexon19 mutation) (P40) were gifts from Professor Joan Massagué (Metastasis Research Center, Memorial Sloan Kettering Cancer Center, New York, NY, USA). The H2030-BrM3 and PC9-BrM3 cell lines are in vivo selection of metastatic derivatives from NCI-H2030 (K-rasG12C mutation) and PC9 cell lines (EGFRΔexon19 mutation). The two cell lines were derived and authenticated by left ventricle inoculation as previously described (29,30). Cells were maintained in RPMI-1640 medium supplemented with 10% heat-inactivated fetal bovine serum (FBS) and streptomycin (100 mg/ml) (UCSF Cell Culture Facility, San Francisco, CA, USA), and cultured at 37°C in a humid incubator with 5% CO2.

Animal studies

All animal studies strictly followed UCSF institutional guidelines (Institutional Animal Care and Use Committee approval no. AN103205-03). Twenty athymic nude (CrTac:NCr-Foxn1nu) female mice, 6–8 weeks of age (body weight at ~20 g) were purchased from Taconic Farms, Inc. (Hudson, NY, USA) and maintained in ventilated cages (with reach food and water) at a constant temperature (20–22°C) and a 12 h light dark cycle in UCSF's Laboratory Animal Resource Center (LARC) facility. The murine experiments were initiated when mice were older than 10 weeks. A metastatic murine model was created by injecting 500,000 H2030-BrM3 cells suspended in 100 µl of PBS into the left ventricle via a percutaneous approach as previously described (29,30). To test whether cucurbitacin E (CuE) (Sigma Aldrich; Merck KGaA) could control NSCLC cell line H2030-BrM3 metastasis in our murine model, mice were divided into two groups of n=10 each. The control group was administered an intraperitoneal (i.p.) injection of 10% dimethyl sulfoxide (DMSO), and the treatment group was administered an i.p. injection of 0.2 mg/kg of CuE. Both treatments began on the day following cell injection and were administered every other day. The mice were euthanized when they showed symptoms of paralysis, appeared extremely sick or a tumor size exceeding 2 cm3, according to the guidelines established by the UCSF LARC and tumor tissues were collected. Survival was assessed from the day of cell injection to the day mice were euthanized.

To monitor the condition of brain metastasis in the mice, bioluminescent imaging (Caliper Life Sciences, Waltham, MA, USA) was used. For this, mice were injected with D-luciferin potassium salt (SYD Labs, Inc., Natick, MA, USA) at a dose of 150 mg/kg i.p., anesthetized by inhalant isoflurane 1–5% in oxygen, and then placed into the Xenogen IVIS® spectrum imaging system (PerkinElmer, Boston, MA, USA) at 10 min after D-luciferin potassium salt injection. Images were recorded with an exposure time of 1 min.

Cell viability assay

H2030-BrM3 and PC9-BrM3 cells were cultured in a 96-well plate and treated with different doses of CuE (Sigma Aldrich; Merck KGaA) (0, 0.01, 0.03, 0.1, 0.3, 1, 3, 10, 30 and 100 µM). After 72 h of incubation, the cells were lysed and CellTiter-Glo Luminescent Cell Viability Assay reagent (Promega) was added to generate luminescent signaling. Luminescent signaling was detected by using the GloMax-96 Microplate Luminometer (Promega, Madison, WI, USA). GraphPad Prism 5.0 software (GraphPad Software, Inc., La Jolla, CA, USA) was used to analyze proportional cell viability and calculate dose-response curves and the half maximal inhibitory concentration (IC50) value.

Luciferase gene transfection

The pGreenFire1-CMV Virus (pTRH1 CMV dscGFP T2A Fluc) positive control was purchased from System Bioscience LLC (Palo Alto, CA, USA). Virus particles were mixed with transfection reagents and medium according to the manufacturer's protocol, and added to the H2030-BrM3 cells cultured in 12-well plates. Fluorescence microscopy was used to determine the efficacy of transfection at 48 h after transfection.

Western blot analysis

The samples were run on 4–20% gradient SDS-polyacrylamide gels (Bio-Rad Laboratories, Inc., Hercules, CA, USA) and then were transferred to Immobilon-P nitrocellulose membranes (Millipore, Bellerica, MA, USA). The membranes were probed with the following primary antibodies: Rabbit anti-YAP (Cell Signaling, cat. no. 4912; dilution 1:1,000), anti-ERK1/2 (Cell Signaling, cat. no. 4912; dilution 1:1,000) and mouse anti-GAPDH (Sigma-Aldrich; Merck KGaA; cat. no. 100242-MM05; dilution 1:10,000) at 4°C overnight after blocking with 5% non-fat milk. The membranes were then incubated with species-specific conjugated secondary antibodies (GE Dharmacon) (cat. no. NA934, anti-rabbit for YAP; cat. no. NA931, anti-mouse for GAPDH) at room temperature for 1 h. Finally, an ECL blotting analysis system (Amersham Pharmacia Biotech, Piscataway, NJ, USA) was used to detect protein expression.

Protein degradation assay

H2030-BrM3 and PC9-BrM3 cells were placed in 6-well plates, and treated with either 0.1 µM CuE or DMSO (control). After 24 h of treatment, the cells were treated with 100 µg/ml cycloheximide (CHX) (Sigma-Aldrich; Merck KGaA), an inhibitor of protein synthesis. Cells were harvested at the time points of 0, 1 and 2 h after CHX treatment. Total proteins were extracted and YAP expression was analyzed by western blot analysis.

Luciferase reporter assay

The cell lines H2030-BrM3 and PC9-BrM3 were placed in 24-well plates and transfected with 8×GTIIC-luciferase plasmid (Addgene, Cambridge, MA, USA) and Renilla luciferase pRL-TK plasmid (Promega, Madison, WI, USA) by using transfection reagent Lipofectamine 2000 (Invitrogen; Thermo Fisher Scientific, Inc.). At 24 h after transfection, the cells were treated with CuE. After a 24 h treatment, the cells were lysed and luciferase activity was assayed by using Dual-Luciferase Reporter Assay system (Promega). Luminescent signaling was detected by using GloMax-96 Microplate Luminometer. All luciferase activities were normalized to Renilla activity. All experiments of luciferase reporter assay were triplicated to achieve statistical significance [DMSO (n=3), CuE 0.05 µM (n=3), CuE 0.1 µM (n=3)].

RNA isolation, cDNA synthesis, and quantitative real-time RT-PCR

Total RNA was extracted from cells by using the High Pure RNA Isolation Kit (Roche, Indianapolis, IN, USA). The iScript cDNA Synthesis Kit (Bio-Rad Laboratories, Inc.) was used to transcribe RNA to cDNA, and the cDNA was used as the template for real-time PCR. Commercially available primers and probe sequences (Applied Biosystems; Thermo Fisher Scientific, Inc.) were used to detect YAP, CYR61, CTGF, EREG and endogenous control gene β-glucuronidase (GUSB) gene expression. Applied Biosystems Relative Quantification software (Version C; Thermo Fisher Scientific, Inc.) was used for mRNA expression analysis. Real-time PCR detection was conducted using TaqMan Technology on an Applied Biosystems 7000 sequence detection system (Thermo Fisher Scientific, Inc.). All experiments of real-time RT-PCR were triplicated to achieve statistical significance [DMSO (n=3), CuE 0.05 µM (n=3), CuE 0.1 µM (n=3)].

Wound-healing and Transwell invasion assay

For the wound-healing migration assay, H2030-BrM3 and PC9-BrM3 cells were subcultured in 6-well plates until the condition of confluence was reached. The plates were scratched using a 200 µl pipette tip, and the cells were grown continuously. In a test of CuE treatment, 0.05 µM was added after the scratch. Phase contrast images were captured at the time of the scratch (0 h) and 18 h later with a Primo Vert microscope (magnification ×100, ×200 and ×400, respectively; Carl Zeiss, Gottingen, Germany), and the distance of cell movement was measured by ImageJ software (National Institutes of Health, Bethesda, MD, USA).

The 6-well plate Transwell system used for the Transwell invasion assay was purchased from Corning Inc. (Corning, NY, USA). The upper chamber of the Transwell plate was coated with 300 µl mMatrigel. Next, H2030-BrM3 and PC9-BrM3 cells were trypsinized and re-suspended in serum-free medium and the cells were seeded on the upper chamber of the Transwell plate. CuE (0.05 µM) was added with serum-free medium to the upper chamber. The lower chamber was infused with 2 ml complete growth medium (10% FBS). The gel in the upper chamber of the Transwell plate was wiped after incubation at 37°C for 48 h. The membrane of the upper chamber was stained by crystal violet for 10 min after methanol fixation. The Primo Vert microscope was again used to observe the cells on the lower side of the membrane, and phase contrast images were captured at ×100, ×200 and ×400 magnification.

Wound-healing and Tanswell invasion assays were triplicated to achieve statistical significance [DMSO (n=3, CuE 0.05 µM (n=3)].

Histologic analysis and immunohistochemistry

After mice were euthanized, the tumor tissues from brain metastases were harvested, immediately fixed in 10% paraformaldehyde for 24 h and then embedded in paraffin. Histological sections of the tissues were stained with hematoxylin and eosin (H&E) stain for general morphological analysis. The morphological analysis to verify brain metastasis in the stained tissues was performed by an experienced pathologist. The YAP immunohistochemistry (IHC) staining was conducted as previously described (27). All images were obtained using a Zeiss Axioscop 2 microscope at ×100, ×200 and ×400 magnification (Carl Zeiss Inc.).

Statistical analysis

All statistical analyses were conducted using GraphPad Prism (version 5.0; GraphPad Software, San Diego, CA, USA). Data are expressed as mean ± standard deviation (SD) from three independent experiments. t-tests were used to compare the differences between two groups, and one-way ANOVA followed by Turkey multiple comparisons was used to compare differences among >2 groups. A Kaplan-Meier survival curve was calculated to determine survival in the animal experiments. The experiments of reporter assay, real-time RT-PCR, wound-healing and Transwell invasion assays were triplicated to achieve statistical significance. All P-values were 2-sided and considered to be statistically significant at P<0.05 (*P<0.05, **P<0.01, ***P<0.001 as indicated in the figures and legends).

Results

CuE decreases YAP protein expression in H2030-BrM3 and PC9-BrM3 cells

The cell viability assay of H2030-BrM3 and PC9-BrM3 cells treated with CuE yielded IC50 values of 0.146 and 0.187 µM, respectively (Fig. 1B and C). Western blotting showed that the level of YAP protein was decreased after treatment with 0.05 and 0.1 µM CuE in the H2030-BrM3 and PC9-BrM3 cells when compared to the DMSO control (Fig. 1D). The YAP protein degradation assay showed that in H2030-BrM3 and PC9-BrM3 cells treated with 0.1 µM CuE, YAP protein expression reached low levels at 2 h when compared to the DMSO-treated controls (Fig. 1E). Real-time PCR showed that YAP mRNA expression was significantly decreased in the H2030-BrM3 and PC9-BrM3 cell lines after treatment with 0.05 (P<0.001 and P<0.01) and 0.1 µM CuE (both P<0.001) when compared with the DMSO control (Fig. S1A and B). Western blot analysis showed that ERK1/2 protein was significantly decreased after treatment with 0.05 and 0.1 µM CuE in the H2030-BrM3 and PC9-BrM3 cell lines (Fig. S1C and D).

CuE decreases GTIIC reporter activity and YAP downstream gene mRNA expression

After verifying that CuE decreases YAP protein expression in H2030-BrM3 and PC9-BrM3 cells, we ascertained whether it could also decrease GTIIC reporter activity and YAP downstream gene mRNA expression. We demonstrated that GTIIC reporter activity in the H2030-BrM3 and PC9-BrM3 cell lines was significantly decreased after 0.05 and 0.1 µM CuE treatment compared to the DMSO control (all P<0.001) (Fig. 2A and B). In both cell lines, mRNA expression of the YAP downstream genes CTGF and CYR61 was significantly decreased after 0.05 and 0.1 µM CuE treatment (P<0.01) (Fig. 2C and D), as was mRNA expression of epidermal growth factor (EGF), epiregulin (EREG) (P<0.05) (Fig. S1E and F).

CuE reduces migration and invasion abilities of H2030-BrM3 and PC9-BrM3 cells

To investigate whether CuE could reduce the migration and invasion abilities of H2030-BrM3 and PC9-BrM3 cells, we used wound closure and Transwell invasion assays. The migration assay showed that the wound closure rate was significantly decreased in the H2030-BrM3 and PC9-BrM3 cells after 0.05 µM CuE treatment (P<0.01 and P<0.001) when compared with the DMSO control (Fig. 3A-D). The Transwell invasion assay showed that the number of cells that invaded the lower side of the membrane was significantly decreased in both cell lines after 0.05 µM CuE treatment (P<0.01) when compared to the DMSO control (Fig. 3E-H) (Fig. S2).

CuE decreases the burden of brain metastasis in an in vivo H2030-BrM3 cell murine model of brain metastasis

To test the efficacy of CuE in suppressing brain metastasis of H2030-BrM3 cells in vivo, mice were inoculated with H2030-BrM3 cells through percutaneous left ventricle injection and then treated with CuE at a dose of 0.2 mg/kg. Control mice were treated with 10% DMSO. Bioluminescent imaging showed that mice treated with CuE had significantly decreased brain metastatic burden compared to the control group at day 35 (week 7) (P=0.034 (Fig. 4A and B). Mice treated with CuE survived significantly longer than those treated with 10% DMSO (P=0.0212) (Fig. 4C).

CuE decreases nuclear YAP immunohistochemical (IHC) staining of mouse brain tumor tissue

After mice were euthanized, brain tissues were collected from the DMSO control and CuE-treated groups for histological analysis. H&E staining was assayed to verify that H2030-BrM3 tumor cell metastasis appeared in the mouse model. Metastasis of H2030-BrM3 tumor cells within brain tissues could be observed by H&E staining (Fig. 5A). To observe the late effect of CuE treatment on YAP in regards to brain metastasis, nuclear YAP IHC staining was performed. The results showed that nuclear YAP IHC staining was decreased in the brain tissue of the CuE-treated group when compared to that observed in the brain tissue of the control DMSO group (Fig. 5B).

Discussion

Although the natural biochemical compound CuE has been shown to inhibit the proliferation, invasion and migration abilities of various types of cancer, the mechanism and the signaling pathway remain unclear, until now. Our study revealed that CuE inhibited YAP protein expression in the NSCLC cell lines H2030-BrM3 and PC9-BrM3, which were derived from NSCLC cell lines NCI-H2030 (K-rasG12C mutation) and PC9 (EGFRΔexon19 mutation), which show very strong brain metastatic potential in murine experiments (27,29). We verified that CuE decreases YAP GTIIC Hippo reporter activity, and expression of the downstream genes connective tissue growth factor (CTGF) and cysteine-rich angiogenic inducer 61 (CYR61). Collectively, these results suggest that CuE inhibits tumorigenesis, invasion and migration thorough inhibition of the YAP signaling pathway.

PC9-BrM3 cells contain an EGFRΔexon19 mutation and depend on the EGFR/mitogen-activated protein kinase (MAPK)/extracellular signal-regulated kinase (ERK) signaling pathway to maintain tumor survival. PC9-BrM3 cells are sensitive to EGFR-tyrosine kinase inhibitor treatment (27,29,30). Although H2030-BrM3 cells have a K-rasG12C mutation, previous studies suggest that these cells do not fully depend on the EGFR/RAS/MAPK/ERK signaling pathway to maintain tumor survival (27,31). In addition, YAP has been suggested to be an oncogenic protein that partly takes over the K-ras mutation in H2030-BrM3 cells (27,31). These findings may explain why YAP protein expression did not decrease equally after CuE treatment in the two cell lines.

Our experimental data showed that CuE treatment decreased YAP mRNA expression, ERK1/2 protein expression, and epidermal growth factor (EGF) EREG mRNA expression in H2030-BrM3 and PC9-BM3 cells. These results suggest that CuE inhibits YAP, at least in part, indirectly through the EGFR/MAPK/ERK signaling pathway. Previous studies have reported crosstalk between the Hippo/YAP and EGFR/MAPK/ERK signaling pathways in human NSCLC cells (22,24,32). Increased cytoplasmic YAP protein translocates into the nucleus to form complexes with transcriptional enhancer factors (TEF; also known as TEAD) leading to activate transcription of downstream genes including CTGF, CYR61 and EGF such as EREG (22,24,27,32). Therefore, YAP regulates these downstream genes, including CTGF, CYR61 and EREG, at the transcriptional mRNA level, not at the protein level. The increased expression of CTGF, CYR61 and EREG may form an autocrine loop with the EGFR/MAPK/ERK signaling pathway to re-enforce YAP protein expression. Two studies demonstrated that CuE inhibits the EGFR/MAPK/ERK signaling pathway in cancer cells (33,34). Recently, CuB was shown to inhibit the Hippo/YAP pathway in human colorectal cancer cell lines (35). These findings lead us to believe that CuE inhibits YAP indirectly through the EGFR/MAPK/ERK and Hippo/YAP signaling pathways in human NSCLC. Further studies in the future to investigate the effects of CuE on ErbB family receptors (EGFR, HER2, ErbB3 and HER4)/MAPK/ERK signaling pathway and Hippo kinase including neurofibromatosis 2 (NF2), large tumor suppressor homolog 1 (LATS1), LATS2, and mammalian sterile-20 like kinase 1 (MST1) in various cancers are suggested according to our findings. The dependence on the EGFR/MAPK/ERK signaling pathway of PC9-BrM3 cells may explain why more YAP protein was decreased and degraded after CuE treatment in that cell line when compared with that observed in the H2030-BrM3 cells.

Several studies have shown that CuE has anticancer activity to NSCLC by inhibiting oncogenic signaling pathways, including the Wnt signaling pathway and the Janus kinase (JAK)/signal transducer and activator of transcription 3 (STAT3) signaling pathway (15,36,37). Wnt and STAT3 signaling pathways were reportedly involved in regulating the expression of Hippo downstream genes CTGF and CYR61 (3841). Collectively, these findings may explain why the decrease of 50–80% in YAP protein by CuE treatment resulted in more than 90% of CTGF and CYR61 mRNA expression in H2030-BrM3 cells in our study.

According to several reports, YAP takes over KRAS as a cancer driver in KRAS-mutant NSCLC, and promotes cancer metastasis in NSCLC (4145). A recent study found that K-ras mutant H2030-BrM3 cells have increased YAP protein and YAP downstream genes CTGF and CYR61 mRNA expressions compared with parental H2030 cells (27). In that study, direct inhibition of YAP by short hairpin RNA suppressed brain metastasis of H2030-BrM3 cells in a mouse model. After verifying that CuE inhibits YAP signaling in vitro, we explored the efficacy of CuE in suppressing brain metastasis of H2030-BrM3 cells in a mouse model (Fig. 4D).

Our study is the first to show that CuE suppresses brain metastasis of human NSCLC in a murine model. A recent study showed that cucurbitacin B (another tetracyclic cucurbitane) inhibits the Hippo-YAP signaling pathway in colorectal cancer cells, but included no in vivo data (35). Another study demonstrated that CuE inhibited breast cancer in a mouse model of metastasis by suppressing cell migration and invasion, but the anti-cancer mechanism of CuE was not investigated (18).

In conclusion, our study revealed that the anti-tumorigenic effects of CuE occur through inhibition of the YAP signaling pathway, and that CuE treatment suppressed brain metastasis of human NSCLC cells in a murine model. More studies to verify the promising efficacy of CuE in inhibiting brain metastasis of NSCLC and various other cancers may be warranted.

Supplementary Material

Supporting Data

Acknowledgements

We thank Professor Joan Massagué (Metastasis Research Center, Memorial Sloan Kettering Cancer Center, New York, NY, USA) for providing the human metastatic cell lines H2030-BrM3 and PC9-BrM3. We thank Pamela Derish in the UCSF Department of Surgery for editorial assistance with the manuscript.

Funding

The present study was supported by the National Institutes of Health (NIH; grant no. R01 CA140654 to LY) and Chang-Gung Medical Research Project (CMRP no. CMRPG3G1661 to PCH and no. CMRPG5H0141 to CTY). We are grateful for support from the Kazan McClain Partner's Foundation; the Estate of Robert Griffiths; the Jeffrey and Karen Peterson Family Foundation; Paul and Michelle Zygielbaum; the Estate of Norman Mancini; and the Barbara Isackson Lung Cancer Research Fund.

Availability of data and materials

The datasets used during the present study are available from the corresponding author upon reasonable request.

Authors' contributions

Conception and design was accomplished by PCH and LY. Development of the research methodology was carried out by PCH, ZX and YLY. Acquisition of data (as well as provision of research animals and facilities) was accomplished by PCH, BT, SL and DMJ. Analysis and interpretation of data (pathologic analysis) was carried out by PCH, BT, ZX and AU. Writing, review and/or revision of the manuscript was carried out by PCH, BT, YLY, CTY and LY. Administrative, technical, or material support (organizing data) was achieved by PCH, BT, YLY, YCW, ZX and LY. Study supervision was carried out by LY. All authors read and approved the manuscript and agree to be accountable for all aspects of the research in ensuring that the accuracy or integrity of any part of the work are appropriately investigated and resolved.

Ethics approval and consent to participate

All animal studies strictly followed UCSF institutional guidelines (Institutional Animal Care and Use Committee approval no. AN103205-03).

Patient consent for publication

Not applicable.

Competing interests

All authors have no competing interest.

Glossary

Abbreviations

Abbreviations:

CuE

cucurbitacin E

NSCLC

non-small cell lung cancer

YAP

Yes-associated protein

CHX

cycloheximide

References

1 

Duell T, Kappler S, Knöferl B, Schuster T, Hochhaus J, Morresi-Hauf A, Huber RM, Tufman A and Zietemann V: Prevalence and risk factors of brain metastases in patients with newly diagnosed advanced non-small-cell lung cancer. Cancer Treat Commun. 4:106–112. 2015. View Article : Google Scholar

2 

Ma X, Zhu H, Guo H, Han A, Wang H, Jing W, Zhang Y, Kong L and Yu J: Risk factors of brain metastasis during the course of EGFR-TKIs therapy for patients with EGFR-mutated advanced lung adenocarcinoma. Oncotarget. 7:81906–81917. 2016. View Article : Google Scholar : PubMed/NCBI

3 

Sørensen JB, Hansen HH, Hansen M and Dombernowsky P: Brain metastases in adenocarcinoma of the lung: Frequency, risk groups, and prognosis. J Clin Oncol. 6:1474–1480. 1988. View Article : Google Scholar : PubMed/NCBI

4 

Dempke WC, Edvardsen K, Lu S, Reinmuth N, Reck M and Inoue A: Brain metastases in NSCLC-are TKIs changing the treatment strategy? Anticancer Res. 35:5797–5806. 2015.PubMed/NCBI

5 

Deng Y, Feng W, Wu J, Chen Z, Tang Y, Zhang H, Liang J, Xian H and Zhang S: The concentration of erlotinib in the cerebrospinal fluid of patients with brain metastasis from non-small-cell lung cancer. Mol Clin Oncol. 2:116–120. 2014. View Article : Google Scholar : PubMed/NCBI

6 

Paz-Ares L, Soulières D, Moecks J, Bara I, Mok T and Klughammer B: Pooled analysis of clinical outcome for EGFR TKI-treated patients with EGFR mutation-positive NSCLC. J Cell Mol Med. 18:1519–1539. 2014. View Article : Google Scholar : PubMed/NCBI

7 

Taniguchi Y, Tamiya A, Nakahama K, Naoki Y, Kanazu M, Omachi N, Okishio K, Kasai T and Atagi S: Impact of metastatic status on the prognosis of EGFR mutation-positive non-small cell lung cancer patients treated with first-generation EGFR-tyrosine kinase inhibitors. Oncol Lett. 14:7589–7596. 2017.PubMed/NCBI

8 

Li K, Yang M, Liang N and Li S: Determining EGFR-TKI sensitivity of G719X and other uncommon EGFR mutations in non-small cell lung cancer: Perplexity and solution (Review). Oncol Rep. 37:1347–1358. 2017. View Article : Google Scholar : PubMed/NCBI

9 

Cao Z, Gao Q, Fu M, Ni N, Pei Y and Ou WB: Anaplastic lymphoma kinase fusions: Roles in cancer and therapeutic perspectives. Oncol Lett. 17:2020–2030. 2019.PubMed/NCBI

10 

Khan M, Lin J, Liao G, Tian Y, Liang Y, Li R, Liu M and Yuan Y: ALK inhibitors in the treatment of ALK positive NSCLC. Front Oncol. 8:5572019. View Article : Google Scholar : PubMed/NCBI

11 

Saber A, van der Wekken A, Hiltermann TJ, Kok K, Van den Berg A and Groen H: Genomic aberrations guiding treatment of non-small cell lung cancer patients. Cancer Treat Commun. 4:23–33. 2015. View Article : Google Scholar

12 

Tan WL, Jain A, Takano A, Newell EW, Iyer NG, Lim WT, Tan EH, Zhai W, Hillmer AM, Tam WL and Tan DSW: Novel therapeutic targets on the horizon for lung cancer. Lancet Oncol. 17:e347–e362. 2016. View Article : Google Scholar : PubMed/NCBI

13 

Torkey HM, Abou-Yousef HM, Abdel Azeiz AZ and Hoda EA: Insecticidal effect of cucurbitacin E glycoside isolated from Citrullus colocynthis against Aphis craccivora. Aust J Basic Appl Sci. 3:4060–4066. 2009.

14 

Ma G, Luo W, Lu J, Ma DL, Leung CH, Wang Y and Chen X: Cucurbitacin E induces caspase-dependent apoptosis and protective autophagy mediated by ROS in lung cancer cells. Chem Biol Interact. 253:1–9. 2016. View Article : Google Scholar : PubMed/NCBI

15 

Feng H, Zang L, Zhao ZX and Kan QC: Cucurbitacin-E inhibits multiple cancer cells proliferation through attenuation of Wnt/β-catenin signaling. Cancer Biother Radiopharm. 29:210–214. 2014. View Article : Google Scholar : PubMed/NCBI

16 

Hsu YC, Huang TY and Chen MJ: Therapeutic ROS targeting of GADD45γ in the induction of G2/M arrest in primary human colorectal cancer cell lines by cucurbitacin E. Cell Death Dis. 5:e11982014. View Article : Google Scholar : PubMed/NCBI

17 

Kong Y, Chen J, Zhou Z, Xia H, Qiu MH and Chen C: Cucurbitacin E induces cell cycle G2/M phase arrest and apoptosis in triple negative breast cancer. PLoS One. 9:e1037602014. View Article : Google Scholar : PubMed/NCBI

18 

Zhang T, Li J, Dong Y, Zhai D, Lai L, Dai F, Deng H, Chen Y, Liu M and Yi Z: Cucurbitacin E inhibits breast tumor metastasis by suppressing cell migration and invasion. Breast Cancer Res Treat. 135:445–458. 2012. View Article : Google Scholar : PubMed/NCBI

19 

Garg S, Kaul SC and Wadhwa R: Cucurbitacin B and cancer intervention: Chemistry, biology and mechanisms (Review). Int J Oncol. 52:19–37. 2018.PubMed/NCBI

20 

Johnson R and Halder G: The two faces of hippo: Targeting the hippo pathway for regenerative medicine and cancer treatment. Nat Rev Drug Discov. 13:63–79. 2014. View Article : Google Scholar : PubMed/NCBI

21 

Zanconato F, Cordenonsi M and Piccolo S: YAP/TAZ at the roots of cancer. Cancer Cell. 29:783–803. 2016. View Article : Google Scholar : PubMed/NCBI

22 

You B, Yang YL, Xu Z, Dai Y, Liu S, Mao JH, Tetsu O, Li H, Jablons DM and You L1: Inhibition of ERK1/2 down-regulates the Hippo/YAP signaling pathway in human NSCLC cells. Oncotarget. 6:4357–4368. 2015. View Article : Google Scholar : PubMed/NCBI

23 

Ye XY, Luo QQ, Xu YH, Tang NW, Niu XM, Li ZM, Shen SP, Lu S and Chen ZW: 17-AAG suppresses growth and invasion of lung adenocarcinoma cells via regulation of the LATS1/YAP pathway. J Cell Mol Med. 19:651–663. 2015. View Article : Google Scholar : PubMed/NCBI

24 

Hsu PC, You B, Yang YL, Zhang WQ, Wang YC, Xu Z, Dai Y, Liu S, Yang CT, Li H, et al: YAP promotes erlotinib resistance in human non-small cell lung cancer cells. Oncotarget. 7:51922–51933. 2016. View Article : Google Scholar : PubMed/NCBI

25 

Janse van Rensburg HJ and Yang X: The roles of the hippo pathway in cancer metastasis. Cell Signal. 28:1761–1772. 2016. View Article : Google Scholar : PubMed/NCBI

26 

Miao J, Hsu PC, Yang YL, Xu Z, Dai Y, Wang Y, Chan G, Huang Z, Hu B, Li H, et al: YAP regulates PD-L1 expression in human NSCLC cells. Oncotarget. 8:114576–114587. 2017. View Article : Google Scholar : PubMed/NCBI

27 

Hsu PC, Miao J, Huang Z, Yang YL, Xu Z, You J, Dai Y, Yeh CC, Chan G, Liu S, et al: Inhibition of yes-associated protein suppresses brain metastasis of human lung adenocarcinoma in a murine model. J Cell Mol Med. 22:3073–3085. 2018. View Article : Google Scholar : PubMed/NCBI

28 

Hsu PC, Yang CT, Jablons DM and You L: The role of yes-associated protein (YAP) in regulating programmed death-ligand 1 (PD-L1) in thoracic cancer. Biomedicines. 6:E1142018. View Article : Google Scholar : PubMed/NCBI

29 

Nguyen DX, Chiang AC, Zhang XH, Kim JY, Kris MG, Ladanyi M, Gerald WL and Massagué J: WNT/TCF signaling through LEF1 and HOXB9 mediates lung adenocarcinoma metastasis. Cell. 138:51–62. 2009. View Article : Google Scholar : PubMed/NCBI

30 

Valiente M, Obenauf AC, Jin X, Chen Q, Zhang XH, Lee DJ, Chaft JE, Kris MG, Huse JT, Brogi E, et al: Serpins promote cancer cell survival and vascular co-option in brain metastasis. Cell. 156:1002–1016. 2014. View Article : Google Scholar : PubMed/NCBI

31 

Kim J, McMillan E, Kim HS, Venkateswaran N, Makkar G, Rodriguez-Canales J, Villalobos P, Neggers JE, Mendiratta S, Wei S, et al: XPO1-dependent nuclear export is a druggable vulnerability in KRAS-mutant lung cancer. Nature. 538:114–117. 2016. View Article : Google Scholar : PubMed/NCBI

32 

He C, Mao D, Hua G, Lv X, Chen X, Angeletti PC, Dong J, Remmenga SW, Rodabaugh KJ, Zhou J, et al: The hippo/YAP pathway interacts with EGFR signaling and HPV oncoproteins to regulate cervical cancer progression. EMBO Mol Med. 7:1426–1449. 2015. View Article : Google Scholar : PubMed/NCBI

33 

Lan T, Wang L, Xu Q, Liu W, Jin H, Mao W and Wang X and Wang X: Growth inhibitory effect of cucurbitacin E on breast cancer cells. Int J Clin Exp Pathol. 6:1799–1805. 2013.PubMed/NCBI

34 

Wu YL, Zhang YJ, Yao YL, Li ZM, Han X, Lian LH, Zhao YQ and Nan JX: Cucurbitacin E ameliorates hepatic fibrosis in vivo and in vitro through activation of AMPK and blocking mTOR-dependent signaling pathway. Toxicol Lett. 258:147–158. 2016. View Article : Google Scholar : PubMed/NCBI

35 

Chai Y, Xiang K, Wu Y, Zhang T, Liu Y, Liu X, Zhen W and Si Y: Cucurbitacin B inhibits the hippo-YAP signaling pathway and exerts anticancer activity in colorectal cancer cells. Med Sci Monit. 24:9251–9258. 2018. View Article : Google Scholar : PubMed/NCBI

36 

Cai Y, Fang X, He C, Li P, Xiao F, Wang Y and Chen M: Cucurbitacins: A systematic review of the phytochemistry and anticancer activity. Am J Chin Med. 43:1331–1350. 2015. View Article : Google Scholar : PubMed/NCBI

37 

Sun C, Zhang M, Shan X, Zhou X, Yang J, Wang Y, Li-Ling J and Deng Y: Inhibitory effect of cucurbitacin E on pancreatic cancer cells growth via STAT3 signaling. J Cancer Res Clin Oncol. 136:603–610. 2010. View Article : Google Scholar : PubMed/NCBI

38 

Surmann-Schmitt C, Sasaki T, Hattori T, Eitzinger N, Schett G, von der Mark K and Stock M: The Wnt antagonist Wif-1 interacts with CTGF and inhibits CTGF activity. J Cell Physiol. 227:2207–2216. 2012. View Article : Google Scholar : PubMed/NCBI

39 

Liu H, Peng F, Liu Z, Jiang F, Li L, Gao S, Wang G, Song J, Ruan E, Shao Z, et al: CYR61/CCN1 stimulates proliferation and differentiation of osteoblasts in vitro and contributes to bone remodeling in vivo in myeloma bone disease. Int J Oncol. 50:631–639. 2017. View Article : Google Scholar : PubMed/NCBI

40 

Ren L, Wang X, Dong Z, Liu J and Zhang S: Bone metastasis from breast cancer involves elevated IL-11 expression and the gp130/STAT3 pathway. Med Oncol. 30:6342013. View Article : Google Scholar : PubMed/NCBI

41 

Liao XH, Wang N, Liu LY, Zheng L, Xing WJ, Zhao DW, Sun XG, Hu P, Dong J and Zhang TC: MRTF-A and STAT3 synergistically promote breast cancer cell migration. Cell Signal. 26:2370–2380. 2014. View Article : Google Scholar : PubMed/NCBI

42 

Singh A, Greninger P, Rhodes D, Koopman L, Violette S, Bardeesy N and Settleman J: A gene expression signature associated with ‘K-Ras addiction’ reveals regulators of EMT and tumor cell survival. Cancer Cell. 15:489–500. 2009. View Article : Google Scholar : PubMed/NCBI

43 

Shao DD, Xue W, Krall EB, Bhutkar A, Piccioni F, Wang X, Schinzel AC, Sood S, Rosenbluh J and Kim JW: KRAS and YAP1 converge to regulate EMT and tumor survival. Cell. 158:171–184. 2014. View Article : Google Scholar : PubMed/NCBI

44 

Kapoor A, Yao W, Ying H, Hua S, Liewen A, Wang Q, Zhong Y, Wu CJ, Sadanandam A, Hu B, et al: Yap1 activation enables bypass of oncogenic Kras addiction in pancreatic cancer. Cell. 158:185–197. 2014. View Article : Google Scholar : PubMed/NCBI

45 

Greten FR: YAP1 takes over when oncogenic K-Ras slumbers. Cell. 158:11–12. 2014. View Article : Google Scholar : PubMed/NCBI

Related Articles

Journal Cover

August-2019
Volume 42 Issue 2

Print ISSN: 1021-335X
Online ISSN:1791-2431

Sign up for eToc alerts

Recommend to Library

Copy and paste a formatted citation
x
Spandidos Publications style
Hsu PC, Tian B, Yang YL, Wang YC, Liu S, Urisman A, Yang CT, Xu Z, Jablons DM, You L, You L, et al: Cucurbitacin E inhibits the Yes‑associated protein signaling pathway and suppresses brain metastasis of human non‑small cell lung cancer in a murine model. Oncol Rep 42: 697-707, 2019
APA
Hsu, P., Tian, B., Yang, Y., Wang, Y., Liu, S., Urisman, A. ... You, L. (2019). Cucurbitacin E inhibits the Yes‑associated protein signaling pathway and suppresses brain metastasis of human non‑small cell lung cancer in a murine model. Oncology Reports, 42, 697-707. https://doi.org/10.3892/or.2019.7207
MLA
Hsu, P., Tian, B., Yang, Y., Wang, Y., Liu, S., Urisman, A., Yang, C., Xu, Z., Jablons, D. M., You, L."Cucurbitacin E inhibits the Yes‑associated protein signaling pathway and suppresses brain metastasis of human non‑small cell lung cancer in a murine model". Oncology Reports 42.2 (2019): 697-707.
Chicago
Hsu, P., Tian, B., Yang, Y., Wang, Y., Liu, S., Urisman, A., Yang, C., Xu, Z., Jablons, D. M., You, L."Cucurbitacin E inhibits the Yes‑associated protein signaling pathway and suppresses brain metastasis of human non‑small cell lung cancer in a murine model". Oncology Reports 42, no. 2 (2019): 697-707. https://doi.org/10.3892/or.2019.7207