miR‑375/Yes‑associated protein axis regulates IL‑6 and TGF‑β expression, which is involved in the cisplatin‑induced resistance of liver cancer cells

  • Authors:
    • Kanru Yu
    • Hao Li
    • Zhongyi Jiang
    • Han-Jen Hsu
    • Hsing-Chun Hsu
    • Yumei Zhang
    • Kunwei Wang
  • View Affiliations

  • Published online on: June 14, 2021     https://doi.org/10.3892/or.2021.8112
  • Article Number: 162
Metrics: Total Views: 0 (Spandidos Publications: | PMC Statistics: )
Total PDF Downloads: 0 (Spandidos Publications: | PMC Statistics: )


Abstract

Chemotherapy resistance is one of the major challenges in the treatment of liver cancer (LC). The present study aimed to investigate the potential roles of Yes‑associated protein (YAP), the core component of the Hippo signaling pathway, in chemoresistance of LC. YAP expression and its function in chemoresistance of LC cells were investigated. It was revealed that the expression levels and nuclear localization of YAP were increased in cisplatin (CDDP)‑resistant LC (LC/CDDP) cells. The targeted inhibition of YAP using small interfering RNA or an inhibitor restored the CDDP sensitivity of LC cells. YAP overexpression was discovered to be essential for the increase of IL‑6 and TGF‑β expression levels in LC/CDDP cells. Furthermore, it was identified that increased mRNA stability was the primary reason for the upregulation of YAP expression in LC/CDDP cells, which was due to the downregulation of microRNA (miR)‑375 expression in LC/CDDP cells. In conclusion, the findings of the present study suggested that the miR‑375/YAP axis may regulate the expression levels of IL‑6 and TGF‑β, which may subsequently be involved in the CDDP resistance of LC cells. The current results indicated that the targeted inhibition of this axis and signaling pathway may be helpful in overcoming CDDP resistance.

Introduction

Liver cancer (LC) was the third leading cause of cancer-associated deaths worldwide in 2016, demonstrating an increasing incidence rate (1). Notably, >50% of LC cases occur in China (2). Liver resection or transplantation is available for early stage LC, while for patients who have reached a stage beyond curative surgery, systematic chemotherapy is the primary treatment option (3). Tyrosine kinase inhibitors (TKIs), such as sorafenib, have been widely used as first-line chemotherapy treatments for LC (4). Cisplatin (CDDP) is another frontline chemotherapeutic drug used for the treatment of LC (5); it can induce the apoptosis of cancer cells via intercalating base pairs of DNA strands and inhibiting DNA/RNA synthesis (6,7). However, chemoresistance is one of the greatest challenges for the chemotherapeutic treatment of LC, leading to limited therapy efficiency and a poor prognosis (8). Therefore, it remains a priority to investigate the mechanisms involved in chemotherapy resistance to overcome this resistance and increase the efficacies of treatments.

The dysregulation of the Hippo signaling pathway has been reported in various types of cancer, including prostate, ovarian, colon, liver, lung and pancreatic cancer (9). Yes-associated protein (YAP) is the core component of the Hippo signaling pathway and is highly conserved from the fruit fly (Drosophila) to mammals (10). The upregulation of YAP expression has been reported in several types of human tumor, such as breast cancer (11), and has been associated with a poor prognosis of cancer progression in breast and lung cancer (1214). Previous studies have indicated that the dysregulation of the YAP and Hippo signaling pathway is involved in the chemoresistance of cancer cells; for example, YAP promotes epithelial-mesenchymal transition and chemoresistance in pancreatic cancer cells (15), and it regulates cellular quiescence to modulate chemoresistance and cancer relapse in colon cancer cells (16). However, whether YAP is involved in the chemoresistance of LC remains to be determined. Therefore, the present study aimed to investigate the potential roles of YAP in LC chemoresistance.

Materials and methods

Cell culture

The human LC cells, HepG2, Huh-6 and Huh-7, were purchased from the American Type Culture Collection. Cells were cultured in DMEM supplemented with 10% FBS (both Gibco; Thermo Fisher Scientific, Inc.) and maintained in a 5% CO2 incubator at 37°C.

To generate CDDP-resistant LC cells, cells were treated with increasing concentrations of CDDP (Sigma-Aldrich; Merck KGaA) over 6 months, with a final concentration of 1 µM, as reported previously (17,18). The resistant cells were named HepG2/CDDP, Huh6/CDDP and Huh7/CDDP, respectively.

Cell proliferation assay

Cells were plated and cultured in 96-well plates in 100 µl medium at a density of 1×103 cells/well. Following treatment with increasing concentrations (0, 0.5, 1, 5, 10, 20 and 50 µM) of CDDP for 48 h at room temperature, 10 µl Cell Counting Kit-8 (Abmole Bioscience Inc.) reagent was added to each well and incubated at 37°C for 2 h. In order to evaluate the effect of YAP, HepG2/CDDP and Huh-7/CDDP cells were pre-treated with or without 4 µM verteporfin (VP; Sigma-Aldrich; Merck KGaA; cat. no. SML0534) for 90 min at room temperature and then further treated with increasing concentrations of CDDP (0, 0.5, 1, 5, 10, 20 and 50 µM) for 48 h at room temperature. In order to investigate whether IL-6 and TGF-β were involved in YAP-regulated chemoresistance of LC cells, HepG2/CDDP cells were pre-treated with 100 ng/ml anti-IL-6 (cat. no. MAB206-SP; R&D Systems, Inc.) or anti-TGF-β (cat. no. BE0057; Bio X Cell) for 2 h at room temperature and then further treated with increasing concentrations of CDDP (0, 0.5, 1, 5, 10, 20 and 50 µM) for 48 h at room temperature. Additionally, HepG2/CDDP or Huh-7/CDDP cells were pre-treated with VP (4 µM) combined with recombinant (r)IL-6 (100 ng/ml; cat. no. 206-IL-010/CF; R&D Systems, Inc.) or rTGF-β (100 ng/ml; cat. no. 240-B-002/CF; R&D Systems, Inc.) for 2 h at room temperature, and then further treated with increasing concentrations of CDDP (0–20 µM) for 48 h at room temperature. The absorbance was measured at 450 nm using a microplate reader (ENSIGHT; PerkinElmer, Inc.) according to the manufacturer's protocol. The cell viability was calculated as the percentage of the viability of untreated control cells. Experiments were repeated ≥3 times.

RNA extraction and reverse transcription-quantitative PCR (RT-qPCR)

Total RNA was extracted from cells using TRIzol® reagent (Invitrogen; Thermo Fisher Scientific, Inc.) and treated with DNase I (Promega Corporation) to remove the DNA contamination. RNA (1 µg) was reverse transcribed into cDNA using the cDNA Synthesis SuperMix (Beijing TransGen Biotech Co., Ltd.) according to the manufacturer's protocol. qPCR was subsequently performed using the SYBR Premix Ex Taq II kit (Takara Biotechnology Co., Ltd.) and a Bio-Rad CFX96 system (Bio-Rad Laboratories, Inc.). The following primer sequences were used: YAP forward, 5′-GGCATACACCTACTCAACTACGG-3′ and reverse, 5′-TGGGCGGTGTAGAATCAGAGTC-3′; precursor-YAP forward, 5′-CCGGCTTGCTCTTATCAAAC-3′ and reverse, 5′-GTCATCGCTTCCCAAACATT-3′; IL-6 forward, 5′-ACTCACCTCTTCAGAACGAATTG-3′ and reverse, 5′-CCATCTTTGGAAGGTTCAGGTTG-3′; IL-10 forward, 5′-TCTCCGAGATGCCTTCAGCAGA-3′ and reverse, 5′-TCAGACAAGGCTTGGCAACCCA-3′; IL-12 forward, 5′-TGCCTTCACCACTCCCAAAACC-3′ and reverse, 5′-CAATCTCTTCAGAAGTGCAAGGG-3′; TNF-α forward, 5′-CTCTTCTGCCTGCTGCACTTTG-3′ and reverse, 5′-ATGGGCTACAGGCTTGTCACTC-3′; TGF-β forward, 5′-TACCTGAACCCGTGTTGCTCTC-3′ and reverse, 5′-GTTGCTGAGGTATCGCCAGGAA-3′; MALAT1 forward, 5′-AAAGCAAGGTCTCCCCACAAG-3′ and reverse, 5′-GGTCTGTGCTAGATCAAAAGGCA-3′; and GAPDH forward, 5′-GGAGCGAGATCCCTCCAAAAT-3′ and reverse, 5′-GGCTGTTGTCATACTTCTCATGG 3′. The PCR cycling conditions were 15 min at 95°C, followed by 40 cycles for 10 sec at 95°C, 30 sec at 60°C and 1 sec at 72°C, and 1 cycle of cooling for 30 sec at 50°C.

To analyze the expression levels of miRNAs, the TaqMan MicroRNA Reverse Transcription kit (Applied Biosystems; Thermo Fisher Scientific, Inc.) was used to generate cDNA according to the manufacturer's protocol. The thermocycling conditions included an initial denaturation at 95°C for 3 min, followed by 40 cycles at 95°C for 15 sec and 60°C for 30 sec. The forward primer is the exact sequence of the mature miRNA (http://www.mirbase.org/search.shtml). The forward primer for U6 was 5′-TGCGGGTGCTCGCTTCGCAGC-3′. The reverse primer was supplied by the aforementioned kit. GAPDH and U6 were used as the internal reference genes for the normalization of mRNA and miRNA, respectively. The gene expression levels were quantified using the 2−ΔΔCq method (19). Each sample was analyzed in triplicate.

Subcellular fractionation

The cytoplasmic and nuclear fractions of cells were prepared using the PARIS™ kit (Ambion; Thermo Fisher Scientific, Inc.) according to the manufacturer's instructions. The protein expression levels within the cytoplasmic and nuclear fractions were analyzed by western blotting. Aliquots of cytoplasmic and nuclear fractions were also subjected to RNA isolation and RT-qPCR, as aforementioned, to analyze the subcellular localization of YAP mRNA. Transcripts of the housekeeping gene GAPDH were used for normalization, while nuclear MALAT1 RNA was selected as endogenous control for the nuclear RNA.

Western blotting

Total protein was extracted from cells using 1X RIPA lysis buffer (50 mM Tris HCl, 150 mM NaCl and 1 mM EDTA) containing a protease inhibitor cocktail (Roche Diagnostics). Total protein was quantified using a bicinchoninic acid assay kit and 20 µg protein/lane was separated by 10% SDS-PAGE. The separated proteins were subsequently transferred onto a nitrocellulose membrane (EMD Millipore) using a wet transfer apparatus. The membranes were blocked with 5% skimmed milk at room temperature for 2 h. Following the incubation with the primary antibodies at 4°C overnight, the membranes were further incubated with the HRP-conjugated secondary antibody (cat. no. ab7090; Abcam; 1:10,000) diluted in 5% skimmed milk. Protein bands were then visualized in a gel imaging system (MG8600; Bio-Rad Laboratories, Inc.). The following primary antibodies (1:1,000; Abcam) were used: Anti-H2A.X (cat. no. ab229914), anti-YAP (cat. no. ab56701), anti-TAZ (cat. no. ab84927), anti-calpain (cat. no. ab39170) and anti-GAPDH (cat. no. ab229914). GAPDH was used as the loading control for normalization. The gray values were analyzed using ImageJ software (version 1.46; National Institutes of Health).

Cell transfection and treatment

The small interfering RNA (siRNA/si) negative control (si-NC; 5′-GCACAACAAGCCGAAUACA-3′), si-YAP (siYAP-1, 5′-GCGUAGCCAGUUACCAACA-3′; siYAP-2, 5′-CAGUGGCACCUAUCACUCU-3′), miRNA control (miR, 5′-UUCUCCGAACGUGUCACGUTT-3′) and miR-375 mimics (5′-UUUGUUCGUUCGGCUCGCGUGA-3′) were synthesized by Shanghai GenePharma Co., Ltd.. Upon cells reaching 50–60% confluence, the transfection was performed using Lipofectamine® 3000 (Invitrogen; Thermo Fisher Scientific, Inc.) according to the manufacturer's instructions with 20 µM of each construct or siRNA. After transfection for 6 h at 37°C, the medium was replaced with fresh complete medium. To investigate the effect of YAP on chemosensitivity, HepG2/CDDP, Huh-6/CDDP and Huh-7/CDDP cells were transfected with si-NC or si-YAP-1 for 12 h and then further treated with increasing concentrations of CDDP (0, 0.5, 1, 5, 10, 20 and 50 µM) for 48 h.

mRNA and protein stability assay

To determine the mRNA stability, cells were treated with 5 µg/ml actinomycin D (Act-D; cat. no. A9415; Sigma-Aldrich; Merck KGaA) at 37°C for 0, 2, 4 or 8 h. Subsequently, total RNA was collected and the target mRNA was analyzed using RT-qPCR, as aforementioned. For the protein stability assay, cells were incubated with 100 µg/ml cycloheximide (CHX) at 37°C for 0, 2, 6 or 12 h and then protein expression was analyzed using western blotting, as aforementioned.

Immunofluorescence

Cells cultured on coverslips were washed with PBS and fixed in 4% paraformaldehyde for 15 min at room temperature. After blocking with 3% BSA in PBS containing 0.3% Triton X-100 solution at 37°C for 1 h, cells were incubated with a primary antibody against YAP (cat. no. ab56701; 1:1,000; Abcam) overnight at 4°C and then treated with an anti-Alexa Fluor 594 secondary antibody (1:200; R&D Systems China Co., Ltd.; cat. no. IC1420T) for 1 h at room temperature. Then, DAPI solution (5 µg/ml) was added to stain the cell nuclei for 5 min at room temperature. The fluorescence signal was observed under a confocal microscope (TCS-SP5; Leica Microsystems GmbH; magnification, ×10).

Statistical analysis

Statistical analysis was performed using SPSS 17.0 software (SPSS Inc.) and presented as the mean ± SD. The comparisons between two groups were analyzed using an unpaired Student's t-test. All experiments were performed ≥3 times independently. P<0.05 was considered to indicate a statistically significant difference.

Results

Establishment of LC/CDDP cells

The CDDP sensitivity of both resistant and parental LC cells was investigated. The results revealed that the established CDDP-resistant cells were more resistant to CDDP treatment compared with their corresponding parental cells (Fig. 1). The IC50 values of CDDP for HepG2/CDDP and HepG2 cells were 22.8 and 3.45 µM, respectively (Fig. 1A), those for Huh-6/CDDP and Huh-6 cells were 30.6 and 5.05 µM, respectively (Fig. 1B), while the IC50 values of CDDP for Huh-7/CDDP and Huh-7 cells were 30.5 and 6.51 µM, respectively (Fig. 1C). The current data confirmed the successful establishment of LC/CDDP cells.

YAP expression is upregulated in CDDP-resistant LC cells

It has been previously reported that the Hippo signaling pathway regulates the progression of LC (20). Thus, the present study analyzed the expression levels of YAP and the transcriptional coactivator with PDZ-binding motif (TAZ), another important member of the Hippo signaling pathway (20), in both parental and CDDP-resistant LC cells. The protein expression levels of YAP, but not TAZ, were significantly upregulated in the HepG2/CDDP, Huh-6/CDDP and Huh-7/CDDP cells compared with in their corresponding parental cells (Fig. 2A). Furthermore, RT-qPCR analysis revealed that the mRNA expression levels of YAP were significantly upregulated in the CDDP-resistant LC cells compared with in their respective parental cells (Fig. 2B). In addition, the amount of YAP localized in both the cytosol and nucleus was increased in HepG2/CDDP cells compared with in HepG2 cells (Fig. 2C), which was confirmed by immunofluorescence staining (Fig. 2D).

YAP is involved in the CDDP resistance of LC cells

To investigate whether YAP was involved in the resistance to CDDP in LC cells, the CDDP-resistant LC cells were transfected with si-YAP-1 and si-YAP-2 (Fig. 3A). si-YAP-1 was used for subsequent experiments since it displayed increased efficiency. The results revealed that si-YAP-1 markedly increased the CDDP sensitivity of HepG2/CDDP (Fig. 3B), Huh-6/CDDP (Fig. 3C) and Huh-7/CDDP (Fig. 3D) cells. Since the results revealed that YAP expression was markedly increased in HepG2/CDDP and Huh-7/CDDP cells, these cell lines were further treated with VP, a suppressor of the YAP-TEAD complex (21). VP increased the sensitivity of CDDP in HepG2/CDDP (Fig. 3E) and Huh-7/CDDP (Fig. 3F) cells.

YAP regulates the expression levels of IL-6 and TGF-β in LC/CDDP cells

It has been previously reported that YAP regulates the expression levels of various cytokines to regulate cancer progression (1214). In the present study, an array of cytokines was analyzed, including IL-6, IL-10, IL-12, TNF-α and TGF-β, in si-YAP-1-transfected LC/CDDP cells. si-YAP-1 significantly downregulated the expression levels of IL-6 and TGF-β in both HepG2/CDDP (Fig. 4A) and Huh-7/CDDP (Fig. 4B) cells. In addition, VP treatment significantly downregulated the expression levels of IL-6 and TGF-β in both HepG2/CDDP (Fig. 4C) and Huh-7/CDDP (Fig. 4D) cells. On the other hand, the expression levels of IL-6 and TGF-β in both HepG2/CDDP (Fig. 4E) and Huh-7/CDDP (Fig. 4F) cells were significantly upregulated compared with in their corresponding control cells. The current results suggested that YAP may regulate the expression levels of IL-6 and TGF-β in LC/CDDP cells.

IL-6 and TGF-β are involved in the YAP-mediated chemoresistance of LC cells

The current study further analyzed whether IL-6 and TGF-β were involved in the YAP-mediated chemoresistance of LC cells. The data demonstrated that neutralization antibodies anti-IL-6 (Fig. 5A) and anti-TGF-β (Fig. 5B) significantly increased the CDDP sensitivity of HepG2/CDDP cells. In addition, rIL-6 (Fig. 5C) and rTGF-β (Fig. 5D) significantly attenuated the VP-induced CDDP sensitivity of HepG2/CDDP cells. All these data indicated that IL-6 and TGF-β may be involved in the YAP-mediated chemoresistance of LC cells.

mRNA stability is responsible for the upregulation of YAP expression in LC/CDDP cells

The potential mechanisms responsible for the upregulation of YAP expression in LC/CDDP cells were subsequently investigated. The protein stability of YAP in HepG2 and HepG2/CDDP cells following CHX treatment was similar to each other (Fig. 6A). Additionally, the expression levels of the precursor mRNA of YAP, analyzed by RT-qPCR, were not significantly different between HepG2 and HepG2/CDDP cells or between Huh-7 and Huh-7/CDDP cells (Fig. 6B). In addition, the nuclear turnover rate of YAP was not significantly different between HepG2 and HepG2/CDDP cells, as analyzed by RT-qPCR (Fig. 6C). However, the data revealed that the mRNA stability of YAP in HepG2/CDDP cells following Act-D treatment was markedly increased compared with in HepG2 cells (Fig. 6D). Consistently, the mRNA stability of YAP in Huh-7/CDDP cells was also increased compared with in Huh-7 cells (Fig. 6E). These results indicated that increased mRNA stability may be responsible for the upregulation of YAP expression in LC/CDDP cells.

miR-375 decreases the mRNA stability of YAP in LC/CDDP cells

miRNAs can decrease mRNA stability via binding to the 3′-untranslated regions of mRNA (22). It has been revealed that miR-375 (23), miR-506 (24), miR-132 (25) and miR-129 (26) directly target YAP mRNA to downregulate its expression. Thus, the expression levels of these miRNAs in both LC/CDDP and LC cells were subsequently analyzed. The data revealed that, among all miRNAs, only the expression levels of miR-375 were significantly downregulated in both HepG2/CDDP (Fig. 7A) and Huh-7/CDDP (Fig. 7B) cells. Furthermore, the overexpression of miR-375 (Fig. 7C) using miR-375 mimics significantly downregulated the mRNA expression levels of YAP in both HepG2/CDDP and Huh-7/CDDP cells (Fig. 7D). This was due to the fact that miR-375 decreased the mRNA stability of YAP (Fig. 7E).

Discussion

Chemotherapy is an important treatment for patients with LC, especially for those with advanced LC (27). Cisplatin has been widely used as a therapeutic agent for patients with LC; however, its application has been significantly limited due to the development of chemoresistance (28). To the best of our knowledge, the molecular mechanisms involved in LC chemoresistance to CDDP are not fully understood. The results of the present study suggested that YAP, an important downstream signaling protein of the Hippo signaling pathway, may mediate the CDDP resistance of LC cells via upregulating IL-6 and TGF-β expression. In addition, the downregulation of miR-375 expression in LC/CDDP cells was responsible for the upregulation of YAP expression. Collectively, these results suggested that the miR-375/YAP axis-induced expression of IL-6 and TGF-β may be critical for the CDDP resistance of LC cells.

The present study discovered that YAP was involved in the CDDP resistance of LC cells. It has been previously revealed that YAP upregulation is strongly associated with the carcinogenesis of LC (29,30). The activation of YAP suppresses the sensitivity of cancer cells to various drugs, such as anti-tubulin drugs and DNA-damaging agents (3134). In LC cells, it has been reported that YAP upregulation confers resistance to doxorubicin (35) and the topoisomerase I inhibitor SN38 (36). The data of the present study illustrated that the expression levels and nuclear localization of YAP were increased in LC/CDDP cells. In addition, the targeted inhibition of YAP via siRNA or an inhibitor restored the CDDP sensitivity of LC cells, which indicated that YAP may be involved in the chemoresistance of LC cells.

The data of the current study also demonstrated that IL-6 and TGF-β were involved in the YAP-mediated chemoresistance of LC cells. It has been previously reported that the activation of YAP stimulates IL-6 gene transcription during colonic tumorigenesis (37). In LC cells, YAP induces IL-6 expression to recruit tumor-associated macrophages (38). Additionally, a recent study has confirmed that YAP can directly bind to the promoter of IL-6 to regulate its transcription (39). As to TGF-β, it has been reported that YAP promotes the TGF-β-induced tumorigenic phenotype in breast cancer cells (40). In addition, YAP/TAZ regulate TGF-β/Smad3 signaling through the induction of Smad7 via activator protein 1 in human skin dermal fibroblasts (41). However, whether YAP can directly activate the transcription of TGF-β requires further investigation.

Furthermore, the present study indicated that the downregulation of miR-375 expression may be responsible for the upregulation of YAP expression in LC/CDDP cells, indicated by the fact that YAP mRNA stability was increased, while miR-375 expression was downregulated, in LC/CDDP cells compared with in LC cells. In gastric cancer cells, the upregulation of miR-375 expression increases the CDDP sensitivity via the regulation of ERBB2 (42). miR-375 is induced in CDDP nephrotoxicity to repress hepatocyte nuclear factor-1β (43). Furthermore, miR-375 can target YAP in LC to inhibit cancer cell viability (23,44). Similarly, miR-375 suppresses YAP expression in lung cancer (45) and mouse pancreatic progenitor (46) cells. All these data suggested that miR-375 may be involved in the CDDP resistance and progression of LC.

In conclusion, the results of the present study revealed that the miR-375/YAP axis may regulate the CDDP resistance of LC via the regulation of IL-6 and TGF-β. Therefore, the targeted inhibition of this axis and signaling pathway may be useful in overcoming the CDDP resistance and enhancing the clinical treatment of patients with LC. Whether the miR-375/YAP axis-induced expression of IL-6 and TGF-β is involved in the TKI resistance of LC requires further investigation in future studies.

Acknowledgements

Not applicable.

Funding

No funding was received.

Availability of data and materials

All data generated or analyzed during this study are included in this published article.

Authors' contributions

KY, KW and HL conceived and designed the study. ZJ, HJH, HCH, YZ and KW acquired the data. KY, KW, HL, ZJ and YZ analyzed and interpreted the data. KY, HCH, YZ and KW wrote and revised the manuscript. The authenticity of the raw data has been assessed by all authors. All authors read and approved the final manuscript.

Ethics approval and consent to participate

Not applicable.

Patient consent for publication

Not applicable.

Competing interests

The authors declare that they have no competing interests.

References

1 

Siegel RL, Miller KD and Jemal A: Cancer statistics, 2016. CA Cancer J Clin. 66:7–30. 2016. View Article : Google Scholar : PubMed/NCBI

2 

Chen W, Zheng R, Baade PD, Zhang S, Zeng H, Bray F, Jemal A, Yu XQ and He J: Cancer statistics in China, 2015. CA Cancer J Clin. 66:115–132. 2016. View Article : Google Scholar : PubMed/NCBI

3 

Raoul JL, Kudo M, Finn RS, Edeline J, Reig M and Galle PR: Systemic therapy for intermediate and advanced hepatocellular carcinoma: Sorafenib and beyond. Cancer Treat Rev. 68:16–24. 2018. View Article : Google Scholar : PubMed/NCBI

4 

Grothey A, Blay JY, Pavlakis N, Yoshino T and Bruix J: Evolving role of regorafenib for the treatment of advanced cancers. Cancer Treat Rev. 86:1019932020. View Article : Google Scholar : PubMed/NCBI

5 

Korita PV, Wakai T, Shirai Y, Matsuda Y, Sakata J, Takamura M, Yano M, Sanpei A, Aoyagi Y, Hatakeyama K and Ajioka Y: Multidrug resistance-associated protein 2 determines the efficacy of cisplatin in patients with hepatocellular carcinoma. Oncol Rep. 23:965–972. 2010.PubMed/NCBI

6 

Plimack ER, Dunbrack RL, Brennan TA, Andrake MD, Zhou Y, Serebriiskii IG, Slifker M, Alpaugh K, Dulaimi E, Palma N, et al: Defects in DNA repair genes predict response to neoadjuvant cisplatin-based chemotherapy in muscle-invasive bladder cancer. Eur Urol. 68:959–967. 2015. View Article : Google Scholar : PubMed/NCBI

7 

Zamble DB and Lippard SJ: Cisplatin and DNA repair in cancer chemotherapy. Trends Biochem Sci. 20:435–439. 1995. View Article : Google Scholar : PubMed/NCBI

8 

Zhu AX: Systemic therapy of advanced hepatocellular carcinoma: How hopeful should we be? Oncologist. 11:790–800. 2006. View Article : Google Scholar : PubMed/NCBI

9 

Steinhardt AA, Gayyed MF, Klein AP, Dong J, Maitra A, Pan D, Montgomery EA and Anders RA: Expression of Yes-associated protein in common solid tumors. Hum Pathol. 39:1582–1589. 2008. View Article : Google Scholar : PubMed/NCBI

10 

Camargo FD, Gokhale S, Johnnidis JB, Fu D, Bell GW, Jaenisch R and Brummelkamp TR: YAP1 increases organ size and expands undifferentiated progenitor cells. Curr Biol. 17:2054–2060. 2007. View Article : Google Scholar : PubMed/NCBI

11 

Guan KLL: Regulation and function of the Hippo-YAP pathway in organ size, tumorigenesis, and metastasis. Cancer Res. 72 (Suppl 8):SY29–03. 2012.PubMed/NCBI

12 

Yu FX, Zhao B and Guan KL: Hippo pathway in organ size control, tissue homeostasis, and cancer. Cell. 163:811–828. 2015. View Article : Google Scholar : PubMed/NCBI

13 

Zhao B, Li L, Lei Q and Guan KL: The Hippo-YAP pathway in organ size control and tumorigenesis: An updated version. Genes Dev. 24:862–874. 2010. View Article : Google Scholar : PubMed/NCBI

14 

Zhang L, Yue T and Jiang J: Hippo signaling pathway and organ size control. Fly (Austin). 3:68–73. 2009. View Article : Google Scholar : PubMed/NCBI

15 

Yuan Y, Li D, Li H, Wang L, Tian G and Dong Y: YAP overexpression promotes the epithelial-mesenchymal transition and chemoresistance in pancreatic cancer cells. Mol Med Rep. 13:237–242. 2016. View Article : Google Scholar : PubMed/NCBI

16 

Corvaisier M, Bauzone M, Corfiotti F, Renaud F, El Amrani M, Monté D, Truant S, Leteurtre E, Formstecher P, Van Seuningen I, et al: Regulation of cellular quiescence by YAP/TAZ and Cyclin E1 in colon cancer cells: Implication in chemoresistance and cancer relapse. Oncotarget. 7:56699–56712. 2016. View Article : Google Scholar : PubMed/NCBI

17 

Qin J, Luo M, Qian H and Chen W: Upregulated miR-182 increases drug resistance in cisplatin-treated HCC cell by regulating TP53INP1. Gene. 538:342–347. 2014. View Article : Google Scholar : PubMed/NCBI

18 

Xu N, Zhang J, Shen C, Luo Y, Xia L, Xue F and Xia Q: Cisplatin-induced downregulation of miR-199a-5p increases drug resistance by activating autophagy in HCC cell. Biochem Biophys Res Commun. 423:826–831. 2012. View Article : Google Scholar : PubMed/NCBI

19 

Livak KJ and Schmittgen TD: Analysis of relative gene expression data using real-time quantitative PCR and the 2(-Delta Delta C(T)) method. Methods. 25:402–408. 2001. View Article : Google Scholar : PubMed/NCBI

20 

Zheng T, Wang J, Jiang H and Liu L: Hippo signaling in oval cells and hepatocarcinogenesis. Cancer Lett. 302:91–99. 2011. View Article : Google Scholar : PubMed/NCBI

21 

Liu-Chittenden Y, Huang B, Shim JS, Chen Q, Lee SJ, Anders RA, Liu JO and Pan D: Genetic and pharmacological disruption of the TEAD-YAP complex suppresses the oncogenic activity of YAP. Genes Dev. 26:1300–1305. 2012. View Article : Google Scholar : PubMed/NCBI

22 

Uddin A and Chakraborty S: Role of miRNAs in lung cancer. J Cell Physiol. Apr 20–2018.(Online ahead of print). View Article : Google Scholar

23 

Liu AM, Poon RT and Luk JM: MicroRNA-375 targets Hippo-signaling effector YAP in liver cancer and inhibits tumor properties. Biochem Biophys Res Commun. 394:623–627. 2010. View Article : Google Scholar : PubMed/NCBI

24 

Wang Y, Cui M, Sun BD, Liu FB, Zhang XD and Ye LH: MiR-506 suppresses proliferation of hepatoma cells through targeting YAP mRNA 3′UTR. Acta Pharmacol Sin. 35:1207–1214. 2014. View Article : Google Scholar : PubMed/NCBI

25 

Lei CJ, Li L, Gao X, Zhang J, Pan QY, Long HC, Chen CZ, Ren DF and Zheng G: Hsa-miR-132 inhibits proliferation of hepatic carcinoma cells by targeting YAP. Cell Biochem Funct. 33:326–333. 2015. View Article : Google Scholar : PubMed/NCBI

26 

Tan G, Cao X, Dai Q, Zhang B, Huang J, Xiong S, Zhang YY, Chen W, Yang J and Li H: A novel role for microRNA-129-5p in inhibiting ovarian cancer cell proliferation and survival via direct suppression of transcriptional co-activators YAP and TAZ. Oncotarget. 6:8676–8686. 2015. View Article : Google Scholar : PubMed/NCBI

27 

Pratama MY, Pascut D, Massi MN and Tiribelli C: The role of microRNA in the resistance to treatment of hepatocellular carcinoma. Ann Transl Med. 7:5772019. View Article : Google Scholar : PubMed/NCBI

28 

Ghosh S: Cisplatin: The first metal based anticancer drug. Bioorg Chem. 88:1029252019. View Article : Google Scholar : PubMed/NCBI

29 

Perra A, Kowalik MA, Ghiso E, Ledda-Columbano GM, Di Tommaso L, Angioni MM, Raschioni C, Testore E, Roncalli M, Giordano S and Columbano A: YAP activation is an early event and a potential therapeutic target in liver cancer development. J Hepatol. 61:1088–1096. 2014. View Article : Google Scholar : PubMed/NCBI

30 

Li L, Wang J, Zhang Y, Zhang Y, Ma L, Weng W, Qiao Y, Xiao W, Wang H, Yu W, et al: MEK1 promotes YAP and their interaction is critical for tumorigenesis in liver cancer. FEBS Lett. 587:3921–3927. 2013. View Article : Google Scholar : PubMed/NCBI

31 

Zhao Y, Khanal P, Savage P, She YM, Cyr TD and Yang X: YAP-induced resistance of cancer cells to antitubulin drugs is modulated by a Hippo-independent pathway. Cancer Res. 74:4493–4503. 2014. View Article : Google Scholar : PubMed/NCBI

32 

Xia Y, Zhang YL, Yu C, Chang T and Fan HY: YAP/TEAD co-activator regulated pluripotency and chemoresistance in ovarian cancer initiated cells. PLoS One. 9:e1095752014. View Article : Google Scholar : PubMed/NCBI

33 

Yoshikawa K, Noguchi K, Nakano Y, Yamamura M, Takaoka K, Hashimoto-Tamaoki T and Kishimoto H: The Hippo pathway transcriptional co-activator, YAP, confers resistance to cisplatin in human oral squamous cell carcinoma. Int J Oncol. 46:2364–2370. 2015. View Article : Google Scholar : PubMed/NCBI

34 

Zhao Y and Yang X: The Hippo pathway in chemotherapeutic drug resistance. Int J Cancer. 137:2767–2773. 2015. View Article : Google Scholar : PubMed/NCBI

35 

Huo X, Zhang Q, Liu AM, Tang C, Gong Y, Bian J, Luk JM, Xu Z and Chen J: Overexpression of Yes-associated protein confers doxorubicin resistance in hepatocellullar carcinoma. Oncol Rep. 29:840–846. 2013. View Article : Google Scholar : PubMed/NCBI

36 

Dai XY, Zhuang LH, Wang DD, Zhou TY, Chang LL, Gai RH, Zhu DF, Yang B, Zhu H and He QJ: Nuclear translocation and activation of YAP by hypoxia contributes to the chemoresistance of SN38 in hepatocellular carcinoma cells. Oncotarget. 7:6933–6947. 2016. View Article : Google Scholar : PubMed/NCBI

37 

Taniguchi K, Moroishi T, de Jong PR, Krawczyk M, Grebbin BM, Luo H, Xu RH, Golob-Schwarzl N, Schweiger C, Wang K, et al: YAP-IL-6ST autoregulatory loop activated on APC loss controls colonic tumorigenesis. Proc Natl Acad Sci USA. 114:1643–1648. 2017. View Article : Google Scholar : PubMed/NCBI

38 

Zhou TY, Zhou YL, Qian MJ, Fang YZ, Ye S, Xin WX, Yang XC and Wu HH: Interleukin-6 induced by YAP in hepatocellular carcinoma cells recruits tumor-associated macrophages. J Pharmacol Sci. 138:89–95. 2018. View Article : Google Scholar : PubMed/NCBI

39 

Wang J, Song T, Zhou S and Kong X: YAP promotes the malignancy of endometrial cancer cells via regulation of IL-6 and IL-11. Mol Med. 25:322019. View Article : Google Scholar : PubMed/NCBI

40 

Hiemer SE, Szymaniak AD and Varelas X: The transcriptional regulators TAZ and YAP direct transforming growth factor β-induced tumorigenic phenotypes in breast cancer cells. J Biol Chem. 289:13461–13474. 2014. View Article : Google Scholar : PubMed/NCBI

41 

Wang L, Lee W, Oh JY, Cui YR, Ryu B and Jeon YJ: Protective effect of sulfated polysaccharides from celluclast-assisted extract of Hizikia fusiforme against ultraviolet B-induced skin damage by regulating NF-κB, AP-1, and MAPKs signaling pathways in vitro in human dermal fibroblasts. Mar Drugs. 16:2392018. View Article : Google Scholar : PubMed/NCBI

42 

Zhou N, Qu Y, Xu C and Tang Y: Upregulation of microRNA-375 increases the cisplatin-sensitivity of human gastric cancer cells by regulating ERBB2. Exp Ther Med. 11:625–630. 2016. View Article : Google Scholar : PubMed/NCBI

43 

Hao J, Lou Q, Wei Q, Mei S, Li L, Wu G, Mi QS, Mei C and Dong Z: MicroRNA-375 is induced in cisplatin nephrotoxicity to repress hepatocyte nuclear factor 1-β. J Biol Chem. 292:4571–4582. 2017. View Article : Google Scholar : PubMed/NCBI

44 

Chang Y, Yan W, He X, Zhang L, Li C, Huang H, Nace G, Geller DA, Lin J and Tsung A: miR-375 inhibits autophagy and reduces viability of hepatocellular carcinoma cells under hypoxic conditions. Gastroenterology. 143:177–187.e8. 2012. View Article : Google Scholar : PubMed/NCBI

45 

Nishikawa E, Osada H, Okazaki Y, Arima C, Tomida S, Tatematsu Y, Taguchi A, Shimada Y, Yanagisawa K, Yatabe Y, et al: miR-375 is activated by ASH1 and inhibits YAP1 in a lineage-dependent manner in lung cancer. Cancer Res. 71:6165–6173. 2011. View Article : Google Scholar : PubMed/NCBI

46 

Zhang ZW, Men T, Feng RC, Li YC, Zhou D and Teng CB: miR-375 inhibits proliferation of mouse pancreatic progenitor cells by targeting YAP1. Cell Physiol Biochem. 32:1808–1817. 2013. View Article : Google Scholar : PubMed/NCBI

Related Articles

Journal Cover

August-2021
Volume 46 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
Yu K, Li H, Jiang Z, Hsu H, Hsu H, Zhang Y and Wang K: miR‑375/Yes‑associated protein axis regulates IL‑6 and TGF‑β expression, which is involved in the cisplatin‑induced resistance of liver cancer cells. Oncol Rep 46: 162, 2021
APA
Yu, K., Li, H., Jiang, Z., Hsu, H., Hsu, H., Zhang, Y., & Wang, K. (2021). miR‑375/Yes‑associated protein axis regulates IL‑6 and TGF‑β expression, which is involved in the cisplatin‑induced resistance of liver cancer cells. Oncology Reports, 46, 162. https://doi.org/10.3892/or.2021.8112
MLA
Yu, K., Li, H., Jiang, Z., Hsu, H., Hsu, H., Zhang, Y., Wang, K."miR‑375/Yes‑associated protein axis regulates IL‑6 and TGF‑β expression, which is involved in the cisplatin‑induced resistance of liver cancer cells". Oncology Reports 46.2 (2021): 162.
Chicago
Yu, K., Li, H., Jiang, Z., Hsu, H., Hsu, H., Zhang, Y., Wang, K."miR‑375/Yes‑associated protein axis regulates IL‑6 and TGF‑β expression, which is involved in the cisplatin‑induced resistance of liver cancer cells". Oncology Reports 46, no. 2 (2021): 162. https://doi.org/10.3892/or.2021.8112