Inhibition of PIKfyve using YM201636 suppresses the growth of liver cancer via the induction of autophagy

  • Authors:
    • Jiu‑Zhou Hou
    • Zhuo‑Qing Xi
    • Jie Niu
    • Wei Li
    • Xiao Wang
    • Chao Liang
    • Hua Sun
    • Dong Fang
    • Song‑Qiang Xie
  • View Affiliations

  • Published online on: December 12, 2018     https://doi.org/10.3892/or.2018.6928
  • Pages: 1971-1979
Metrics: Total Views: 0 (Spandidos Publications: | PMC Statistics: )
Total PDF Downloads: 0 (Spandidos Publications: | PMC Statistics: )


Abstract

Liver cancer is among the most common types of cancer worldwide. The aim of the present study was to investigate whether the phosphatidylinositol‑3‑phosphate 5‑kinase (PIKfyve) inhibitor, YM201636, exerts anti‑proliferative effects on liver cancer. The methods used in the present study included MTT assay, flow cytometry, western blot analysis and an allograft mouse model of liver cancer. The results revealed that YM201636 inhibited the proliferation of HepG2 and Huh‑7 cells in a dose‑dependent manner. HepG2 and Huh‑7 cells exhibited strong monodansylcadaverine staining following treatment with YM201636. Accordingly, YM201636 treatment increased the expression of the autophagosome‑associated marker protein microtubule‑associated 1A/1B light chain 3‑II in HepG2 and Huh‑7 cells. The autophagy inhibitor 3‑methyladenine attenuated the inhibitory effects of YM201636 on liver cancer cell proliferation. Further in vivo analysis revealed that YM201636 (2 mg/kg) inhibited tumor growth without notable systemic toxicity. Mechanistic experiments demonstrated that YM201636 induced‑autophagy is dependent upon epidermal growth factor receptor (EGFR) overexpression in HepG2 and Huh‑7 cells. Collectively, these results suggested that the PIKfyve inhibitor YM201636 may inhibit tumor growth by promoting EGFR expression. This indicates that PIKfyve may be a potential therapeutic target for the treatment of liver cancer.

Introduction

Liver cancer is one of the most common types of cancer worldwide, ranking as the third leading cause of cancer-associated mortality (1). Despite the great advances in the use of modern surgical techniques in combination with chemotherapy, the overall 5-year survival rate for patients with liver cancer remains poor (2). Therefore, novel strategies for the anticancer therapy of liver cancer are urgently required.

Phosphatidylinositol-3-phosphate 5-kinase (PIKfyve) is a lipid kinase that phosphorylates phosphatidylinositol-3-phosphate (PI3P) to generate phosphatidylinositol 3,5-bisphosphate [PtdIns(3,5)P2] or phosphatidylinositol 5-phosphate (PtdIns5P) (3,4). PtdIns(3,5)P2 and PtdIns5P have been proposed to be involved in several cellular functions, including vesicular trafficking, ion channel activation and epidermal growth factor receptor (EGFR) signaling (57). In addition, accumulating evidence has suggested that PIKfyve is involved in oncogenesis and cancer cell migration (8,9), and knockdown of PIKfyve resulted in a significant decrease in cancer cell migration (10). Therefore, the potential role of PIKfyve inhibition in anticancer therapy was explored in the present study. Recent studies demonstrated that inhibition of PIKfyve activity using the inhibitor YM201636 led to a strong reduction in cell proliferation in multiple cancers (8,11); however, whether PIKfyve inhibition could be applied for anticancer therapy of liver cancer remains unknown. Therefore, the aim of the present study was to investigate the antitumor activity of the PIKfyve inhibitor, YM201636, in liver cancer.

Materials and methods

Reagents and antibodies

RPMI-1640 and fetal bovine serum (FBS) were purchased from Gibco (Thermo Fisher Scientific, Inc., Waltham, MA, USA). MTT and monodansylcadaverine (MDC) were purchased from Sigma-Aldrich (Merck KGaA, Darmstadt, Germany). YM201636 (cat. no. sc-204193) was obtained from Santa Cruz Biotechnology, Inc. (Dallas, TX, USA). Monoclonal mouse anti-human EGFR antibody (cat. no. sc-71034; 1:200), monoclonal mouse anti-human phospho (p)-EGFR antibody (cat. no. sc-81490; 1:200), monoclonal mouse anti-human β-actin antibody (cat. no. sc-47778; 1:1,000) were all purchased from Santa Cruz Biotechnology, Inc. Polyclonal rabbit anti-human microtubule associated protein 1 light chain 3 (LC3) A/B antibody (cat. no. 14600-1-AP; 1:1,000) was purchased from ProteinTech Group, Inc. (Chicago, IL, USA). The horseradish peroxidase (HRP)-conjugated secondary antibodies including goat anti-rabbit IgG (cat. no. sc-2054; 1:1,000) and goat anti-mouse IgG (cat. no. sc-2973; 1:1,000) were also purchased from Santa Cruz Biotechnology, Inc.

Cell lines and cell culture

All cell lines (HepG2, Huh-7 and H22) used in the present study were purchased from the Cell Bank of Type Culture Collection of Chinese Academy of Sciences (Shanghai, China). These cells were cultured in RPMI-1640 medium supplemented with 10% FBS, 100 U/ml penicillin and 100 µg/ml streptomycin, at 37°C in a humidified incubator containing 5% CO2.

Transient transfections

HepG2 and Huh-7 cells were transiently transfected with pcDNA3.1-epidermal growth factor receptor (EGFR) plasmid or control pcDNA3.1 plasmid (Shanghai GeneChem Co., Ltd., Shanghai, China) using Lipofectamine® 2000 reagent (Invitrogen; Thermo Fisher Scientific, Inc.) according to the manufacturer's instructions. Briefly, the cells were seeded in 6-well plates at 5×105/well and cultured at 37°C in a humidified atmosphere with 5% CO2. The subsequent day, when the cells were ~70% confluent, they were transfected with a mixture of 3 µl Lipofectamine® 2000 and 2 µg plasmids. At 4 h post-transfection, the cell culture medium was replaced with RPMI-1640 medium. After 24 h, these transfected cells were collected to perform MDC staining or western blot analysis.

MTT assay

Cells were seeded in 96-well plates at an initial density of 4×103 cells/well in 90 µl medium and cultured overnight. As cells reached 30% confluence, various concentrations of YM201636 (0.1, 0.2, 0.5, 1, 2 and 5 µM) were added to the cells, which were then incubated for 24 h. Subsequently, 50 µl 1 mg/ml of MTT (Sigma-Aldrich; Merck KGaA) was added to each well for 4 h. Then, the supernatant was removed and 100 µl DMSO was subsequently added to solubilize the crystal products at room temperature for 10 min. The optical density (OD) was measured at a wavelength of 490 nm using a microplate reader (BioTek Instruments, Inc., Winooski, VT, USA). The growth inhibition ratio was calculated as follows: Growth inhibition ratio (%) = (ODcontrol - ODdrug)/ODcontrol × 100; where ODcontrol is the OD of the group treated with vehicle. The experiments were repeated at least 3 times. To verify whether the mechanism of autophagy promoted cell survival or cell death, HepG2 and Huh-7 cells were pretreated with 5 mM 3-methyladenine (3-MA) for 30 min followed by cotreatment with 2 µM YM201636 for 24 h. Then, an MTT assay was performed. The growth inhibition ratio was calculated as follows: Growth inhibition ratio (%) = (ODcontrol - ODdrug)/ODcontrol × 100; where ODcontrol is the OD of the group treated with vehicle. To examine the effects of EGFR overexpression on cell growth, HepG2 and Huh-7 cells (80% confluence) seeded in a 6-well plate were transfected with pcDNA3.1-EGFR or control pcDNA3.1 plasmids using Lipofectamine® 2000. After 24 h, the cells were harvested and seeded into 96-well plates at a density of 4×103 cells/well for another 24 h. An MTT assay was performed to determine the OD value of each group. During incubation, EGF was not added to the culture medium to avoid EGFR activation.

Flow cytometry

MDC, a specific marker for autophagic vacuoles, was used to examine whether YM201636 induced autophagy. HepG2 and Huh-7 cells were grown on coverslips in a 6-well plate overnight at 3×105/well, and then treated with 2 or 5 µM of YM201636 for 24 h. The cells were collected and washed with ice-cold PBS 3 times, then incubated with 50 µM MDC at 37°C for 30 min. The stained cells were washed, fixed with 4% paraformaldehyde for 10 min at room temperature and analyzed with the imaging flow cytometer FlowSight® (Amnis®; Merck KGaA). A laser set at 405 nm was used for excitation. Bright field and MDC images (green fluorescence) were collected in channels 1 and 8, respectively. The stained cells (1×104/sample) were analyzed using IDEAS version 6.0 software (Merck KGaA).

Western blot analysis

The total proteins were isolated from cancer cells or allograft tumors using radio immunoprecipitation assay buffer (Beyotime Institute of Biotechnology, Haimen, China) following the manufacturer's instructions. The protein concentration was determined using a bicinchoninic acid assay kit (Pierce; Thermo Fisher Scientific, Inc.). Samples were denatured in 5X SDS-sample buffer at 95°C for 5 min. Total proteins (40 µg/well) were separated using SDS-PAGE on 10% gels for EGFR detection or 12% gels for LC3 detection. Following separation, the protein was transferred onto polyvinylidene difluoride membranes. Subsequently, the membranes were blocked using 5% non-fat milk in TBS-Tween (TBS-T) at room temperature for 1 h. Following blocking, membranes were incubated with corresponding primary antibodies overnight at 4°C, then washed 3 times with TBST and incubated with the appropriate HRP-conjugated secondary antibody, and then washed 3 times with TBST. Proteins were detected using the enhanced chemiluminescence plus reagents (Beyotime Institute of Biotechnology). The western blots were visualized using a FluroChem E Imager (Protein Simple, San Jose, CA, USA). Quantity One Software (Quantity One 462; Bio-Rad Laboratories, Inc., Hercules, CA, USA) was used to calculate the alteration of corresponding protein expression.

Evaluation of antitumor effects in vivo

Male BALB/c mice (weighing 20–25 g; aged 5 weeks) were purchased from the Laboratory Animal Center of Henan (Zhengzhou, China). All animal procedures were performed with the approval of the Institutional Animal Care and Use Committee of Henan University (Kaifeng, China). For the development of solid tumors, mice were subcutaneously injected with 2×106 H22 cells. At 1 day after inoculation, the mice were randomly divided into the control and YM201636 groups, which were treated with 5% DMSO or 2 mg/kg YM201636, respectively, via oral administration once daily for 7 consecutive days. The mice were anesthetized using ether for ~30 sec via the respiratory route, and the heart rate and respiratory rate were monitored to ensure the animals were simply anesthetized. Then the mice were euthanized by cervical dislocation, and solid tumors were isolated and weighed. Meanwhile, the heart, liver, kidney, lung, and spleen of the mice were collected and weighed on the last day. The organ weight index was investigated for systemic toxicity evaluation as follows: Organ index (%) = (organ weight/bodyweight) × 100.

Statistical analysis

Statistical analyses were performed using GraphPad Prism 5 software for Windows (GraphPad Software, Inc., La Jolla, CA, USA). All data are expressed as the mean ± standard error. A two-tailed unpaired t-test was used for the comparison of the mean values between two groups. One-way analysis of variance (ANOVA) followed by a Dunnett's test or two-way ANOVA followed by a Bonferroni post hoc test was used for multiple comparisons. P<0.05 was considered to indicate a statistically significant difference.

Results

Effects of YM201636 on HepG2 and Huh-7 cell viability

In order to determine whether the PIKfyve inhibitor YM201636 affects the cell viability of hepatoma, HepG2 and Huh-7 cells were cultured and incubated in the presence of increasing concentrations of the drug for 24 h. Cell viability rates were then detected using an MTT assay. Following incubation of HepG2 and Huh-7 cells with 0.1,0.2, 0.5, 1, 2 and 5 µM YM201636 for 24 h, YM201636 reduced the HepG2 and Huh-7 cell viability in a dose-dependent manner (Fig. 1). The data indicated that YM201636 may inhibit the cell growth of liver cancer cell lines.

YM201636 induces autophagy in liver cancer cell lines

Previous studies have demonstrated that knockdown or inhibition of PIKfyve induces secretory autophagy in multiple cell lines (12,13). Therefore, the present study investigated whether decreased cell viability was associated with autophagy in liver cancer cell lines using an MDC staining assay. Following treatment with 2 or 5 µM YM201636 for 24 h, HepG2 and Huh-7 cells exhibited strong MDC staining (Fig. 2), with enhanced fluorescence in cells treated with 5 µM YM201636. Additionally, the percentage of autophagic cells in the population was increased in HepG2 and Huh-7 cells treated with YM201636 for 24 h (Fig. 2). To further confirm whether YM201636 induces autophagy in liver cancer cell lines, the ratio of LC3I to LC3II was determined, an established autophagosome-related marker (14,15). The results demonstrated that YM201636 significantly promoted the conversion of LC3I to LC3II in HepG2 and Huh-7 cells (Fig. 3). To verify whether the mechanism of autophagy promoted cell survival or cell death, the autophagy inhibitor 3-MA was applied to inhibit YM201636 induced-autophagy in HepG2 and Huh-7 cells; 3-MA inhibits autophagy by blocking autophagosome formation via the inhibition of type III phosphatidylinositol 3-kinases (PI3Ks) (16,17). Co-treatment with 5 mM 3-MA for 24 h attenuated the inhibitory effects of YM201636 on liver cancer cell lines (Fig. 4). Collectively, these results suggested that YM201636 inhibits the growth of HepG2 and Huh-7 cells via the induction of autophagy.

YM201636 induced-autophagy depends on EGFR overexpression

It has been previously reported that PIKfyve inhibition blocks the lysosomal degradation of EGFR, resulting in increased expression levels of EGFR in MCF-10A cells (18), and overexpression of inactive EGFR has been demonstrated to be associated with autophagy (19). Therefore, the present study investigated whether YM201636-induced autophagy is dependent upon EGFR overexpression. To test this hypothesis, the total protein expression of EGFR was determined in HepG2 and Huh-7 cells treated with YM201636. Western blotting demonstrated that the total protein expression levels of EGFR were significantly upregulated in HepG2 and Huh-7 cells treated with 2 µM YM201636 for 24 h (Fig. 5). To investigate whether the increased EGFR was activated in HepG2 and Huh-7 cells treated with YM201636, the phosphorylation levels of EGFR at Tyr1068, an indicator of EGFR activation, were examined. As shown in Fig. 5, the phosphorylation levels of EGFR at Tyr1068 were notably unaffected in HepG2 and Huh-7 cells treated with YM201636. The results suggested that the increased total protein constituted inactive EGFR.

To further investigate the hypothesis that inactive EGFR mediates autophagy in HepG2 and Huh-7 cells, HepG2 and Huh-7 cells were transfected with the pcDNA3.1-EGFR plasmid with no EGF treatment. At 24 h after transfection, the expression of EGFR was increased significantly (Fig. 6). Subsequently, whether overexpression of EGFR was able to affect autophagy in liver cancer cells was examined. As demonstrated in Fig. 7, HepG2 and Huh-7 cells transfected with EGFR exhibited strong MDC staining and an increase in the proportion of autophagic cells. Additionally, overexpression of EGFR also promoted the transformation of LC3I into LC3II in HepG2 and Huh-7 cells (Fig. 8). Furthermore, the effects of EGFR overexpression on liver cancer cell growth were determined. Overexpression of EGFR in HepG2 and Huh-7 cells inhibited cell proliferation (Fig. 9). The results indicated that YM201636 induced-autophagy was dependent on EGFR overexpression in HepG2 and Huh-7 cells.

YM201636 inhibits the in situ growth of hepatoma in vivo

To further evaluate the antitumor activity of the PIKfyve inhibitor, YM201636, in vivo, an allograft model of mouse liver cancer was established by subcutaneously injecting H22 cells (mouse hepatoma cell line) into BALB/c mice. Transplantation of H22 cells into BALB/c mice induced the in situ formation of tumors. Following treatment with 2 mg/kg YM201636 for 7 consecutive days, the tumor weight of the YM201636 group was lower than that of the negative control group. For example, the mean weight of tumors in the YM201636-treated group was 0.31±0.05 g, compared with 0.67±0.07 g in the control group (Fig. 10A and B). Additionally, YM201636 promoted the expression of EGFR total protein in allograft tumors; however, the phosphorylation levels of EGFR at Tyr1068 were not notably altered (Fig. 10C and D). In addition, visceral organ indices were also examined to evaluate adverse effects of YM201636. No significant differences in the visceral organ indexes (heart, liver, spleen, lung and kidney) were observed in the YM201636 group compared with the control group (Fig. 10E). These results suggested that YM201636 may inhibit tumor growth without notable systemic toxicity at the dose applied in the present study.

Discussion

PIKfyve is an enzyme that is crucial for the synthesis of PtdIns(3,5)P2 and has been associated with various membrane transport events (20,21). Perturbations in the functions of PIKfyve lead to the formation of swollen endosomal/lysosomal structures in a variety of mammalian cell lines (22,23). Additionally, PIKfyve has been proposed to be involved in oncogenesis and cancer cell migration. For example, PIKfyve promotes cell migration and invasion through the activation of Ras-related protein Rac1in lung carcinoma, osteosarcoma and rhabdomyosarcoma cells (8,24); knockdown of PIKfyve resulted in significant decreases in cell migration velocity and persistence (10). Consistent with these observations, in the present study, it was demonstrated that pharmacological inhibition of PIKfyve using YM201636 resulted in an inhibitory effect on tumor cell growth via the induction of autophagy in hepatoma cell lines.

The present study demonstrated that EGFR upregulation required for autophagy induced by the inhibition of PIKfyve. In support of the results, Er et al (18) reported that suppression of PIKfyve activity reduces the rate of EGFR degradation in MCF-10A cells. EGFR is part of the ErbB family of receptor tyrosine kinases and is overexpressed in many human cancers (25). Abnormal hyperactivation of EGFR is associated with unregulated proliferation, malignant transformation and metastasis in cancer cells (26,27). It has been demonstrated that EGFR activation inhibits autophagy, dependent upon its kinase domain, by maintaining high activation levels of the PI3K/protein kinase B/mammalian target of rapamycin signaling pathway; knockdown or inhibition of EGFR signaling induces autophagy in various epithelial cancers (28,29). However, in the present study, overexpression of EGFR induced autophagy in liver cancer cells. The results suggested that the increased EGFR may be inactive, as Tan et al (19) reported that inactive EGFR complexes collaborate with lysosomal-associated transmembrane protein 4B and exocyst complex component 2 at endosomes to disassociate Rubicon from Beclin 1, which in turn initiates autophagy. Autophagy exhibits a dual role that can induce a cell survival or cell death response (30,31). In the present study, EGFR-mediated autophagy was proposed to underlie cancer cell death.

Numerous studies have suggested EGFR overexpression promotes tumor cell proliferation (26,32); however, the findings of the present study indicated that PIKfyve inhibitor YM201636-induced autophagy was dependent on EGFR overexpression. As mentioned above, the increased EGFR total protein was inactive EGFR, and inactive EGFR complexes could initiate autophagy (19). In support of the present results, Lanaya et al (33) demonstrated that mice lacking EGFR in hepatocytes develop more hepatocellular carcinoma (33). However, there are limitations in this study. Although the results suggested the increased EGFR total protein was inactive EGFR, the expression of proteins downstream of EGFR, such as Akt, p-Akt, ERK, p-ERK, was not analyzed. The phosphorylation of Akt and ERK are indicators of EGFR activation (34,35), therefore, the expression of these downstream proteins will be examined in future studies.

In the present study, it was speculated that YM201636 induced total EGFR expression through reducing the rate of EGFR degradation in HepG2 and Huh-7 cells. Since EGF stimulation causes EGFR degradation by delivery to the lysosomes (18), therefore, it was assumed that the unchanged phosphorylation levels of EGFR at Tyr1068, was due to lack of EGF stimulation.

In conclusion, the results of the present study suggested that the PIKfyve inhibitor YM201636 may possess therapeutic potential for the treatment of liver cancer. In addition, EGFR overexpression induced by a PIKfyve inhibitor contributes to autophagy, thus leading to an inhibitory effect on cancer cell growth.

Acknowledgements

Not applicable.

Funding

This study was supported by the National Science Foundation of China (grant nos. 81573465, 81772832 and 81701110), the Natural Science Foundation of Henan (grant no. 162300410039); and the Program for Science and Technology of the Department of Education of Henan Province (grant no. 16A350013).

Availability of data and materials

All data generated or analysed during this study are available from the corresponding author on reasonable request.

Authors' contributions

DF and SQX participated in the design of the study, data analysis and manuscript writing. JZH, ZQX, JN conducted MTT assays and flow cytometry, participated in the design of the study and data analysis. WL, XW, CL and HS performed the assays and analysis. All authors read and approved the final manuscript.

Ethics approval and consent to participate

All experiments were approved by the Ethics Committee of University of Henan University (Kaifeng, China).

Patient consent for publication

Not applicable.

Competing interests

The authors declare that they have no competing interests.

References

1 

Lozano R, Naghavi M, Foreman K, Lim S, Shibuya K, Aboyans V, Abraham J, Adair T, Aggarwal R, Ahn SY, et al: Global and regional mortality from 235 causes of death for 20 age groups in 1990 and 2010: A systematic analysis for the global burden of disease study 2010. Lancet. 380:2095–2128. 2012. View Article : Google Scholar : PubMed/NCBI

2 

Yang Y, Nagano H, Ota H, Morimoto O, Nakamura M, Wada H, Noda T, Damdinsuren B, Marubashi S, Miyamoto A, et al: Patterns and clinicopathologic features of extrahepatic recurrence of hepatocellular carcinoma after curative resection. Surgery. 141:196–202. 2007. View Article : Google Scholar : PubMed/NCBI

3 

Sbrissa D, Ikonomov OC and Shisheva A: PIKfyve, a mammalian ortholog of yeast Fab1p lipid kinase, synthesizes 5-phosphoinositides. Effect of insulin. J Biol Chem. 274:21589–21597. 1999. View Article : Google Scholar : PubMed/NCBI

4 

Shisheva A: PIKfyve: The road to PtdIns 5-P and PtdIns 3,5-P2. Cell Biol Int. 25:1201–1206. 2001. View Article : Google Scholar : PubMed/NCBI

5 

Takasuga S and Sasaki T: Phosphatidylinositol-3,5-bisphosphate: Metabolism and physiological functions. J Biochem. 154:211–218. 2013. View Article : Google Scholar : PubMed/NCBI

6 

McCartney AJ, Zolov SN, Kauffman EJ, Zhang Y, Strunk BS, Weisman LS and Sutton MA: Activity-dependent PI(3,5)P2 synthesis controls AMPA receptor trafficking during synaptic depression. Proc Natl Acad Sci USA. 111:E4896–E4905. 2014. View Article : Google Scholar : PubMed/NCBI

7 

Kim J, Jahng WJ, Di Vizio D, Lee JS, Jhaveri R, Rubin MA, Shisheva A and Freeman MR: The phosphoinositide kinase PIKfyve mediates epidermal growth factor receptor trafficking to the nucleus. Cancer Res. 67:9229–9237. 2007. View Article : Google Scholar : PubMed/NCBI

8 

Oppelt A, Haugsten EM, Zech T, Danielsen HE, Sveen A, Lobert VH, Skotheim RI and Wesche J: PIKfyve, MTMR3 and their product PtdIns5P regulate cancer cell migration and invasion through activation of Rac1. Biochem J. 461:383–390. 2014. View Article : Google Scholar : PubMed/NCBI

9 

Ikonomov OC, Filios C, Sbrissa D, Chen X and Shisheva A: The PIKfyve-ArPIKfyve-Sac3 triad in human breast cancer: Functional link between elevated Sac3 phosphatase and enhanced proliferation of triple negative cell lines. Biochem Biophys Res Commun. 440:342–347. 2013. View Article : Google Scholar : PubMed/NCBI

10 

Oppelt A, Lobert VH, Haglund K, Mackey AM, Rameh LE, Liestol K, Schink KO, Pedersen NM, Wenzel EM, Haugsten EM, et al: Production of phosphatidylinositol 5-phosphate via PIKfyve and MTMR3 regulates cell migration. EMBO Rep. 14:57–64. 2013. View Article : Google Scholar : PubMed/NCBI

11 

Dupuis-Coronas S, Lagarrigue F, Ramel D, Chicanne G, Saland E, Gaits-Iacovoni F, Payrastre B and Tronchere H: The nucleophosmin-anaplastic lymphoma kinase oncogene interacts, activates, and uses the kinase PIKfyve to increase invasiveness. J Biol Chem. 286:32105–32114. 2011. View Article : Google Scholar : PubMed/NCBI

12 

Sano O, Kazetani K, Funata M, Fukuda Y, Matsui J and Iwata H: Vacuolin-1 inhibits autophagy by impairing lysosomal maturation via PIKfyve inhibition. FEBS Lett. 590:1576–1585. 2016. View Article : Google Scholar : PubMed/NCBI

13 

Hessvik NP, Overbye A, Brech A, Torgersen ML, Jakobsen IS, Sandvig K and Llorente A: PIKfyve inhibition increases exosome release and induces secretory autophagy. Cell Mol Life Sci. 73:4717–4737. 2016. View Article : Google Scholar : PubMed/NCBI

14 

Galluzzi L, Baehrecke EH, Ballabio A, Boya P, Bravo-San Pedro JM, Cecconi F, Choi AM, Chu CT, Codogno P, Colombo MI, et al: Molecular definitions of autophagy and related processes. EMBO J. 36:1811–1836. 2017. View Article : Google Scholar : PubMed/NCBI

15 

Lee YK and Lee JA: Role of the mammalian ATG8/LC3 family in autophagy: Differential and compensatory roles in the spatiotemporal regulation of autophagy. BMB Rep. 49:424–430. 2016. View Article : Google Scholar : PubMed/NCBI

16 

Bhat P, Kriel J, Shubha Priya B, Basappa, Shivananju NS and Loos B: Modulating autophagy in cancer therapy: Advancements and challenges for cancer cell death sensitization. Biochem Pharmacol. 147:170–182. 2018. View Article : Google Scholar : PubMed/NCBI

17 

Heckmann BL, Yang X, Zhang X and Liu J: The autophagic inhibitor3-methyladenine potently stimulates PKA-dependent lipolysis in adipocytes. Br J Pharmacol. 168:163–171. 2013. View Article : Google Scholar : PubMed/NCBI

18 

Er EE, Mendoza MC, Mackey AM, Rameh LE and Blenis J: AKT facilitates EGFR trafficking and degradation by phosphorylating and activating PIKfyve. Sci Signal. 6:ra452013. View Article : Google Scholar : PubMed/NCBI

19 

Tan X, Thapa N, Sun Y and Anderson RA: A kinase-independent role for EGF receptor in autophagy initiation. Cell. 160:145–160. 2015. View Article : Google Scholar : PubMed/NCBI

20 

Hirano T, Munnik T and Sato MH: Inhibition of phosphatidylinositol 3,5-bisphosphate production has pleiotropic effects on various membrane trafficking routes in Arabidopsis. Plant cell Physiol. 58:120–129. 2017.PubMed/NCBI

21 

Ikonomov OC, Fligger J, Sbrissa D, Dondapati R, Mlak K, Deeb R and Shisheva A: Kinesin adapter JLP links PIKfyve to microtubule-based endosome-to-trans-Golgi network traffic of furin. J Biol Chem. 284:3750–3761. 2009. View Article : Google Scholar : PubMed/NCBI

22 

Jin N, Jin Y and Weisman LS: Early protection to stress mediated by CDK-dependent PI3,5P2 signaling from the vacuole/lysosome. J Cell Biol. 216:2075–2090. 2017. View Article : Google Scholar : PubMed/NCBI

23 

Bissig C, Hurbain I, Raposo G and van Niel G: PIKfyve activity regulates reformation of terminal storage lysosomes from endolysosomes. Traffic. 18:747–757. 2017. View Article : Google Scholar : PubMed/NCBI

24 

Dayam RM, Sun CX, Choy CH, Mancuso G, Glogauer M and Botelho RJ: The lipid kinase PIKfyve coordinates the neutrophil immune response through the activation of the Rac GTPase. J Immunol. 199:2096–2105. 2017. View Article : Google Scholar : PubMed/NCBI

25 

Du Z and Lovly CM: Mechanisms of receptor tyrosine kinase activation in cancer. Mol Cancer. 17:582018. View Article : Google Scholar : PubMed/NCBI

26 

Ye QH, Zhu WW, Zhang JB, Qin Y, Lu M, Lin GL, Guo L, Zhang B, Lin ZH, Roessler S, et al: GOLM1 modulates EGFR/RTK cell-surface recycling to drive hepatocellular carcinoma metastasis. Cancer Cell. 30:444–458. 2016. View Article : Google Scholar : PubMed/NCBI

27 

Sainsbury JR, Farndon JR, Needham GK, Malcolm AJ and Harris AL: Epidermal-growth-factor receptor status as predictor of early recurrence of and death from breast cancer. Lancet. 1:1398–1402. 1987.PubMed/NCBI

28 

Li X and Fan Z: The epidermal growth factor receptor antibody cetuximab induces autophagy in cancer cells by downregulating HIF-1alpha and Bcl-2 and activating the beclin 1/hVps34 complex. Cancer Res. 70:5942–5952. 2010. View Article : Google Scholar : PubMed/NCBI

29 

Li H, You L, Xie J, Pan H and Han W: The roles of subcellularly located EGFR in autophagy. Cell Signal. 35:223–230. 2017. View Article : Google Scholar : PubMed/NCBI

30 

Chen Y, Henson ES, Xiao W, Huang D, McMillan-Ward EM, Israels SJ and Gibson SB: Tyrosine kinase receptor EGFR regulates the switch in cancer cells between cell survival and cell death induced by autophagy in hypoxia. Autophagy. 12:1029–1046. 2016. View Article : Google Scholar : PubMed/NCBI

31 

Jacob JA, Salmani JMM, Jiang Z, Feng L, Song J, Jia X and Chen B: Autophagy: An overview and its roles in cancer and obesity. Clin Chim Acta. 468:85–89. 2017. View Article : Google Scholar : PubMed/NCBI

32 

Sato H, Yamamoto H, Sakaguchi M, Shien K, Tomida S, Shien T, Ikeda H, Hatono M, Torigoe H, Namba K, et al: Combined inhibition of MEK and PI3K pathways overcomes acquired resistance to EGFR-TKIs in non-small cell lung cancer. Cancer Sci. 109:3183–3196. 2018. View Article : Google Scholar : PubMed/NCBI

33 

Lanaya H, Natarajan A, Komposch K, Li L, Amberg N, Chen L, Wculek SK, Hammer M, Zenz R, Peck-Radosavljevic M, et al: EGFR has a tumour-promoting role in liver macrophages during hepatocellular carcinoma formation. Nature Cell Biol. 16:972–977. 2014. View Article : Google Scholar : PubMed/NCBI

34 

Zhang Y, Wang L, Zhang M, Jin M, Bai C and Wang X: Potential mechanism of interleukin-8 production from lung cancer cells: An involvement of EGF-EGFR-PI3K-Akt-Erk pathway. J Cell Physiol. 227:35–43. 2012. View Article : Google Scholar : PubMed/NCBI

35 

Zhang L, Li Y, Lan L, Liu R, Wu Y, Qu Q and Wen K: Tamoxifen has a proliferative effect in endometrial carcinoma mediated via the GPER/EGFR/ERK/cyclin D1 pathway: A retrospective study and an in vitro study. Mol Cell Endocrinol. 437:51–61. 2016. View Article : Google Scholar : PubMed/NCBI

Related Articles

Journal Cover

March-2019
Volume 41 Issue 3

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
Hou JZ, Xi ZQ, Niu J, Li W, Wang X, Liang C, Sun H, Fang D and Xie SQ: Inhibition of PIKfyve using YM201636 suppresses the growth of liver cancer via the induction of autophagy. Oncol Rep 41: 1971-1979, 2019
APA
Hou, J., Xi, Z., Niu, J., Li, W., Wang, X., Liang, C. ... Xie, S. (2019). Inhibition of PIKfyve using YM201636 suppresses the growth of liver cancer via the induction of autophagy. Oncology Reports, 41, 1971-1979. https://doi.org/10.3892/or.2018.6928
MLA
Hou, J., Xi, Z., Niu, J., Li, W., Wang, X., Liang, C., Sun, H., Fang, D., Xie, S."Inhibition of PIKfyve using YM201636 suppresses the growth of liver cancer via the induction of autophagy". Oncology Reports 41.3 (2019): 1971-1979.
Chicago
Hou, J., Xi, Z., Niu, J., Li, W., Wang, X., Liang, C., Sun, H., Fang, D., Xie, S."Inhibition of PIKfyve using YM201636 suppresses the growth of liver cancer via the induction of autophagy". Oncology Reports 41, no. 3 (2019): 1971-1979. https://doi.org/10.3892/or.2018.6928