Open Access

Diosmetin inhibits cell proliferation and induces apoptosis by regulating autophagy via the mammalian target of rapamycin pathway in hepatocellular carcinoma HepG2 cells

  • Authors:
    • Jie Liu
    • Hao Ren
    • Bin Liu
    • Qingyu Zhang
    • Mingyi Li
    • Runzhi Zhu
  • View Affiliations

  • Published online on: October 19, 2016     https://doi.org/10.3892/ol.2016.5301
  • Pages: 4385-4392
  • Copyright: © Liu 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

Hepatocellular carcinoma (HCC), which is a type of malignant tumor, is the fifth most common cancer in men and ninth in women worldwide. The aim of the present study was to investigate the antitumor effect of diosmetin (DIOS) in hepatocellular carcinoma HepG2 cells. The proliferation, apoptosis and autophagy rates of HepG2 cells were measured following treatment with DIOS. The effects of DIOS treatment on HepG2 cell proliferation and apoptosis rates were analyzed using MTT assays and Annexin V staining, respectively. The effect of DIOS treatment on autophagy levels was assessed using transmission electron microscopy, green fluorescent protein (GFP)‑microtubule‑associated protein 1 light chain (LC3) transfection and LysoTracker Red staining. Furthermore, bafilomycin A1 (BA1), an autophagy inhibitor, was used to assess the association between DIOS and cell autophagy, proliferation and apoptosis. In addition, the expression of autophagy‑related proteins [mammalian target of rapamycin (mTOR), phosphatidylinositol 3‑kinase, P70S6K, phosphoinositide‑dependent kinase‑1, extracellular signal‑regulated kinase, 5'‑AMP‑activated protein kinase and Akt] and apoptosis‑related proteins [B‑cell lymphoma (Bcl)‑2‑associated X protein, Bak, p53, Bcl‑2 and caspase‑3] were analyzed by western blotting. The results revealed that DIOS significantly inhibited proliferation (P<0.01) and induced apoptosis (P<0.001) in HepG2 cells. It was also demonstrated that DIOS triggered autophagy by regulating the mTOR pathway in HepG2 cells. Notably, following treatment of HepG2 cells with the autophagy inhibitor, BA1, the expression of apoptosis‑related proteins, including Bax, Bak and p53, were significantly decreased (P<0.05), and cell viability was recovered to a certain extent. In conclusion, DIOS inhibits cell proliferation and induces apoptosis in HepG2 cells via regulation of the mTOR pathway. Thus, the results of the current study indicate that DIOS may present a potential therapeutic agent for HCC treatment.

Introduction

Hepatocellular carcinoma (HCC) is a relatively common type of malignant tumor in some less developed regions, especially in Eastern Asia (1,2). In 2012, it was estimated that HCC was the fifth most common type of cancer in males, accounting for 7.5% of all cancers, and the ninth most common type of cancer in females, accounting for 3.4% of all cancers (2). HCC exhibits one of the highest mortality rates among malignant tumors worldwide (the ratio of mortality to incidence is 0.95) and a low 5-year survival rate (4.26%) when compared with other cancer types (3,4). Because of the lack of specific symptoms in the early stage, a large proportion of patients are not diagnosed with HCC patients until metastasis has occurred (5). At present, common clinical and pathological parameters, including α-fetoprotein and tumor differentiation, are used to diagnose HCC, and hepatectomy is combined with chemotherapy as the standard treatment (1). However, treatment is often ineffective due to the high incidence of recurrence and metastasis (6), and, thus, it is important to develop an effective novel therapeutic strategy to treat HCC (7,8).

Autophagy and apoptosis are two processes that remove aged or altered cells from the body (9). Autophagy is an evolutionarily conserved catabolic process that occurs in eukaryotic cells, which results in the lysosomal breakdown of organelles in response to stressful stimuli (10). It has previously been reported that anticancer therapies including cytotoxic chemotherapy, (11) radiation (12) and kinase inhibitors (13), are able to induce autophagy in tumors and cause cell death in tumor tissues. Apoptosis, also termed programmed cell death, is a complex process that is associated with the expression of apoptosis-related proteins, including p53, caspases, Bcl-2 and Bax (14). Cell apoptosis may be induced due as a result of physiological regulation and chemical stimulation (7).

Diosmetin (DIOS; Fig. 1A), which is obtained from the legume Acacia farnesiana and the leaves of Olea europaea L., is an aglycone of the flavonoid glycoside, diosmin (15). DIOS has been demonstrated to exhibit antibacterial (16), antimicrobial (17), anti-inflammatory (18) and antioxidant (19) effects. Additionally, it has been demonstrated that DIOS exhibits anticancer activity in breast cancer cells by inducing cell cycle arrest (20). The present study aimed to investigate the effect of DIOS treatment on cell invasion and autophagy in HCC cells and to evaluate whether DIOS treatment triggers cell apoptosis.

Materials and methods

Reagents

DIOS (Sigma-Aldrich; Merck Millipore Darmstadt, Germany) was dissolved in 10 mg/ml dimethylsulfoxide (DMSO), diluted in RPMI-1640 medium (Thermo Fisher Scientific, Inc., Waltham, MA, USA) and stored at −20°C in the dark prior to use. MTT was purchased from Amresco LCC (Solon, OH, USA) and the fluorescein isothiocyanate (FITC)-Annexin V Apoptosis Detection kit I was obtained from BD Biosciences (Franklin Lakes, NJ, USA). LysoTracker Red was purchased from Beyotime Institute of Biotechnology (Haimen, China) and the green fluorescent protein (GFP)-microtubule-associated protein 1 light chain (LC3) plasmid was provided by the Institute of Neurology, Guangdong Medical College (Guangdong, China). Lipofectamine™ 2000 transfection reagent was purchased from Thermo Fisher Scientific, Inc. Antibodies targeting Bak (cat. no. 12105), B-cell lymphoma-2 (Bcl-2; cat. no. 2876S), Bcl-2-associated X protein (Bax; cat. no. 2772S), p53 (cat. no. 9282S), caspase-3 (cat. no. 9665), LC3 (cat. no. 12741), Beclin-1 (cat. no. 4122S), mammalian target of rapamycin (mTOR; cat. no. 2972), phosphorylated (p)-mTOR (cat. no. 5536), phosphatidylinositol 3-kinase (PI3K p85, cat. no. 4257; PI3K p110, cat. no. 4255), phosphoinositide-dependent kinase-1 (PDK1; cat. no. 13037), Akt (cat. no. 9272), p-Akt (cat. no. 13038), extracellular signal-regulated kinase (ERK; cat. no. 4695S), p-ERK (cat. no. 4377S), 5′-AMP-activated protein kinase (AMPK; cat. no. 2795S), p-AMPK (cat. no. 2537), p70-S6 kinase (S6k; cat. no. 2708), p-S6k (cat. no. 9234) and GAPDH (cat. no. 2118) were purchased from Cell Signaling Technology, Inc. (Danvers, MA, USA). All antibodies were diluted to 1:1,000. Bafilomycin A1 (BA1) was purchased from Sigma-Aldrich; Merck Millipore.

Cell culture

The human HCC HepG2 cell line was provided by the Shanghai Maternal and Child Health Hospital (Shanghai, China). Cells were maintained in RPMI-1640 medium supplemented with 10% fetal bovine serum (Gibco; Thermo Fisher Scientific, Inc.), cultured at a 37°C in an atmosphere of 5% CO2, and passaged to 80% confluence.

Cell proliferation assay

Cell proliferation was assessed by MTT assay. A total of 1×104 cells/well were seeded in 100 µl RPMI-1640 culture medium in a 96-well plate. Following 24 h in adhesive culture, the medium was removed and 1, 2, 5, 10, 15 or 20 µg/ml DIOS was added. Untreated cells served as the control group. After 24 h, 20 µl MTT (5 mg/ml) was added to each well. Following incubation for 3 h at 37°C, the medium was removed and 200 µl DMSO was added to each well. Subsequently, absorbance was measured using a microplate reader (PerkinElmer, Inc., Waltham, MA, USA) at a wavelength of 570 nm. Each experiment was performed in triplicate.

Annexin V staining

Annexin V staining was used to assess the apoptotic rate of HCC cells following treatment with DIOS. The Annexin V Apoptosis Detection kit I was used, according to the manufacturer's instructions. Briefly, cells (5×104/well) were seeded into a 6-well plate at 37°C overnight followed by treatment with 5, 10 and 20 µg/ml DIOS. Untreated cells served as the control. Cells were washed twice with cold phosphate-buffered saline (PBS) and resuspended in 100 µl binding buffer. The cell suspensions were then treated with 5 µl FITC-Annexin V and 10 µl propidium iodide (PI) and incubated for 15 min at room temperature in the dark. Next, 400 µl binding buffer was added to terminate the reaction and the apoptosis rate of the cells was analyzed by flow cytometry (BD FACSCanto II; BD Biosciences) at a 488-nm excitation wavelength; the emission wavelengths of FITC and PI were observed at 520 nm and 617 nm, respectively.

Transmission electron microscopy

Cells (1×106) were fixed in 2.5% glutaraldehyde (Ted Pella Inc., Redding, CA, USA) and 0.1 mol/l phosphate buffer (pH 7.4; Jinuo, Hangzhou, China) for 30 min at room temperature followed by fixation in 1% osmium tetraoxide (Ted Pella Inc.) and 0.1 mol/l phosphate buffer (pH 7.4) for 30 min. Cells were then subjected to graded ethanol dehydration with epoxy propane (Ted Pella Inc.), embedded in epoxy resin and segmented using a UC7 ultrastructural slicer (Leica Microsystems GmbH, Wetzlar, Germany). Sections (90-nm) were subsequently stained with 5% uranyl acetate (SPI-Chem, West Chester, PA, USA) and 5% lead citrate (SPI-Chem) prior to analysis using a JEM1400 transmission electron microscope (JEOL Ltd., Tokyo, Japan).

Cell transfection

Cells at 80% confluence were seeded in 6 wells plate and transfected with the GFP-LC3 plasmid using Lipofectamine™ 2000, according to the manufacturer's instructions. Cells were treated with various concentrations (0, 5, 10 and 20 µg/ml) of DIOS 24 h after transfection and assessed using a fluorescence microscope (Leica Microsystems GmbH). Green fluorescence indicated that plasmids had successfully been transfected into the cells.

Lyso-Tracker Red staining

Cells were treated with 0, 5, 10 and 20 µg/ml DIOS for 24 h followed by incubation with 75 nmol/l Lyso-Tracker Red for 2–3 h at 37°C. Next, LysoTracker Red was removed and cells were stained with Hoechst 33342 (Beyotime Institute of Biotechnology) for 10 min at 37°C. Cells were washed three times in PBS, incubated with fresh culture medium and observed using a fluorescence microscope (Leica Microsystems GmbH). Red fluorescence was measured using 544 nm excitation and 590 nm long pass emission filters.

Western blotting

Cells were washed twice with PBS and suspended in lysis buffer (Beyotime Institute of Biotechnology) for 30 min on ice. Lysates were centrifuged at 13,000 × g for 10 min at 4°C and transferred to polyvinylidene difluoride membranes (EMD Millipore, Billerica, USA) prior to 10% SDS-PAGE. Membranes were then blocked with 5% bovine serum albumin (Biosharp, Hefei, China) in Tris-buffered saline (Beyotime Institute of Biotechnology) containing Tween-20 (TBST; Sangon Biotech Co., Ltd., Shanghai, China) for 1 h at room temperature. After three washes with TBST, membranes were incubated with primary antibodies at 4°C overnight. Membranes were then washed three times with TBST prior to incubation with secondary antibody (cat. no. E030120; 1:1,000; EarthOX Life Sciences, Millbrae, CA, USA) for 2 h at room temperature. The protein bands were exposed in a dark room and analyzed using AlphaView SA 3.4.0. software (ProteinSimple, San Jose, CA, USA). Protein expression was normalized to GAPDH.

Statistical analysis

Data were obtained from at least three independent experiments and all results are expressed as the mean ± standard error of the mean. Differences between the groups were assessed using the Student's t-test and all statistical analysis was performed using SPSS 18.0 statistical software (SPSS, Inc., Chicago, IL, USA). P<0.05 was considered to indicate a statistically significant difference.

Results

DIOS inhibits HepG2 cell proliferation

MTT assay was performed to assess the effect of DIOS on HepG2 cell proliferation. The results demonstrated that cell proliferation was significantly inhibited following treatment with ≥5 µg/ml DIOS (P<0.01; Fig. 1B) with a half maximal inhibitory concentration of 11.60±1.71 µg/ml at 24 h. In addition, morphological changes were observed under a microscope: Cells treated with 10 and 20 µg/ml DIOS were distorted and cell proliferation was markedly inhibited compared with controls (Fig. 1C).

DIOS promotes apoptosis via activation of caspase-3 in HepG2 cells

FITC-Annexin V/PI double staining was used to detect apoptosis in HepG2 cells following DIOS treatment. Following treatment with 10 and 20 µg/ml DIOS, the rate of apoptosis significantly increased compared with the control (25.6±4.8 and 37.6±6.1 vs. 8.8±0.7, respectively; P<0.001; Fig. 2A). These results indicated that DIOS treatment promotes apoptosis in HepG2 cells in a dose-dependent manner. Furthermore, western blot analysis demonstrated that DIOS downregulated Bcl-2 expression and upregulated Bak, Bax, p53 and casapse-3 protein expression in a dose-dependent manner (Fig. 2B).

DIOS induces autophagy in HepG2 cells

Transmission electron microscopy demonstrated that DIOS induced the generation of autophagosomes in HepG2 cells. As shown in Fig. 3A, cells treated with 5 µg/ml DIOS exhibited enlarged mitochondria and fragmented cristae. Cells treated with ≥10 µg/ml DIOS exhibited increased numbers of autophagosomes in the cytoplasm. To confirm the progression of autophagy, the distribution of GFP-LC3 was detected following transfection of the GFP-LC3 plasmid into the cytoplasm. The cytosolic form (LC3-I), viewed as dispersed green fluorescence under the microscope, is converted to the autophagosome-associating form (LC3-II), viewed as bright fluorescent spots under the microscope, as autophagy occurs (21). Following treatment of cells with 0, 5, 10 and 20 µg/ml DIOS for 24 h, the GFP-LC3 in the cytoplasm changed from the cytosolic form into the autophagosome-associating form (Fig. 3B), indicating that DIOS treatment converted LC3-I to LC3-II, which is associated with the formation of autophagosomes. In addition, LysoTracker Red staining was used to count the number of lysosomes in the HepG2 cells following treatment with DIOS. The results revealed that the number of active lysosomes in HepG2 cells increased in a dose-dependent manner following DIOS treatment (Fig. 3C). These results indicated that DIOS induced cell autophagy in HepG2 cells in a dose-dependent manner.

DIOS induces cell autophagy via the mTOR-related signaling pathway

mTOR and proteins associated with the signaling pathway, including PI3K, PDK1, Akt, ERK1/2, AMPK S6k and Beclin-1, are autophagy regulatory proteins (22,23). LC3-II is critical for autophagosome formation and thus is a marker of autophagy (24). In the current study, expression of the aforementioned proteins in HepG2 cells was measured to evaluate whether the autophagy mechanism was induced following DIOS treatment. Western blot analysis revealed that following treatment with DIOS for 24 h, LC3-II and Beclin-1 expression increased in a dose-dependent manner (Fig. 3D). In addition AMPK protein expression was significantly increased (P<0.001) and mTOR, PI3K p85 and S6k protein expression were significantly decreased in a dose-dependent manner (P<0.05; Fig. 4). No marked changes in the expression of Akt and ERK1/2 were observed (P>0.05); however, the phosphorylation of Akt and ERK1/2 were significantly decreased (P<0.001; Fig. 4).

Autophagy inhibitor BA1 reverses DIOS-induced cell proliferation inhibition and apoptosis

To determine the effect of DIOS-induced autophagy, HepG2 cells were treated with BA1 to attenuate the autophagy process. The results revealed that treatment with DIOS alone resulted in significantly decreased cell proliferation when compared with cells treated with DIOS and BA1 (Fig. 5A and B). Furthermore, western blotting demonstrated that the expression of pro-apoptosis proteins Bak, Bax and p53 was higher following treatment with DIOS alone when compared with combined treatment with DIOS and BA1 (Fig. 5C and D).

Discussion

HCC is one of the most common malignant tumors worldwide, accounting for the second highest number of cancer-associated mortalities (9.1% of all cancer-associated deaths); the mortality rate reached 95% in 2012 (2,3). It has been reported that several flavonoid compounds, including baicalein (25), dihydromyricetin (26,27) and resveratrol (28) may exhibit antitumor effects. DIOS is a flavonoid that has been demonstrated to exhibit antibacterial, antimicrobial, anti-inflammatory and antioxidant effects (29). Previously, it has been demonstrated that DIOS induces cell cycle arrest in the G2/M phase (30); however, the association between DIOS and cell proliferation inhibition, apoptosis and autophagy in HCC cells remains unclear.

Apoptosis induction is considered to be an effective strategy for cancer treatment (27). The present study indicated that exposing HepG2 cells to >5 ug/ml DIOS for 24 h significantly inhibited cell proliferation Furthermore, treatment with 10 and 20 µg/ml DIOS also increased apoptosis rates in HepG2 cells via the upregulation of apoptosis-related proteins, p53 and Bak, alteration of the Bax/Bcl-2 ratio and subsequent activation of caspase-3. p53 is one of the most important proteins involved in the death receptor and mitochondrial pathways (26). It regulates the expression of Bax, Bak and Bcl-2 and converts pro-caspase-3 into cleaved caspase-3 to stimulate apoptosis (8). Bax is a pro-apoptotic protein, which relocates to the mitochondria following the induction of apoptosis to induce mitochondria depolarization. By contrast, Bcl-2 is an anti-apoptotic protein that inhibits the function of Bax (31). Therefore, the ratio of Bax/Bcl-2 is associated with apoptosis (32). The current study indicated that DIOS induces HepG2 cell apoptosis by upregulating the protein expression of Bax and downregulating Bcl-2 protein expression.

Autophagy is a degradation process that delivers cytoplasmic material to lysosomes via autophagosomes. Autophagy is considered to exhibit two separate effects on tumor development: It has the ability to suppress the growth of precancerous cells and maintain a stable genome and it may also prevent the survival of tumor cells (33). Therefore, whether autophagosomes protect cancer cells from external factors or induce cell apoptosis in cancer therapy remains controversial (34). Previous studies have indicated that pharmacological inhibition of autophagy or silencing of autophagy related genes, such as ATG5, may reduce the risk of drug resistance arising following chemotherapy treatment (3537). It has been reported that autophagy induces cell death by depleting the cell of organelles and critical proteins, a process termed autophagic cell death (3840). mTOR is considered a critical inhibitor protein in the regulation of autophagy; therefore, inhibition of mTOR is associated with triggering autophagy in tumor cells (41). Pathways that regulate mTOR expression include the PI3K/Akt (42), mitogen-activated protein kinase/ERK 1/2 (41) and the AMPK (43) pathways. mTOR activity is stimulated by Akt and ERK 1/2 phosphorylation and inhibited by AMPK phosphorylation. S6 kinase is another essential protein involved in mTOR phosphorylation and it has been suggested that S6 kinase negatively regulates autophagy (44).

The results of the present study indicated that DIOS does not affect Akt, Erk1/2 and AMPK expression, however, it was demonstrated that DIOS treatment decreased the phosphorylation of Akt and Erk1/2 and stimulated AMPK phosphorylation, subsequently inhibiting mTOR expression. Furthermore, DIOS suppressed the phosphorylation of S6 kinase, which also contributed to the inhibition of mTOR phosphorylation. We hypothesize that the DIOS-induced inhibition of mTOR expression and phosphorylation triggered autophagy in HepG2 cells.

Autophagy and apoptosis are important biological processes that maintain cell stability, structure, function and development (45). In the present study, autophagy was blocked by BA1 (41) to investigate whether DIOS-triggered apoptosis was affected. Notably, the inhibition of HepG2 cell proliferation caused by DIOS treatment was partially reversed and the expression of apoptosis-related proteins was altered: p53, Bak and Bax expression decreased, however, Bcl-2 expression increased marginally. Several studies have demonstrated that a number of molecules involved in autophagy are also involved in apoptosis signal transduction (4648). For example, the autophagy-related protein Beclin-1 generates a pro-apoptotic protein fragment, which interacts with the mitochondrial apoptosis pathway via the Bcl-2 family (48). Furthermore, it has been demonstrated that Bcl-2 functions as an anti-apoptotic protein and an anti-autophagy protein via its inhibitory interaction with Beclin-1 (49). However, the exact association between autophagy and apoptosis remains unclear and requires further investigation.

In conclusion, the present study demonstrated that DIOS effectively inhibits proliferation and induces apoptosis in HCC cells, and these effects were a result of autophagy induction via mTOR-related pathways. Therefore, DIOS may present a novel therapeutic treatment for HCC.

Acknowledgements

The present study was supported by The Zhanjiang Key Laboratory of Hepatobiliary Diseases (grant no. 2013A402-4), The Research Funding of Guangdong Medical University (grant no. M2014020) and the Yangfan Plan of Talents Recruitment Grant, Guangdong, China (grant no. YueRenCaiBan [2016] 6).

References

1 

Mu H, Lin KX, Zhao H, Xing S, Li C, Liu F, Lu HZ, Zhang Z, Sun YL, Yan XY, et al: Identification of biomarkers for hepatocellular carcinoma by semiquantitative immunocytochemistry. World J Gastroenterol. 20:5826–5838. 2014. View Article : Google Scholar : PubMed/NCBI

2 

Ferlay J, Soerjomataram I, Dikshit R, Eser S, Mathers C, Rebelo M, Parkin DM, Forman D and Bray F: Cancer incidence and mortality worldwide: Sources, methods and major patterns in GLOBOCAN 2012. Int J Cancer. 136:E359–E386. 2015. View Article : Google Scholar : PubMed/NCBI

3 

Jin Y, Chen J, Feng Z, Fan W, Wang Y, Li J and Tong D: The expression of Survivin and NF-κB associated with prognostically worse clinicopathologic variables in hepatocellular carcinoma. Tumour Biol. 35:9905–9910. 2014. View Article : Google Scholar : PubMed/NCBI

4 

Kim HY and Park JW: Clinical trials of combined molecular targeted therapy and locoregional therapy in hepatocellular carcinoma: Past, present and future. Liver cancer. 3:9–17. 2014. View Article : Google Scholar : PubMed/NCBI

5 

Chen L, Wu LL, Zhang ZL, Hu J, Tang M, Qi CB, Li N and Pang DW: Biofunctionalized magnetic nanospheres-based cell sorting strategy for efficient isolation, detection and subtype analyses of heterogeneous circulating hepatocellular carcinoma cells. Biosens Bioelectron. 85:633–640. 2016. View Article : Google Scholar : PubMed/NCBI

6 

Mano Y, Aishima S, Kubo Y, Tanaka Y, Motomura T, Toshima T, Shirabe K, Baba S, Maehara Y and Oda Y: Correlation between biological marker expression and fluorine-18 fluorodeoxyglucose uptake in hepatocellular carcinoma. Am J Clin Pathol. 142:391–397. 2014. View Article : Google Scholar : PubMed/NCBI

7 

Zhang QY, Li R, Zeng GF, Liu B, Liu J, Shu Y, Liu ZK, Qiu ZD, Wang DJ, Miao HL, et al: Dihydromyricetin inhibits migration and invasion of hepatoma cells through regulation of MMP-9 expression. World J Gastroenterol. 20:10082–10093. 2014. View Article : Google Scholar : PubMed/NCBI

8 

Liu J, Shu Y, Zhang Q, Liu B, Xia J, Qiu M, Miao H, Li M and Zhu R: Dihydromyricetin induces apoptosis and inhibits proliferation in hepatocellular carcinoma cells. Oncol Lett. 8:1645–1651. 2014.PubMed/NCBI

9 

Guglielmotto M, Monteleone D, Piras A, Valsecchi V, Tropiano M, Ariano S, Fornaro M, Vercelli A, Puyal J, Arancio O, et al: Abeta1-42 monomers or oligomers have different effects on autophagy and apoptosis. Autophagy. 10:1827–1843. 2014. View Article : Google Scholar : PubMed/NCBI

10 

Kongsuphol P, Mukda S, Nopparat C, Villarroel A and Govitrapong P: Melatonin attenuates methamphetamine-induced deactivation of the mammalian target of rapamycin signaling to induce autophagy in SK-N-SH cells. J Pineal Res. 46:199–206. 2009. View Article : Google Scholar : PubMed/NCBI

11 

Fu J, Shao CJ, Chen FR, Ng HK and Chen ZP: Autophagy induced by valproic acid is associated with oxidative stress in glioma cell lines. Neuro Oncol. 12:328–340. 2010. View Article : Google Scholar : PubMed/NCBI

12 

Yao KC, Komata T, Kondo Y, Kanzawa T, Kondo S and Germano IM: Molecular response of human glioblastoma multiforme cells to ionizing radiation: Cell cycle arrest, modulation of cyclin-dependent kinase inhibitors, and autophagy. J Neurosurg. 98:378–384. 2003. View Article : Google Scholar : PubMed/NCBI

13 

Ertmer A, Huber V, Gilch S, Yoshimori T, Erfle V, Duyster J, Elsässer HP and Schätzl HM: The anticancer drug imatinib induces cellular autophagy. Leukemia. 21:936–942. 2007.PubMed/NCBI

14 

Wu S, Liu B, Zhang Q, Liu J, Zhou W, Wang C, Li M, Bao S and Zhu R: Dihydromyricetin reduced Bcl-2 expression via p53 in human hepatoma HepG2 cells. PloS one. 8:e768862013. View Article : Google Scholar : PubMed/NCBI

15 

Spanakis M, Kasmas S and Niopas I: Simultaneous determination of the flavonoid aglycones diosmetin and hesperetin in human plasma and urine by a validated GC/MS method: In vivo metabolic reduction of diosmetin to hesperetin. Biomed Chromatogr. 23:124–131. 2009. View Article : Google Scholar : PubMed/NCBI

16 

Chan BC, Ip M, Gong H, Lui SL, See RH, Jolivalt C, Fung KP, Leung PC, Reiner NE and Lau CB: Synergistic effects of diosmetin with erythromycin against ABC transporter over-expressed methicillin-resistant Staphylococcus aureus (MRSA) RN4220/pUL5054 and inhibition of MRSA pyruvate kinase. Phytomedicine. 20:611–614. 2013. View Article : Google Scholar : PubMed/NCBI

17 

Meng JC, Zhu QX and Tan RX: New antimicrobial mono-and sesquiterpenes from Soroseris hookeriana subsp. Planta Med. 66:541–544. 2000. View Article : Google Scholar : PubMed/NCBI

18 

Chandler D, Woldu A, Rahmadi A, Shanmugam K, Steiner N, Wright E, Benavente-García O, Schulz O, Castillo J and Münch G: Effects of plant-derived polyphenols on TNF-alpha and nitric oxide production induced by advanced glycation endproducts. Mol Nutr Food Res. 54:(Suppl 2). S141–S150. 2010. View Article : Google Scholar : PubMed/NCBI

19 

Liao W, Ning Z, Chen L, Wei Q, Yuan E, Yang J and Ren J: Intracellular antioxidant detoxifying effects of diosmetin on 2, 2-azobis(2-amidinopropane) dihydrochloride (aaph)-induced oxidative stress through inhibition of reactive oxygen species generation. J Agric Food Chem. 62:8648–8654. 2014. View Article : Google Scholar : PubMed/NCBI

20 

Androutsopoulos VP, Mahale S, Arroo RR and Potter G: Anticancer effects of the flavonoid diosmetin on cell cycle progression and proliferation of MDA-MB 468 breast cancer cells due to CYP1 activation. Oncol Rep. 21:1525–1528. 2009.PubMed/NCBI

21 

Kabeya Y, Mizushima N, Yamamoto A, Oshitani-Okamoto S, Ohsumi Y and Yoshimori T: LC3, GABARAP and GATE16 localize to autophagosomal membrane depending on form-II formation. J Cell Sci. 117:2805–2812. 2004. View Article : Google Scholar : PubMed/NCBI

22 

Saiki S, Sasazawa Y, Imamichi Y, Kawajiri S, Fujimaki T, Tanida I, Kobayashi H, Sato F, Sato S, Ishikawa K, et al: Caffeine induces apoptosis by enhancement of autophagy via PI3K/Akt/mTOR/p70S6K inhibition. Autophagy. 7:176–187. 2011. View Article : Google Scholar : PubMed/NCBI

23 

Hanahan D and Weinberg RA: Hallmarks of cancer: The next generation. Cell. 144:646–674. 2011. View Article : Google Scholar : PubMed/NCBI

24 

Mauthe M, Jacob A, Freiberger S, Hentschel K, Stierhof YD, Codogno P and Proikas-Cezanne T: Resveratrol-mediated autophagy requires WIPI-1-regulated LC3 lipidation in the absence of induced phagophore formation. Autophagy. 7:1448–1461. 2011. View Article : Google Scholar : PubMed/NCBI

25 

Chen H, Gao Y, Wu J, Chen Y, Chen B, Hu J and Zhou J: Exploring therapeutic potentials of baicalin and its aglycone baicalein for hematological malignancies. Cancer Lett. 354:5–11. 2014. View Article : Google Scholar : PubMed/NCBI

26 

Zhang Q, Liu J, Liu B, Xia J, Chen N, Chen X, Cao Y, Zhang C, Lu C, Li M and Zhu R: Dihydromyricetin promotes hepatocellular carcinoma regression via a p53 activation-dependent mechanism. Sci Rep. 4:46282014.PubMed/NCBI

27 

Zeng G, Liu J, Chen H, Liu B, Zhang Q, Li M and Zhu R: Dihydromyricetin induces cell cycle arrest and apoptosis in melanoma SK-MEL-28 cells. Oncol Rep. 31:2713–2719. 2014.PubMed/NCBI

28 

Liu B, Zhou Z, Zhou W, Liu J, Zhang Q, Xia J, Liu J, Chen N, Li M and Zhu R: Resveratrol inhibits proliferation in human colorectal carcinoma cells by inducing G1/S-phase cell cycle arrest and apoptosis through caspase/cyclin-CDK pathways. Mol Med Rep. 10:1697–1702. 2014.PubMed/NCBI

29 

Androutsopoulos VP, Mahale S, Arroo RR and Potter G: Anticancer effects of the flavonoid diosmetin on cell cycle progression and proliferation of MDA-MB 468 breast cancer cells due to CYP1 activation. Oncol Rep. 21:1525–1528. 2009.PubMed/NCBI

30 

Androutsopoulos VP and Spandidos DA: The flavonoids diosmetin and luteolin exert synergistic cytostatic effects in human hepatoma HepG2 cells via CYP1A-catalyzed metabolism, activation of JNK and ERK and P53/P21 up-regulation. J Nutr Biochem. 24:496–504. 2013. View Article : Google Scholar : PubMed/NCBI

31 

Li G, Ma R, Huang C, Tang Q, Fu Q, Liu H, Hu B and Xiang J: Protective effect of erythropoietin on beta-amyloid-induced PC12 cell death through antioxidant mechanisms. Neurosci Lett. 442:143–147. 2008. View Article : Google Scholar : PubMed/NCBI

32 

Hao C, Gao L, Zhang Y, Wang W, Yu G, Guan H, Zhang L and Li C: Acetylated chitosan oligosaccharides act as antagonists against glutamate-induced pc12 cell death via bcl-2/bax signal pathway. Mar Drugs. 13:1267–1289. 2015. View Article : Google Scholar : PubMed/NCBI

33 

Cheng Y, Ren X, Hait WN and Yang JM: Therapeutic targeting of autophagy in disease: Biology and pharmacology. Pharmacol Rev. 65:1162–1197. 2013. View Article : Google Scholar : PubMed/NCBI

34 

Lorenzi PL, Claerhout S, Mills GB and Weinstein JN: A curated census of autophagy-modulating proteins and small molecules: Candidate targets for cancer therapy. Autophagy. 10:1316–1326. 2014. View Article : Google Scholar : PubMed/NCBI

35 

Zhong W, Zhu H, Sheng F, Tian Y, Zhou J, Chen Y, Li S and Lin J: Activation of the MAPK11/12/13/14 (p38 MAPK) pathway regulates the transcription of autophagy genes in response to oxidative stress induced by a novel copper complex in HeLa cells. Autophagy. 10:1285–1300. 2014. View Article : Google Scholar : PubMed/NCBI

36 

Pan X, Zhang X, Sun H, Zhang J, Yan M and Zhang H: Autophagy inhibition promotes 5-fluorouraci-induced apoptosis by stimulating ROS formation in human non-small cell lung cancer A549 cells. PLoS One. 8:e566792013. View Article : Google Scholar : PubMed/NCBI

37 

Sui X, Chen R, Wang Z, Huang Z, Kong N, Zhang M, Han W, Lou F, Yang J, Zhang Q, et al: Autophagy and chemotherapy resistance: A promising therapeutic target for cancer treatment. Cell Death Dis. 4:e8382013. View Article : Google Scholar : PubMed/NCBI

38 

Yu L, Alva A, Su H, Dutt P, Freundt E, Welsh S, Baehrecke EH and Lenardo MJ: Regulation of an ATG7-beclin 1 program of autophagic cell death by caspase-8. Science. 304:1500–1502. 2004. View Article : Google Scholar : PubMed/NCBI

39 

Amaravadi RK and Thompson CB: The roles of therapy-induced autophagy and necrosis in cancer treatment. Clin Cancer Res. 13:7271–7279. 2007. View Article : Google Scholar : PubMed/NCBI

40 

Yuan J, Lei Z, Wang X, Zhu F and Chen D: Ruthenium complex Λ-WH0402 induces hepatocellular carcinoma LM6 (HCCLM6) cells death by triggering Beclin-1-dependent autophagy pathway. Metallomics. 7:896–907. 2015. View Article : Google Scholar : PubMed/NCBI

41 

Xia J, Guo S, Fang T, Feng D, Zhang X, Zhang Q, Liu J, Liu B, Li M and Zhu R: Dihydromyricetin induces autophagy in HepG2 cells involved in inhibition of mTOR and regulating its upstream pathways. Food Chem Toxicol. 66:7–13. 2014. View Article : Google Scholar : PubMed/NCBI

42 

Niu NK, Wang ZL, Pan ST, Ding HQ, Au GH, He ZX, Zhou ZW, Xiao G, Yang YX, Zhang X, et al: Pro-apoptotic and pro-autophagic effects of the Aurora kinase A inhibitor alisertib (MLN8237) on human osteosarcoma U-2 OS and MG-63 cells through the activation of mitochondria-mediated pathway and inhibition of p38 MAPK/PI3K/Akt/mTOR signaling pathway. Drug Des Devel Ther. 9:1555–1584. 2015.PubMed/NCBI

43 

Shi L, Zhang T, Liang X, Hu Q, Huang J, Zhou Y, Chen M, Zhang Q, Zhu J and Mi M: Dihydromyricetin improves skeletal muscle insulin resistance by inducing autophagy via the AMPK signaling pathway. Mol Cell Endocrinol. 409:92–102. 2015. View Article : Google Scholar : PubMed/NCBI

44 

Shin JH, Min SH, Kim SJ, Kim YI, Park J, Lee HK and Yoo OJ: TAK1 regulates autophagic cell death by suppressing the phosphorylation of p70 S6 kinase 1. Sci Rep. 3:15612013. View Article : Google Scholar : PubMed/NCBI

45 

Zhang J, Ma K, Qi T, Wei X, Zhang Q, Li G and Chiu JF: P62 regulates resveratrol-mediated Fas/Cav-1 complex formation and transition from autophagy to apoptosis. Oncotarget. 6:789–801. 2014. View Article : Google Scholar

46 

Jakhar R, Paul S, Bhardwaj M and Kang SC: Astemizole-Histamine induces Beclin-1-independent autophagy by targeting p53-dependent crosstalk between autophagy and apoptosis. Cancer Lett. 372:89–100. 2016. View Article : Google Scholar : PubMed/NCBI

47 

Rah B, ur Rasool R, Nayak D, Yousuf SK, Mukherjee D, Kumar LD and Goswami A: PAWR-mediated suppression of BCL2 promotes switching of 3-azido withaferin A (3-AWA)-induced autophagy to apoptosis in prostate cancer cells. Autophagy. 11:314–331. 2015. View Article : Google Scholar : PubMed/NCBI

48 

Luo S and Rubinsztein DC: Apoptosis blocks Beclin 1-dependent autophagosome synthesis: An effect rescued by Bcl-xL. Cell Death Differ. 17:268–277. 2010. View Article : Google Scholar : PubMed/NCBI

49 

Pattingre S, Tassa A, Qu X, Garuti R, Liang XH, Mizushima N, Packer M, Schneider MD and Levine B: Bcl-2 antiapoptotic proteins inhibit Beclin 1-dependent autophagy. Cell. 122:927–939. 2005. View Article : Google Scholar : PubMed/NCBI

Related Articles

Journal Cover

December-2016
Volume 12 Issue 6

Print ISSN: 1792-1074
Online ISSN:1792-1082

Sign up for eToc alerts

Recommend to Library

Copy and paste a formatted citation
x
Spandidos Publications style
Liu J, Ren H, Liu B, Zhang Q, Li M and Zhu R: Diosmetin inhibits cell proliferation and induces apoptosis by regulating autophagy via the mammalian target of rapamycin pathway in hepatocellular carcinoma HepG2 cells. Oncol Lett 12: 4385-4392, 2016
APA
Liu, J., Ren, H., Liu, B., Zhang, Q., Li, M., & Zhu, R. (2016). Diosmetin inhibits cell proliferation and induces apoptosis by regulating autophagy via the mammalian target of rapamycin pathway in hepatocellular carcinoma HepG2 cells. Oncology Letters, 12, 4385-4392. https://doi.org/10.3892/ol.2016.5301
MLA
Liu, J., Ren, H., Liu, B., Zhang, Q., Li, M., Zhu, R."Diosmetin inhibits cell proliferation and induces apoptosis by regulating autophagy via the mammalian target of rapamycin pathway in hepatocellular carcinoma HepG2 cells". Oncology Letters 12.6 (2016): 4385-4392.
Chicago
Liu, J., Ren, H., Liu, B., Zhang, Q., Li, M., Zhu, R."Diosmetin inhibits cell proliferation and induces apoptosis by regulating autophagy via the mammalian target of rapamycin pathway in hepatocellular carcinoma HepG2 cells". Oncology Letters 12, no. 6 (2016): 4385-4392. https://doi.org/10.3892/ol.2016.5301