Upregulation of Stat1-HDAC4 confers resistance to etoposide through enhanced multidrug resistance 1 expression in human A549 lung cancer cells

  • Authors:
    • Chutima Kaewpiboon
    • Ratakorn Srisuttee
    • Waraporn Malilas
    • Jeong Moon
    • Sangtaek Oh
    • Hye Gwang Jeong
    • Randal N. Johnston
    • Wanchai Assavalapsakul
    • Young‑Hwa Chung
  • View Affiliations

  • Published online on: November 14, 2014     https://doi.org/10.3892/mmr.2014.2949
  • Pages: 2315-2321
Metrics: Total Views: 0 (Spandidos Publications: | PMC Statistics: )
Total PDF Downloads: 0 (Spandidos Publications: | PMC Statistics: )


Abstract

Despite efforts to develop efficient chemotherapeutic drug strategies to treat cancer, acquired drug resistance is a commonly encountered problem. In the present study, to investigate this phenomenon, human A549 lung cancer cells resistant to the topoisomerase inhibitor etoposide (A549RT‑eto) were used and compared with A549 parental cells. A549RT‑eto cells demonstrated increased resistance to etoposide‑induced apoptosis when compared with A549 parental cells. Notably, A549RT‑eto cells were observed to exhibit greater levels of histone deacetylase 4 (HDAC4), phospho‑Stat1 and P‑glycoprotein [P‑gp; encoded by the multidrug resistance 1 (MDR1) gene], compared with A549 cells. To address whether HDAC4 protein is involved in etoposide resistance in A549 cells, A549RT‑eto cells were treated with trichostatin A (TSA; an HDAC inhibitor) during etoposide treatment. The combined treatment was demonstrated to enhance etoposide‑induced apoptosis and reduce expression levels of HDAC4, P‑gp and phospho‑Stat1. In addition, the suppression of Stat1 with siRNA enhanced etoposide‑induced apoptosis and reduced the expression levels of HDAC4 and P‑gp, suggesting that Stat1 is essential in the regulation of resistance to etoposide, and in the upregulation of P‑gp. Notably, TSA treatment reduced P‑gp transcript levels but Stat1 siRNA treatment did not, suggesting that P‑gp is regulated by HDAC at the transcriptional level and by Stat1 at the post‑transcriptional level. These results suggest that the upregulation of Stat1 and HDAC4 determines etoposide resistance through P‑gp expression in human A549 lung cancer cells.

Introduction

The transcription factor Stat1 is established as an important antiviral agent, acting via IFN-associated intracellular signaling, but evidence has indicated that Stat1 also serves an antioncogenic role. This occurs in part via the upregulation of caspases (1,2) and cyclin-dependent kinase inhibitor 1A (3), by the IFN-regulatory factor 1 (IRF1)/p53 pathway (4) and by the downregulation of Bcl-2 family members (5). By contrast, a number of studies have indicated that in certain cellular contexts, the IFN/Stat1 pathway may facilitate tumor cell growth (6,7). One study reported that resistance to ionizing radiation and IFNs is associated with constitutive overactivity of the IFN/Stat1 pathway in radioresistant tumor cells (6). Previous studies have also demonstrated that constitutive overexpression of Stat1 is positively correlated with the protection of tumor cells from genotoxic stress evoked by treatment with doxorubicin (8) or cisplatin (9). In addition, since overactivity of the IFN/Stat1 pathway is associated with poor prognosis in various types of cancer, IFN-associated genes have been suggested as predictive markers for patients with breast cancer that is resistant to adjuvant chemotherapy (7).

Histone deacetylases (HDACs) are essential for the regulation of the acetylation state of histones, and thus are required for the maintenance and function of chromatin (10). Previous studies suggested that HDACs regulate the acetylation state of various non-histone targets (1113). In particular, HDAC4 (a class IIa HDAC) has been recognized as a notable enzyme, due to its involvement in multiple biological processes (12). One study demonstrated that HDAC4 binds to HIF-1α, in addition to identifying that HDAC4 suppression with siRNA augments HIF-1α acetylation. This leads to destabilization of HIF-1α and downregulation of HIF-1α-targeted gene transcription (11). Another study indicated that HDAC4 directly interacts with and reduces levels of acetylation of FOXO1, leading to the upregulation of FOXO1 transcriptional activity (12). In addition, previous studies demonstrated the ability of HDAC inhibition or suppression as a method to activate Stat3 via acetylation (14,15). However, in ovarian cancer cells resistant to cisplatin, HDAC4 emerges as an activator of Stat1 (13).

Cancer cells commonly acquire anticancer drug resistance during the administration of chemotherapy. To explore one mechanism by which cancer cells develop this resistance, the features of human A549 lung cancer cells resistant to etoposide were investigated. In the current study, Stat1 and HDAC4 were demonstrated to be upregulated and involved in etoposide resistance in A549 cells through P-glycoprotein (P-gp), which is encoded by the multidrug resistance 1 (MDR1) gene. Based on this result, Stat1 and HDAC4 were proposed as potential therapeutic targets for the treatment of chemotherapy-resistant lung cancer cells that overexpress P-gp.

Materials and methods

Cell cultures

A549 cells and etoposide-resistant A459 cells (A549RT-eto) were developed and provided by the Laboratory of Biochemistry at Chulabhorn Research Institute (Bangkok, Thailand), as previously described (16). These cells were cultured in RPMI-1640 medium (Gibco Life Technologies, Grand Island, NY, USA) which was supplemented with 10% fetal bovine serum (FBS), 1% penicillin and 1% streptomycin (all from Gibco Life Technologies) at 37°C in a humidified atmosphere with 5% CO2. Cells were observed under a light microscope (Nikon ELIPSE TS100, Nikon Corporation, Tokyo, Japan).

Reagents and antibodies

Antibodies against phospho-Stat1 (rabbit anti-human polyclonal, #9171) Stat1 (rabbit anti-human polyclonal, #9172), HDAC4 (mouse anti-human monoclonal, #5392), PARP (mouse anti-human monoclonal, #9549), caspase 9 (mouse anti-human monoclonal, #9508) and anti-P-gp (mouse anti-human monoclonal, #517310) were obtained from EMD Millipore, Billerica, MA, USA. Anti-β-actin (mouse anti-human monoclonal, reactive to human, sc-47778) antibodies were also obtained from Santa Cruz Biotechnology, Inc. (Santa Cruz, CA, USA). Trichostatin A (TSA) and etoposide were purchased from Sigma-Aldrich (St. Louis, MO, USA).

Immunoblotting

Lysis buffer (150 mM NaCl; 1% NP-40; 50 mM Tris-HCl, pH 7.5) containing 0.1 mM Na2VO3, 1 mM NaF and Protease Inhibitor Cocktail (Sigma-Aldrich) was used to lyse the cells subsequent to harvest. Proteins from whole cell lysates were resolved using 10 or 12% SDS-PAGE (Bio-Rad, Hercules, CA, USA) using a power-supply (PowerPac, Bio-Rad) and then transferred to nitrocellulose membranes (Whatman, Dassel, Germany) in order to complete the immunoblotting. Primary antibodies were used at a dilution of 1:1,000 (phospho-Stat1 or Stat1) or 1:2,000 (HDAC4, PARP, caspase 9 or P gp), and secondary antibodies (goat anti-mouse Ig, sc-2031, or goat anti-rabbit Ig sc-2030, Santa Cruz Biotechnology, Inc.), which were conjugated with horseradish peroxidase, were used at a dilution of 1:2,000 in 5% nonfat dry milk. Subsequent to final washing with a washing buffer (137 mM NaCl; 0.1% Tween-20; 20 mM Tris-HCl, pH 7.6; Cell Signaling Technology, Inc. Danvers, MA, USA), the nitrocellulose membranes were exposed using the ImageQuant LAS 4000 Mini (GE Healthcare, Cleveland, OH, USA) for an enhanced chemiluminescence assay.

Short interference (si)RNA transfection

Cells were trypsinized (Gibco Life Technologies) and incubated overnight in order to achieve 60–70% confluence prior to transfection with siRNA. Stat1 siRNA [pre-made commercially at Bioneer Corporation, Daejeon, Korea; 200 nM sense, 5′-CUG ACU UCC AUG CGG UUG A(dTdT)-3′ and antisense, 5′-UCA ACC GCA UGG AAG UCA G(dTdT)-3′] or negative control siRNA (Bioneer Corporation) were mixed with Lipofectamine 2000 (Invitrogen Life Technologies, Carlsbad, CA, USA). The cells underwent a 6-h incubation with the transfection mixture and were then rinsed with RPMI-1640 medium containing 10% FBS. The cells were incubated for 48 h prior to harvest.

Reverse transcription-polymerase chain reaction (RT-PCR)

Total RNA was extracted from cells using the RNeasy Mini Kit (Qiagen, Valencia, CA, USA) in accordance with the manufacturer’s instructions. Total RNA (3 μg) was converted into cDNA using SuperScript II Reverse Transcriptase (Invitrogen Life Technologies) and PCR was performed using the following specific primers: Human MDR1 sense, 5′-CCC ATC ATT GCA ATA GCA GG-3′ and antisense, 5′-GTT CAA ACT TCT GCT CCT GA-3′; MRP2 sense, 5′-ACA GAG GCT GGT GGC AAC C-3′ and antisense, 5′-ACC ATT ACC TTG TCA CTG TCC-3′; and breast cancer resistance protein (BCRP) sense, 5′-GAT CAC AGT CTT CAA GGA GAT C-3′ and antisense, 5′-CAG TCC CAG TAC GAC TGT GAC A-3′ (all from Bioneer Corporation). The cDNAs of each sample were diluted and PCR was run at the optimal cycle number (95°C for 1 min, 58°C for 1 min and 72°C for 1 min; 28 cycles for MDR1, and 30 cycles for MRP2 and BCRP). β-Actin mRNA was measured as an internal standard. Subsequent to amplification, the products were subjected to electrophoresis on a 2.0% agarose gel and detected using ethidium bromide (Sigma-Aldrich) staining. Stained band intensity was measured using Multi Gauge software version 2.1 (FujiFilm, Tokyo, Japan).

3-(4,5-dimethylthiazol-2-yl)-2,5-diphenyltetrazolium bromide (MTT) assay

MTT assay (CellTiter 96 Non-Radioactive Cell Proliferation assay; Promega Corporation, Madison, WI, USA) was used to measure cell survival, as previously described (17). Dye solutions containing tetrazolium were added to the cells in the 96-well plate and incubated for 2 h. The absorbance of the formazan produced by living cells was measured at a wavelength of 570 nm (Victor3, PerkinElmer, Waltham, MA, USA). The relative percentage of cell survival was calculated by the mean absorbance of the treated cells (ODT) and the mean absorbance of control cells (ODC) with the following formula: % Cell survival = (ODT/ODC).

Luciferase reporter assay

A549 and A549RT-eto cells were transfected with hMDR1-luciferase (18) or pGL3 empty vector (Promega Corporation) as a control luciferase vector. To normalize transfection efficiency, a pGK-β-gal vector (19) expressing β-galactosidase from a phosphoglucokinase promoter was included in the transfection mixture. At 48 h subsequent to transfection, cells were washed with cold phosphate-buffered saline and lysed in lysis solution [25 mM Tris (pH 7.8), 2 mM EDTA, 2 mM DTT, 10% glycerol, and 1% Triton X-100; Sigma-Aldrich]. Luciferase activity was measured with a luminometer (Lumat LB9507; Berthold Technologies, Oak Ridge, TN, USA) using a luciferase kit (ONE-Glo Luciferase System; Promega Corporation).

Statistical analysis

Data are presented as the mean ± standard deviation. Student’s t-test was used for statistical analysis (SigmaPlot 9, Systat Software Inc., San Jose, CA, USA) and P<0.05 was considered to indicate a statistically significant difference.

Results

A549RT-eto cells exhibit higher levels of HDAC4, phospho-Stat1 and P-gp compared with A549 parental cells

The first experiments aimed to determine the concentration of etoposide required to affect the cell viability of the A549 and A549RT-eto cells. The A549 parental cells were sensitive to growth inhibition at a lower concentration of etoposide (5 μg/ml) while A549RT-eto cells were relatively resistant to growth inhibition (data not shown). The majority of A549 cells died following 24–48 h exposure to 50 μg/ml etoposide, whereas the majority of A549RT-eto cells under similar conditions survived (Fig. 1A). A549 cells were more sensitive to apoptosis during etoposide treatment than A549RT-eto cells, as illustrated by the detection of cleaved PARP, a substrate of active caspase-3 and -7 (Fig. 1B).

Subsequent to confirmation of the resistance of A549RT-eto cells to etoposide, the differences between A549RT-eto and A549 parental cells, which may explain this resistance, were investigated. When expression levels of Stat1, HDAC4 and P-gp were compared, it was noted that the levels of HDAC4 and P-gp proteins were significantly enhanced in A549RT-eto cells compared with those in the A549 parental cells (Fig. 1C). Total Stat1 protein levels in A549RT-eto cells were not identified to be significantly different from those in A549 cells, but the active form of phospho-Stat1 was more strongly expressed in the A549RT-eto cells compared with the corresponding control level (Fig. 1C). Based on these results, the enhanced levels of phospho-Stat1, HDAC4 and P-gp were predicted to be involved in A549 cell etoposide resistance.

HDAC inhibition enhances susceptibility to etoposide in A549RT-eto cells

Enhanced HDAC4 levels in A549RT-eto cells were demonstrated to be involved in etoposide resistance in A549 cells. Thus, A549 and A549RT-eto cells were treated with the HDAC inhibitor TSA (6.25 nM) and cell viability was examined by MTT assay. TSA treatment alone did not influence cell growth in the two cell types (Fig. 2A). As expected, A549 cells exhibited greater sensitivity to etoposide, leading to reduced cell survival compared with the A549RT-eto cells (P<0.05). Additionally, the combined treatment of etoposide and TSA was observed to inhibit cell growth by ~85% and ~65%, in A549 and A549RT-eto cells, respectively.

Cleaved PARP and caspase-9 (indicators of intrinsic apoptotic cell death) were detected during the combined treatments, suggesting that the TSA and etoposide treatment sensitizes A549 and A549RT-eto cells to apoptosis, compared with etoposide or TSA alone (Fig. 2B). In addition, when the protein levels of HDAC4, P-gp and phospho-Stat1 were observed, TSA treatment was demonstrated to reduce HDAC4 and P-gp expression levels in the two types of cell. Protein levels of Stat1 were not significantly altered in A549RT-eto cells, while there was a clear reduction in A549 cell Stat1 protein levels during TSA treatment (Fig. 2B). It was also observed that TSA treatment alone inhibited the activation of phospho-Stat1 in the two cell types. Etoposide treatment alone diminished protein levels of HDAC4 in the two cell lines (Fig. 2B). It was observed that etoposide treatment alone reduced Stat1 protein levels in A549 cells but not in A549RT-eto cells (Fig. 2B). The combined treatment reduced protein levels of HDAC4, P-gp and phospho-Stat1 in A549RT-eto cells. These results suggest that HDAC4 inhibition sensitizes A549RT-eto cells to etoposide-induced apoptosis through a reduction in P-gp protein levels.

Suppression of Stat1 with siRNA enhances susceptibility to etoposide in A549RT-eto cells

Since enhanced levels of phospho-Stat1 were observed in A549RT-eto cells, it was hypothesized that elevated Stat1 may be involved in resistance to etoposide. To assess this, Stat1 siRNA was introduced to suppress Stat1 levels in A549RT-eto cells. The optimal Stat1 siRNA concentration for the suppression of Stat1 expression was identified according to the results obtained from transfection using Lipofectamine 2000, and according to the results, 100 nM Stat1 siRNA was used (data not shown). Suppression of Stat1 was observed to induce a reduction in HDAC4 and P-gp expression levels, indicating that HDAC4 and Stat1 cross-talk with each other (Fig. 3A). Etoposide treatment alone reduced HDAC4, P-gp and phospho-Stat1 protein levels, but was not sufficient to induce significant apoptosis in A549RT-eto cells (Fig. 3B). The combined treatment with Stat1 siRNA and etoposide induced a reduction of HDAC4, P-gp and phospho-Stat1 protein levels (Fig. 3A), resulting in the acceleration of apoptosis by the activation of caspase-3, -7 and -9 in A549RT-eto cells. Together, these results suggest that Stat1 is involved in P-gp expression, resulting in resistance to etoposide.

Etoposide resistance is attributed to the enhancement of MDR1 transcript levels and its transcriptional activity in A549RT-eto cells

Other genes, such as those encoding MRP2 and BCRP, are established to be involved in drug resistance (20,21). Therefore, the levels of these transcripts (MDR1, MRP2 and BCRP genes) were examined in A549 and A549RT-eto cells in the present study. A549RT-eto cells were observed to exhibit increased levels of MDR1 mRNA, whereas low levels were observed in A549 cells (Fig. 4A). However, levels of MRP2 and BCRP transcript were similar between A549RT-eto and A549 parental cells. This result indicates that the MDR1 gene is specifically activated to create etoposide resistance in A549 cells. Furthermore, MDR1 promoter activity was examined in A549RT-eto and A549 cells. An increase in MDR1 promoter activity was observed in A549RT-eto cells compared with that of the A549 parental cells (Fig. 4B), in concordance with the higher levels of MDR1 mRNA observed in A549RT-eto cells.

TSA treatment inhibits P-gp expression at the transcriptional level in A549RT-eto cells but Stat1 siRNA treatment does not

Next, the effect of TSA or Stat1 siRNA treatment on MDR1, MRP2 and BCRP mRNA levels in A549RT-eto cells was investigated. TSA and etoposide treatments alone reduced transcriptional levels of MDR1, while neither treatment reduced the levels of MRP2 or BCRP (Fig. 5A). The combined treatment of TSA and etoposide also reduced transcriptional levels of MDR1. When Stat1 siRNA was administered to the A549RT-eto cells, there was no reduction in MDR1 mRNA levels, yielding results similar to those treated with the control siRNA (Fig. 5B). The combined treatment of Stat1 siRNA and etoposide significantly reduced MDR1 mRNA levels due to the action of etoposide. Based on these results, it can be theorized that HDAC4 affects the expression of P-gp protein at the transcriptional level, while Stat1 influences its expression at the post-transcriptional level.

Discussion

The majority of patients with non-small cell lung cancer display intrinsic chemoresistance, which limits the possibility of successful treatment with chemotherapy (22). The most prevalent form observed in these patients is multidrug resistance, which has been associated with the overexpression of the ATP-binding cassette superfamily of transporters, including P-gp and MRP (23,24). In the current study, A549 cells with acquired etoposide resistance were observed to specifically exhibit the upregulation of P-gp but not other MDR-associated genes, such as MRP2 and BCRP, which are frequently overexpressed in patients with cancer that exhibits chemotherapeutic resistance. In addition, human H460 lung cancer cells resistant to etoposide have exhibited overexpression of lung resistance protein (25). It is thus concluded that the upregulation of various specific MDR gene family members is associated with the context of the cell origin and chemotherapeutic drug exposure.

In the present study, it has been reported that the expression of P-gp is modulated by HDAC4 and Stat1. Suppression of HDAC4 activity and Stat1 levels induced a reduction of P-gp expression levels, leading to sensitization to apoptosis, suggesting that HDAC4 and Stat1 are responsible for etoposide resistance. Notably, it was also observed that inhibition of HDAC4 activity by TSA reduced Stat1 activity, indicating that HDAC4 may regulate Stat1 activity. The results of the current study are supported by another that suggested that HDAC4 interacts with Stat1, resulting in a reduction of Stat1 acetylation, which eventually enhances Stat1 phosphorylation in cisplatin-resistant cancer cells (13). Furthermore, when expression of Stat1 was suppressed by its siRNA, HDAC4 protein levels were also diminished in etoposide-resistant A549 cells, leading to sensitization of etoposide-induced apoptosis through the downregulation of P-gp. Based on these results, it is proposed that Stat1 and HDAC4 coregulate each other.

However, it was observed that the transcript levels of P-gp were reduced during Stat1 siRNA treatment. Thus, it is hypothesized that the suppression of Stat1 may influence protein levels of P-gp at a post-transcriptional level, or indirectly through other proteins. Supporting this hypothesis, a previous study demonstrated that FBXO15/Fbx15, an F-box protein in the ubiquitin E3 ligase complex, regulates P-gp expression levels through the ubiquitin-proteosome pathway (26). However, it remains unclear how Stat1 suppression is associated with the upregulation of FBXO15/Fbx15. Thus, future studies should focus upon the investigation of the detailed mechanisms by which the suppression of Stat1 induces a reduction in protein levels of P-gp.

Acknowledgements

The current study was supported by a grant from the World Class University Program (R31-2008-000-20004-0) through the National Research Foundation funded by the Korean government; the Office of the Higher Education Commission, Thailand, under the Strategic Scholarships Fellowships Frontier Research Networks (specifically for the southern region) for the Joint PhD Thai Doctoral Degree Program, a CHE-SSR-PhD SW Scholarship to Ms. Chutima Kaewpiboon.

References

1 

Chin YE, Kitagawa M, Kuida K, Flavell RA and Fu XY: Activation of the STAT signaling pathway can cause expression of caspase 1 and apoptosis. Mol Cell Biol. 17:5328–5337. 1997.PubMed/NCBI

2 

Kumar A, Commane M, Flickinger TW, Horvath CM and Stark GR: Defective TNF-alpha-induced apoptosis in STAT1-null cells due to low constitutive levels of caspases. Science. 278:1630–1632. 1997. View Article : Google Scholar : PubMed/NCBI

3 

Chin YE, Kitagawa M, Su WC, You ZH, Iwamoto Y and Fu XY: Cell growth arrest and induction of cyclin-dependent kinase inhibitor p21 WAF1/CIP1 mediated by STAT1. Science. 272:719–722. 1996. View Article : Google Scholar : PubMed/NCBI

4 

Townsend PA, Scarabelli TM, Davidson SM, Knight RA, Latchman DS and Stephanou A: STAT-1 interacts with p53 to enhance DNA damage-induced apoptosis. J Biol Chem. 279:5811–5820. 2004. View Article : Google Scholar

5 

Stephanou A, Brar BK, Knight RA and Latchman DS: Opposing actions of STAT-1 and STAT-3 on the Bcl-2 and Bcl-x promoters. Cell Death Differ. 7:329–330. 2000. View Article : Google Scholar : PubMed/NCBI

6 

Khodarev NN, Beckett M, Labay E, Darga T, Roizman B and Weichselbaum RR: STAT1 is overexpressed in tumors selected for radioresistance and confers protection from radiation in transduced sensitive cells. Proc Natl Acad Sci USA. 101:1714–1719. 2004. View Article : Google Scholar : PubMed/NCBI

7 

Weichselbaum RR, Ishwaran H, Yoon T, et al: An interferon-related gene signature for DNA damage resistance is a predictive marker for chemotherapy and radiation for breast cancer. Proc Natl Acad Sci USA. 105:18490–18495. 2008. View Article : Google Scholar : PubMed/NCBI

8 

Fryknäs M, Dhar S, Oberg F, et al: STAT1 signaling is associated with acquired crossresistance to doxorubicin and radiation in myeloma cell lines. Int J Cancer. 120:189–195. 2007. View Article : Google Scholar

9 

Roberts D, Schick J, Conway S, et al: Identification of genes associated with platinum drug sensitivity and resistance in human ovarian cancer cells. Br J Cancer. 92:1149–1158. 2005. View Article : Google Scholar : PubMed/NCBI

10 

Yang XJ and Seto E: HATs and HDACs: from structure, function and regulation to novel strategies for therapy and prevention. Oncogene. 26:5310–5318. 2007. View Article : Google Scholar : PubMed/NCBI

11 

Geng H, Harvey CT, Pittsenbarger J, et al: HDAC4 protein regulates HIF1α protein lysine acetylation and cancer cell response to hypoxia. J Biol Chem. 286:38095–38102. 2011. View Article : Google Scholar : PubMed/NCBI

12 

Mihaylova MM, Vasquez DS, Ravnskjaer K, et al: Class IIa histone deacetylases are hormone-activated regulators of FOXO and mammalian glucose homeostasis. Cell. 145:607–621. 2011. View Article : Google Scholar : PubMed/NCBI

13 

Stronach EA, Alfraidi A, Rama N, et al: HDAC4-regulated STAT1 activation mediates platinum resistance in ovarian cancer. Cancer Res. 71:4412–4422. 2011. View Article : Google Scholar : PubMed/NCBI

14 

Sun Y, Chin YE, Weisiger E, et al: Cutting edge: Negative regulation of dendritic cells through acetylation of the nonhistone protein STAT-3. J Immunol. 182:5899–5903. 2009. View Article : Google Scholar : PubMed/NCBI

15 

Yuan ZL, Guan YJ, Chatterjee D and Chin YE: Stat3 dimerization regulated by reversible acetylation of a single lysine residue. Science. 307:269–273. 2005. View Article : Google Scholar : PubMed/NCBI

16 

Kanintronkul Y, Worayuthakarn R, Thasana N, et al: Overcoming multidrug resistance in human lung cancer with novel benzo[a]quinolizin-4-ones. Anticancer Res. 31:921–927. 2011.PubMed/NCBI

17 

Kaewpiboon C, Lirdprapamongkol K, Srisomsap C, et al: Studies of the in vitro cytotoxic, antioxidant, lipase inhibitory and antimicrobial activities of selected Thai medicinal plants. BMC Complement Altern Med. 12:2172012. View Article : Google Scholar : PubMed/NCBI

18 

Kim HG, Hien TT, Han EH, et al: Metformin inhibits P-glycoprotein expression via the NF-κB pathway and CRE transcription activity through AMPK activation. Br J Pharmacol. 162:1096–1108. 2011. View Article : Google Scholar :

19 

Cho IR, Jeong S, Jhun BH, et al: Activation of non-canonical NF-kappaB pathway mediated by STP-A11, an oncoprotein of Herpesvirus saimiri. Virology. 359:37–45. 2007. View Article : Google Scholar

20 

Sugimoto Y, Tsukahara S, Ishikawa E and Mitsuhashi J: Breast cancer resistance protein: molecular target for anticancer drug resistance and pharmacokinetics/pharmacodynamics. Cancer Sci. 96:457–465. 2005. View Article : Google Scholar : PubMed/NCBI

21 

Young LC, Campling BG, Cole SP, Deeley RG and Gerlach JH: Multidrug resistance proteins MRP3, MRP1, and MRP2 in lung cancer: correlation of protein levels with drug response and messenger RNA levels. Clin Cancer Res. 7:1798–1804. 2001.PubMed/NCBI

22 

Ihde DC and Minna JD: Non-small cell lung cancer. Part II: Treatment. Curr Probl Cancer. 15:105–154. 1991.PubMed/NCBI

23 

Borst P, Evers R, Kool M and Wijnholds J: A family of drug transporters: the multidrug resistance-associated proteins. J Natl Cancer Inst. 92:1295–1302. 2000. View Article : Google Scholar : PubMed/NCBI

24 

Chan HS, Lu Y, Grogan TM, et al: Multidrug resistance protein (MRP) expression in retinoblastoma correlates with the rare failure of chemotherapy despite cyclosporine for reversal of P-glycoprotein. Cancer Res. 57:2325–2330. 1997.PubMed/NCBI

25 

Lee E and Lim SJ: The association of increased lung resistance protein expression with acquired etoposide resistance in human H460 lung cancer cell lines. Arch Pharm Res. 29:1018–1023. 2006. View Article : Google Scholar : PubMed/NCBI

26 

Katayama K, Noguchi K and Sugimoto Y: FBXO15 regulates P-glycoprotein/ABCB1 expression through the ubiquitin-proteasome pathway in cancer cells. Cancer Sci. 104:694–702. 2013. View Article : Google Scholar : PubMed/NCBI

Related Articles

Journal Cover

March-2015
Volume 11 Issue 3

Print ISSN: 1791-2997
Online ISSN:1791-3004

Sign up for eToc alerts

Recommend to Library

Copy and paste a formatted citation
x
Spandidos Publications style
Kaewpiboon C, Srisuttee R, Malilas W, Moon J, Oh S, Jeong HG, Johnston RN, Assavalapsakul W and Chung YH: Upregulation of Stat1-HDAC4 confers resistance to etoposide through enhanced multidrug resistance 1 expression in human A549 lung cancer cells. Mol Med Rep 11: 2315-2321, 2015
APA
Kaewpiboon, C., Srisuttee, R., Malilas, W., Moon, J., Oh, S., Jeong, H.G. ... Chung, Y. (2015). Upregulation of Stat1-HDAC4 confers resistance to etoposide through enhanced multidrug resistance 1 expression in human A549 lung cancer cells. Molecular Medicine Reports, 11, 2315-2321. https://doi.org/10.3892/mmr.2014.2949
MLA
Kaewpiboon, C., Srisuttee, R., Malilas, W., Moon, J., Oh, S., Jeong, H. G., Johnston, R. N., Assavalapsakul, W., Chung, Y."Upregulation of Stat1-HDAC4 confers resistance to etoposide through enhanced multidrug resistance 1 expression in human A549 lung cancer cells". Molecular Medicine Reports 11.3 (2015): 2315-2321.
Chicago
Kaewpiboon, C., Srisuttee, R., Malilas, W., Moon, J., Oh, S., Jeong, H. G., Johnston, R. N., Assavalapsakul, W., Chung, Y."Upregulation of Stat1-HDAC4 confers resistance to etoposide through enhanced multidrug resistance 1 expression in human A549 lung cancer cells". Molecular Medicine Reports 11, no. 3 (2015): 2315-2321. https://doi.org/10.3892/mmr.2014.2949