Suppression of Dicer Increases Sensitivity to Gefitinib in Human Lung Cancer Cells Jui-Chieh Chen, PhD1, Yen-Hao Su, MD2,3, Yang-Hao Yu, MD, PhD4,5, Yi-Wen Chang, PhD6,7, Ching-Feng Chiu, PhD1, Chi-Feng Tseng, MS1, Hsin-An Chen, MD, PhD2,3, and Jen-Liang Su, PhD1,8,9,10 1 National Institute of Cancer Research, National Health Research Institutes, Zhunan, Taiwan; 2Graduate Institute of Clinical Medicine, College of Medicine, Taipei Medical University, Taipei, Taiwan; 3Department of Surgery, Division of General Surgery, Shuang Ho Hospital, Taipei Medical University, Taipei, Taiwan; 4 Department of Internal Medicine, Divisions of Pulmonary and Critical Care Medicine, China Medical University Hospital, Taichung, Taiwan; 5Graduate Institute of Clinical Medical Science, China Medical University, Taichung, Taiwan; 6Graduate Institute of Biochemistry and Molecular Biology, National Yang-Ming University, Taipei, Taiwan; 7Genomics Research Center, Academia Sinica, Taipei, Taiwan; 8 Graduate Institute of Cancer Biology, China Medical University, Taichung, Taiwan; 9 Department of Biotechnology, Asia University, Taichung, Taiwan; 10 Center for Molecular Medicine, China Medical University Hospital, Taichung, Taiwan 1 Correspondence author: Jen-Liang Su, PhD; National Institute of Cancer Research, National Health Research Institutes, No.35 Keyan Road, Zhunan, Miaoli County 35053, Taiwan; e-mail: jlsu@nhri.org.tw; tel: + 886-37-246-166 (ext. 35142); fax: + 886-37-586-401 The running head: Dicer regulate gefitinib sensitivity in NSCLC Disclosure of potential conflict of interest: No potential conflicts of interest were disclosed. 2 SYNOPSIS In the present study, we found that Dicer-upregulated miR-30b/c and miR-221/222 promote gefitinib resistance of NSCLC cells by downregulation of caspase-3. We speculate that suppression of Dicer might have a potential for future applications in gefitinib resistance of NSCLCs. 3 ABSTRACT Background. Accumulating evidence is revealing an important role of microRNA (miRNA) in tumor progression and chemotherapeutic resistance. Dicer is a cytoplasmic endoribonuclease Type III crucial for production of mature microRNAs. The aberrant expression of Dicer has also been reportedly associated with clinical aggressiveness, prognosis and patient survival in various cancer types. However, the molecular mechanisms of Dicer in acquired gefitinib resistance are still not clear. Methods. In this study, we analyzed the protein level of Dicer between gefitinib-sensitive (PC9) and gefitinib-resistant (PC9/GR) non-small cell lung cancer (NSCLC) cell lines by Western blot analysis. Silence and overexpression of the Dicer were respectively performed to investigate the effects on gefitinib sensitivity, as assessed by MTT assay and sub-G1 assay of flow cytometry. To further explore the mechanism of chemoresistance, we examined whether Dicer knockdown leading to modulating specific miRNAs and and its microRNA target genes. Results. Dicer expression was significantly increased in PC9/GR compared with PC9 cells. Knockdown of Dicer restores gefitinib sensitivity in resistant cells, and overexpression of Dicer enhances resistance to gefitinib in sensitive cells. Silencing of Dicer induces sensitive to gefitinib in NSCLC cells through the down-regulation of miR-30b/c and miR-221/222 to increase the protein level of caspase-3 resulting in an 4 increase in gefitinib-induced apoptosis. Conclusions. Dicer contributes to the resistance to gefitinib in lung cancer. These results indicate that Dicer may be a potential target for diagnosis and therapy of patients with resistance to gefitinib. 5 INTRODUCTION Global cancer statistics indicate that lung cancer is the leading cause of cancer deaths worldwide.1 Approximately 85% of all lung cancers are non-small cell lung cancers (NSCLCs), which include adenocarcinoma, squamous cell carcinoma and large cell carcinoma.2 Gefitinib, an EGFR tyrosine kinase inhibitor (TKI), is a commonly used therapeutic agent for advanced NSCLCs.3,4 Nevertheless, despite a positive response to initial treatment, many patients eventually develop resistance to gefitinib after varying periods of time.5 Therefore, novel approaches to the treatment of NSCLCs must be developed. Micro-RNAs (miRNAs) are a class of endogenous non-coding RNAs consisting of approximately 22 nucleotides in length and are capable of repressing gene expression by binding to their complementary sequence in the 3' untranslated region (UTR) of mRNA targets.6 MiRNA biogenesis is required for sequential processing by Drosha, AGO, and Dicer.7 Recently, emerging evidence is revealing an important role of miRNAs in anticancer drug resistance and miRNA may be a target for the development of novel therapeutic strategies to overcome drug resistance.8-11 Dicer is cytoplasmic RNase III-type endoribonuclease that processes precursor microRNAs into mature microRNAs. Deregulated expression of Dicer has been reportedly associated with drug resistance in different cancer types.12-16 Nevertheless, 6 whether or not Dicer is involved in the gefitinib resistance of NSCLC cells has not yet been reported. A previous study indicated that epidermal growth factor receptor (EGFR) and MET (the receptor for hepatocyte growth factor) could induce gefitinib resistance in NSCLCs via upregulation of miR-30b, miR-30c, miR-221, and miR-222. In addition, the downregulation of miR-103 in NSCLCs has also been shown to increase these microRNAs production to induce resistance to gefitinib treatment.17 A recent report showed that miR-103 could attenuate miRNA biosynthesis by targeting Dicer.18 In the present study, we speculate that expressions of Dicer might play important role in the development of gefitinib resistance in NSCLCs. 7 MATERIALS AND METHODS Reagents and antibodies Gefitinib was obtained from AstraZeneca Pharmaceuticals (Wilmington, DE). FuGENE 6 was purchased from Roche (Basel, CH). Polyethyleneimine (PEI), polybrene, RNase A, propidium iodide, protease inhibitor cocktail and 3-(4,5-dimethylthiazol-2-yl)-2,5-diphenyltetrazolium bromide (MTT) were purchased from Sigma-Aldrich (St Louis, MO). The caspase-3 inhibitor Z-DEVD-FMK was purchased from R&D Systems Inc. (Minneapolis). All cell culture-related reagents were purchased from Invitrogen (Carlsbad, CA). The sources of primary antibodies were as follows: AGO2, Dicer and Drosha, were purchased from Abcam Ltd. (Cambridge, UK). MET was purchased from Santa Cruz Biotechnology (Santa. Cruz, CA). Cleaved caspase-3, EGFR and pEGFR were from Cell Signaling Technology (Beverly, MA). Anti-α-tubulin antibody was purchased from Sigma. All secondary antibodies were purchased from Jackson Immuno Research (West Grove, PA). Cell culture Cells were grown in RPMI (PC9/WT, PC9/GR) or DMEM (A549, H1299) supplemented with 10% (vol/vol) FBS, 2 mM of L-glutamine and 100 units/ml penicillin/streptomycin at 37°C in a humidified atmosphere with 5% CO2. In order to generate a resistant cell, PC9 cells were initially treated with 0.2 μM gefitinib for 48 h. 8 After exposure to gefitinib, cells were washed and cultured in drug-free medium. The surviving cells were continuously exposed to increasing dosages and finally to a concentration of 10 μM. PC9 resistant cells were termed PC9/GR. For drug treatment studies, gefitinib was diluted in serum free medium and added to cultures to give the desired final concentrations. Cytotoxicity assays MTT assay was used to evaluate relative cell viability. For in vitro studies with a caspase-3 inhibitor, cells were pre-incubated with 10 μM of Z-DEVD-FMK at 37°C for 90 min, followed by treatment with 20 μM gefitinib. Absorbance of the dye was measured at a wavelength of 570 nm with background subtraction at 630 nm. Western blot Cells were lysed in RIPA buffer containing protease inhibitor cocktail. Protein concentration was determined by the BCA assay (Pierce). The equal amount of total protein was resolved by SDS-PAGE and transferred to PVDF membranes (Millipore, Bedford, MA). Membranes were blocked with tris-buffered saline/0.1% Tween 20 (TBST) containing 5% skim milk and then incubated overnight at 4°C with the indicated primary antibodies. Membranes were washed with TBST and incubated for 1 h at room temperature with the appropriate secondary antibodies conjugated to horseradish peroxidase. Membranes were then washed and bound antibodies were 9 visualized using ECL reagents (PerkinElmer, MA) and autoradiography. Semi-quantitative analysis of band intensities was performed by densitometry using image analysis software ImageJ. Lentivirus infection and shRNA knockdown The pLKO.1-puro-based lentiviral vectors: TRCN0000290426 (shDicer#1) and TRCN0000290489 (shDicer#2), and the pLKO.1-shscramble were purchased from the National RNAi Core Facility at Academia Sinica, Taipei, Taiwan. Recombinant lentiviruses were packaged according to the manufacturer's instruction. Cultured cells were incubated with lentivirus containing 8 μg/ml polybrene for 24 h, replaced fresh medium and incubated for another 48 h. For stable cell lines, cells were selected by puromycin. RNA isolation, reverse transcription and real-time PCR quantification Total RNA was isolated using Trizol reagent (Invitrogen) and reverse transcribed into cDNA using M-MLV reverse transcriptase (Invitrogen) according to the manufacturer's instructions. Real-time PCR was performed using the Roche LightCycler 480 (Roche). The relative mRNA levels of the target gene were normalized to the mean of internal GAPDH. The mature miRNA sequences were obtained from the Sanger Center miRNA Registry and the stem-loop RT primers were designed according to Chen and Ridzon et al.19 Expression levels of miRNA genes 10 were based on the amount of the target message relative to that of the RNU6B transcript, used as a control to normalize the initial input of total RNA. The relative levels of gene expression were represented as ΔCt = Ct gene – Ct reference, and the fold change of gene expression was calculated by the 2−ΔΔCt Method. All primers sequences are shown in Supplemental Table 1. Cloning and expression of Dicer in PC9 cells The PCR products were digested with BamH I and EcoR I and cloned into pcDNA6.1 to generate pcDN6.1-Dicer plasmid. Empty pcDNA6.1 plasmid was used as control. Cells were transiently transfected using FuGENE 6 as suggested by the manufacturer. Stable clones were selected for blasticidin resistance using standard protocols. Gefitinib-induced cell death Aliquots of 1 × 106 cells were collected and washed once with PBS and then fixed with 70% ethanol overnight. Fixed cells were then washed twice in PBS and stained with 1 µg/ml propidium iodide in PBS containing 10 µg/ml RNase A for 1 h. The DNA content was measured by flow cytometry. For all assays, 10,000 cells were counted. 11 RESULTS Characterization of PC9 and PC9/GR cells To study molecular mechanisms of acquired gefitinib resistance in NSCLCs, we established a gefitinib-resistant PC9 subline designated PC9/GR. After isolating, PC9/GR cells were not growth inhibited by exposure to gefitinib at concentrations up to 7.5 μM (IC50 = 19.64 μM), whereas PC9 cells were very sensitive to gefitinib, with an IC50 value of 0.009974 μM (Fig. 1a). As shown in Fig. 1b, expression of Dicer, but not Drosha or AGO2, was higher in PC9/GR cells than PC9 cells. When serially passed in the absence of gefitinib for 25 passages, PC9/GR cells regained partial sensitivity to gefitinib corresponded with a decrease in the expression of Dicer (Fig. 1c). In a previous study, EGFR and MET were addressed to induce gefitinib resistance in lung cancers through regulation of expression of specific miRNAs. Consistent with the previous study, we observed an increase in expression levels of EGFR and MET in lysates from PC9/GR cells treated with gefitinib. However, EGFR and MET were not obviously increased in PC9 cells at the same treatment (Fig. 1d). Knockdown of Dicer restores gefitinib sensitivity in gefitinib-resistant cells To clarify whether Dicer expression is associated with sensitivity to gefitinib in PC9/GR cells, we knocked down Dicer expression by lentivirus-mediated delivery of Dicer shRNA. Knockdown efficiency of Dicer was determined by q-PCR (Fig. 2a) 12 and Western blot (Fig. 2b). As shown in Fig. 2c, silencing Dicer expression resulted in elevated sensitivity to gefitinib in a dose-dependent fashion, suggesting that Dicer is a key regulator of drug resistance. Furthermore, knockdown of Dicer in PC9/GR had significantly higher proportion of apoptotic cells under treatment with gefitinib compared with PC9/GR/shScramble cells, as demonstrated by an increased sub-G1 fraction (Fig. 2d). To extend our observations, we introduced specific shRNAs to silence Dicer in A549 and H1299 cells, which are insensitive to gefitinib in vitro (IC50 > 9 μM), and the expression of Dicer was further validated by Western blot analysis (Fig. 3a, d). The results revealed that knockdown of Dicer conferred increased sensitivity to gefitinib compared with control shRNA (shScramble), as assessed by MTT assay (Fig. 3b, e). Moreover, gefitinib-induced apoptotic population was also increased significantly in A549/shDicer cells compared with A549/shScramble cells, as evidenced by an increased sub-G1 fraction (Fig. 3c). Overexpression of Dicer enhances resistance to gefitinib in gefitinib-sensitive cells Next, to confirm whether Dicer expression mediates gefitinib sensitivity, we generated stable PC9 cells that expressed either control vector or Dicer expression vector. The Dicer expression level was analyzed by q-PCR (Fig. 4a) and Western blot (Fig. 4b). The results revealed that overexpression of Dicer in PC9 significantly increased resistance to gefitinib, as assessed by MTT assay (Fig. 4c). HCC827 cells, 13 which are sensitive to gefitinib in vitro, were also transfected with Dicer-expressing vectors or control vectors, and Dicer expression was determined by Western blot (Fig. 4d). A similar result is obtained in Dicer stably overexpressed HCC827 cells that render cells more resistant to gefitinib compared with cells transfected with a control vector (Fig. 4e). Silencing of Dicer expression by shRNA suppresses miR-30b/c and miR-221/222 in human NSCLC cells Previous research had shown that miR-30b/c and miR-221/222 involved in gefitinib resistance, we investigated whether Dicer could modulate amounts of these miRNAs in NSCLC cells. Analysis of four miRNAs from Dicer-silenced PC9/GR cells revealed reduced amounts of mature miRNAs when compared with PC9/GR/shScramble cells (Fig. 5a). Similar results were also found in Dicer-silenced A549 cells (Fig. 5b). These results indicated that Dicer is required for maintenance of the specific miRNAs expression level, which involved in resistance to gefitinib in lung cancer. Identification of a potential target of miR-30b/c and miR-221/222 involved in gefitinib resistance To identify cellular targets of miR-30b/c and -221/222, we performed two independent algorithms, including miRanda and TargetScan. We found that 14 miRs-221/222 and -30b/30c are partially complementary to regions in the 3’UTR of caspase-3 (Fig. 5c). In addition, we observed a marked up-regulation of the protein level of cleaved caspase-3 in gefitinib-treated PC9/GR/shDicer cells compared with PC9/GR/shScramble cells (Fig. 5d), suggesting that depletion of Dicer reduces the expression of miR-30b/c and miR-221/222 resulting in elevated protein levels of cleaved caspase-3 to induce apoptosis and increase gefitinib sensitivity. To further confirm that gefitinib-induced activation of caspase-3 is involved in the induction of apoptotic cell death, the PC9/GR/shDicer cells were pre-treated with the caspase-3 inhibitor (z-DEVD-fmk) and apoptotic cells were analyzed using flow cytometry. As shown in Fig. 5e, pre-treatment of PC9/GR/shDicer cells with the caspase-3 inhibitor significantly blocked the gefitinib-induced apoptosis of these cells. Taken together, these results suggest that Dicer knockdown induces sensitive to gefitinib treatment in NSCLC cells through the down-regulation of miR-30b/c and miR-221/222, and elevation of these miRNAs target, caspase-3, leading to an increase in sensitivity to gefitinib-induced apoptosis. 15 DISCUSSION In this study, we investigated whether altered Dicer expression affects sensitivity to gefitinib in human lung cancer cells. Here, our results indicate that miR-30b/c and miR-221/222 are downregulated after Dicer silencing, which induce apoptosis in gefitinib-resistant NSCLC cells by increases in the amount and activity of caspase-3. Although a previous study has demonstrated that the reduced expression of Dicer in a significant fraction of lung cancers is associated with shorter postoperative survival,20 it remains unknown whether Dicer is sufficient to make lung cancer cells resistant to gefitinib and is correlated significantly with clinical response and survival following gefitinib treatment in patients. Deregulated expression of Dicer has been reportedly associated with drug resistance in different cancer types. In human breast carcinoma cells, a study showed that knockdown of Dicer in MCF-7 cells leads to significant G1 arrest and increased sensitivity to cisplatin.12 Moreover, Tamoxifen (Tam) is the most prescribed hormonal agent for the treatment of breast cancer patients with estrogen receptor α (ERα)-positive, but acquired Tam resistance frequently occur. Dicer overexpression leads to an increase in the breast cancer resistance protein (BCRP) resulting in Tam resistance in MCF-7 cells.14 With regard to the roles of Dicer in the relationship between tumor cells and immune cells, Dicer is responsible for the generation of the 16 mature miR-222 and -339 to suppress ICAM-1 (intercellular cell adhesion molecule) expression on tumor cells, which promotes resistance of cancer cells to be killed by cytotoxic T-lymphocytes (CTLs). However, disruption of Dicer leading to up-regulation of ICAM-1 can enhance the susceptibility of tumor cells to antigen specific lysis by CTLs.13 A recent study has also shown that decreasing the expression of Dicer through specific RNAi can sensitize resistant adenocarcinoma cells to cisplatin.15 Accumulating evidence suggests that Dicer depletion leads to the general decrease in the amount of mature miRNAs.21,22 It has also been observed that miRNA expression in normal tissues is globally higher than in tumors. Moreover, poorly differentiated tumors had lower global levels of miRNA expression compared with the more-differentiated samples.23 This might potentially account for the generation or maintenance of cancer stem cells, recently proposed to be responsible for the source of resistance to gefitinib therapy in lung cancer.24 A study has reported microRNA-128b is a regulator of EGFR in NSCLC cell lines. The loss of heterozygosity of the microRNA-128b is frequent in tumor samples and correlated significantly with clinical response and survival after gefitinib treatment.25 Moreover, Dicer expression was found inversely correlated with expression levels of mature let-7 in a panel of human cancer cell lines.26 Reduced 17 let-7 significantly over-expressed the expression of Dicer resulting in increased cell proliferation of oral cancer cells.27 Interestingly, a study has shown that overexpression of let-7 in lung adenocarcinoma cells inhibited cancer cell growth in vitro.28 Furthermore, our study has revealed that knockdown of Dicer in cultured NSCLC cells triggers activation of caspase-3 and leads to increased sensitivity to gefitinib. Caspase-3 is a cysteine protease that plays a critical role in the process of apoptotic cell death. Previously, the caspase-3 was also reported to be associated with chemotherapeutic resistance in cancer. Overexpression of caspase-3 restores sensitivity for drug-induced apoptosis in cancer cells with acquired drug resistance.29,30 Conversely, loss of caspase-3 expression may render cancer cells resistant to chemotherapy.31,32 In NSCLC patients, caspase-3-positive tumors had a tendency for the most favorable prognosis.33 Moreover, a member of the inhibitor of apoptosis family of proteins, survivin, has been shown to inhibit caspases and to prevent caspase mediated cell death. Targeting of survivin has the potential to overcome EGFR–TKI resistance in NSCLCs.34 In conclusion, we have demonstrated that Dicer is required for maintenance of the specific miRNAs expression level, which involved in resistance to gefitinib in lung cancer. These findings will be important for the further development of novel 18 therapeutic strategy and a biomarker to early diagnosis for gefitinib resistance in lung cancer patients. 19 ACKNOWLEDGMENT This work was supported by grants from the National Science Council, Taiwan (NSC 102-2314-B-039-200, NSC 102-2314-B-038-028-MY3, NSC 101-2320-B-400-016-MY3; from National Health Research Institutes, Taiwan (CA-102-PP-41); from China Medical University Hospital (DMR-101-014); and from China Medical University (CMU99-TC-22, CMU100-S-22). 20 REFERENCES 1. Jemal A, Bray F, Center MM, Ferlay J, Ward E, Forman D. Global cancer statistics. CA Cancer J Clin. 2011;61(2):69-90. 2. Molina JR, Yang P, Cassivi SD, Schild SE, Adjei AA. Non-small cell lung cancer: epidemiology, risk factors, treatment, and survivorship. Mayo Clin Proc. 2008;83(5):584-94. 3. Muhsin M, Graham J, Kirkpatrick P. Gefitinib. Nat Rev Drug Discov. 2003;2(7):515-6. 4. Fukuoka M, Yano S, Giaccone G, et al. Multi-institutional randomized phase II trial of gefitinib for previously treated patients with advanced non-small-cell lung cancer (The IDEAL 1 Trial) [corrected]. J Clin Oncol. 2003;21(12):2237-46. 5. Jackman D, Pao W, Riely GJ, et al. Clinical definition of acquired resistance to epidermal growth factor receptor tyrosine kinase inhibitors in non-small-cell lung cancer. J Clin Oncol. 2010;28(2):357-60. 6. He L, Hannon GJ. MicroRNAs: small RNAs with a big role in gene regulation. Nat Rev Genet. 2004;5(7):522-31. 7. Kim VN. MicroRNA biogenesis: coordinated cropping and dicing. Nat Rev Mol Cell Biol. 2005;6(5):376-85. 21 8. Zheng T, Wang J, Chen X, Liu L. Role of microRNA in anticancer drug resistance. Int J Cancer. 2010;126(1):2-10. 9. Hummel R, Hussey DJ, Haier J. MicroRNAs: predictors and modifiers of chemo- and radiotherapy in different tumour types. Eur J Cancer. 2010;46(2):298-311. 10. Melo SA, Kalluri R. Molecular pathways: microRNAs as cancer therapeutics. Clin Cancer Res. 2012;18(16):4234-9. 11. Garofalo M, Croce CM. MicroRNAs as therapeutic targets in chemoresistance. Drug Resist Updat. 2013. 12. Bu Y, Lu C, Bian C, et al. Knockdown of Dicer in MCF-7 human breast carcinoma cells results in G1 arrest and increased sensitivity to cisplatin. Oncol Rep. 2009;21(1):13-7. 13. Ueda R, Kohanbash G, Sasaki K, et al. Dicer-regulated microRNAs 222 and 339 promote resistance of cancer cells to cytotoxic T-lymphocytes by down-regulation of ICAM-1. Proc Natl Acad Sci U S A. 2009;106(26):10746-51. 14. Selever J, Gu G, Lewis MT, et al. Dicer-mediated upregulation of BCRP confers tamoxifen resistance in human breast cancer cells. Clin Cancer Res. 2011;17(20):6510-21. 22 15. Pouliot LM, Shen DW, Suzuki T, Hall MD, Gottesman MM. Contributions of microRNA dysregulation to cisplatin resistance in adenocarcinoma cells. Exp Cell Res. 2013;319(4):566-74. 16. Vincenzi B, Zoccoli A, Schiavon G, et al. Dicer and Drosha expression and response to Bevacizumab-based therapy in advanced colorectal cancer patients. Eur J Cancer. 2013;49(6):1501-8. 17. Garofalo M, Romano G, Di Leva G, et al. EGFR and MET receptor tyrosine kinase-altered microRNA expression induces tumorigenesis and gefitinib resistance in lung cancers. Nat Med. 2012;18(1):74-82. 18. Martello G, Rosato A, Ferrari F, et al. A MicroRNA targeting dicer for metastasis control. Cell. 2010;141(7):1195-207. 19. Chen C, Ridzon DA, Broomer AJ, et al. Real-time quantification of microRNAs by stem-loop RT-PCR. Nucleic Acids Res. 2005;33(20):e179. 20. Karube Y, Tanaka H, Osada H, et al. Reduced expression of Dicer associated with poor prognosis in lung cancer patients. Cancer Sci. 2005;96(2):111-5. 21. Cummins JM, He Y, Leary RJ, et al. The colorectal microRNAome. Proc Natl Acad Sci U S A. 2006;103(10):3687-92. 22. Koscianska E, Starega-Roslan J, Krzyzosiak WJ. The role of Dicer protein partners in the processing of 23 microRNA precursors. PLoS One. 2011;6(12):e28548. 23. Lu J, Getz G, Miska EA, et al. MicroRNA expression profiles classify human cancers. Nature. 2005;435(7043):834-8. 24. Huang CP, Tsai MF, Chang TH, et al. ALDH-positive lung cancer stem cells confer resistance to epidermal growth factor receptor tyrosine kinase inhibitors. Cancer Lett. 2013;328(1):144-51. 25. Weiss GJ, Bemis LT, Nakajima E, et al. EGFR regulation by microRNA in lung cancer: correlation with clinical response and survival to gefitinib and EGFR expression in cell lines. Ann Oncol. 2008;19(6):1053-9. 26. Tokumaru S, Suzuki M, Yamada H, Nagino M, Takahashi T. let-7 regulates Dicer expression and constitutes a negative feedback loop. Carcinogenesis. 2008;29(11):2073-7. 27. Jakymiw A, Patel RS, Deming N, et al. Overexpression of dicer as a result of reduced let-7 MicroRNA levels contributes to increased cell proliferation of oral cancer cells. Genes Chromosomes Cancer. 2010;49(6):549-59. 28. Takamizawa J, Konishi H, Yanagisawa K, et al. Reduced expression of the let-7 microRNAs in human lung cancers in association with shortened postoperative survival. Cancer Res. 2004;64(11):3753-6. 29. Friedrich K, Wieder T, Von Haefen C, et al. Overexpression of caspase-3 24 restores sensitivity for drug-induced apoptosis in breast cancer cell lines with acquired drug resistance. Oncogene. 2001;20(22):2749-60. 30. Yang XH, Sladek TL, Liu X, Butler BR, Froelich CJ, Thor AD. Reconstitution of caspase 3 sensitizes MCF-7 breast cancer cells to doxorubicin- and etoposide-induced apoptosis. Cancer Res. 2001;61(1):348-54. 31. Devarajan E, Sahin AA, Chen JS, et al. Down-regulation of caspase 3 in breast cancer: a possible mechanism for chemoresistance. Oncogene. 2002;21(57):8843-51. 32. Yang X, Zheng F, Xing H, et al. Resistance to chemotherapy-induced apoptosis via decreased caspase-3 activity and overexpression of antiapoptotic proteins in ovarian cancer. J Cancer Res Clin Oncol. 2004;130(7):423-8. 33. Yoo J, Kim CH, Song SH, et al. Expression of caspase-3 and c-myc in non-small cell lung cancer. Cancer Res Treat. 2004;36(5):303-7. 34. Okamoto K, Okamoto I, Hatashita E, et al. Overcoming erlotinib resistance in EGFR mutation-positive non-small cell lung cancer cells by targeting survivin. Mol Cancer Ther. 2012;11(1):204-13. 25 FIGURE LEGENDS FIG. 1 Establishment and characterization of the gefitinib-acquired resistant PC9 cells. (a). Serum-starved PC-9 and PC9/GR cells were exposed to various doses of gefitinib for 24 h and assayed for survival by MTT assay. Data shown are mean ± SEM of six independent experiments performed in triplicate. ***p < 0.001. (b). The protein expression levels of Dicer, Drosha and AGO2 in PC9 and PC9/GR cells were analyzed by Western blot. (c). Associations between dicer expression and gefitinib sensitivity were investigated in PC9/GR cells cultured without gefitinib for 25 passages. Cells were treated with gefitinib at the indicated doses for 24 h and assayed for survival by MTT assay. Protein expression of Dicer at different passages was analyzed by Western blot. The bars represent the means ± SEM of three independent experiments, performed using different passages of PC9/GR cells in triplicates. **p < 0.01. (d). A western blot showing dose-dependent effects of gefitinib on the expression of dicer, MET, p-EGFR, and EGFR. α-tubulin was used as a loading control. FIG. 2 Silencing of Dicer in PC9/GR cells by shRNA lentivirus resulted in increased gefitinib sensitivity. Dicer knockdown efficiency was verified by using quantitative real-time PCR (a) and western blot analysis (b). Bars represent means ± 26 SEM from three independent experiments in triplicates. α-tubulin was used as a loading control. (c). Cells were exposed to different concentrations of gefitinib for 24 h and subsequently evaluated by MTT assay. Percentage of cell viability is shown relative to untreated control. Data are representative of at least three independent experiments performed in triplicate. ***p < 0.001. (d). Cells were treated with or without 20 μM gefitinib for 24 h. Sub-G1 cell populations were determined by flow cytometry in PC9/GR. Bars represent means ± SEM from at least three independent experiments. *p < 0.05. FIG. 3 Dicer modulating gefitinib sensitivity also exists in other types of non-small cell lung cancer (NSCLC) cells. (a). Western blot analysis of Dicer expression in A549 infected with shDicer and shScramble by lentivirus system. (b). A549/shDicer and A549/shScramble cells were treated with 10 μM of gefitinib for 48 h and then analyzed for cell viability by a MTT assay. (c). Dicer was required for resistance of A549 cells to gefitinib-induced apoptosis. Apoptotic cells with sub-G1 DNA content were determined by flow cytometry. Bars represent means ± SEM from three independent experiments in triplicates. *p < 0.05. (d). Western blot analysis of Dicer expression in H1299 infected with shDicer and shScramble by lentivirus system. (e). H1299/shDicer and H1299/shScramble cells were treated with 10 μM gefitinib for 48 27 h. Sub-G1 cell populations were determined by flow cytometry. Bars represent means ± SEM from at least three independent experiments. *p < 0.05. FIG. 4 Overexpression of dicer induces gefitinib resistance in PC9 and HCC827 cells. Dicer-expressing vector or control vector was transfected into PC9 cells, followed by analysis of Dicer expression using real-time PCR (a) and Western blot (b). Bars represent means ± SEM from three independent experiments in triplicates. (c). PC9/pcDNA and PC9/Dicer cells were exposed to 5 μM of gefitinib for 24 h and assayed for survival by MTT assay. Data are representative of at least three independent experiments performed in triplicate. ***p < 0.001. (d). Western blot analysis of Dicer in HCC827 cells transfected with Dicer-expressing vector or control. (e). HCC827/pcDNA and HCC827/Dicer cells were treated with 5 μM of gefitinib for 24 h and measured the function of Dicer in gefitinib-suppressed viability by MTT assay. Data are representative of at least three independent experiments performed in triplicate. *p < 0.05. FIG. 5 Knockdown of Dicer in NSCLC cells decreases expression of miR-30b/c and miR-221/222 leading to up-regulation of caspase-3 and increased gefitinib sensitivity. Knockdown of Dicer in PC9/GR cells (a) and A549 cells (b) decreases 28 expression of miR-30b/c and miR-221/222. All reactions were normalized to wild-type cells. Error bars indicate the SEM of three independent experiments. *p < 0.05, **p < 0.01. (c). The miR-30b/c and miR-221/222 are partially complementary to regions in the caspase 3 mRNAs 3’UTR. (d). PC9/GR/shDicer and PC9/GR/Scramble cells were untreated or treated with gefitinib and analyzed the cleavaged caspase-3 by Western blot analysis. (e). The effect of the caspase 3 inhibitor on viability of Dicer-silenced PC9/GR cells after treated with gefitinib. The cell viability was measured by MTT assay. Data are representative of at least three independent experiments performed in triplicate. *p < 0.05. 29