DOI QR코드

DOI QR Code

The MicroRNA-551a/MEF2C Axis Regulates the Survival and Sphere Formation of Cancer Cells in Response to 5-Fluorouracil

  • Kang, Hoin (Department of Biochemistry, The Catholic University of Korea College of Medicine) ;
  • Kim, Chongtae (Department of Biochemistry, The Catholic University of Korea College of Medicine) ;
  • Ji, Eunbyul (Department of Biochemistry, The Catholic University of Korea College of Medicine) ;
  • Ahn, Sojin (Department of Biochemistry, The Catholic University of Korea College of Medicine) ;
  • Jung, Myeongwoo (Department of Biochemistry, The Catholic University of Korea College of Medicine) ;
  • Hong, Youlim (Department of Biochemistry, The Catholic University of Korea College of Medicine) ;
  • Kim, WooK (Department of Molecular Science and Technology, Ajou University) ;
  • Lee, Eun Kyung (Department of Biochemistry, The Catholic University of Korea College of Medicine)
  • Received : 2018.07.09
  • Accepted : 2018.12.12
  • Published : 2019.02.28

Abstract

microRNAs regulate a diverse spectrum of cancer biology, including tumorigenesis, metastasis, stemness, and drug resistance. To investigate miRNA-mediated regulation of drug resistance, we characterized the resistant cell lines to 5-fluorouracil by inducing stable expression of miRNAs using lenti-miRNA library. Here, we demonstrate miR-551a as a novel factor regulating cell survival after 5-FU treatment. miR-551a-expressing cells (Hep3B-lenti-miR-551a) were resistant to 5-FU-induced cell death, and after 5-FU treatment, and showed significant increases in cell viability, cell survival, and sphere formation. It was further shown that myocyte-specific factor 2C is the direct target of miR-551a. Our results suggest that miR-551a plays a novel function in regulating 5-FU-induced cell death, and targeting miR-551a might be helpful to sensitize cells to anti-cancer drugs.

Keywords

INTRODUCTION

Acquisition of resistance to anti-cancer drugs is one of the major obstacles leading to the failure of cancer treatment (Gottesman, 2002; Hasan et al., 2018). Cancer recurrence and the growth of resistant tumors reduce the survival rate of cancer patients; therefore, it is important to understand the underlying molecular mechanisms of drug resistance. It has been known that resistance to anti-cancer drugs can be acquired by several mechanisms, such as enhanced DNA damage repair, cell death inhibition, drug inactivation, altered cell proliferation, and activation of cancer stem cells (CSCs) (Cree and Charlton, 2017; Housman et al., 2014; Prieto-Vila et al., 2017). Although several efforts have made to overcome the multi-drug resistance of cancer cells, how drug resistance arises and how cancer cells acquire drug resistance have yet been fully elucidated.

microRNAs (miRNAs) are small non-coding RNAs and function as a pivotal regulator in gene expression by inhibiting translation or facilitating the degradation of target mRNAs (Ambros, 2004; Bartel, 2004). miRNAs have been known to be involved in diverse regulation of cellular processes, such as cell growth, death, differentiation, and selfrenewal; therefore, differential expression of certain miRNAs is linked to the pathogenesis of diseases (He and Hannon, 2004; McDermott et al., 2011). Accumulating evidence demonstrates that miRNAs execute active roles in regulating the drug resistance of cancer cells by affecting stress response, cell survival, autophagy, and the absortion or digestion of anti-cancer drugs (An et al., 2017; Fojo, 2007; Wang et al., 2015). In previous studies, we have established miRNA-overexpressing cell lines resistant to 5-fluorouracil (5-FU), a widely used medication to treat a range of cancers, to find miRNAs regulating the acquisition of drug resistance, and demonstrated the role of miR-200a-3p and miR-195 in 5- FU-induced cell death (Kim et al., 2018; Lee et al., 2017).

In this study, we investigate the mechanism of resistance to 5-FU-induced cell death using one of the resistant clones established in our previous study (Lee et al., 2017). We identified miR-551a as a novel factor regulating cell survival after 5-FU treatment. miR-551a-expressing cells were resistant to 5-FU-induced cell death and inhibition of miR-551a sensitized cell to 5-FU. We showed that miR-551a increased cell survival after 5-FU treatment, by enhancing myocyte-specific factor 2C (MEF2C)-mediated sphere formation. Our results suggest that miR-551a plays a novel function in regulating 5-FU-induced cell death, and that targeting miR-551a might be helpful to sensitizing cells to anti-cancer drugs.

MATERIALS AND METHODS

Cell culture, transfection of plasmids and small interfering RNAs

Stable cell lines expressing miRNAs were established using a Lenti-miRTM Library as described in our previous studies (Kim et al., 2018; Lee et al., 2017). In brief, cells were transduced with Lenti-miRTM Library (Cat. No. PMIRHPLVAHT-1, System Biosciences, USA) (the multiplicity of infection = 10) and incubated with 10 μM of 5-FU for two weeks to establish the resistant clones. Hep3B cells were maintained with Dulbecco’s modified Eagle’s medium (Invitrogen, USA) containing 10% fetal bovine serum and 1% antibiotics. All transient transfections including miR-551a precursor, inhibitor (antimiR-551a) (Bioneer, Korea), the plasmid encoding GFPtagged MEF2C (pMEF2C) (Sino-Biological, China) and enhanced green fluorescent protein (EGFP) reporter plasmids were performed using LipofectamineTM 2000 (Invitrogen, USA). EGFP reporters were constructed by inserting 3’UTR of MEF2C mRNA into pEGFP-C1 vector (BD Bioscience, Germany) or site-directed mutagenesis.

Identification of miR-551a in the stable clone

Genomic DNA (gDNA) was isolated from the stable clone using AccuPrep® Genomic DNA Extraction Kit (Bioneer, Korea) according to the manufacturer’s instructions. The miRNA sequences integrated into genomic DNA were amplified by polymerase chain reaction (PCR) with specific primers and the nucleotide sequence was determined by sequencing as described in Ref. (Kim et al., 2018; Lee et al., 2017).

Cell viability assay

A colorimetric assay using the tetrazolium salt, 3-(4, 5- dimethylthiazol-2-yl)-2, 5-diphenyltetrazolium bromide (MTT) was used to assess cell viability. Cells (1 × 104 cells) were plated on each well of a 96-well plate and treated with 5-FU, oxaliplatin (OXP), cisplatin (CDDP), or doxorubicin (DOX) for 72 h and then further inculated with 0.5 mg/ml of MTT for 3 h. Formazan crystals were solubilized with isopropanol and the absorbance was measured using a Victor 3 microplate reader (Perkin Elmer, Finland).

RNA analysis

Total RNAs were isolated from cell lines or sphere using TRIzol® Reagent (Life Technologies, USA). For the analysis of mRNA, complementary DNA (cDNA) was synthesized by reverse transcription using a ReverTra Ace® RT Kit (Toyobo, Japan). For miRNA analysis, cDNA was prepared using the MiR-XTM miRNA First-Strand cDNA synthesis kit (Clontech, USA) according to the manufacturer’s instructions. The relative abundance of each transcript was assessed by real-time quantitative polymerase chain reaction (RT-qPCR) using the Bioline SYBR Fast qPCR kit (Bioline, UK) and specific primer sets on the StepOne PlusTM system (Applied Biosystems, USA).

Western blotting analysis

Whole cell lysates were prepared using RIPA buffer containing 10 mM Tris–HCl (pH 7.4), 150 mM NaCl, 1% NP-40, 1 mM EDTA and 0.1% sodium dodecyl sulfate separated by electrophoresis in SDS-containing polyacrylamide gels (SDSPAGE), and transferred onto poly-vinylidene difluoride (PVDF) membranes (Millipore, USA). The blots were incubated with the following antibodies against GFP, MEF2C (Santa Cruz Biotechnology, USA), and β-actin (Abcam, USA), then sequentially incubated with the appropriate secondary antibodies conjugated with horseradish peroxidase (HRP) (Santa Cruz Biotechnology, USA). Chemo-luminescent signals were visualized using NEW ClarityTM ECL substrate (BioRad, USA).

Sphere-forming assay

Cells (1 × 103 cells) were seeded in low attachment 96-well plate, and cultured in serum-free medium. After one week, spheres were observed and manually counted. The number of spheres was analyzed in triplicate for each cell type, and at least three independent experiments were carried out.

RESULTS

Hep3B clone expressing miR-551a is resistant to 5-fluorouracil-induced cell death

To identify miRNAs involved in the acquisition of anti-cancer drug resistance to 5-FU, we established stable cell lines expressing certain miRNAs using lenti-miR library with sequential exposure to 5-FU as shown in Fig. 1A (Lee et al., 2017). The specific miRNA expressed in the GFP-positive survival clone was identified as miR-551a by genomic DNA PCR and sequencing analysis of PCR amplicon (Fig. 1B). To analyze the relative expression of miR-551a between miR-551aexpressing clone (Hep3B-lenti-miR-551a) and control cell (Hep3B-lenti-miR-Ctrl), miR-551a level was determined by miRNA RT-qPCR and the result showed higher expression of miR-551a in Hep3B-lenti-miR-551a cell (Fig. 1C). Because Hep3B-lenti-miR-551a clone survived sequential exposure to 5-FU, the relative response of Hep3B-lenti-miR-551a and Hep3B-lenti-miR-Ctrl cells to 5-FU was assessed by MTT assay. Figure 1D shows that the cell viability of Hep3B-lentimiR-551a cell was higher than that of Hep3B-lenti-miR-Ctrl. These results indicate that Hep3B-lenti-miR-551a cell was resistant after 5-FU treatment, and that miR-551a has a role in the regulation of 5-FU-induced cell death.

E1BJB7_2019_v42n2_175_f0001.png 이미지

Fig. 1. Hep3B cells stably expressing miR-551a are resistant to 5-FU-induced cell death. (A) After infection with a lentiviral miRNA library, Hep3B cells were exposed to 10 μM of 5-FU until all control cells (Hep3B-lenti-miR-Ctrl) were dead. Surviving clones were isolated and established as 5-FU-resistant Hep3B clones. (B) GFP expression of Hep3B-lenticlones was observed using fluorescence microscopy. The insertion of miRNA gene integrated from lentivirus was analyzed by sequencing of PCR products amplified from genomic DNA (gDNA) using specific primers, and identified as miR-551a in Hep3B-lentimiR-551a clone. (C) Relative levels of miR551a between Hep3B-lenti-miR-Ctrl and Hep3B-lenti-miR-551a were quantified by miRNA RT-qPCR. U6 RNA was used for normalization. (D) Hep3B-lenti-miR-Ctrl and Hep3B-lenti-miR-551a cells were exposed to 5-FU (1, 5, 10, 20, and 50 μM) for 72 h, and cell viability was determined by MTT assay. Data represent mean ± SEM from three independent experiments. *P < 0.05

miR-551a increases cell viability in response to anti-cancer drugs

To investigate whether miR-551a is an essential factor in regulating cell survival after anti-cancer drug treatment, cell viability between Hep3B-lenti-miR-551a and Hep3B-lentimiR-Ctrl cells was assessed by MTT assay. No significant change of cell viability of these cell lines at basal level was observed; however, Hep3B-lenti-miR-551a cells had higher viability in response to multiple anti-cancer drugs including 5-FU, oxaliplatin (OXP), cisplatin (CDDP), and doxorubicin (DOX) compared to Hep3B-lenti-Ctrl cells (Figs. 2A and 2B). To determine whether a high level of miR-551a is responsible for an increase of cell survival in Hep3B-lenti-miR-551 cell, we investigated the effect of miR-551a inhibition using antimiR-551a. Ectopic expression of anti-miR-551a resulted in a significant reduction of cell viability after treatment of 5-FU, CDDP, and DOX (Fig. 2C). These results indicate that miR551a has a role in promoting cell survival in response to anticancer drugs.

E1BJB7_2019_v42n2_175_f0002.png 이미지

Fig. 2. Inhibition of miR-551a sensitizes Hep3B-lenti-miR-551a cells to several anticancer agents. (A and B) Cell viability was determined by MTT assay at the basal level (A), or after incubation with 5-FU, oxaliplatin (OXP), cisplatin (CDDP), and doxorubicin (DOX) for 72 h (B). (C) After transfection of anti-miR-551a or control miRNA, Hep3B-lenti-miR-551a cells were incubated with 5-FU, OXP, CDDP, and DOX for 72 h and cell viability was determined by MTT assay. Data represent mean ± SEM from three independent experiments. *P < 0.05

miR-551a promotes the sphere formation of Hep3B cells

We observed that Hep3B-lenti-miR-551a cells are resistant to 5-FU-induced cell death, thereby increasing cell survival after 5-FU treatment. Several studies have shown that sphere formation is one of the essential characteristics affecting the drug resistance of cancer cells (Cao et al., 2011; Chen et al., 2012; Cioce et al., 2010; Fujii et al., 2009). Therefore, we investigated the sphere-forming ability of Hep3B-lenti-miR551a cells. Figure 3A shows that Hep3B-lenti-miR-551a cells displayed enhanced sphere-forming ability, compared to Hep3B-lenti-miR-Ctrl cells. In addition, the relative expression of common stemness regulators, including OCT4, NANOG, KLF4, and SOX2, was slightly upregulated in Hep3B-lentimiR-551a cells. To further investigate whether miR-551a has the potential to regulate sphere-forming ability, the number of spheres was analyzed after ectopic expression of miR551a in Hep3B cells. Ectopic expression of miR-551a increased the number of spheres and the levels of common stemness regulators in Hep3B cells (Fig. 3B). In addition, sphere-forming ability between Hep3B-lenti-miR-551a cells and Hep3B-lenti-miR-Ctrl cells after 5-FU treatement was further analyzed. While Hep3B-lenti-miR-Ctrl cells failed to form spheres after 5-FU treatment, Hep3B-lenti-miR-551a cells survived, and generated spheres, even in the presence of 5-FU (Fig. 3C). Taken together, these results indicate that miR-551a promotes the sphere formation of Hep3B cells, thereby increasing resistance to 5-FU-induced cell death.

E1BJB7_2019_v42n2_175_f0003.png 이미지

Fig. 3. miR-551a increases the sphere formation of Hep3B cells. (A and B) The number of spheres and relative gene expressions were analyzed in both Hep3B-lenti-miR clones (A) or miR-transfected cells (B). Cells were seeded in a low attachment 96-well plate and cultured in serum-free medium. One week later, the number of spheres was counted from three different areas. Relative levels of OCT4, NANOG, KLF4, and SOX2 mRNA were analyzed by RT-qPCR. (C) Seventy-two h after 5-FU treatment, the number of spheres and relative gene expression were analyzed between Hep3B-lenti-miRCtrl and Hep3B-lenti-miR-551a cells. Images are representative from three independent experiments. Data represent mean ± SEM from three independent experiments. *P < 0.05miR-551a downregulates MEF2C expression

To determine a potential target of miR-551a, in silico analysis using Targetscan and miRDB was done and identified MEF2C mRNA. Figure 4A shows that MEF2C mRNA has the binding site for miR-551a at its 3’UTR (2145-2151 nt). Ectopic expression of miR-551a decreased MEF2C expression at both mRNA and protein level in Hep3B cells (Fig. 4B). In addition, a lower expression of MEF2C was observed in Hep3B-lenti-miR-551a compared to Hep3B-lenti-miR-Ctrl cell (Fig. 4C). These results indicate that MEF2C expression is regulated by miR-551a. To further determine whether MEF2C is the direct target of miR-551a, we generated EGFP reporter contructs containing 3’UTR of MEF2C mRNA (2015- 2246 nt)(pEGFP-MEF2C 3U) or mutant 3’UTR missing the seed region binding site for miR-551a (pEGFP-MEF2C 3UM) and relative EGFP expression was assessed by Western blotting (Fig. 4D). Overexpression of miR-551a reduced EGFPMEF2C 3U reporter but did not change the level of EGFP control or EGFP-MEF2C 3UM (Fig. 4D). These results suggest that MEF2C is the direct target of miR-551a.

E1BJB7_2019_v42n2_175_f0004.png 이미지

Fig. 4. miR-551a downregulates MEF2C expression. (A) Schematic of MEF2C mRNA. miR-551a binding site in MEF2C mRNA 3’UTR (2145- 2151bp) is shown as a white box. (B and C) Relative expression of MEF2C proteins and mRNA was determined by Western blotting and RT-qPCR in both Hep3B-lenti-miR clones (B) or miR-transfected cells (C). (D) EGFP reporter (pEGFP-MEF2C 3U) containing MEF2C mRNA 3’UTR (2106- 2249bp) and mutant reporter lacking miRNA binding site were constructed. (E) Hep3B cells were sequentially transfected with miRNAs and EGFP reporters and relative expressions of EGFP were determined by Western blotting. β-actin was used as loading control. Images are representative from three independent experiments. Data are mean ± SEM or representative from three independent experiments. *P < 0.05

Knockdown of MEF2C enhances the sphere formation of Hep3B cells

We showed that miR-551a-expressing cells increased the number of spheres, and that MEF2C is the direct target of miR-551a (Fig. 3 and Fig. 4). To investigate whether sphere forming ability is affected by MEF2C, Hep3B cells were transfected with MEF2C siRNA or pMEF2C plasmid (Fig. 5A), then cell viability and sphere forming ability were assessed as described previously. Knockdown of MEF2C increased, while MEF2C overexpression decreased both cell viability and the number of spheres after 5-FU treatment (Figs. 5B and 5C). To further elucidate whether the miR-551a-mediated MEF2C downregulation is responsible for the resistant phenotypes in response to 5-FU, MEF2C was ectopically expressed in Hep3B-lenti-miR-551a cells and cells were further incubated with 5-FU. As shown in Figs. 5D and 5E, MEF2C expression reduced cell viability and the number of spheres in Hep3B-lenti-miR-551a cells after 5-FU treatment. These results indicate that MEF2C overexpression sensitized Hep3B-lenti-miR-551a cells in response to 5-FU and the miR551a-MEF2C axis has a role in 5-FU-induced cell death by regulating sphere formation.

E1BJB7_2019_v42n2_175_f0005.png 이미지

Fig. 5. Knockdown of MEF2C enhances the sphere formation of Hep3B cells. (A-C) Hep3B cells were transfected with MEF2C siRNA or pMEF2C plasmid. Relative levels of MEF2C mRNA (A), cell viability (B), and the number of spheres (C) were determined by RT-qPCR, MTT assay, and sphere formation assay after 5-FU treatment. (D and E) After transfection of pMEF2C plasmid, Hep3Blenti-miR-551a cells were further incubated with 5-FU, then cell viability (D) and the number of spheres (E) were analyzed. Spheres were counted from at least three different fields of each set of experiment. Data are mean ± SEM from three independent experiments. *P < 0.05

DISCUSSION

Although numerous efforts have made to understand how cancer cells acquire anti-cancer drug resistance, it still remains a major obstacle to effective cancer therapy. Drug inactivation, drug target alteration, DNA damage response, cell death inhibition, EMT development or an increase of stemness contribute to the anti-cancer drug resistance of cancer cells (Hasan et al., 2018; Housman et al., 2014). In this study, we demonstrate miR-551a as a novel factor regulating cell survival after 5-FU treatment. We identified miR-551a in one of the 5-FU resistant Hep3B clones established using lentimiR library (Lee et al., 2017). We showed that miR-551aexpressing cells are resistant to 5-FU-induced cell death, and that inhibition of miR-551a sensitized Hep3B cells to 5-FU by regulating MEF2C-mediated sphere formation. Our results suggest that miR-551a plays a novel function in regulating 5-FU-induced cell death, and that targeting miR-551a might decrease the resistance of cancer cells to anti-cancer drugs.

miRNAs execute a major part in several aspects of cancer progression including tumorigenesis, metastasis, differentiation of cancer stem cells, and drug resistance (Caliskan et al., 2017; Deng et al., 2016; Van Roosbroeck and Calin, 2017). Aberrant expressions of certain miRNAs execute tumor suppressive or oncogenic roles, and could be a useful biomarker or drugable target (Hayes et al., 2014; Jansson and Lund, 2012). In this study, we demonstrated the role of miR-551a as a novel factor regulating anti-cancer drug-induced cell death. miR-551a is known to have a tumor-suppressive function by targeting phosphatase of regenerating liver-3 (PRL-3), insulin receptor substrate 2 (IRS2), and creatin kinase brain-type (CKB) in certain types of cancer, including gastric, colon, and ovarian cancer (Du and Sha, 2017; Li et al., 2012; Loo et al., 2015). Although aberant expression of miR-551a has been reported so far, little is known about the cellular function and molecular target of miR-551a. We demonstrate that miR-551a promotes cell survival, by increasing the sphere formation of cancer cells after 5-FU treatment (Figs. 2 and 3). In this study, we did not investigate the level of miR-551a in the tissues from the patients who have drug resistance; and further studies are required to provide the clinical significance of miR-551a in the regulation of drug resistance.

MEF2C is a member of the MEF2 gene family and regulates cell differentiation, self renewal, and stemness (Abad et al., 2017; Cho et al., 2011; Ignatius et al., 2017; Li et al., 2008; Rocha et al., 2016). In addition, MEF2C is also known to control cell migration and the proliferation of cancer cells (Wang et al., 2016; Zhang et al., 2018). Therefore, dysregulation of MEF2C expression affects the developmental process or cancer progression. Several miRNAs, including miR21, -27, -135, -155,- 204, -214, and -448 have been reported to target MEF2C mRNA (Cheng et al., 2018; Chinchilla et al., 2011; Ling et al., 2017; Sahoo et al., 2016; Xu et al., 2015; Yelamanchili et al., 2010; Zhang et al., 2017); and we also showed that miR-551 is one of MEF2C-targeting miRNAs (Fig. 4). We observed that MEF2C directly regulates the sphere-forming ability of Hep3B cells (Fig. 5). The detailed mechanism of MEF2C-mediated sphere formation needs to be further elucidated.

In this study, we characterized Hep3B-lenti-miR-551a cells as a 5-FU resistant clone. The miR-551a-expressing clone survived sequential exposure to 5-FU (Figs. 1 and 2). In Hep3B-lenti-miR-551a cells, sphere-forming ability and the expression of stemness marker genes, which are typical characteristics of cancer-initiating cells (Facompre et al., 2012), were increased even after 5-FU treatment (Fig. 3). These results suggest that miR-551a is the mediator of acquisition of drug resistance, by enhancing the sphereformation ability of cells. Sphere formation is one of the stem-like properties of cancer-initiating cells, and is known to be related to high levels of self-renewal, tumorigenecity, anti-cancer drug resistance, and invasive phenotype of stem cells (Dieter et al., 2011; Lee et al., 2016). Several studies have demonstrated the essential roles of miRNAs in the regulation of drug resistance, by affecting the sphere-forming ability of cells (Hasegawa et al., 2014; Jeon et al., 2011; Park et al., 2014; Tung et al., 2017; Wang et al., 2017). Therefore, targeting certain miRNAs to reduce the sphere-forming ability of cancer cells could be a useful technique for effective cancer treatment. Our study provides novel insights into the function of miR-551a, which enable to the reduction of anticancer drug resistance, thereby increasing the efficiency of chemotherapy.

ACKNOWLEDGMENTS

This work is supported by the National Research Foundation of Korea (NRF) grant funded by the Korea government (2017R1A2B2009381 and 2012R1A5A2047939).

References

  1. Abad, M., Hashimoto, H., Zhou, H., Morales, M.G., Chen, B., Bassel-Duby, R., and Olson, E.N. (2017). Notch inhibition enhances cardiac reprogramming by increasing MEF2C transcriptional activity. Stem Cell Rep. 8, 548-560. https://doi.org/10.1016/j.stemcr.2017.01.025
  2. Ambros, V. (2004). The functions of animal microRNAs. Nature 431, 350-355. https://doi.org/10.1038/nature02871
  3. An, X., Sarmiento, C., Tan, T., and Zhu, H. (2017). Regulation of multidrug resistance by microRNAs in anti-cancer therapy. Acta. Pharm. Sin. B 7, 38-51. https://doi.org/10.1016/j.apsb.2016.09.002
  4. Bartel, D.P. (2004). MicroRNAs: genomics, biogenesis, mechanism, and function. Cell 116, 281-297. https://doi.org/10.1016/S0092-8674(04)00045-5
  5. Caliskan, M., Guler, H., and Bozok Cetintas, V. (2017). Current updates on microRNAs as regulators of chemoresistance. Biomed. Pharmacother 95, 1000-1012. https://doi.org/10.1016/j.biopha.2017.08.084
  6. Cao, L., Zhou, Y., Zhai, B., Liao, J., Xu, W., Zhang, R., Li, J., Zhang, Y., Chen, L., Qian, H., et al. (2011). Sphere-forming cell subpopulations with cancer stem cell properties in human hepatoma cell lines. BMC Gastroenterol. 11, 71. https://doi.org/10.1186/1471-230X-11-71
  7. Chen, S.F., Chang, Y.C., Nieh, S., Liu, C.L., Yang, C.Y., and Lin, Y.S. (2012). Nonadhesive culture system as a model of rapid sphere formation with cancer stem cell properties. PLoS One 7, e31864. https://doi.org/10.1371/journal.pone.0031864
  8. Cheng, X., Du, J., Shen, L., Tan, Z., Jiang, D., Jiang, A., Li, Q., Tang, G., Jiang, Y., Wang, J., et al. (2018). MiR-204-5p regulates C2C12 myoblast differentiation by targeting MEF2C and ERRgamma. Biomed. Pharmacother 101, 528-535. https://doi.org/10.1016/j.biopha.2018.02.096
  9. Chinchilla, A., Lozano, E., Daimi, H., Esteban, F.J., Crist, C., Aranega, A.E., and Franco, D. (2011). MicroRNA profiling during mouse ventricular maturation: a role for miR-27 modulating Mef2c expression. Cardiovasc. Res. 89, 98-108. https://doi.org/10.1093/cvr/cvq264
  10. Cho, E.G., Zaremba, J.D., McKercher, S.R., Talantova, M., Tu, S., Masliah, E., Chan, S.F., Nakanishi, N., Terskikh, A., and Lipton, S.A. (2011). MEF2C enhances dopaminergic neuron differentiation of human embryonic stem cells in a parkinsonian rat model. PLoS One 6, e24027. https://doi.org/10.1371/journal.pone.0024027
  11. Cioce, M., Gherardi, S., Viglietto, G., Strano, S., Blandino, G., Muti, P., and Ciliberto, G. (2010). Mammosphere-forming cells from breast cancer cell lines as a tool for the identification of CSC-like- and early progenitor-targeting drugs. Cell Cycle 9, 2878-2887. https://doi.org/10.4161/cc.9.14.12371
  12. Cree, I.A., and Charlton, P. (2017). Molecular chess? Hallmarks of anti-cancer drug resistance. BMC Cancer 17, 10. https://doi.org/10.1186/s12885-016-2999-1
  13. Deng, Y., Zhu, G., Luo, H., and Zhao, S. (2016). MicroRNA-203 As a Stemness Inhibitor of Glioblastoma Stem Cells. Mol. Cells 39, 619-624. https://doi.org/10.14348/molcells.2016.0118
  14. Dieter, S.M., Ball, C.R., Hoffmann, C.M., Nowrouzi, A., Herbst, F., Zavidij, O., Abel, U., Arens, A., Weichert, W., Brand, K., et al. (2011). Distinct types of tumor-initiating cells form human colon cancer tumors and metastases. Cell Stem Cell 9, 357-365. https://doi.org/10.1016/j.stem.2011.08.010
  15. Du, Z., and Sha, X. (2017). Demethoxycurcumin inhibited human epithelia ovarian cancer cells' growth via up-regulating miR-551a. Tumour Biol. 39, 1010428317694302.
  16. Facompre, N., Nakagawa, H., Herlyn, M., and Basu, D. (2012). Stemlike cells and therapy resistance in squamous cell carcinomas. Adv. Pharmacol. 65, 235-265. https://doi.org/10.1016/B978-0-12-397927-8.00008-7
  17. Fojo, T. (2007). Multiple paths to a drug resistance phenotype: mutations, translocations, deletions and amplification of coding genes or promoter regions, epigenetic changes and microRNAs. Drug Resist. Updat. 10, 59-67. https://doi.org/10.1016/j.drup.2007.02.002
  18. Fujii, H., Honoki, K., Tsujiuchi, T., Kido, A., Yoshitani, K., and Takakura, Y. (2009). Sphere-forming stem-like cell populations with drug resistance in human sarcoma cell lines. Int. J. Oncol. 34, 1381-1386.
  19. Gottesman, M.M. (2002). Mechanisms of cancer drug resistance. Annu. Rev. Med. 53, 615-627. https://doi.org/10.1146/annurev.med.53.082901.103929
  20. Hasan, S., Taha, R., and Omri, H.E. (2018). Current opinions on chemoresistance: an overview. Bioinformation 14, 80-85. https://doi.org/10.6026/97320630014080
  21. Hasegawa, S., Eguchi, H., Nagano, H., Konno, M., Tomimaru, Y., Wada, H., Hama, N., Kawamoto, K., Kobayashi, S., Nishida, N., et al. (2014). MicroRNA-1246 expression associated with CCNG2-mediated chemoresistance and stemness in pancreatic cancer. Br. J. Cancer 111, 1572-1580. https://doi.org/10.1038/bjc.2014.454
  22. Hayes, J., Peruzzi, P.P., and Lawler, S. (2014). MicroRNAs in cancer: biomarkers, functions and therapy. Trends. Mol. Med. 20, 460-469. https://doi.org/10.1016/j.molmed.2014.06.005
  23. He, L., and Hannon, G.J. (2004). MicroRNAs: small RNAs with a big role in gene regulation. Nat. Rev. Genet. 5, 522-531. https://doi.org/10.1038/nrg1379
  24. Housman, G., Byler, S., Heerboth, S., Lapinska, K., Longacre, M., Snyder, N., and Sarkar, S. (2014). Drug resistance in cancer: an overview. Cancers (Basel) 6, 1769-1792. https://doi.org/10.3390/cancers6031769
  25. Ignatius, M.S., Hayes, M.N., Lobbardi, R., Chen, E.Y., McCarthy, K.M., Sreenivas, P., Motala, Z., Durbin, A.D., Molodtsov, A., Reeder, S., et al. (2017). The NOTCH1/SNAIL1/MEF2C Pathway Regulates Growth and Self-Renewal in Embryonal Rhabdomyosarcoma. Cell Rep. 19, 2304-2318. https://doi.org/10.1016/j.celrep.2017.05.061
  26. Jansson, M.D., and Lund, A.H. (2012). MicroRNA and cancer. Mol. Oncol. 6, 590-610. https://doi.org/10.1016/j.molonc.2012.09.006
  27. Jeon, H.M., Sohn, Y.W., Oh, S.Y., Kim, S.H., Beck, S., Kim, S., and Kim, H. (2011). ID4 imparts chemoresistance and cancer stemness to glioma cells by derepressing miR-9*-mediated suppression of SOX2. Cancer Res. 71, 3410-3421. https://doi.org/10.1158/0008-5472.CAN-10-3340
  28. Kim, C., Hong, Y., Lee, H., Kang, H., and Lee, E.K. (2018). MicroRNA-195 desensitizes HCT116 human colon cancer cells to 5-fluorouracil. Cancer Lett. 412, 264-271. https://doi.org/10.1016/j.canlet.2017.10.022
  29. Lee, C.H., Yu, C.C., Wang, B.Y., and Chang, W.W. (2016). Tumorsphere as an effective in vitro platform for screening anticancer stem cell drugs. Oncotarget 7, 1215-1226. https://doi.org/10.18632/oncotarget.6261
  30. Lee, H., Kim, C., Kang, H., Tak, H., Ahn, S., Yoon, S.K., Kuh, H.J., Kim, W., and Lee, E.K. (2017). microRNA-200a-3p increases 5-fluorouracil resistance by regulating dual specificity phosphatase 6 expression. Exp. Mol. Med. 49, e327. https://doi.org/10.1038/emm.2017.33
  31. Li, H., Radford, J.C., Ragusa, M.J., Shea, K.L., McKercher, S.R., Zaremba, J.D., Soussou, W., Nie, Z., Kang, Y.J., Nakanishi, N., et al. (2008). Transcription factor MEF2C influences neural stem/progenitor cell differentiation and maturation in vivo. Proc. Natl. Acad. Sci. USA 105, 9397-9402. https://doi.org/10.1073/pnas.0802876105
  32. Li, Z., Cao, Y., Jie, Z., Liu, Y., Li, Y., Li, J., Zhu, G., Liu, Z., Tu, Y., Peng, G., et al. (2012). miR-495 and miR-551a inhibit the migration and invasion of human gastric cancer cells by directly interacting with PRL-3. Cancer Lett. 323, 41-47. https://doi.org/10.1016/j.canlet.2012.03.029
  33. Ling, X., Yao, D., Kang, L., Zhou, J., Zhou, Y., Dong, H., Zhang, K., Zhang, L., and Chen, H. (2017). Involment of RAS/ERK1/2 signaling and MEF2C in miR-155-3p inhibition-triggered cardiomyocyte differentiation of embryonic stem cell. Oncotarget 8, 84403-84416. https://doi.org/10.18632/oncotarget.21218
  34. Loo, J.M., Scherl, A., Nguyen, A., Man, F.Y., Weinberg, E., Zeng, Z., Saltz, L., Paty, P.B., and Tavazoie, S.F. (2015). Extracellular metabolic energetics can promote cancer progression. Cell 160, 393-406. https://doi.org/10.1016/j.cell.2014.12.018
  35. McDermott, A.M., Heneghan, H.M., Miller, N., and Kerin, M.J. (2011). The therapeutic potential of microRNAs: disease modulators and drug targets. Pharm. Res. 28, 3016-3029. https://doi.org/10.1007/s11095-011-0550-2
  36. Park, E.Y., Chang, E., Lee, E.J., Lee, H.W., Kang, H.G., Chun, K.H., Woo, Y.M., Kong, H.K., Ko, J.Y., Suzuki, H., et al. (2014). Targeting of miR34a-NOTCH1 axis reduced breast cancer stemness and chemoresistance. Cancer Res. 74, 7573-7582. https://doi.org/10.1158/0008-5472.CAN-14-1140
  37. Prieto-Vila, M., Takahashi, R.U., Usuba, W., Kohama, I., and Ochiya, T. (2017). Drug resistance driven by cancer stem cells and their niche. Int. J. Mol. Sci. 18. 2574. https://doi.org/10.3390/ijms18122574
  38. Rocha, H., Sampaio, M., Rocha, R., Fernandes, S., and Leao, M. (2016). MEF2C haploinsufficiency syndrome: Report of a new MEF2C mutation and review. Eur. J. Med. Genet. 59, 478-482. https://doi.org/10.1016/j.ejmg.2016.05.017
  39. Sahoo, S., Meijles, D.N., Al Ghouleh, I., Tandon, M., Cifuentes-Pagano, E., Sembrat, J., Rojas, M., Goncharova, E., and Pagano, P.J. (2016). MEF2C-MYOCD and leiomodin1 suppression by miRNA-214 promotes smooth muscle cell phenotype switching in pulmonary arterial hypertension. PLoS One 11, e0153780. https://doi.org/10.1371/journal.pone.0153780
  40. Tung, S.L., Huang, W.C., Hsu, F.C., Yang, Z.P., Jang, T.H., Chang, J.W., Chuang, C.M., Lai, C.R., and Wang, L.H. (2017). miRNA-34c-5p inhibits amphiregulin-induced ovarian cancer stemness and drug resistance via downregulation of the AREG-EGFR-ERK pathway. Oncogenesis 6, e326. https://doi.org/10.1038/oncsis.2017.25
  41. Van Roosbroeck, K., and Calin, G.A. (2017). Cancer hallmarks and MicroRNAs: the therapeutic connection. Adv. Cancer Res. 135, 119-149. https://doi.org/10.1016/bs.acr.2017.06.002
  42. Wang, J., Yang, M., Li, Y., and Han, B. (2015). The role of MicroRNAs in the chemoresistance of breast cancer. Drug Dev. Res. 76, 368-374. https://doi.org/10.1002/ddr.21275
  43. Wang, W., Li, Y., Liu, N., Gao, Y., and Li, L. (2017). MiR-23b controls ALDH1A1 expression in cervical cancer stem cells. BMC Cancer 17, 292. https://doi.org/10.1186/s12885-017-3192-x
  44. Wang, W., Org, T., Montel-Hagen, A., Pioli, P.D., Duan, D., Israely, E., Malkin, D., Su, T., Flach, J., Kurdistani, S.K., et al. (2016). MEF2C protects bone marrow B-lymphoid progenitors during stress haematopoiesis. Nat. Commun. 7, 12376. https://doi.org/10.1038/ncomms12376
  45. Xu, Z., Han, Y., Liu, J., Jiang, F., Hu, H., Wang, Y., Liu, Q., Gong, Y., and Li, X. (2015). MiR-135b-5p and MiR-499a-3p promote cell proliferation and migration in atherosclerosis by directly targeting MEF2C. Sci. Rep. 5, 12276. https://doi.org/10.1038/srep12276
  46. Yelamanchili, S.V., Chaudhuri, A.D., Chen, L.N., Xiong, H., and Fox, H.S. (2010). MicroRNA-21 dysregulates the expression of MEF2C in neurons in monkey and human SIV/HIV neurological disease. Cell Death Dis. 1, e77. https://doi.org/10.1038/cddis.2010.56
  47. Zhang, H., Liu, W., Wang, Z., Meng, L., Wang, Y., Yan, H., and Li, L. (2018). MEF2C promotes gefitinib resistance in hepatic cancer cells through regulating MIG6 transcription. Tumori, 300891618765555.
  48. Zhang, R., Sui, L., Hong, X., Yang, M., and Li, W. (2017). MiR-448 promotes vascular smooth muscle cell proliferation and migration in through directly targeting MEF2C. Environ. Sci. Pollut. Res. Int. 24, 22294-22300. https://doi.org/10.1007/s11356-017-9771-1

Cited by

  1. Discovery and validation of methylated-differentially expressed genes in Helicobacter pylori-induced gastric cancer vol.27, pp.6, 2019, https://doi.org/10.1038/s41417-019-0125-7