Journal list menu

Volume 6, Issue 3 p. 299-310
Paper
Open Access

EGFR and myosin II inhibitors cooperate to suppress EGFR-T790M-mutant NSCLC cells

Huan-Chih Chiu

Huan-Chih Chiu

Institute of Biotechnology and Pharmaceutical Research, National Health Research Institutes, Miaoli, Taiwan, ROC

Search for more papers by this author
Teng-Yuan Chang

Teng-Yuan Chang

Institute of Biotechnology and Pharmaceutical Research, National Health Research Institutes, Miaoli, Taiwan, ROC

Search for more papers by this author
Chin-Ting Huang

Chin-Ting Huang

Institute of Biotechnology and Pharmaceutical Research, National Health Research Institutes, Miaoli, Taiwan, ROC

Search for more papers by this author
Yu-Sheng Chao

Yu-Sheng Chao

Institute of Biotechnology and Pharmaceutical Research, National Health Research Institutes, Miaoli, Taiwan, ROC

Search for more papers by this author
John T.-A. Hsu

Corresponding Author

John T.-A. Hsu

Institute of Biotechnology and Pharmaceutical Research, National Health Research Institutes, Miaoli, Taiwan, ROC

Department of Biological Science and Technology, NationalChiao Tung University, Hsinchu, Taiwan, ROC

Corresponding author. Institute of Biotechnology and Pharmaceutical Research, National Health Research Institutes, No.35, Keyan Road, Zhunan Town, Miaoli County 350, Taiwan, ROC. Tel.: +886 37 246166 35717; fax: +886 37 586456.Search for more papers by this author
First published: 10 February 2012
Citations: 10

Abstract

An acquired mutation (T790M) in the epidermal growth factor receptor (EGFR) accounts for half of all relapses in non-small cell lung cancer (NSCLC) patients who initially respond to EGFR kinase inhibitors. In this study, we demonstrated for the first time that EGFR-T790M interacts with the cytoskeletal components, myosin heavy chain 9 (MYH9) and β-actin, in the nucleus of H1975 cells carrying the T790M-mutant EGFR. The interactions of EGFR with MYH9 and β-actin were reduced in the presence of blebbistatin, a specific inhibitor for the MYH9-β-actin interaction, suggesting that the EGFR interaction with MYH9 and β-actin is affected by the integrity of the cytoskeleton. These physical interactions among MYH9, β-actin, and EGFR were also impaired by CL-387,785, a kinase inhibitor for EGFR-T790M. Furthermore, CL-387,785 and blebbistatin interacted in a synergistic fashion to suppress cell proliferation and induce apoptosis in H1975 cells. The combination of CL-387,785 and blebbistatin enhanced the down-regulation of cyclooxygenase-2 (COX-2), a transcriptional target of nuclear EGFR. Overall, our findings demonstrate that disrupting EGFR interactions with the cytoskeletal components enhanced the anti-cancer effects of CL-387,785 against H1975 cells, suggesting a novel therapeutic approach for NSCLC cells that express the drug-resistant EGFR-T790M.

1 Introduction

Lung cancer has been the leading cause of cancer-related deaths worldwide. Because of the poor survival rates, researchers continue to pursue more effective strategies for the treatment of lung cancer. Clinical studies have shown that 80% of lung cancer patients are diagnosed with NSCLC (Sharma et al., 2007). Overexpression of EGFR has been found in 40–80% of NSCLC patients and correlates with a poorer prognosis (Shigematsu et al., 2005). An in-frame deletion in exon 19 (Del) and a missense mutation in exon 21 (L858R) account for a majority of the NSCLC-related EGFR mutations frequently found in certain subsets of patients (Kobayashi et al., 2005a; Pao et al., 2005). NSCLC patients with such mutations in EGFR demonstrate a superior clinical response to the EGFR inhibitors, gefitinib (Iressa) and erlotinib (Tarceva) (Shigematsu et al., 2005). Despite an initial promising response to these EGFR inhibitors, drug resistance eventually emerges (Kobayashi et al., 2005a; Pao et al., 2005). A secondary EGFR mutation, T790M, has been identified in NSCLC patients with an acquired resistance to the EGFR inhibitors. The EGFR double-mutants L858R/T790M and Del/T790M have enhanced oncogenic activity in vivo and in vitro compared to wild-type EGFR and EGFR with a single mutation (Godin-Heymann et al., 2007).

EGFR, a member of the ErbB family, functions as a receptor tyrosine kinase to transduce signals by extracellular growth factors, such as epidermal growth factor (EGF) and transforming growth factor-α (TGF-α). Following homo- or hetero-dimerization with other ErbB family members, membrane-bound EGFR is autophosphorylated and subsequently phosphorylates downstream targets including phosphoinositide 3-kinase (PI-3K), phospholipase C-γ (PLC-γ), extracellular signal-regulated kinase (Erk), and signal transducers and activators of transcription (STATs) to promote cell proliferation and survival. Compared to the cytoplasmic EGFR pathway, the nuclear EGFR pathway is relatively less understood. Clinical studies have indicated a significant inverse correlation between high levels of nuclear EGFR and overall survival rates in breast and ovarian cancers, suggesting that nuclear EGFR may play important roles in the pathogenesis of cancer (Lo et al., 2005b; Xia et al., 2009). In addition to the conventional role EGFR plays as a tyrosine kinase, nuclear EGFR also functions as a transcriptional co-activator to activate the expression of numbers of genes, such as CCND1, B-MYB, inducible nitric oxide synthase (iNOS), and COX-2 (Hanada et al., 2006; Lin et al., 2001; Lo et al., 2005a, 2010). Due to the lack of a DNA-binding domain, nuclear EGFR executes the transactivation activity through cooperation with transcription factors that have a DNA-binding domain. In this context, EGFR in concert with Stat3 induces the expression of iNOS and COX-2; likewise, EGFR cooperates with E2F1 to activate B-MYB.

The superfamily of actin-based myosin motor protein includes at least 25 members. The myosin II subfamily, which consists of cardiac, skeletal, and smooth muscle myosins and non-muscle myosin II (NM-II), is involved in several cellular activities, such as phagocytosis, cell adhesion, cell polarity, and intracellular particle trafficking (Berg et al., 2001; Hodge and Cope, 2000). The myosin II molecules are hexamers containing 2 heavy chains and 4 light chains. More than 15 different types of myosin II isoforms exist in vertebrates and each has a diverse myosin II heavy chain. In the NM-II subclass, NM-IIA, NM-IIB, and NM-IIC contain MYH9, MYH10, and MYH14 as the heavy chain, respectively. Myosin II binds to actin filaments to catalyze actin-based ATP hydrolysis leading to the conversion of chemical energy into mechanical force and movement.

In this study, we identified MYH9 as a novel EGFR-associating protein. A relatively strong EGFR binding intensity to MYH9 and β-actin was observed predominately in the nuclei of H1975 cells, an NSCLC-derived cell line carrying the L858R/T790M mutation. H1975 cells are commonly used to investigate the signaling consequence of NSCLC cells with acquired resistance rendered by the presence of the T790M mutation (Pao et al., 2005). Blebbistatin, a MYH9-specific inhibitor, was used to elucidate the importance of cytoskeleton integrity to the EGFR interaction with MYH9 and β-actin. Moreover, CL-387,785, an EGFR-T790M inhibitor that has shown efficacy in inducing growth arrest and apoptosis in H1975 cells, was used to determine whether EGFR kinase activity is required for the interactions among EGFR, MYH9, and β-actin (Kobayashi et al., 2005b, 2006). Because both CL-387,785 and blebbistatin had similar impacts on the interplay of these 3 proteins, it was examined whether blebbistatin could enhance the sensitivity of H1975 cells to CL-387,785. Taken together, the results from this study suggest a novel therapeutic concept that a network target may be a more effective strategy for the treatment of NSCLC compared to a single-targeted therapy.

2 Materials and methods

2.1 Cell lines and chemical agents

H322, H358 (courtesy of Dr. Jeou-Yuan Chen), H1650, and H1975 (American Type Culture Collection, Manassas, VA) cells were maintained in RPMI1640 medium (Invitrogen, Carlsbad, CA) containing 10% fetal bovine serum (FBS) (Sigma–Aldrich, St. Louis, MO) and penicillin/streptomycin (Invitrogen, Carlsbad, CA) at 37 °C. Gefitinib (Ryss Lab, Inc., Union City, CA), CL-387,785 (EMD Chemicals, Gibbstown, NJ), and blebbistatin (Tocris, Ellisville, MO) were commercially obtained. All chemicals were dissolved in DMSO for a final concentration of 10 μM.

2.2 Cellular fractionation

The cells were rinsed twice with PBS and resuspended in hypotonic lysis buffer (10 mM HEPES (pH 7.9), 10 mM KCl, 0.1 mM EDTA, 0.4% Triton X-100). The cells were lysed in lysis buffer with continuous rotation at 4 °C for 30 min. The lysate was centrifuged at top speed at 4 °C for 10 min. The supernatant was collected as the cytosolic fraction and the nuclear pellet was resuspended in hypertonic buffer (20 mM HEPES (pH 7.9), 0.4 M NaCl, 1 mM EDTA, 10% glycerol) with pipetting followed by vigorous shaking at 4 °C for 2 h. The sample was centrifuged at top speed for 10 min and the supernatant was collected as the nuclear extract.

2.3 Western blotting

The cells were lysed in lysis buffer (150 mM NaCl, 1% Triton X-100, 50 mM Tris, pH 8.0). The cell extracts were resolved on 10% SDS-PAGE gels and transferred to PVDF membranes (PerkinElmer, Waltham, MA). The membrane was blocked with 5% skim milk in TBS with 0.1% Tween-20 for 30 min at room temperature prior to overnight incubation with primary antibodies recognizing tubulin (Sigma–Aldrich, St. Louis, MO), MYH9 (Abnova, Taiwan), EGFR, Lamin B, COX-2 (all from Santa Cruz, Santa Cruz, CA), β-actin (Millipore, Billerica, CA), p-EGFR (Tyr1068), p-EGFR (Tyr845), p-Stat3 (Tyr705), Stat3, p-Akt (Ser473), Akt, p-Erk (Thr202/Tyr204), and Erk (all from Cell Signaling, Danvers, MA). The membranes were washed thrice in TBST and incubated with horseradish peroxidase-conjugated secondary antibodies in TBST for 1 h. After consecutive washes, the membranes were visualized with a chemiluminescence kit (PerkinElmer, Waltham, MA).

2.4 Immunoprecipitation

The lysates collected as previously described were incubated with an anti-EGFR or anti-MYH9 (Santa Cruz, Santa Cruz, CA) antibody overnight at 4 °C. The next day, the reactions were incubated with protein A/G plus-agarose (Santa Cruz, Santa Cruz, CA) at 4 °C. After 3 h, the beads were washed twice with washing buffer (150, 300, or 500 mM NaCl, 10% glycerol, 0.1% NP-40, and 0.1% Triton X-100). The pull-down complexes were eluted by boiling the beads in SDS-sample dye and were subjected to Western blotting or Coomassie blue staining.

2.5 Cell proliferation analysis

The cells were incubated with the indicated compounds for 3 days. The cell proliferation rate was determined by the MTS assay. RPMI 1640/MTS (Promega, Madison, WI)/PMS (Sigma–Aldrich, St. Louis, MO) (8: 2: 0.1) medium was added to each well, and the cells were incubated for 1.5 h. The absorbance was measured at 490 nm by PowerWavex (BioTEK instruments, Winooski, VT). The rate of cell proliferation was calculated as the absorbance ratio of treated to vehicle-treated cells.

2.6 Flow cytometry

The cells were treated with the indicated compounds for 3 days, harvested by trypsin-EDTA, and stained with Annexin V-FITC (BD Pharmingen, San Diego, CA) and propidium iodide (PI; Sigma–Aldrich, St. Louis, MO) in binding buffer containing 2.5 mM Ca+2. After 10 min, the samples were subjected to flow cytometry on a FACSCalibur machine (BD Bioscience, Franklin Lakes, NJ) for quantification of the apoptotic population using the CellQuest Pro software (BD Bioscience, Franklin Lakes, NJ). The cells in the early apoptotic stage were Annexin V-FITC positive and PI negative, whereas the cells in the late apoptotic stage were positive for both stains.

2.7 RNA interference

H1975 cells were transfected with control or MYH9 siRNA using Lipofectamine RNAiMAX (Invitrogen, Carlsbad, CA), following the manufacturer's instructions. The MYH9 siRNA target sequence (cgt tac tac tca ggg ctc atc tac a) was designed and synthesized by Invitrogen using BLOCK-iT™ RNAi Designer. The negative universal control (46–2001) siRNA was purchased from Invitrogen.

2.8 Real-time polymerase chain reaction

The total RNA was extracted by TRIzol reagent (Invitrogen, Carlsbad, CA) and was reverse-transcribed by M-MLV reverse transcriptase (Promega, Madison, WI). The cDNA of COX-2 was analyzed by real-time PCR in a LightCycler 1.0 system (Roche, Indianapolis, IN) using a KAPA SYBR® FAST qPCR Kit (Kapa Biosystems, Woburn, MA) following the manufacturer's recommendations. The primers used for the amplification of COX-2 were as follows: forward 5′-ttaatgagtaccgcaaacgc-3′ and reverse 5′- accagaagggcaggatacag-3′. The PCR amplification consisted of 40 cycles (95 °C for 2 s, and 60 °C for 30 s) after an initial denaturation step (95 °C for 1 min). Each sample was normalized to the individual GAPDH mRNA content.

3 Results

3.1 The identification of the cytoskeletal proteins MYH9 and β-actin as EGFR-associating proteins in H1975 cells

To determine whether the EGFR-T790M mutation alters the specificity of EGFR for substrates, certain EGFR-associating proteins were investigated in H1975 cells, which have been widely used to investigate molecular mechanisms of acquired drug resistance. EGFR-interacting partners were isolated by co-immunoprecipitation (co-IP) using an anti-EGFR antibody in cell lysates from H1975 and H322 cells, which are EGFR wt and were used as a control. Three unique bands were detected in lysates prepared from H1975 cells in a Coomassie blue-stained gel (Figure 1A). Two of the bands with a higher molecular weight were identified as MYH9 and the third band was identified as β-actin by in-gel trypsin digestion followed by matrix-assisted laser desorption/ionization-time of flight (MALDI-TOF) analysis. Hits which had a non-specific interaction with EGFR or an incorrect molecular weight were considered as insignificant hits. To confirm the identity of these proteins, co-IP was performed in H1975 and H358 (EGFR wt) cells followed by Western blotting analysis. Both cell lines expressed similar levels of EGFR, MYH9, and β-actin (Figure 1C). Increased levels of MYH9 and β-actin were detected in the anti-EGFR pull-down products in H1975 cells, whereas background levels of MYH9 were pulled down by the anti-EGFR antibody in H358 cells (Figure 1B). The resistance of EGFR-MYH9 and EGFR-β-actin interactions to a wash with high-salt buffer and the increase of salt concentration in the wash buffer magnifying the binding intensity of MYH9 and β-actin with EGFR suggested that a strong and hydrophobic interaction may exist between EGFR and MYH9 and β-actin.

Details are in the caption following the image
MYH9 and β-actin are identified as EGFR-associated proteins. (A) Whole cell extracts from H322 and H1975 cells were subjected to co-IP analysis using an anti-EGFR antibody. The proteins were separated on SDS-PAGE gels and detected by Coomassie blue staining. Filled star: MYH9; open star: β-actin. (B) An anti-EGFR co-IP was performed using crude lysates prepared from H358 and H1975 cells. During the co-IP procedure, agarose beads were washed twice with the wash buffer containing 150, 300, or 500 mM NaCl. Pull-down products were analyzed by Western blotting. (C) Levels of EGFR, MYH9, and β-actin were determined by Western blotting in H358 and H1975 cells.

3.2 EGFR interacts with MYH9 and β-actin in the nuclei of H1975 cells

To understand the role of MYH9 and β-actin in EGFR-involved pathways, we first investigated the location of the EGFR-MYH9 and EGFR-β-actin interactions. Co-IP was conducted to examine EGFR interactions with MYH9 and β-actin in the cytoplasmic and nuclear fractions. An additional EGFR-mutant NSCLC cell line, H1650 (EGFR-Del), was used to determine whether EGFR interactions with MYH9 and β-actin universally occur in EGFR-mutant NSCLC cells. As shown in Figure 2A, neither the nuclear nor the cytoplasmic EGFR showed detectable physical interactions with MYH9 and β-actin in H322, H358, or H1650 cells. The failure to detect EGFR interactions with MYH9 and β-actin in H1650 cells suggests that these interactions may uniquely occur in H1975 cells. Moreover, EGFR appeared to associate with MYH9 and β-actin predominately in the nuclei of H1975 cells, implying that MYH9 and β-actin may be involved in the EGFR nuclear pathway in H1975 cells. To understand why EGFR has different degrees of binding intensity to MYH9 and β-actin in diverse cell lines, the protein levels of all 3 proteins from cytoplasmic and nuclear fractions were examined in these cell lines (Figure 2B). Tubulin, a cytoplasmic marker, was detected in the cytoplasmic fractions, and lamin B, a nuclear marker, was only present in the nuclear fractions, indicating a clean separation of the cytoplasmic and nuclear fractions. The cytoplasmic EGFR levels of H358 and H1650 cells were relatively low compared to those of H322 and H1975 cells. There were no large differences in the protein levels of cytoplasmic MYH9 and β-actin among the 4 cell lines. Notably, relatively higher levels of EGFR were observed in the nuclei of H1975 cells compared to those in H322, H358, and H1650 cells, suggesting that abundant nuclear EGFR may at least partially contribute to the stronger binding intensity of EGFR with MYH9 and β-actin in H1975 cells.

Details are in the caption following the image
The EGFR interactions with MYH9 and β-actin occur in the nuclei of H1975 cells. (A) H322, H358, H1650, and H1975 cells were fractionated and the cytoplasmic and nuclear fractions were subjected to co-IP using an anti-EGFR antibody. (B) The cytoplasmic and nuclear fractions from (A) were separated by SDS-PAGE and detected by Western blotting. Lamin B and β-tubulin served as nuclear and cytoplasmic markers, respectively. (C) H1975 cells were serum-starved overnight and treated with EGF (100 ng/ml) for 0, 5, 30, or 45 min. The fractionated lysates were analyzed by Western blotting to determine the levels of EGFR in the cytoplasm and nucleus. (D) Overnight serum-starved H322 and H1975 cells were treated with EGF (100 ng/ml) for 0 or 45 min. Levels of MYH9 and β-actin were determined by Western blotting. An anti-EGFR co-IP was performed using fractionated lysates prepared from both cell lines.

Considering that the EGF ligand initiates EGFR endocytosis and subsequent EGFR accumulation in the nuclei of different cancer cell lines (Lo et al., 2006; Wang and Hung, 2009), we were curious whether EGF treatment could increase the levels of nuclear EGFR and enhance the EGFR interaction with MYH9 and β-actin in H1975 cells. However, the levels of nuclear EGFR in H1975 cells were not significantly altered by the addition of EGF up to 45 min (Figure 2C). Moreover, EGF treatment did not affect the EGFR interaction with MYH9 and β-actin in either H322 or H1975 cells as evidenced by the observation that those interactions did not change in response to EGF (Figure 2D).

3.3 Disrupted cytoskeleton integrity by blebbistatin reduces the EGFR interaction with MYH9 and β-actin

MYH9 is a heavy chain of myosin IIA that interacts with β-actin to maintain cytoskeleton integrity. To determine whether a fully-functioning cytoskeleton is required for EGFR-MYH9 and EGFR-β-actin interactions, blebbistatin was used to disrupt cytoskeletal integrity. Blebbistatin has been identified as a small molecule inhibitor for myosin IIA and has been shown to inhibit actin-activated MgATPase activity of myosin IIA (Straight et al., 2003). Subsequent characterizations revealed its selectivity and high affinity for several classes of myosin II (Limouze et al., 2004). Kinetic analysis indicated that blebbistatin binds to the myosin-ADP-inorganic phosphate complex with high affinity and disrupts the release of phosphate which traps myosin in a state with low actin affinity (Kovacs et al., 2004). In this study, the MTS assay was used first to determine the effects of blebbistatin on the proliferation of H1975 cells. As shown in Figure 3A, blebbistatin exhibited anti-proliferation effects against H1975 cells with an EC50 value of gt; 40 μM. The EC50 value is the effective concentration required for a certain compound to inhibit cell growth by 50%. In addition, a feature of dendritic morphology was observed in H1975 cells in the presence of 10 and 100 μM blebbistatin, suggesting its efficacy in H1975 cells (Figure 3B). As previously described (Kovacs et al., 2004), blebbistatin interrupted the physical interactions between MYH9 and β-actin as determined by the observation that reduced β-actin was detected in the pull-down products of an anti-MYH9 antibody in H1975 cells treated with blebbistatin for 2 h (Figure 3C).An increase of blebbistatin up to 100 μM did not further inhibit the interactions of MYH9 and β-actin. Further studies showed that higher concentrations of blebbistatin impaired not only the MYH9-β-actin interaction but also the EGFR interaction with MYH9 and β-actin, as shown by the decreased levels of MYH9 and β-actin pulled down by an anti-EGFR antibody in cells treated with 100 μM blebbistatin (Figure 3D). It was observed that a 2-hr treatment of blebbistatin did not significantly change the protein levels of EGFR, MYH9, and β-actin in the nuclei (Figure 3E); therefore, we excluded the possibility that the disruption of the EGFR interactions with MYH9 and β-actin by blebbistatin results from reduced levels of these 3 proteins. It is evident that blebbistatin can physically disrupt EGFR interaction with MYH9 and β-actin.

Details are in the caption following the image
Blebbistatin functions to weaken EGFR-MYH9 and EGFR-β-actin interactions. (A). The cell proliferation was determined by the MTS assay in H1975 cells treated with blebbistatin for 3 days. The proliferation of vehicle-treated cells was set at 100%. (B) The morphology of H1975 cells treated with vehicle, 10 μM, or 100 μM blebbistatin for 2 h was observed by light microscopy. The nuclear lysates prepared from H1975 cells treated with vehicle, 10, or 100 μM blebbistatin for 2 h were subjected to co-IP using (C) anti-MYH9 or (D) anti-EGFR antibodies. The pull-down products were detected by Western blotting analysis using anti-MYH9 and anti-β-actin antibodies. (E) The nuclear extracts from (C) (D) were subjected to SDS-PAGE, followed by Western blotting analysis. V: vehicle; Bars: 50 μm.

3.4 The down-regulation of MYH9 improves the sensitivity of H1975 cells to CL-387,785

The compound CL-387,785 was used to determine whether EGFR kinase activity is required for nuclear EGFR interactions with MYH9 and β-actin. CL-387,785 is an irreversible inhibitor that covalently binds to the ATP-binding region of EGFR (Discafani et al., 1999). Consistent with previous studies, CL-387,785 effectively inhibited the phosphorylation of EGFR and its downstream targets, ERK and AKT (Kobayashi et al., 2006), whereas gefitinib had a minimal effect on EGFR-mediated pathways in H1975 cells (Figure 4A). Co-IP was performed to examine the interactions among EGFR, MYH9 and, β-actin in H1975 cells treated with 0.1, 1, or 10 μM CL-387,785 for 2 h. CL-387,785 efficiently inhibited EGFR phosphorylation at Tyr1068 in the nuclei of H1975 cells but did not significantly affect the levels of nuclear MYH9 and β-actin after a 2-hr treatment (Figure 4B). EGFR kinase activity was not required for the EGFR interactions with MYH9 and β-actin because nuclear EGFR still associated with residual MYH9 and β-actin, despite EGFR-Tyr1068 being completely inhibited by 1 μM CL-387,785 (Figure 4B, C). Compared to low concentrations of CL-387,785, increasing CL-387,785 to 10 μM led to a further disruption of the EGFR-MYH9 and EGFR-β-actin interactions (Figure 4C). Surprisingly, CL-387,785, which is known as an EGFR kinase inhibitor, also functioned to weaken the interaction between MYH9 and β-actin, similar to blebbistatin (Figure 4D).

Details are in the caption following the image
The sensitivity of H1975 cells to CL-387,785 is enhanced by the down-regulation of MYH9. (A) Serum-starved H1975 cells were treated with gefitinib or CL-387,785 for 2 h prior to the treatment of EGF (100 ng/ml) for 5 min. The lysates were analyzed by Western blotting using the indicated antibodies. (B) The nuclear extracts from H1975 cells treated with CL-387,785 for 2 h were analyzed by Western blotting. The nuclear lysates prepared from (B) were subjected to co-IP using an (C) anti-EGFR or (D) anti-MYH9 antibody. (E) H1975 cells were transfected with 10 or 50 nM MYH9 siRNA. The lysates collected at the indicated time were subjected to Western blotting analysis to determine the protein levels of MYH9. (F) H1975 cells transfected with control siRNA or 50 nM MYH9 siRNA were seeded into 96-well plates. Twenty-four hours later, the cells were treated with vehicle or CL-387,785 for 72 h. The cell proliferation rate was determined by the MTS analysis. The proliferation of vehicle-treated cells from each group was set at 100%. V: vehicle; Ctrl: control.

Our results indicate physical interactions among EGFR, MYH9, and β-actin in H1975 cells; however, it remains unclear whether these interactions have functional significance. To elucidate this question, the functional significance of MYH9 in EGFR-mediated pathways was determined by ascertaining whether the down-regulation of MYH9 improved the efficacy of CL-387,785 in H1975 cells. Small-interfering RNA (siRNA) was used to knock-down MYH9 expression. Modest down-regulation of MYH9 was observed in H1975 cells 48 h after transfection with 10 nM MYH9 siRNA (Figure 4E). A higher concentration of MYH9 siRNA (50 nM) resulted in further down-regulation of MYH9. We examined the effects of MYH9 down-regulation on the sensitivity of H1975 cells to CL-387,785. H1975 cells were transfected with control or 50 nM MYH9 siRNA. After 24 h, cells were treated with vehicle (DMSO) or various concentrations of CL-387,785 for 72 h. MYH9 siRNA alone only caused minor cytotoxicity (<10%, data not shown). Because the proliferation of vehicle-treated cells in each group (transfected with control or MYH9 siRNA) was set at 100%, the cytotoxicity caused by MYH9 siRNA has been excluded. Treatment with CL-387,785 inhibited the cell proliferation of control siRNA-transfected H1975 cells; this inhibition was more prominent when MYH9 was down-regulated by siRNA treatment (Figure 4F), suggesting that the EGFR interaction with MYH9 is functionally significant and important for the proliferation of H1975 cells.

3.5 The disruption of cytoskeleton integrity by blebbistatin induces higher levels of apoptosis in CL-387,785-treated H1975 cells

Drug combinations have demonstrated clinical advantages in the treatment of different cancers; therefore, we were interested in understanding whether there is a cooperative interaction between an EGFR kinase inhibitor, CL-387,785, and a myosin inhibitor, blebbistatin. The pharmacological interactions between CL-387,785 and blebbistatin were determined by the isobologram and combination index (CI) methods. The data obtained from the MTS analysis were used to perform these analyses by the median-effect principle (CalcuSyn analysis) (Chou and Talalay, 1983). The individual doses of CL-387,785 and blebbistatin to achieve 90% growth inhibition (ED90; effective dose 90), 75% growth inhibition (ED75), and 50% growth inhibition (ED50) were plotted on the x- and y-axes. CI values that fall on the line represent an additive interaction between drugs, whereas CI values below or above the line represent a synergistic or antagonistic interaction, respectively. Figure 5A indicates that the combined treatment of CL-387,785 and blebbistatin synergistically inhibited the proliferation of H1975 cells by 50%, 75%, and 90%. To determine whether apoptosis was triggered synergistically by the combination of CL-387,785 and blebbistatin, the apoptosis signals were examined by flow cytometry analysis 3 days after treatment. Moderate doses of CL-387,785 (0.5 μM) and blebbistatin (20 μM) were selected to examine the combined effects on apoptosis in H1975 cells. The results from the flow cytometry analysis indicated that the combination of CL-387,785 and blebbistatin enhanced the accumulation of apoptotic cells compared to each agent alone (Figure 5B). Taken together, these observations suggest that a synergistic effect exists between CL-387,785 and blebbistatin for the inhibition of cell proliferation and the induction of apoptosis in H1975 cells. Because of the observation that each agent effectively disrupts the interaction of EGFR with MYH9 and β-actin, we next examined whether the cooperation of CL-387,785 and blebbistatin has synergistic effects on the physical interactions of these proteins. The protein interactions from cells treated with either agent or the combination were analyzed by co-IP. Although neither the single treatments nor the combined treatment caused significant changes in levels of MYH9 and β-actin, a synergistic reduction in the EGFR interactions with MYH9 and β-actin was observed in cells treated with the combined treatment compared to those with a single treatment (Figure 5C).

Details are in the caption following the image
Blebbistatin induces more apoptosis in H1975 cells treated with CL-387,785. (A) An isobologram analysis of the combination of CL-387,785 and blebbistatin in H1975 cells. The individual doses of CL-387,785 and blebbistatin to achieve 50%, 75%, and 90% growth inhibition were plotted on the x- and y-axes. (B) H1975 cells were treated with vehicle (DMSO), 0.5 μM CL-387,785, 20 μM blebbistatin, or the combination (0.5 μM CL-387,785 and 20 μM blebbistatin) for 3 days. The levels of apoptosis were analyzed by flow cytometry. The data represent 1 of the 3 sets of experiments. (C) An anti-EGFR co-IP was performed using nuclear extracts from H1975 cells treated with the indicated compounds for 2 h. The input extracts were analyzed by Western blotting to determine the levels of MYH9 and β-actin. The lysates prepared from H1975 cells treated with (D) the indicated concentration of CL-387,785 or blebbistatin (E) 0.1 μM CL-387,785, 5 μM blebbistatin, or the combination (0.1 μM CL-387,785 and 5 μM blebbistatin) for 8 h were analyzed by Western blotting. Real-time PCR was used to quantify the mRNA levels of COX-2. The transcript level of vehicle-treated cells was set at 100%. The Student's unpaired t-test was used to evaluate the differences between cells treated with blebbistatin and the combined treatment. V: vehicle.

In order to investigate whether the enhanced cytotoxicity of CL-387,785 and blebbistatin results from the suppression of the EGFR nuclear pathway, the levels of COX-2, a transcriptional target of nuclear EGFR, were examined. COX-2 catalyzes the biosynthesis of prostaglandins and is implicated in the invasiveness and angiogenesis of a number of cancers (Greenhough et al., 2009; Lo et al., 2010). A COX-2 inhibitor, celecoxib, has demonstrated anti-tumor effects in glioma xenografts (Nam et al., 2004). Our results showed that CL-387,785 alone dramatically inhibited the protein levels of COX-2, whereas blebbistatin had no effects on levels of COX-2 (Figure 5D). It has been proposed that nuclear EGFR activates the transcription of COX-2; therefore, we sought to understand whether the reduced protein levels of COX-2 resulted from a decrease in transcript levels of COX-2. As shown in Figure 5D, blebbistatin did not effectively inhibit the transcription of COX-2, whereas a tremendous reduction of COX-2 transcript was observed in cells treated with CL-387,785, implying that CL-387,785 functions to suppress the transcriptional activity of nuclear EGFR. Co-treatment with blebbistatin resulted in a further decline in transcript and protein levels of COX-2 in cells treated with CL-387,785 (Figure 5E).

4 Discussion

It is well studied that MYH9, as the heavy chain of non-muscle myosin IIA, binds directly to actin filaments to catalyze the ATP hydrolysis leading to the conversion of chemical energy into mechanical movement. In addition to MYH9, EGFR is also found to have a direct interaction with actin in a previous report (den Hartigh et al., 1992). A more recent study indicates that the interaction between EGFR and actin occurs mainly in lysosomes, suggesting that the EGFR association with actin may be involved in the internalization of EGFR/EGF complex into lysosomes (Song et al., 2008). In this study, EGFR was found to interact with MYH9 and β-actin in H1975 cells harboring an oncogenic mutation in EGFR but not in H322 and H358 cells with wild-type EGFR. Moreover, our findings suggested a possible new role the cytoskeletal elements MYH9 and β-actin may play in EGFR-involved pathways. Because the interaction of EGFR with MYH9 and β-actin predominately occurred in the nucleus of H1975 cells, we hypothesized that MYH9 and β-actin may be implicated in the EGFR nuclear pathway.

Evidence showing that ErbB family members can be transported from the plasma membrane to the nucleus has been reported (Wang and Hung, 2009). Nuclear EGFR in a full-length form has been discovered in a variety of cancer cells, such as breast, bladder, ovary, and oral cavity cancers (Lo, 2010). Clinical studies have indicated a significant inverse correlation between high levels of nuclear EGFR and the overall survival rate in patients with breast and ovarian cancers, suggesting that nuclear EGFR is a prognostic indicator for poor clinical outcome (Lo et al., 2005b; Xia et al., 2009). Despite the lack of investigations on the clinical significance of nuclear EGFR in NSCLC, it has been revealed that nuclear EGFR contributes to the resistance of NSCLC cells to cetuximab, an EGFR-blocking antibody used to treat patients with head and neck cancers and colorectal cancers (Li et al., 2009). Increased expression levels of EGFR ligands, nuclear EGFR, and Src family kinase (SFK) are detected in cetuximab-resistant NSCLC cells. Li et al. suggest that SFK is responsible for the EGF-induced EGFR nuclear transport, which may be an essential indicator of resistance to cetuximab therapy. A more recent study in NIH3T3 fibroblasts suggests a role for the nuclear EGFR in modulating the repair of damaged DNA following cisplatin and ionizing radiation treatment (Liccardi et al., 2011).

Nuclear EGFR has been shown to act as a transcriptional co-factor in concert with Stat3, E2F1, and Muc1 to activate the transcription of iNOS, B-MYB, and CCND1, respectively (Bitler et al., 2010; Hanada et al., 2006; Lo et al., 2005a). Likewise, the importance of nuclear β-actin and nuclear myosin I (NMI) has been demonstrated in transcription. β-actin plays a fundamental role in transcription as determined by the observations that β-actin interacts with RNA polymerase II and co-localizes with transcription sites in the early mouse embryo (Grummt, 2006; Nguyen et al., 1998; Smith et al., 1979). Hofmann et al. revealed that β-actin is recruited to the promoter of the interferon-γ-inducible MHC2TA and G1P3 genes in the presence of interferon-γ and the depletion of β-actin prevents the formation of pre-initiation complexes (Hofmann et al., 2004). Like β-actin, NMI, a monomeric and single-headed myosin, is also found to co-localize with RNA polymerase II and is required for RNA polymerase II-mediated transcription (Pestic-Dragovich et al., 2000). It has been speculated that β-actin and NMI are involved in the initiation and transition into an elongation complex during the transcription process (de Lanerolle et al., 2005).

Myosin II has been found to be part of the transcriptional machinery along with NMI (Li and Sarna, 2009). In human colonic circular smooth muscle cells, myosin II was found to have a physical interaction with RNA polymerase II and transcription factor II B (TFIIB) and to bind to the promoter of the ICAM-1 gene, which is involved in smooth muscle dysfunction during colonic inflammation (Li and Sarna, 2009; Pazdrak et al., 2004). Overexpression of both smooth muscle and non-muscle light chains results in an increase in the transcriptional activity of the ICAM-1 gene. Taken together, accumulating evidence suggests the implication of myosin II in transcriptional activity. In this work, we found that the EGFR-MYH9-β-actin complex was formed predominately in the nucleus and that perturbation of the EGFR-MYH9-β-actin interaction by blebbistatin and CL-387,785 enhanced the reduction of transcript and protein levels of an EGFR nuclear target, COX-2 (Figure 5E). Considering the present understanding of the role each protein plays in the transcription, we speculate that actin-based myosin IIA may facilitate EGFR-mediated transcription of target genes in H1975 cells. Certainly, additional investigations are needed to further characterize the interplays between EGFR and the actin-based myosin IIA in transcription activation.

Lung cancer is a threat to public health worldwide due to its poor prognosis. Although progress has been made in the treatment of NSCLC through the advent of targeted therapies such as EGFR kinase inhibitors, treatments that are more effective are still urgently needed due to the emergence of acquired resistance in patients who initially respond to current EGFR inhibitors. Presently, it is clear that diseases like cancer are substantially more complex than initially anticipated because multiple molecular abnormalities result in the pathogenesis instead of a single deficiency. A growing perception is that effective strategies for the treatment of cancers should target not just a single protein but rather a global network of molecular pathways (Pujol et al., 2010). In this study, we demonstrate for the first time that blebbistatin, an agent that functions to perturb the cytoskeletal integrity, enhanced the sensitivity of H1975 cells to an EGFR kinase inhibitor. Our findings suggest a promising avenue for the development of a more effective treatment with the cytoskeletal network as the target of a combination of anti-cancer agents.

Conflict of interest

None declared.

Acknowledgements

This work was supported by grants from the National Health Research Institutes in Taiwan and the Translational Medicine Grant contract NSC 100-2321-B-400-006 from National Science Council, Taiwan.