Open Access

Silencing Aurora A leads to re-sensitization of breast cancer cells to Taxol through downregulation of SRC-mediated ERK and mTOR pathways

  • Authors:
    • Yan Li
    • Wanqi Zhou
    • Ke Tang
    • Xiaoguang Chen
    • Zhiqiang Feng
    • Jindong Chen
  • View Affiliations

  • Published online on: August 14, 2017     https://doi.org/10.3892/or.2017.5908
  • Pages: 2011-2022
  • Copyright: © Li et al. This is an open access article distributed under the terms of Creative Commons Attribution License.

Metrics: Total Views: 0 (Spandidos Publications: | PMC Statistics: )
Total PDF Downloads: 0 (Spandidos Publications: | PMC Statistics: )


Abstract

While Taxol has been reported to improve the clinical survival of breast cancer patients, subsequently developed drug-resistance of the cancer cells limits its final efficacy and applications. Previous studies suggested that Aurora A is involved in the development of the Taxol-resistance of breast cancer. We established Taxol-resistant breast cancer MCF-7/T cells and xenograft models to explore the role of Aurora A in Taxol resistant ER-positive breast cancer. Compared with their parental MCF-7/C cells, the Taxol-resistant MCF-7/T cells exhibited enhanced colony formation, less cell death and higher invasive ability. The resistant cells presented overexpressed Aurora A, elevated phosphorylated SRC and upregulated Ras/Raf/ERK and Akt/mTOR pathways. Silencing of Aurora A reduced the activity of SRC and downregulated the ERK and Akt/mTOR pathways, which led to re-sensitization of the resistant MCF-7/T cells to Taxol in vitro. These results suggested that the activation of Aurora A and the subsequent upregulation of ERK and Akt through SRC induced Taxol-resistance in breast cancer cells, and inhibiting Aurora A and the related SRC/EKT/Akt pathway could restore the sensitivity of breast cancer cells to Taxol. These results might shed light on the development of strategies to circumvent Taxol-related chemoresistance in breast cancer clinical practice.

Introduction

Breast cancer is one of the major causes of cancer-related death in women. Moreover, the incidence of breast cancer continues to rise in the world. In the treatment of breast cancer, Taxol/Paclitaxel is still regarded as one of the most effective drugs (1), which could prolong the survival of breast cancer patients in combination with other chemotherapy drugs (2). Unfortunately, continuous use of Taxol results in the acquired resistance of tumor cells to this drug. However, the molecular mechanisms of resistance to Taxol in breast cancer are not fully understood (35).

Aurora kinase has been observed highly expressed and amplified in breast cancer, which is related to survival and tumorigenesis (6,7). Moreover, overexpression of Aurora A induces chemoresistance in breast cancer cells and ovarian cancer cells (8). In the clinical trials for breast cancer, the expression level of Aurora A was related with the resistance of cancer cells to Taxol. The exact role of Aurora A in Taxol-resistant breast cancer remains unclear. A previous report has shown that inhibition of Aurora A expression could result in the enhancement of Taxol-induced apoptosis in a variety of cancer cell lines, such as head and neck, esophageal and breast cancer cell lines (9,10).

It has been established that Taxol inhibits signal pathways related to cell survival and growth, such as Ras/ERK and Akt kinase pathway (1113). However, the Ras/ERK and Akt pathways were overexpressed in drug-resistant ovarian and colorectal cancers (14,15). Furthermore, the overexpression of Akt and Ras/Raf/ERK pathways might be associated with the phosphorylation of SRC (16). As Aurora A was reported to promote proliferation and invasion through activation of the SRC and upregulate downstream pathways such as Ras/Raf/ERK and Akt/mTOR pathway (17,18), we speculated that activated Aurora A might induce Taxol-resistance of breast cancer cells through SRC/ERK or SRC/Akt pathway. In the present study, by using our MCF-7/T cell and xenograft tumor models, we aimed to determine whether inhibition of Aurora A expression in Taxol-resistant breast cancer could downregulate the expression of SRC and downstream signaling pathways such as Ras/ERK and Akt and would suppress the tumor cell growth and restore cancer cell chemosensitivity to Taxol.

Materials and methods

Cell lines

MCF-7 cells were obtained from the Cell Center of CAMS & PUMC (Beijing, China). The cells were maintained in Dulbeccos modified Eagles medium (DMEM; Invitrogen, Carlsbad CA, USA) (19) supplemented with 10% fetal bovine serum (FBS; Invitrogen), 100 IU/ml penicillin and 100 µg/ml streptomycin (Invitrogen) in a humidified incubator containing 5% CO2 at 37°C.

Establishment of the Taxol-resistant MCF-7 model in vitro and in vivo

In the present study, MCF-7 resistant cell line MCF-7/T was developed through continued administration of sub-lethal concentrations of Taxol to the cells. A highly invasive and low colony was obtained from the MCF-7/T cells. In brief, MCF-7/T colonies were selected after 18 pulse drug treatments with Taxol. The majority of the cells were dead following 24 h of exposure to Taxol. The survived cells were then washed with 0.01 mol/l phosphate-buffered saline (PBS) and cultured in Taxol growth medium. After 1–2 days, the dead cells were washed out with PBS and fresh Taxol medium was added. Once the cells reached 70–80% confluence, they were preserved for further study. The Taxol-resistant cell line was stabilized for ~6 months after treatment initiation and the resistant phenotype was developed. For the maintenance of Taxol-resistant cells, the MCF-7/T cells were grown in the presence of 0.001 µmol/l Taxol. Prior to experimentation, MCF-7/T cells were maintained in a Taxol-free culture medium and subcultured for at least three generations. The drug-resistant characteristics of the MCF-7/T cells were tested using various concentrations of Taxol.

Taxol-resistant cells lose the resistant characteristics after injection into nude mice

To obtain an in vivo Taxol-resistant MCF-7 xenograft model (MCF-7/T) for studying resistance mechanisms frequently observed in a human therapeutic setting, MCF-7 cells were injected subcutaneously into the left flanks of the athymic nude mice (Balb/c-nu/nu females, 6–8 weeks old) which were purchased from Vital River Laboratory Animal Technology, Co., Ltd. (Beijing, China) and housed in the controlled environment at 25°C on a 12-h light/dark cycle. Mice were maintained following the rules of the National Institute of Health Guide for the Care and Use of Laboratory Animals. Taxol-resistant MCF-7 xenograft tumors were achieved after ten passages of Taxol treatment. For each passage, mice were treated with 30.0 mg/kg Taxol 24 h before sacrifice. Then, xenograft tumors were collected and transplanted into the new athymic nude mice. After ten passages (~12 months), drug-resistant characteristics of the xenografts were determined by the absence of tumor regression after treatment of Taxol.

Tissue culture from MCF-7 and Taxol-resistant MCF-7 xenograft model

Tumor tissues from a Taxol-resistant MCF-7 and a parent MCF-7 xenograft model were cut into small pieces with surgical scissors and minced with sterile razor blades until the explants size was <1 mm3. The explants were transferred to flasks, trypsinized (Invitrogen) for 30 min, covered with complete medium and incubated in an atmosphere of 5% CO2 and 95% air at 37°C. The medium was replaced after 48 h.

Generation of the stable knockdown Aurora A in the MCF-7 and MCF-7/T cell

MCF-7 and MCF-7/T cell was seeded at a density of 1×105 cells/well in 6-well plates and incubated overnight or until cells reached 50% confluence. They were then transfected with either the AurA microRNAs, or control microRNA vector (BLOCK-iT™ Pol II miR RNAi Expression Vector kit with EmGFP, purchased from Invitrogen) through Lipofectamine 2000 (Invitrogen) following the manufacturer's protocol. The transfected cells were initially selected in DMEM medium containing 8 µg/ml Blasticidin S HCl (Invitrogen). Selective pressure was maintained in a medium containing 8 µg/ml Blasticidin S HCl for two weeks. Clones with green fluorescence were collected for further culture in regular media. Then, cells were harvested for western blot analysis of Aurora A expression. Two stable transfected cell clones with AurA microRNAs, were designated as MCF-7/T/AurA1 and MCF-7/T/AurA2. Stable transfected cells with control microRNA were designated as MCF-7/C and MCF-7/T/C (10). Since Aurora A-miRNA silencing constructs express GFP (BLOCK-iT™ Pol II miR RNAi Expression Vector kit with EmGFP) which would interfere with the flow cytometry (Annexin V-FITC) and TUNEL assay, we did not use flow cytometry (Annexin V-FITC) and TUNEL assay to detect apoptosis in the following experiments.

Analysis of cell proliferation and viability

MCF-7/C, MCF-7/T/C, MCF-7/T/AurA1 and MCF-7/T/AurA2 cells were seeded in 96-well plates in DMEM medium supplemented with 10% fetal bovine serum (FBS). The proliferation of the cells was monitored by CCK-8 assay every day for 14 days. MCF-7/C, MCF-7/T/C, MCF-7/T/AurA1 and MCF-7/T/AurA2 cells were seeded in 96-well plates in DMEM medium supplemented with 10% FBS. Cells were treated with dimethyl sulfoxide (DMSO) or Taxol for 72 h and then cell viability was measured with CCK-8 assay.

Colony formation assay

MCF-7/C, MCF-7/T/C, MCF-7/T/AurA1 and MCF-7/T/AurA2 cells were trypsinized to single-cell suspensions. Then, cells were diluted in DMEM culture medium containing 10% FBS, and 300–600 cells were plated in each well of the 6-well plates. Cells were incubated with 5% CO2 at 37°C for 14 days, and colonies were washed with PBS, fixed and stained with 0.005% crystal violet in methanol. Numbers of colonies were manually counted. Experiments were performed in triplicate and were repeated thrice.

Cell death and cell cycle analysis

MCF-7/C, MCF-7/T/C, MCF-7/T/AurA1 and MCF-7/T/AurA2 cells were treated with either Taxol, or 0.1% DMSO for 72 h, washed in PBS, and fixed with ice-cold 70% ethanol overnight. Cells were then suspended in PBS containing RNase A (100 µg/ml) and propidium iodide (50 µg/ml) and 0.1% Triton X-100, and incubated in the dark for at least 1 h (20). Cell cycle profiles and death population were determined by flow cytometric (FCM) analysis.

In vitro invasion assay

Invasion was determined using a variation of the Boyden chamber assay as previously described (21). Briefly, cells were trypsinized and counted; next, 1×106 cells (MCF-7/C, MCF-7/T/C, MCF-7/T/AurA1 and MCF-7/T/AurA2) and cells cultured from tumor tissue (MCF-7, MCF-7/T xenograft model) suspended in 200 µl of DMEM containing 0.1% BSA. The cells were seeded into the upper compartment (Costar, Costar, Cambridge, MA, USA) coated polycarbonate filter with a pore size of 8.0 µm in a 24-well plate. Each polycarbonate filter was coated with 10 µl of 0.5% Matrigel before the addition of cells. DMEM medium (500 µl) containing 10% FBS was added to the lower compartment as a chemoattractant. After 18 h of incubation at 37°C with 5% CO2, the cells on the lowerside of the filter were fixed to the membrane using methanol for 10 min. Filters were stained with hematoxylin and eosin (H&E) at room temperature. Cells in the upper compartment were removed using a cotton swab, leaving only the cells on the underside of the filter, representing those cells that had successfully invaded across the Matrigel-coated filter. The chambers were then photographed to compare the amount of invasive cells on the underside of the membrane. Five visual fields were photographed in every membrane and nuclear-stained cells were manually counted. All samples were run in triplicate.

Cell lysates and western blot analysis

Lysates (from MCF-7/C, MCF-7/T/C, MCF-7/T/AurA1 and MCF-7/T/AurA2 cells) were prepared following the method previously described. Portion of three randomly selected tumors from each group (MCF-7 and MCF-7/T) were homogenized for lysate preparation as previously described (22). For western blot analysis, samples were transferred to a nitrocellulose membrane by semi-wet electrophoresis (Invitrogen), and incubated with primary antibodies (rabbit anti-Aurora A, rabbit anti-phosphorylated Aurora A (Thr288), rabbit anti-phosphorylated SRC (Tyr416), rabbit anti-SRC, rabbit anti-phosphorylated Akt (Ser473), rabbit anti-Akt, rabbit anti-phosphorylated mTOR, rabbit anti-mTOR, rabbit anti-phosphorylated p70s6 (Thr389), rabbit anti-phosphorylated c-Raf (Ser259), rabbit anti-c-Raf, rabbit anti-phosphorylated MEK, rabbit anti-MEK, rabbit anti-phosphorylated p-ERK, rabbit anti-ERK, mouse anti-cyclin B, mouse anti-cdc2, rabbit anti-Bcl-2, rabbit anti-c-caspase 3, rabbit anti-c-PARP, rabbit anti-MMP9, rabbit anti-MMP2 and rabbit anti-β-actin) overnight at 4°C, detected with horseradish peroxidase-conjugated anti-rabbit or anti-mouse IgG (Santa Cruz Biotechnology, Santa Cruz, CA, USA) and developed using an ECL Western blot detection and analysis system (Applygen Technologies, Inc., Beijing, China). Membranes were tested for equal loading by probing for β-actin.

ELISA assay

Lysates (portion of three randomly selected tumors from MCF-7 and MCF-7/T) were mixed and homogenized for lysate preparation as previously described (22). For ELISA assay, the protocol of Phospho-Aurora A (Thr288) sandwich ELISA kit (#7114; Cell Signaling Technology, Danvers, MA, USA) was followed.

Statistical analysis

All values were expressed as mean ± SD. Values were compared using the Student's t-test. P<0.05 was considered significant.

Results

Activation of Aurora A and SRC in Taxol-resistant MCF-7 model (MCF-7/T) in vitro and in vivo

To examine whether Aurora A activation was related to the resistance of breast cancer to Taxol, we established a Taxol-resistant breast cancer cell clone MCF-7/T in vitro and developed two xenograft tumor models, the Taxol-resistant MCF-7/T and the Taxol-sensitive parental MCF-7. When a Taxol dose of 5.0 mg/kg was adopted, the parental Taxol-sensitive MCF-7 tumors exhibited significant growth inhibition while the MCF-7/T xenografts presented resistance to Taxol. The relative tumor volume (RTV) of MCF-7 was 5.06±2.46 (control) vs. 0.34±0.22 (Taxol 5.0 mg/kg) (Table I and Fig. 1A and B), whereas the RTV of MCF-7/T was 12.19±2.78 (control) vs. 12.59±3.17 (Taxol 5.0 mg/kg) (Table II and Fig. 1A and C). The results indicated that MCF-7/T tumors had developed resistance to Taxol in vivo and was suitable for further study.

Table I.

Effects of Taxol on MCF-7 tumors in athymic mice.

Table I.

Effects of Taxol on MCF-7 tumors in athymic mice.

Body weight (g)Tumor sizeTumor weight



Dose (mg/kg)No. of animals (n)StartEndVolume (mm3)RTVT/C (%)(g)Inhibition (%)
Control 5/522.6±0.626.0±1.21163.8±503.25.06±2.46 1.02±0.46
Taxol5.05/521.0±1.622.8±2.3 68.3±32.5a 0.34±0.22a6.75 0.04±0.02a96.36

a P<0.01 vs. control. T/C (%) = TRTV/CRTV × 100; RTV, relative tumor volume.

Table II.

Effects of Taxol on MCF-7/T tumors in athymic mice.

Table II.

Effects of Taxol on MCF-7/T tumors in athymic mice.

Body weight (g)Tumor sizeTumor weight



Dose (mg/kg)No. of animals (n)StartEndVolume (mm3)RTVT/C (%)(g)Inhibition (%)
Control 5/518.0±1.418.6±2.41373.5±363.112.19±2.78 1.26±0.24
Taxol5.05/416.8±1.917.5±2.11871.0±981.512.59±3.17103.31.36±0.48

[i] T/C (%) = TRTV/CRTV × 100; RTV, relative tumor volume.

We further examined the expression of Aurora A in MCF-7/C and MCF-7/T/C cell lines, as well as MCF-7 and MCF-7/T xenograft tumors. Western blot analysis revealed that both p-Aurora A and Aurora A were highly expressed in Taxol-resistant MCF-7/T cells and xenograft tumors analyzed (Fig. 1D and E). ELISA further confirmed that the Aurora A was activated in MCF-7/T xenograft tumors (Fig. 1F). Given previous report (23) suggested that Aurora A might function through phosphorylation of SRC at tyrosine 416, we herein validated that the level of p-SRC and SRC was upregulated in MCF-7/T cells and tumors as well (Fig. 1D and E). These results implied that the activation of Aurora A and SRC might be linked to the resistance of the breast cancer to Taxol.

Specific silencing of Aurora A in MCF-7/T cells led to down-regulation of SRC phosphorylation

To further determine the partnership between Aurora A and SRC in resistance to Taxol, MCF-7/T cells were transfected with either vector-containing human-specific Aurora A microRNAs or a vector-containing control microRNA. Two Aurora A microRNA stable-expressing clones (named as MCF-7/T/AurA1 and MCF-7/T/AurA2) were chosen for subsequent experiments. Our western blot analyses displayed that Aurora A as well as p-Aurora A were downregulated significantly in MCF-7/T/AurA1 and MCF-7/T/AurA2 cells compared to control microRNA-expressing cells (MCF-7/T/C) (Fig. 2A). Concurrently, SRC phosphorylation significantly decreased while the total of SRC was not influenced (Fig. 2A). These data indicated that SRC activity might be regulated by Aurora A in MCF-7/T cells.

Knockdown of Aurora A inhibits MCF-7/T cell proliferation

To explore whether high expression of Aurora A contributes to cell proliferation, the MCF-7/T/AurA1 and MCF-7/T/AurA2 cells were monitored in vitro by CCK-8 assay for 14 days. While there was no significant cell viability difference between MCF-7/C and MCF-7/T/C (Fig. 2Ba), the MCF-7/T cell proliferation significantly decreased upon silencing of Aurora A (MCF-7/T/AurA1 and MCF-7/T/AurA2) in a time-dependent manner compared with the negative MCF-7/T/C controls, and the highest inhibitory rates were 65.1 and 47.5% (Fig. 2Bb) on the 8th day, respectively. In addition, our colony formation assay indicated that the number of colonies in the MCF-7/T/C cells was higher than in MCF-7/C cells (average colony number, 305.7±4.0 and 260.3±6.4, respectively) (Fig. 2Ca), whereas the number of colonies in the MCF-7/T/AurA1 and MCF-7/T/AurA2 cells (average colony number, 221.3±31.9 and 330.7±10.07, respectively) were significantly decreased compared to the MCF-7/T/C control cells (average colony number, 544.7±7.6) (Fig. 2Cb).

Silencing of Aurora A induces MCF-7/T cells death

To study the impact of Aurora A expression on cell death in Taxol resistant breast cancer cells, we examined the percentage of SubG1 phase in MCF-7/T sublines (MCF-7/T/C, MCF-7/T/AurA1 and MCF-7/T/AurA2) by FCM. The results revealed, that when both MCF-7/C and MCF-7/T/C cells were cultured for 72 h, the percentage of MCF-7/T/C cells in the SubG1 (3.8±0.9%) was lower than the parental MCF-7/C population (13.4±1.1%) (Fig. 3Aa). Moreover, in the silenced MCF-7/T/AurA1 and MCF-7/T/AurA2 cells, the SubG1 phase percentage was increased by 20.2±2.3 and 19.5±2.0% compared with control MCF-7/T/C cells (3.8±0.9%) (Fig. 3Ab). These results demonstrated that downregulation of Aurora A could induce MCF-7/T cell death.

Knockdown of Aurora A causes G2/M arrest in MCF-7/T cells

As Aurora A could exert positive effects on the cell cycle in several cell lines in a dose-dependent manner, we further examined whether knockdown of Aurora A would lead to cell cycle arrest in MCF-7/T. The results showed that the percentage of G2/M phase in the Taxol-resistant MCF-7/T/C cells was lower than that in the MCF-7/C control cells. The percentages of the G2/M cells were 14.3±1.3% for MCF-7/C and 20.9±1.3 for MCF-7/T/C (Fig. 3Ba). We observed that inhibition of Aurora A by microRNAs led to cell G2/M-phase accumulation (Fig. 3Bb) and G1-phase decrease in the Taxol-resistant cells. The cell percentages of G2/M phase in MCF-7/T/AurA1 and MCF-7/T/AurA2 cells increased by 26.4±0.8 and 25.4±0.7%, respectively, compared with 20.8±1.5% in MCF-7/T/C control cells (Fig. 3Bb).

Silencing of Aurora A inhibits MCF-7/T cell invasion in vitro

To determine the effect of elevated Aurora A on MCF-7/T invasive capacity, we further conducted invasion assay. The MCF-7/T sublines and MCF-7/C control cells were allowed to invade through a membrane coated with Matrigel, toward a chemo-attractant (10% FBS) for 18 h. While only 32.5±2.1/well MCF-7/C cells exhibited invasion, there were 327.5±9.2/well MCF-7/T/C cells invaded through the membrane, indicating MCF-7/T/C cells presented an enhanced invasive ability compared with MCF-7/C cells (Fig. 4A and B). Moreover, the xenograft-originated Taxol-resistant MCF-7 cells [MCF-7/T(vivo)] were more invasive than the parent xenograft-originated MCF-7 cells [MCF-7(vivo)], [MCF-7/T(vivo), 141.0±9.9 vs. MCF-7(vivo), 42.5±3.5] (Fig. 4A and B). After knockdown of Aurora A by microRNAs, the invaded cells decreased by 80.7% (MCF-7/T/AurA1, 60.5±4.9/well) and 71.0% (MCF-7/T/AurA2, 91.0±12.7/well) compared to the untreated control MCF-7/T/C cells (313.5±9.20/well) (Fig. 4C and D), suggesting that inhibition of Aurora could considerably reduce the invasive ability of MCF-7/T cells.

Suppression of Aurora A downregulates the Raf/ERK and Akt pathway in MCF-7/T cells

Aurora A interacts with several proteins related to proliferation, survival, cell cycle, apoptosis, and invasive signaling pathways such as Raf/ERK and Akt (2426). We examined whether knockdown of Aurora A would downregulate the ERK and Akt pathways in the MCF-7 and MCF-7/T subline cells and xenograft tumors. As shown in Fig. 5Aa, 5Ab, 5Ba and 5Bb, Akt, mTOR, p70s6, c-Raf, MEK and ERK were highly phosphorylated in the Taxol resistant MCF-7 cells and tumors. Whereas, the phosphorylation and basal levels of these proteins were remarkably reduced in MCF-7/T/AurA1 and MCF-7/T/AurA2 cells compared with the negative control MCF-7/T/C (Fig. 5Ac and 5Bc), indicating inhibition of Aurora A significantly downregulated the Ras/Raf/ERK and Akt pathway.

Since cyclin B/cdc2 complex regulates cell cycle (27), we herein found that the cyclin B and cdc2 were overexpressed in MCF-7/T cells and xenograft tumors (Fig. 5Ca and 5Cb). Nevertheless, the expressions of cyclin B and cdc2 were decreased in the MCF-7/T/AurA1 and MCF-7/T/AurA2 cells though the decrease of cyclin B was not significant in the MCF-7/T/AurA1 cells (Fig. 5Cc), suggesting that Aurora A regulates cell cycle through cyclin B/cdc2 in MCF-7/T cells. In addition, knockdown of Aurora A downregulated Bcl-2 and induced the cleavage of caspase-3 and PARP in MCF-7/T/AurA1 and MCF-7/T/AurA2 cells, suggesting that knockdown of Aurora A might enhance apoptosis (Fig. 5Cc).

MMPs play an important role in degrading extracellular matrix components and promoting invasion of tumor cells (28). We observed that the expression of MMP2 and MMP9 was much higher in MCF-7/T/C cells than that in the MCF-7/T/AurA1 and MCF-7/T/AurA2 cells (Fig. 5Da and Db), implying that silencing Aurora A might inhibit the invasion of MCF-7/T cells.

Inhibition of Aurora A restores the sensitivity of MCF-7/T/C to Taxol in vitro

To investigate the chemosensitivity of MCF-7/T/C cells to Taxol after knockdown of Aurora A, we performed dose-response experiments to measure IC50 values with Taxol treatment for 72 h. As expected, the MCF-7/T/C cells showed lower sensitivity to Taxol (IC50=85.72±3.41 nmol/l) than MCF-7/C cells (IC50=0.43±0.022 nmol/l) (Fig. 6A). In contrast, inhibition of Aurora A increased the susceptibility of MCF-7/T/C to Taxol, the IC50 values for MCF-7/T/AurA1 and MCF-7/T/AurA2 reduced to 12.67±0.20 and 3.25±0.77 nmol/l, respectively. The sensitivity to Taxol advanced 6.8- and 26.4-fold in MCF-7/T/AurA1 and MCF-7/T/AurA2 cells, respectively, as compared with that in the MCF-7/T/C cells (Fig. 6A). These results indicated that inhibition of Aurora A resulted in enhanced chemosensitivity in Taxol-resistant breast cancer cells.

Inhibition of Aurora A enhances the Taxol-induced MCF-7/T cell death

The MCF-7/T cells were administered with Taxol for 72 h and cell death (SubG1) was evaluated after PI staining by FCM. Taxol induced death in a dose-dependent manner. When the cells were treated with 1.0 nmol/l Taxol, the percentages of death cells in MCF-7/C and Taxol-resistant MCF-7/T/C cells were 42.53±2.87 and 7.78±0.92%, respectively. The percentage of dead MCF-7/T/C cells elevated to 24.83±3.33% at a dose of 10.0 nmol/l of Taxol (Fig. 6B). Whereas, the MCF-7/T/AurA2 knockdown cells presented much higher dead cell percentages after Taxol treatment, 25.57±2.82% (1.0 nmol/l) and 48.03±2.58% (10.0 nmol/l), suggesting that inhibition of Aurora A in MCF-7/T cells could enhance Taxol-mediated death.

Discussion

Taxol resistance is a major obstacle to successful chemotherapy in cancer patients. In our previous study, we detected that p-Aurora A, along with p-gp, was highly expressed in MX-1/T and MCF-7/T cells, and demonstrated that kinase Aurora A was related with tumor resistance in Taxol resistance MX-1/T partly through p-ERK/P-gp (10). However, that study focused on the three triple-negative breast cancer cells, and the mechanism of drug-resistance was only involved in the P-gp and p-ERK. As is known, besides p-gp, Taxol resistance might be attributed to the upregulation of several factors including the Class III β-tubulin (TUBB3) in lung cancer (19,29), PI3K/Akt/mTOR (3033) in breast and renal tumors, NF-κB, FGFR2 and CHK2 (34) in gastric neoplasia (35,36) and SRC in ovarian carcinomas (16,37). In addition, overexpression of Aurora A has been identified in the chemo-resistant breast and ovarian cancer cells (8,38). Thus, we want to further elicit the function of Aurora A in Taxol-resistant ER-positive breast cancer, and investigate the mechanism of resistance related with Aurora A except p-gp.

In this study, we demonstrated that overexpressed Aurora A was associated with the proliferation, invasion, and Taxol-resistance of the breast cancer cell line MCF-7/T in vitro and in vivo. Although both Aurora A and p-Aurora A may play a role in the chemoresistance of MCF-7/T cells, p-Aurora A is the dominate factor because the pathway is supposed to be activated through phosphorylation of Aurora A, and phosphorylated Aurora A plays a more active role. Knockdown of the Aurora A concomitantly decreases the p-Aurora A in the same cell. In this study, silencing of Aurora A by microRNA could efficiently inhibit MCF-7/T growth, colony formation, and invasion. The knockdown of the Aurora A led to a significant accumulation of G2/M phase cells and death in vitro, which further confirmed that Aurora A played an important part in the Taxol-resistance of breast cancer cells. In addition, inhibition of Aurora A significantly enhanced the chemosensitivity of the resistant breast cancer cells to Taxol by increasing cell death. These results further elucidated that Aurora A contributed to the Taxol-resistance of breast cancer cells.

Although it has been established that Taxol inhibits cell survival and growth by downregulating Ras/ERK and Akt kinase pathway (1113), several studies have demonstrated that the Ras/ERK and Akt pathways were overexpressed in drug-resistant ovarian and colorectal cancers (14,15), and the overexpression of Akt and Ras/Raf/ERK pathways might be related to the phosphorylation of SRC (16). Following these previous findings, we performed similar experiments and found that Aurora A, along with SRC, Ras/Raf/ERK and Akt pathway, was highly upregulated in the Taxol-resistant breast cancer cells and xenograft models. Silencing of Aurora A significantly reduced SRC phosphorylation and downregulated the Ras/Raf/ERK and Akt pathway in the Taxol-resistant MCF-7/T cells. Studies indicated that inhibition of SRC tyrosine kinase could enhance the chemosensitivity to Taxol through downregulating several pathways such as Ras/Raf/ERK and PI3K/Akt (39,40), thus, re-sensitize resistant ovarian cancer cells to Taxol (41,42). Consistent with reported findings in ovarian cancer cells, our results showed that Aurora A was related to the Taxol-resistance of breast cancer cells through SRC, and these pathways might regulate the chemoresistance in the Taxol-resistant breast cancer cells. SRC might be a potent molecular target for the therapy of Taxol resistant breast cancer.

In addition, studies have revealed that the growth inhibition was more effective by blocking both Ras/Raf/ERK and PI3K/Akt/mTOR pathways simultaneously, which reduced Taxol resistance in several types of cancer (25,26,43). Likewise, Taxol-resistant MCF-7/T cells displayed simultaneous activation of SRC, Akt and ERK accompanied with high expression of Aurora A. Silencing Aurora A expression in MCF-7/T cells using microRNA targeting Aurora A led to the decrease of p-SRC Tyr416. Moreover, the silenced MCF-7/T cells showed not only significant low level of activated ERK and Akt, but also low malignancy and high sensitivity to Taxol.

Aurora kinase was the main regulator of cell cycle (44), and inhibition of Aurora A could induce G2/M phase cells. Our results showed the percentage of the G2/M phase in MCF-7/T cells significantly increased after silencing Aurora A. It has been proposed that inhibition of Aurora A could increase cleavage of procaspase-3 and reduce Bcl-2 expression through inhibition of Akt (45). Similarly, we observed that the cleaved caspase-3 and PARP proteins significantly increased while Bcl-2 and p-Akt decreased in silenced Aurora A MCF-7/T cells. Therefore, we confirmed that silencing of Aurora A led to Akt-related tumor cell growth inhibition and cell death.

In addition, Ras/Raf/ERK and Akt/mTOR pathways have been reported to mediate critical signals for the regulation of MMP secretion and expression (28,32,46,47). In this study, we demonstrated that MMP2 and MMP9 were highly expressed in Taxol-resistant breast cancer cells, and inhibition of Aurora A caused the decrease of MMP2 and MMP9, accompanied with the downregulation of ERK and Akt activity.

In summary, our data demonstrated that the activated Aurora A plays a critical role in Akt and ERK-mediated survival in Taxol-resistant human MCF-7 cells through p-SRC. Inhibition of Aurora A could enhance the sensitivity of drug-resistant MCF-7/T cells to Taxol. The findings provided evidence that Aurora A might provide an additional and effective target for small molecule chemotherapeutic intervention in drug-resistant breast cancer. In addition, pathways such as Ras/Raf/ERK and Akt/mTOR have been well-studied and related inhibitors are available (25,32,33), targeting Aurora A and Ras/Raf/ERK, and Akt/mTOR simultaneously may improve the sensitivity of breast cancer cells to Taxol, which could be a promising strategy for the treatment of patients with Taxol-resistant breast cancer. The insights into the mechanisms of Taxol resistance and sensitization would represent a useful basis for further development of strategies to circumvent Taxol-related chemoresistance in breast cancer clinical practice.

Acknowledgements

The present study was supported by the National Natural Science Foundation of China (Approval no. 81102025) and the CAMS Initiative for Innovative Medicine (Approval no. 2016-12-M-1-008).

References

1 

Schettini F, Giuliano M, De Placido S and Arpino G: Nab-paclitaxel for the treatment of triple-negative breast cancer: Rationale, clinical data and future perspectives. Cancer Treat Rev. 50:129–141. 2016. View Article : Google Scholar : PubMed/NCBI

2 

Wan Y, Huang A, Yang Y, Xie G, Chen X, Hu J, Chen X, Yang L, Li J, Chen L, et al: A vector-based short hairpin RNA targeting Aurora A inhibits breast cancer growth. Int J Oncol. 36:1121–1128. 2010.PubMed/NCBI

3 

Chen S, Dong Q, Hu S, Cai J, Zhang W, Sun J, Wang T, Xie J, He H, Xing J, et al: Proteomic analysis of the proteins that are associated with the resistance to paclitaxel in human breast cancer cells. Mol Biosyst. 10:294–303. 2014. View Article : Google Scholar : PubMed/NCBI

4 

Lu J, Tan M, Huang WC, Li P, Guo H, Tseng LM, Su XH, Yang WT, Treekitkarnmongkol W, Andreeff M, et al: Mitotic deregulation by survivin in ErbB2-overexpressing breast cancer cells contributes to Taxol resistance. Clin Cancer Res. 15:1326–1334. 2009. View Article : Google Scholar : PubMed/NCBI

5 

Lai D, Ho KC, Hao Y and Yang X: Taxol resistance in breast cancer cells is mediated by the hippo pathway component TAZ and its downstream transcriptional targets Cyr61 and CTGF. Cancer Res. 71:2728–2738. 2011. View Article : Google Scholar : PubMed/NCBI

6 

Lens SM, Voest EE and Medema RH: Shared and separate functions of polo-like kinases and aurora kinases in cancer. Nat Rev Cancer. 10:825–841. 2010. View Article : Google Scholar : PubMed/NCBI

7 

Mountzios G, Terpos E and Dimopoulos MA: Aurora kinases as targets for cancer therapy. Cancer Treat Rev. 34:175–182. 2008. View Article : Google Scholar : PubMed/NCBI

8 

Zou Z, Yuan Z, Zhang Q, Long Z, Chen J, Tang Z, Zhu Y, Chen S, Xu J, Yan M, et al: Aurora kinase A inhibition-induced autophagy triggers drug resistance in breast cancer cells. Autophagy. 8:1798–1810. 2012. View Article : Google Scholar : PubMed/NCBI

9 

Terakawa T, Miyake H, Kumano M and Fujisawa M: Growth inhibition and enhanced chemosensitivity induced by down-regulation of Aurora-A in human renal cell carcinoma Caki-2 cells using short hairpin RNA. Oncol Lett. 2:713–717. 2011.PubMed/NCBI

10 

Li Y, Tang K, Zhang H, Zhang Y, Zhou W and Chen X: Function of Aurora kinase A in Taxol-resistant breast cancer and its correlation with P-gp. Mol Med Rep. 4:739–746. 2011.PubMed/NCBI

11 

Bergstralh DT, Taxman DJ, Chou TC, Danishefsky SJ and Ting JP: A comparison of signaling activities induced by Taxol and desoxyepothilone B. J Chemother. 16:563–576. 2004. View Article : Google Scholar : PubMed/NCBI

12 

Li W, Liu J, Jackson K, Shi R and Zhao Y: Sensitizing the therapeutic efficacy of taxol with shikonin in human breast cancer cells. PLoS One. 9:e940792014. View Article : Google Scholar : PubMed/NCBI

13 

Zhang Q, Si S, Schoen S, Chen J, Jin XB and Wu G: Suppression of autophagy enhances preferential toxicity of paclitaxel to folliculin-deficient renal cancer cells. J Exp Clin Cancer Res. 32:992013. View Article : Google Scholar : PubMed/NCBI

14 

Wang NN, Zhao LJ, Wu LN, He MF, Qu JW, Zhao YB, Zhao WZ, Li JS and Wang JH: Mechanistic analysis of taxol-induced multidrug resistance in an ovarian cancer cell line. Asian Pac J Cancer Prev. 14:4983–4988. 2013. View Article : Google Scholar : PubMed/NCBI

15 

Xu R, Nakano K, Iwasaki H, Kumagai M, Wakabayashi R, Yamasaki A, Suzuki H, Mibu R, Onishi H and Katano M: Dual blockade of phosphatidylinositol 3-kinase and mitogen-activated protein kinase pathways overcomes paclitaxel-resistance in colorectal cancer. Cancer Lett. 306:151–160. 2011. View Article : Google Scholar : PubMed/NCBI

16 

Le XF and Bast RC Jr: Src family kinases and paclitaxel sensitivity. Cancer Biol Ther. 12:260–269. 2011. View Article : Google Scholar : PubMed/NCBI

17 

Liao CH, Sang S, Ho CT and Lin JK: Garcinol modulates tyrosine phosphorylation of FAK and subsequently induces apoptosis through down-regulation of Src, ERK, and Akt survival signaling in human colon cancer cells. J Cell Biochem. 96:155–169. 2005. View Article : Google Scholar : PubMed/NCBI

18 

Qin B, Ariyama H, Baba E, Tanaka R, Kusaba H, Harada M and Nakano S: Activated Src and Ras induce gefitinib resistance by activation of signaling pathways downstream of epidermal growth factor receptor in human gallbladder adenocarcinoma cells. Cancer Chemother Pharmacol. 58:577–584. 2006. View Article : Google Scholar : PubMed/NCBI

19 

Gan PP, Pasquier E and Kavallaris M: Class III beta-tubulin mediates sensitivity to chemotherapeutic drugs in non small cell lung cancer. Cancer Res. 67:9356–9363. 2007. View Article : Google Scholar : PubMed/NCBI

20 

Li Y, Tang K, Zhang L, Li C, Niu F, Zhou W, Yang H, Feng Z and Chen X: The molecular mechanisms of a novel multi-kinase inhibitor ZLJ33 in suppressing pancreatic cancer growth. Cancer Lett. 356:392–403. 2015. View Article : Google Scholar : PubMed/NCBI

21 

Li Y, Zhou W, Wei L, Jin J, Tang K, Li C, Teh BT and Chen X: The effect of Aurora kinases on cell proliferation, cell cycle regulation and metastasis in renal cell carcinoma. Int J Oncol. 41:2139–2149. 2012. View Article : Google Scholar : PubMed/NCBI

22 

Li Y, Zhang ZF, Chen J, Huang D, Ding Y, Tan MH, Qian CN, Resau JH, Kim H and Teh BT: VX680/MK-0457, a potent and selective Aurora kinase inhibitor, targets both tumor and endothelial cells in clear cell renal cell carcinoma. Am J Transl Res. 2:296–308. 2010.PubMed/NCBI

23 

Do TV, Xiao F, Bickel LE, Klein-Szanto AJ, Pathak HB, Hua X, Howe C, OBrien SW, Maglaty M, Ecsedy JA, et al: Aurora kinase A mediates epithelial ovarian cancer cell migration and adhesion. Oncogene. 33:539–549. 2014. View Article : Google Scholar : PubMed/NCBI

24 

Mendiola M, Barriuso J, Mariño-Enríquez A, Redondo A, Domínguez-Cáceres A, Hernández-Cortés G, Pérez-Fernández E, Sánchez-Navarro I, Vara JA, Suárez A, et al: Aurora kinases as prognostic biomarkers in ovarian carcinoma. Hum Pathol. 40:631–638. 2009. View Article : Google Scholar : PubMed/NCBI

25 

Kawaguchi W, Itamochi H, Kigawa J, Kanamori Y, Oishi T, Shimada M, Sato S, Shimogai R, Sato S and Terakawa N: Simultaneous inhibition of the mitogen-activated protein kinase kinase and phosphatidylinositol 3-kinase pathways enhances sensitivity to paclitaxel in ovarian carcinoma. Cancer Sci. 98:2002–2008. 2007. View Article : Google Scholar : PubMed/NCBI

26 

Knuefermann C, Lu Y, Liu B, Jin W, Liang K, Wu L, Schmidt M, Mills GB, Mendelsohn J and Fan Z: HER2/PI-3K/Akt activation leads to a multidrug resistance in human breast adenocarcinoma cells. Oncogene. 22:3205–3212. 2003. View Article : Google Scholar : PubMed/NCBI

27 

Li JP, Yang YX, Liu QL, Pan ST, He ZX, Zhang X, Yang T, Chen XW, Wang D, Qiu JX, et al: The investigational Aurora kinase A inhibitor alisertib (MLN8237) induces cell cycle G2/M arrest, apoptosis, and autophagy via p38 MAPK and Akt/mTOR signaling pathways in human breast cancer cells. Drug Des Devel Ther. 9:1627–1652. 2015.PubMed/NCBI

28 

Wang X, Lu N, Niu B, Chen X, Xie J and Cheng N: Overexpression of Aurora-A enhances invasion and matrix metalloproteinase-2 expression in esophageal squamous cell carcinoma cells. Mol Cancer Res. 10:588–596. 2012. View Article : Google Scholar : PubMed/NCBI

29 

Gan PP, McCarroll JA, Pouha ST, Kamath K, Jordan MA and Kavallaris M: Microtubule dynamics, mitotic arrest, and apoptosis: Drug-induced differential effects of betaIII-tubulin. Mol Cancer Ther. 9:1339–1348. 2010. View Article : Google Scholar : PubMed/NCBI

30 

Almhanna K, Cubitt CL, Zhang S, Kazim S, Husain K, Sullivan D, Sebti S and Malafa M: MK-2206, an Akt inhibitor, enhances carboplatinum/paclitaxel efficacy in gastric cancer cell lines. Cancer Biol Ther. 14:932–936. 2013. View Article : Google Scholar : PubMed/NCBI

31 

Clark AS, West K, Streicher S and Dennis PA: Constitutive and inducible Akt activity promotes resistance to chemotherapy, trastuzumab, or tamoxifen in breast cancer cells. Mol Cancer Ther. 1:707–717. 2002.PubMed/NCBI

32 

Chen J, Huang D, Rubera I, Futami K, Wang P, Zickert P, Khoo SK, Dykema K, Zhao P, Petillo D, et al: Disruption of tubular Flcn expression as a mouse model for renal tumor induction. Kidney Int. 88:1057–1069. 2015. View Article : Google Scholar : PubMed/NCBI

33 

Wu M, Si S, Li Y, Schoen S, Xiao GQ, Li X, Teh BT, Wu G and Chen J: Flcn-deficient renal cells are tumorigenic and sensitive to mTOR suppression. Oncotarget. 6:32761–32773. 2015. View Article : Google Scholar : PubMed/NCBI

34 

Gutiérrez-González A, Belda-Iniesta C, Bargiela-Iparraguirre J, Dominguez G, Alfonso García P, Perona R and Sanchez-Perez I: Targeting Chk2 improves gastric cancer chemotherapy by impairing DNA damage repair. Apoptosis. 18:347–360. 2013. View Article : Google Scholar : PubMed/NCBI

35 

Haruki K, Shiba H, Fujiwara Y, Furukawa K, Iwase R, Uwagawa T, Misawa T, Ohashi T and Yanaga K: Inhibition of nuclear factor-κB enhances the antitumor effect of paclitaxel against gastric cancer with peritoneal dissemination in mice. Dig Dis Sci. 58:123–131. 2013. View Article : Google Scholar : PubMed/NCBI

36 

Qiu H, Yashiro M, Zhang X, Miwa A and Hirakawa K: A FGFR2 inhibitor, Ki23057, enhances the chemosensitivity of drug-resistant gastric cancer cells. Cancer Lett. 307:47–52. 2011. View Article : Google Scholar : PubMed/NCBI

37 

George JA, Chen T and Taylor CC: SRC tyrosine kinase and multidrug resistance protein-1 inhibitions act independently but cooperatively to restore paclitaxel sensitivity to paclitaxel-resistant ovarian cancer cells. Cancer Res. 65:10381–10388. 2005. View Article : Google Scholar : PubMed/NCBI

38 

Ding YH, Zhou ZW, Ha CF, Zhang XY, Pan ST, He ZX, Edelman JL, Wang D, Yang YX, Zhang X, et al: Alisertib, an Aurora kinase A inhibitor, induces apoptosis and autophagy but inhibits epithelial to mesenchymal transition in human epithelial ovarian cancer cells. Drug Des Devel Ther. 9:425–464. 2015.PubMed/NCBI

39 

Puls LN, Eadens M and Messersmith W: Current status of SRC inhibitors in solid tumor malignancies. Oncologist. 16:566–578. 2011. View Article : Google Scholar : PubMed/NCBI

40 

Wheeler DL, Iida M and Dunn EF: The role of Src in solid tumors. Oncologist. 14:667–678. 2009. View Article : Google Scholar : PubMed/NCBI

41 

Pengetnze Y, Steed M, Roby KF, Terranova PF and Taylor CC: Src tyrosine kinase promotes survival and resistance to chemotherapeutics in a mouse ovarian cancer cell line. Biochem Biophys Res Commun. 309:377–383. 2003. View Article : Google Scholar : PubMed/NCBI

42 

Chen T, Pengetnze Y and Taylor CC: Src inhibition enhances paclitaxel cytotoxicity in ovarian cancer cells by caspase-9-independent activation of caspase-3. Mol Cancer Ther. 4:217–224. 2005.PubMed/NCBI

43 

Kim SH, Juhnn YS and Song YS: Akt involvement in paclitaxel chemoresistance of human ovarian cancer cells. Ann N Y Acad Sci. 1095:82–89. 2007. View Article : Google Scholar : PubMed/NCBI

44 

Fu J, Bian M, Jiang Q and Zhang C: Roles of Aurora kinases in mitosis and tumorigenesis. Mol Cancer Res. 5:1–10. 2007. View Article : Google Scholar : PubMed/NCBI

45 

Long M, Yin G, Liu L, Lin F, Wang X, Ren J, Wei J, Dong K and Zhang H: Adenovirus-mediated Aurora A shRNA driven by stathmin promoter suppressed tumor growth and enhanced paclitaxel chemotherapy sensitivity in human breast carcinoma cells. Cancer Gene Ther. 19:271–281. 2012. View Article : Google Scholar : PubMed/NCBI

46 

Marshall CJ: MAP kinase kinase kinase, MAP kinase kinase and MAP kinase. Curr Opin Genet Dev. 4:82–89. 1994. View Article : Google Scholar : PubMed/NCBI

47 

Tseng YS, Lee JC, Huang CY and Liu HS: Aurora-A overexpression enhances cell-aggregation of Ha-ras transformants through the MEK/ERK signaling pathway. BMC Cancer. 9:4352009. View Article : Google Scholar : PubMed/NCBI

Related Articles

Journal Cover

October-2017
Volume 38 Issue 4

Print ISSN: 1021-335X
Online ISSN:1791-2431

Sign up for eToc alerts

Recommend to Library

Copy and paste a formatted citation
x
Spandidos Publications style
Li Y, Zhou W, Tang K, Chen X, Feng Z and Chen J: Silencing Aurora A leads to re-sensitization of breast cancer cells to Taxol through downregulation of SRC-mediated ERK and mTOR pathways. Oncol Rep 38: 2011-2022, 2017
APA
Li, Y., Zhou, W., Tang, K., Chen, X., Feng, Z., & Chen, J. (2017). Silencing Aurora A leads to re-sensitization of breast cancer cells to Taxol through downregulation of SRC-mediated ERK and mTOR pathways. Oncology Reports, 38, 2011-2022. https://doi.org/10.3892/or.2017.5908
MLA
Li, Y., Zhou, W., Tang, K., Chen, X., Feng, Z., Chen, J."Silencing Aurora A leads to re-sensitization of breast cancer cells to Taxol through downregulation of SRC-mediated ERK and mTOR pathways". Oncology Reports 38.4 (2017): 2011-2022.
Chicago
Li, Y., Zhou, W., Tang, K., Chen, X., Feng, Z., Chen, J."Silencing Aurora A leads to re-sensitization of breast cancer cells to Taxol through downregulation of SRC-mediated ERK and mTOR pathways". Oncology Reports 38, no. 4 (2017): 2011-2022. https://doi.org/10.3892/or.2017.5908