+TIP EB1 downregulates paclitaxel‑induced proliferation inhibition and apoptosis in breast cancer cells through inhibition of paclitaxel binding on microtubules

  • Authors:
    • Geethu Emily Thomas
    • Jamuna S. Sreeja
    • K. K. Gireesh
    • Hindol Gupta
    • Tapas K. Manna
  • View Affiliations

  • Published online on: October 7, 2014     https://doi.org/10.3892/ijo.2014.2701
  • Pages: 133-146
Metrics: Total Views: 0 (Spandidos Publications: | PMC Statistics: )
Total PDF Downloads: 0 (Spandidos Publications: | PMC Statistics: )


Abstract

Microtubule plus‑end‑binding protein (+TIP) EB1 has been shown to be upregulated in breast cancer cells and promote breast tumor growth in vivo. However, its effect on the cellular actions of microtubule‑targeted drugs in breast cancer cells has remained poorly understood. By using cellular and biochemical assays, we demonstrate that EB1 plays a critical role in regulating the sensitivity of breast cancer cells to anti‑microtubule drug, paclitaxel (PTX). Cell viability assays revealed that EB1 expression in the breast cancer cell lines correlated with the reduction of their sensitivity to PTX. Knockdown of EB1 by enzymatically‑prepared siRNA pools (esiRNAs) increased PTX‑induced cytotoxicity and sensitized cells to PTX‑induced apoptosis in three breast cancer cell lines, MCF‑7, MDA MB‑231 and T47D. Apoptosis was associated with activation of caspase‑9 and an increase in the cleavage of poly(ADP‑ribose) polymerase (PARP). p53 and Bax were upregulated and Bcl2 was downregulated in the EB1‑depleted PTX‑treated MCF‑7 cells, indicating that the apoptosis occurs via a p53‑dependent pathway. Following its upregulation, the nuclear accumulation of p53 and its association with cellular microtubules were increased. EB1 depletion increased PTX‑induced microtubule bundling in the interphase cells and induced formation of multiple spindle foci with abnormally elongated spindles in the mitotic MCF‑7 cells, indicating that loss of EB1 promotes PTX‑induced stabilization of microtubules. EB1 inhibited PTX‑induced microtubule polymerization and diminished PTX binding to microtubules in vitro, suggesting that it modulates the binding sites of PTX at the growing microtubule ends. Results demonstrate that EB1 downregulates inhibition of PTX‑induced proliferation and apoptosis in breast cancer cells through a mechanism in which it impairs PTX‑mediated stabilization of microtubule polymerization and inhibits PTX binding on microtubules.

Introduction

Breast cancer is among the leading causes of cancer mortality and accounts for more than 400,000 deaths annually worldwide (1). The introduction of new chemotherapeutic regimens in recent years has modestly improved the survival rate of patients, but there are still major obstacles that must be overcome before this disease can be successfully treated (2,3). One of the major clinical issues is the development of drug resistance, which accounts for a poor response and reduces the overall survival rate in patients (4,5). Among the drugs used for treating breast cancer, the ones most commonly used belong to the taxane group of agents, which kill tumor cells by targeting cellular microtubules (6,7). Similar to other classes of agents, however, a major drawback of taxane chemotherapy is that tumor cells can develop resistance to these drugs (5,810).

Paclitaxel (PTX) is a prototype of microtubule-stabilizing agents that disrupt cellular microtubule function by stabilizing the microtubule assembly dynamics (6). Specifically, it suppresses the growth and shortening dynamics of microtubules through its interaction with the plus-end of the microtubule (11). As a result, it disrupts the normal spindle architecture of the cell, which leads to chromosome segregation defects, mitotic arrest and eventually apoptosis (12,13). Earlier studies have demonstrated that there is a close relationship between microtubule dynamics and PTX resistance in cancer cells (1420).

Microtubules associate with a specialized class of proteins, known as the plus-end-binding proteins (+TIPs), that localize to the growing plus-ends of microtubules and regulate numerous plus-end-mediated processes. Recent studies have emphasized the central role of end-binding (EB) family proteins among the +TIPs (2125). Mammals contain three EBs, EB1, EB2, and EB3 with ~57–66% sequence identity and are encoded by separate genes (26). EBs play critical roles in the regulation of microtubule dynamics (2730). Recently, analysis using reconstituted microtubules showed that EBs can modulate the dynamics of PTX-treated microtubules. Specifically, EB3 has been shown to stimulate catastrophe events and to rescue growth of PTX-treated microtubules, indicating that it suppresses PTX’s ability to stabilize microtubule dynamics (31). Among the EB family proteins, EB1 was focused on previously as it is highly conserved compared to other EB members and is ubiquitously expressed in all cell types (26). High resolution structural analysis showed that EB1 alters the structure of microtubule plus-ends (32). Consistent with its ability to modulate structure-function of microtubules, EB1 plays essential roles in mitosis (3336). Recently, overexpression of EB1 has been shown to stimulate cell growth in cultured human breast cancer cells, indicating that it plays an important role in stimulation of cell proliferation (37,38).

In addition to its microtubule regulatory roles, EB1 has been strongly linked to tumorigenesis. It binds to adenomatous polyposis coli (APC), a major tumor suppressor in colon. Mutations in the EB1-binding region of APC are commonly found in colon cancer cells (39). Recent proteomic and biochemical analyses have shown that there is a high EB1 expression level in various human carcinomas and breast cancer cells (38,40,41). Consistent with a higher expression level in breast cancer cells, EB1 has also been shown to be upregulated in breast cancer patients (38). Specifically, its expression level appears to be correlated with the clinicopathological signature of the tumor malignancy in patients (38). This study has also shown that EB1 enhances breast tumor growth in nude mice (38).

Because of the close relationship between the regulation of microtubule dynamics by EB1 and PTX, and the correlation between EB1 expression and breast cancer, we speculated that EB1 could regulate PTX-induced cytotoxicity and PTX-mediated microtubule stabilization in breast cancer cells. In the present study, we investigated the role of EB1 in regulating PTX-induced cytotoxicity and microtubule stabilization in breast cancer cells. Our results demonstrate that suppression of EB1 sensitizes cells to PTX-induced cytotoxicity and apoptosis. Additional results revealed details of the underlying mechanism associated with the regulation of PTX sensitivity by EB1.

Materials and methods

Reagents and antibodies

PTX, vinblastine (VIN), cisplatin, propidium iodide (PI), DAPI, GTP, PIPES, EGTA, reagents for the methylthiotetrazol (MTT) assay and the Annexin V apoptosis detection kit were obtained from Sigma (St. Louis, MO, USA). Dulbecco’s modified Eagle’s medium (DMEM), Leibovitz (L-15) medium, RPMI-1640 medium, DMEM-F12 (1:1) medium supplemented with HEPES and fetal bovine serum (FBS) were purchased from HiMedia Laboratories (Mumbai, India). Rhodamine labeled tubulin was obtained from Cytoskeleton, Inc. (Denver, CO, USA), and Oregon green 488-labeled PTX (Ore-PTX) was obtained from Invitrogen Life Technologies (Carlsbad, CA, USA). Monoclonal antibodies against EB1, actin and BubR1 were obtained from BD Biosciences (San Jose, CA, USA). Mouse monoclonal antibodies against α-tubulin and p53 and the rabbit polyclonal anti-EB1 were obtained from Sigma. Mouse monoclonal anti-Bax, rat monoclonal anti-α-tubulin and rabbit polyclonal anti-Bcl2, anti-caspase-9 and anti-PARP were obtained from Abcam (Cambridge, MA, USA). Rabbit polyclonal p21 antibody was obtained from Cell Signaling Technology, Inc. (Danvers, MA, USA). FITC, TRITC and peroxidase conjugated secondary antibodies were obtained from Jackson ImmunoResearch (West Grove, PA, USA).

Cell culture, transfection with ribonuclease III-prepared siRNA pools (esiRNA)

MCF-7, MDA MB-231 and T47D cells originally obtained from ATCC (Manassas, VA, USA) were cultured in DMEM, L-15, and RPMI-1640 media, respectively, which were supplemented with 10% FBS, 2 mM L-glutamine, 1.5 mg/ml sodium bicarbonate, 100 μg/ml penicillin and 100 μg/ml streptomycin. MCF-10 A cells were grown as a monolayer culture in DMEM-F12 medium supplemented with 5% donor horse serum (Invitrogen Life Technologies), 20 ng/ml epidermal growth factor (EGF), 10 μg/ml insulin, 0.5 μg/ml hydrocortisone, 100 ng/ml cholera toxin (all from Sigma), 100 μg/ml penicillin and 100 μg/ml streptomycin (42).

To downregulate protein expression, we used ribonuclease III-prepared small interfering RNA pools, which ensure the efficient knockdown of proteins with minimum off-target effects compared with single siRNAs (43,44). esiRNA consists of enzymatically-prepared siRNA pools comprised of a heterogeneous mixture of siRNAs which target the same mRNA sequence of the gene (43). Cells at ~50% confluence were transfected with either EB1 esiRNA (cat. no. EHU045671; Sigma) against the 331–807 nucleotide region of EB1 (NM_012325.2) or luciferase control esiRNA (cat. no. EHUFLUC; Sigma) using either oligofectamine (for MCF-7 cells) or lipofectamine (for T47D and MDA MB-231 cells) as the vehicle.

Plasmids and proteins

pEGFP-EB1 was used as the template for PCR amplification of the complete coding sequence of the human EB1 (NM_012325.2) gene. The amplified product was ligated into the pET-28a (Novagen, Inc., Madison, WI, USA) expression vector and transformed into BL21-(DE3) cells. The 6-His-tagged EB1 was expressed under 1 mM IPTG and then purified through Nickel-NTA (28). Tubulin was purified from goat brains by repetitive cycles of polymerization and depolymerization (45). Protein concentrations were estimated by the Bradford method using BSA as the standard (46).

Cell proliferation and apoptosis assays

The effect of PTX on cell viability was measured by MTT assay as previously described (47). The cells at ~60% confluence were treated with gradients of PTX concentrations (1–200 nM), and allowed to grow for the next 48 h prior to measuring cell viability using MTT assay in the Multi-Mode Microplate Reader, SpectraMax 5 (Molecular Devices, Sunnyvale, CA, USA). The percentage of viable cells as a function of drug concentration was plotted to determine the drug concentration needed to inhibit cell viability by 50%, half inhibitory concentrations (IC50). For determination of IC50 values in the esiRNA-treated cells, the cells after 24 h of transfection with esiRNA were treated with gradients of PTX concentrations (5–200 nM) and then incubated for another 48 h prior to measuring cell viability using the MTT assay. For the apoptosis assay, after 24 h of transfection with esiRNA, the cells were treated with PTX (10 nM) for the next 48 h. Apoptosis was measured using the Annexin V-FITC apoptosis assay kit followed by flow cytometric analysis using the FACSAria III (BD Biosciences). The quadrants of the raw sample data were determined by comparing the data of unstained cells. Data were analyzed using FlowJo software. Percentages of apoptosis were quantified from the sum of first (Q1) and second (Q2) quadrant. Total cell death was quantified from the sum of Q1, Q2 and the third quadrant (Q3). Apoptosis was also detected using immunofluorescence-based imaging. The cells were grown on coverslips, washed with PBS and then treated with binding buffer containing Annexin V FITC and PI for 10 min (48). The relative levels of Annexin V and PI staining were determined using confocal microscopy.

Microtubule pull-down assay from cell lysate

MCF-7 cells were lysed in BRB80 buffer, pH 6.8 supplemented with a 1X protease inhibitor cocktail and 0.1% Triton X-100 and then centrifuged at 15,000 rpm for 15 min at 4°C. Exogenously purified bovine tubulin (2 μM) and 1 mM GTP were then added to the cell lysates, and the reaction mixture was polymerized at 37°C for 15 min (49). Microtubules were pelleted at 100,000 × g for 40 min at 35°C and washed with warm BRB80 buffer. The pellet was treated with 4X SDS sample buffer, and proteins were detected by western blot analysis.

Western blot analysis

Protein samples and cell lysates were run on SDS-PAGE, and the protein bands were transferred onto polyvinylidene fluoride (PVDF) membranes. Target proteins were detected by incubating the membranes in the appropriate primary antibodies and then incubating them in the appropriate HRP-conjugated secondary antibodies. Densitometric analyses of the blots were performed using the Quantity One software (Bio-Rad, Hercules, CA, USA).

Immunofluorescence microscopy and image analysis

Cells fixed in either methanol at −20°C or 3.7% paraformaldehyde were washed with PBS containing 2% bovine serum albumin and 0.5% Triton X-100. The cells were then incubated in primary antibody for 1 h and then incubated in secondary antibody and DAPI for 45 and 1 min, respectively. Coverslips were mounted using ProLong Gold (Invitrogen Life Technologies), and the images (63X) were captured using a Leica SP5 laser confocal microscope. The intensity measurements were taken using the system run software provided by Leica. Briefly, intensity per pixel was quantified by selecting regions of interest (ROI) of equal size in all the images for comparison.

Microtubule polymerization and PTX microtubule-binding assay

Reaction mixtures containing tubulin (15 μM) and the desired concentration of EB1 were polymerized in BRB80 buffer (80 mM PIPES, 2 mM EGTA and 1 mM MgCl2), pH 6.8, containing 1 mM GTP and 0.1 μM PTX. Polymerization was monitored by measuring the turbidity of the reaction mixtures at 360 nm. For the quantitative estimation of microtubules, the reaction mixtures were passed through a 15% glycerol cushion by centrifugation at 100,000 g after 30 min of polymerization. The pellets were resuspended in cold buffer, and the proteins were run on 10% SDS-PAGE and then subjected to Coomassie staining. For the quantitative analysis of PTX binding to microtubules, tubulin (10 μM) was polymerized into microtubules in vitro in BRB80 buffer containing 10% glycerol and 1 mM GTP for 15 min at 37°C. Next, Ore-PTX (20 nM) was added either to the control microtubules or the microtubules pre-incubated with EB1 (20–200 nM) for 15 min. The reaction mixtures were incubated for another 15 min prior to centrifugation through a 15% sucrose cushion at 100,000 g. The microtubule pellets were resuspended in ice-cold BRB80 buffer. The binding of Ore-PTX to the microtubules was determined by measuring the fluorescence intensity of Ore-PTX present in the microtubule solutions. Fluorescence measurements were taken using a fluorescence spectrophotometer (Horiba FluoroLog-3) with an excitation at 488 nm and an emission in the 500–560 nm range. The percentage of Ore-PTX binding on microtubules inhibited by EB1 was estimated by comparing the fluorescence values at 525 nm of the EB1 containing Ore-PTX-treated samples with the Ore-PTX only treated control sample. All fluorescence measurements were taken using a 1-cm path length cuvette. The pelleted microtubule solutions were also run through 10% SDS-PAGE.

Statistical analysis

Results are presented as the mean ± standard error (SE). Statistical significance was set at p<0.05. One-way analysis of variance (ANOVA) was used for statistical analysis on EB1 expression levels and IC50 values in different cell lines. The comparison of differences between control and the treated groups was performed using a two-tailed Student’s t-test. The data were plotted using Origin 8.

Results

EB1 expression negatively correlates with PTX sensitivity in breast cancer cell lines

We examined the expression of EB1 by immunoblotting in three breast cancer cell lines, the human breast adenocarcinoma cells MCF-7, the metastatic human breast adenocarcinoma MDA MB-231 cells, and the human ductal breast epithelial tumor T47D cells. Two non-tumorigenic cell lines, mammary MCF-10 A and the kidney epithelial HEK-293 were used as control. A significantly high EB1 expression level was observed in all three breast cancer cell lines compared with the normal MCF-10 A and HEK-293 cells (Fig. 1A and B). EB1 expressions in the breast cancer cell lines were several folds higher as compared with the MCF-10 A and HEK-293 cells. Statistical analyses were performed based on the results of two independent western blot analysis images, as described in Materials and methods. These results were in good agreement with previously published results on EB1 expression in breast cancer cells (38). We then examined if the EB1 expression level correlates with the sensitivity of these cells to PTX. To determine the sensitivity of these cells to PTX, we performed in vitro cell viability assays. Cells treated with gradients of PTX concentrations (1–200 nM) were analyzed for cell viability by the MTT assay. The IC50 values of PTX, which stand for the concentration of PTX needed to suppress cell viability by 50%, in the cell lines were then determined. Strikingly, we noted that MCF-7, MDA MB-231 and T47D cells had much higher IC50 values than the control MCF-10 A cells (Fig. 1C). Consistent with the previously published data (50), MCF-10 A cells exhibited a very low IC50 of ~6 nM. However, the IC50 values for MCF-7, MDA MB-231 and T47D cells were increased to 40, 28 and 62 nM, respectively with a range of 5–10-fold increase compared to MCF-10 A. Thus, the expression levels of EB1 correlated with the IC50 values of PTX. Unlike PTX, we did not observe any significant correlation between EB1 expression level in the cell lines and the cytotoxicity of VIN, a microtubule destabilizing agent (not shown).

EB1 depletion increased PTX’s ability to inhibit cell viability in breast cancer cells

Next, we investigated the effect of EB1 depletion on PTX-induced cytotoxicity in the breast cancer cells. For efficient knockdown of EB1, we used ribonuclease III-prepared pool of siRNAs, as described in Materials and methods. Cells after treatment with EB1 esiRNA or control luciferase esiRNA for 24 h, were incubated with gradients of PTX concentrations (5–200 nM) for another 48 h prior to measuring cell viability. EB1 was effectively depleted (90±6%) in all three breast cancer cell lines during this period (Fig. 2A). The ability of PTX to inhibit cell viability increased significantly in the EB1 knockdown (EB1-KD) cells compared with the control cells. We observed that the IC50 of PTX in all three breast cancer cell lines were significantly reduced (Fig. 2B). The IC50 values for the MCF-7, MDA MB-231 and T47D control cells were 38, 28 and 59 nM, respectively, and the IC50 values for the EB1-KD MCF-7, MDA MB-231 and T47D cells were 23, 11 and 43 nM, respectively. These results indicate a 40, 60 and 28% reduction in the IC50 in the EB1-KD MCF-7, MDA MB-231 and T47D cells, respectively, compared with the control cells. EB1-KD alone, in the absence of PTX, did not significantly inhibit cell viability. After 72 h of EB1 depletion, the percentage of viable cells was reduced only by 17% (Fig. 2C). Additionally, we also examined the effect of EB1-KD on cytotoxic effects of VIN and the microtubule unrelated drug cisplatin in MCF-7 cells. EB1-KD did not have any effect on either VIN- or cisplatin-induced effects on cell viability in MCF-7 cells (data not shown). We also found that EB1-KD did not significantly enhance PTX sensitivity in the control MCF-10 A and HEK-293 cells (data not shown), indicating that the effect was specific to the breast cancer cells.

EB1 depletion sensitizes breast cancer cells to PTX-induced apoptosis via activation of p53 and its downstream apoptosis regulators

Next, we investigated the effect of EB1 depletion on PTX-induced apoptosis in the breast cancer cell lines. After a 24-h esiRNA transfection, the EB1-KD and control cells were treated with a low dose of PTX (10 nM, ~1/4th of IC50 of PTX in control MCF-7 cells) for the next 48 h prior to measuring apoptosis using the Annexin V assay. In all three breast cancer cell lines, there was a significant amount of apoptosis induced in the EB1-depleted cells under PTX treatment (Fig. 3A). While the PTX treatment in the control MCF-7, MDA MB-231 and T47D cells induced apoptosis in ~21, 18 and 14% cells, respectively, PTX treatment in the EB1-KD MCF-7, MDA MB-231 and T47D cells induced apoptosis in ~30, 30 and 22% cells, respectively (Fig. 3B). These results were equivalent to a 1.4-, 1.6- and 1.5-fold increase in apoptosis in the MCF-7, MDA MB-231 and T47D cells, respectively. The total cell death increased from 26, 26 and 16% in the PTX-treated control MCF-7, MDA MB-231 and T47D cells, respectively, to 43, 37 and 31% in the PTX-treated EB1-KD MCF-7, MDA MB-231 and T47D cells, respectively (Fig. 3A), as described in Materials and methods. Furthermore, EB1-KD alone, in the absence of PTX, did not significantly induce apoptosis in any of the three cell lines as compared with control cells not treated with PTX (Fig. 3A and B). These results were confirmed by the Annexin V and PI immunofluorescence staining results (Fig. 3C).

To elucidate the molecular mechanism underlying apoptosis induced in cells depleted of EB1 and treated with PTX, we analyzed the expression patterns of various apoptosis-associated proteins. The PTX-treated EB1-KD MCF-7 cells demonstrated a time-dependent increase in the caspase-9 expression level and poly(ADP-ribose) polymerase (PARP) cleavage compared with the PTX-treated control cells (Fig. 4A). In addition, the p53 and Bax expression levels were remarkably higher (Fig. 4A), and the Bcl2 expression level was reduced in the PTX-treated EB1 deficient cells compared with the PTX-treated control cells (Fig. 4B). The p21 expression level, however, appeared similar under both of these conditions (Fig. 4A). Furthermore, there was no significant change in the expression levels of p53, Bax, caspase-9 and PARP in the control EB1-KD cells in the absence of PTX, but the level of p21 was slightly increased in these cells (Fig. 4C).

EB1 depletion induced nuclear accumulation of p53 and increased p53 association with cellular microtubules in PTX-treated MCF-7 cells

Next, we examined the cellular localization of p53 in EB1-KD vs. control cells. Consistent with the increase in p53 expression, the amount of p53 that accumulated in the nucleus was significantly higher in the PTX-treated EB1-KD MCF-7 cells (Fig. 5A). Furthermore, we also observed an increase in the number of multinucleated cells in PTX-treated EB1-KD cells. Approximately 56% interphase cells were multinucleated, containing two or more nuclei (bar graph, Fig. 5A). Multinucleated cells were also observed in the PTX-treated control cells; however, they were less abundant (31%). The majority of the multinucleated cells in EB1-KD condition demonstrated a higher accumulation of p53 in their nuclei, whereas the PTX-treated control cells did not demonstrate a higher nuclear accumulation of p53 (bar graph, Fig. 5A).

Next, we investigated whether the association between p53 and the cellular microtubules was altered in the PTX-treated EB1-KD cells. The cellular microtubules were assembled in microtubule-stabilizing buffer and then centrifuged. Finally, the amount of p53 associated with the microtubules was determined using a biochemical analysis, as described in Materials and methods. As expected and consistent with previous studies (51), the PTX-treated control cells demonstrated a moderate increase in the amount of p53 associated with microtubules compared with the cells not treated with PTX (Fig. 5B). However, when compared with PTX-treated EB1-KD cells, we observed a robust increase (~3-fold) in the amount of p53 associated with the cellular microtubules in the EB1-KD cells (Fig. 5B). Unlike the PTX-treated cells, the EB1-KD control cells did not exhibit any noticeable increase in the amount of p53 associated with the microtubules (Fig. 5B). Consistent with the biochemical analysis results, the immunofluorescence images also showed an increase in the amount of p53 localized to the microtubules in the EB1-depleted PTX-treated cells compared with the PTX-treated control and EB1-depleted control cells (Fig. 5C). The co-localization of p53 and the microtubules was evident from the increase in co-staining (yellow) of α-tubulin and p53 on microtubules in the merged images (enlarged images, Fig. 5C).

EB1 depletion induced formation of multiple spindle foci in PTX-treated mitotic MCF-7 cells

We investigated the effect of EB1 depletion on the effects of PTX in mitotic cells. First, we determined how the EB1-KD expression affects the levels of the spindle assembly checkpoint (SAC) proteins in MCF-7 cells. There was a higher BubR1 level in the control EB1-KD cells, which was stable and persistent, even after 72 h of esiRNA treatment (Fig. 6A). A similar change was observed in the Mad2 expression level (Fig. 6A). In contrast, the EB1-KD and control cells demonstrated time-dependent decreases in the BubR1 expression level, albeit at different rates, when treated with PTX (Fig. 6B). While the BubR1 level quickly decreased soon after 12 h of PTX treatment in the control cells, the rate of BubR1 loss was relatively slower in the EB1-KD cells (Fig. 6B).

Next, we examined the cellular phenotypes induced under these different conditions. MCF-7 cells were immunostained to visualize the chromosomes and spindle microtubules. EB1 depletion alone induced chromosome congressional defects in the metaphase cells (Fig. 6C). The chromosomes appeared to be unorganized and poorly aligned along the metaphase plate. Although the spindle poles appeared to be intact, their positions along the spindle pole axis were disrupted. In the majority of these cells, the poles were displaced from the axis, which resulted in a loss of spindle symmetry. These findings are consistent with previous findings in other cell lines (33,34). We then investigated whether PTX treatment in the EB1-KD cells intensifies the EB1 depletion-induced mitotic defects. PTX treatment in the EB1-KD cells induced multiple spindle foci formation with severe defects in the spindle organization and chromosome congression (Fig. 6C). The chromosomes appeared to be scattered around the spindle foci. The bipolar arrangement of the spindle microtubules was lost. The spindles appeared to be unusually long and curvy (Fig. 6C). About 60% mitotic cells had multiple spindle foci in the EB1 deficient PTX-treated condition (Fig. 6D). There was a strong correlation between these defects and the level of EB1 depletion in the cells. In contrast to the phenotypes observed in the EB1-depleted cells, the PTX-treated control MCF-7 cells appeared to be predominantly bipolar (Fig. 6C).

EB1 depletion potentiates PTX-induced microtubule stabilization and stabilizes PTX localization in MCF-7 cells

To investigate whether EB1-KD affects the ability of PTX to induce microtubule stabilization in cells, we assessed the stabilization of microtubules in PTX-treated control and EB1-KD cells. We used microtubule bundling (lateral association of microtubules) in an immunofluorescence assay to determine the PTX-induced stabilization of microtubules because microtubule bundle formation is the result of PTX-mediated microtubule polymer stabilization and therefore, represents the cellular activity of taxanes (15) (shown by arrows, Fig. 7A). Consistent with the previous studies (15), we observed formation of microtubule bundles in MCF-7 cells upon PTX treatment (Fig. 7A). However, the relative size and the number density of the bundles were significantly increased in the EB1-KD cells compared with the PTX-treated control cells (Fig. 7A). Based on an analysis of a large population of cells, it was found that the average microtubule intensity within the bundles was ~40% higher in the PTX-treated EB1-KD cells compared with the PTX-treated control cells (Fig. 7B). We also found that the average intensity of microtubules that are not associated with the bundles was similar in the control vs. the EB1-KD cells (not shown). To determine if this increase in microtubule bundling is the result of an increase in PTX localization in cells, we treated EB1-KD and control esiRNA-treated cells with fluorescent Ore-PTX and then determined the intracellular level of fluorescent PTX in the MCF-7 cells by live imaging. The level of intracellular fluorescent PTX was visibly higher in the PTX-treated EB1-KD cells compared with the PTX-treated control cells (Fig. 7C).

EB1 inhibited PTX-induced microtubule polymerization and PTX binding on microtubules

Next, we investigated whether EB1 regulates PTX-induced microtubule polymerization. PTX-induced microtubule polymerization was carried out in vitro by adding 0.1 μM PTX to purified tubulin. The turbidity curve indicates that 0.1 μM PTX was very efficient in inducing microtubule polymerization (square, Fig. 8A). Polymerization was attenuated when PTX was added to the pre-incubated mixture of tubulin and EB1. It was reduced by ~40% with only 0.1 μM EB1 (1:1 molar ratio of EB1 and PTX) (lower triangle, Fig. 8A). A 2-fold molar excess of EB1 over PTX further suppressed the polymerization by ~60% (circle, Fig. 8A). By using an unrelated histidine-tagged protein, we also confirmed that the inhibition of polymerization was not due to any non-specific effect of the 6-His tag attached to EB1 (data not shown). The inhibition of polymerization by EB1 was further confirmed by an SDS-PAGE analysis of the PTX-assembled microtubules after separating the polymerized proteins from the unpolymerized fractions through glycerol cushion followed by centrifugation (inset, Fig. 8A), as described in Materials and methods. The SDS-PAGE data also showed that the association between EB1 and the microtubules increased in a concentration-dependent manner (Fig. 8A). We also found that EB1 did not inhibit microtubule polymerization when the microtubules were polymerized from tubulin using DMSO as inducer (not shown), indicating that the inhibitory effect of EB1 is very much specific to the PTX-induced microtubule polymerization.

We then investigated whether EB1 affects PTX binding on microtubules. For this, we performed a quantitative fluorescence-based assay. The fluorescent Ore-PTX was allowed to bind to pre-polymerized steady-state microtubules in vitro in the absence or presence of increasing concentrations of EB1. The amount of PTX bound to the microtubules was determined by measuring the fluorescence intensity of the Ore-PTX associated with the microtubules, as described in Materials and methods. The Ore-PTX fluorescence in the microtubule solutions was reduced in the presence of EB1 indicating that EB1 inhibited the association between Ore-PTX and the microtubules in a dose-dependent manner (Fig. 8B). At 100 nM EB1 which corresponds to the molar ratio of PTX:EB1 as 1:5, the Ore-PTX binding was inhibited by ~30%. The inhibition was further increased to ~40% at 200 nM EB1 (Fig. 8B and C). Increase of EB1 >200 nM did not show any additional increase in the inhibition of PTX binding beyond 40%. We also confirmed by SDS-PAGE analysis that EB1 did not significantly alter the total microtubule polymer mass in the range of concentrations (20–200 nM) used in the PTX-binding experiment (inset, Fig. 8B) (28). This result confirms that the reduction of PTX binding to the microtubules in the presence of EB1 was not due to any decrease in the total microtubule polymer mass, but rather it was due to inhibition of PTX binding by EB1 to the microtubules. Altogether, these results indicate that EB1 inhibits PTX-mediated microtubule polymerization by preventing PTX binding to the microtubules.

Discussion

EB1 is a major +TIP and has previously been shown to promote proliferation of breast cancer cells (22,38). However, its effects on the sensitivity of breast cancer cells to microtubule-targeted drugs have not been clearly understood. In the present study, we demonstrate that EB1 plays an important role in regulating the sensitivity of breast cancer cells to microtubule-stabilizing drug, PTX. We show that PTX sensitivity correlates negatively with the EB1 expression level in breast cancer cell lines (Fig. 1). The EB1-KD increases PTX sensitivity in breast cancer cells by stimulating PTX-induced apoptosis and inhibition of cell viability (Figs. 24). Our findings support a mechanism in which EB1 downregulates PTX sensitivity by impairing PTX-induced stabilization of microtubule polymerization and inhibiting PTX binding to the microtubules (Figs. 68). During the preparation of this manuscript, we found a related report published by Luo et al (52), in which they investigated the role of EB1 on PTX sensitivity in breast cancer cells. However, it did not provide a clear conclusion due to a number of reasons. First, EB1 depletion was not significant in the study. Furthermore, GFP-fused EB1 was used to examine the effect of EB1 overexpression on PTX sensitivity. Fusion of GFP either to the N- or the C-terminus of EB1 has previously been shown to inhibit EB1’s ability to bind to the microtubules and to other +TIPs (53). The results of Luo et al (52) also could not clearly address the effect of EB1 on PTX-induced microtubule polymerization as the EB1 protein used in their assay was tagged with glutathione S-transferase (GST). Since GST is a bulky molecule and it itself exerts an effect on microtubule polymerization (52), the results could not clearly describe the exclusive role of EB1 in PTX-induced microtubule polymerization. In this study, we used esiRNAs, which ensured >90% suppression of EB1 expression in all the breast cancer cell lines used in this study (Fig. 2A). In the biochemical assays, we used purified recombinant human EB1, which clearly describes the exclusive effect of EB1 on PTX-induced microtubule polymerization (Fig. 8).

It has been previously reported that an increase in the nuclear translocation of p53 in cancer cells occurs in response to treatment with microtubule-targeting drugs, including PTX (51,56). It is generally accepted that the drug-induced stabilization of microtubules is the primary mechanism responsible for the increase in the p53 association with microtubules and its nuclear export (51,54,55). Consistently, we found that the association between p53 and cellular microtubules increased in response to PTX treatment in the control cells (51) (Fig. 5B and C). However, the association between p53 and cellular microtubules was further increased in the PTX-treated EB1-KD cells (Fig. 5B and C). The increased p53-microtubule association in PTX-treated EB1-KD cells could be the result of an increase in the stabilization of microtubules in the PTX-treated EB1-KD cells compared with the PTX-treated control cells. In support of this mechanism, we showed that microtubule bundling was greater in the PTX-treated EB1 deficient interphase cells compared with the PTX-treated control interphase cells (Fig. 7A). Consistently, we also observed an increase in the abundance of multiple spindle foci with abnormally long spindles in the PTX-treated EB1-KD mitotic cells (Fig. 6D). We also found that the level of intracellular PTX was higher in the EB1 deficient cells, which suggests that the association between PTX and the cellular microtubules is increased in the absence of EB1 (Fig. 7B). Altogether, these results support a mechanism in which EB1 inhibits the microtubule-stabilizing function of PTX. The results also support the previously proposed hypothesis that microtubule stabilization and p53 activation are closely linked (51,56).

EB1 mediates several key interactions that are essential for stabilizing spindle-chromosome attachment and activation of the SAC. For example, it has been shown that the disruption of its interaction with the plus-end protein TIP150 activates the SAC in HeLa cells (35). Consistently, we found that the EB1-KD resulted in a prolonged and stable accumulation of BubR1 and Mad2 in the control cells (Fig. 6A). It suggests that the EB1-deficient control cells undergo a stable mitotic arrest. On the contrary, EB1-deficient cells treated with PTX exhibited a time-dependent decrease in the BubR1 level, which indicates that the SAC became less effective or was suppressed by PTX treatment (Fig. 6B). It has been shown previously that mitotic cells with weak SAC activation may undergo apoptosis through either of these mechanisms: apoptosis may be triggered at the mitotically arrested stage, or the cells may exit mitosis as viable cells but then die at a multinucleated G1-stage (57). Our findings that PTX treatment increased apoptosis (Fig. 4A) and induced multinucleation with increased nuclear accumulation of p53 in the EB1-KD cells (Fig. 5A), suggest that apoptosis was triggered at the multinucleated stage after exiting mitosis rather than at the mitosis. We also found that the loss of BubR1 occurred relatively more rapidly in the PTX-treated control cells than in the PTX-treated EB1-KD cells (Fig. 6B). This indicates that the SAC activation was relatively weaker and transient in the PTX-treated control cells than the PTX-treated EB1-KD cells. As the PTX-treated control cells did not exhibit significant apoptosis like the PTX-treated EB1-KD cells (Fig. 3), a likely possibility is that these cells proceeded to the next round of division. In support of this possibility, we found that PTX-treated control cells displayed higher cell viability (higher IC50 of PTX) as compared with the PTX-treated EB1-KD cells (Fig. 2).

Recent high resolution electron microscopy analysis of interaction between the Schizosaccharomyces pombe EB1 homolog Mal3p and microtubules showed that EB1 binds between the microtubule protofilaments at and near the plus-ends (58,59). Combination of in vitro reconstitution and the sub-pixel level fluorescence analysis with human recombinant EB1 further revealed that EB1 binding accelerates the conformational maturation of microtubules at the polymerizing ends (32). Because EB1 is involved in conformational maturation of the ends, it may affect the assembly of protofilaments during PTX-mediated microtubule polymerization. Consistent with this mechanism, we found that PTX-induced microtubule polymerization was inhibited by EB1 (Fig. 8). Although EB1 effectively inhibited microtubule polymerization induced by PTX, it did not inhibit DMSO-induced microtubule polymerization (not shown), or it did not depolymerize the pre-polymerized steady-state microtubules in vitro (Fig. 8B) (28). These findings suggest that EB1 specifically inhibits the action of PTX on the microtubules, presumably by preventing PTX to bind to the tubulin subunits on the microtubules and to induce the assembly of the protofilaments.

How could EB1 being a +TIP inhibit the anti-mitotic action of PTX, which is known to bind all along the microtubule protofilament? It has been demonstrated previously that PTX exerts its anti-mitotic action in cancer cells at a very sub-stoichiometric level, the concentrations at which it does not significantly affect microtubule polymer mass, but suppresses microtubule dynamics very efficiently (11,12). This suggested that the anti-mitotic action of PTX is predominantly attributed to its binding at the microtubule plus-ends rather than its binding along the microtubule surface (6). Thus, the increased accumulation of EB1 on plus-ends of microtubules could specifically inhibit binding of PTX to the ends, but PTX binding along the surface of microtubule may not be affected under such condition. Consistent with this possibility, we observed that EB1 inhibited binding of PTX on the microtubules to a maximum extent of 40%, and the remaining 60% PTX was still bound to the microtubules (Fig. 8B). The inhibitory effect of EB1 on PTX-induced microtubule polymerization is also in good support with the effects of the EB proteins on the dynamics of PTX-treated microtubules shown by Mohan et al (31) in which they showed that the overall dynamicity of PTX-treated microtubules is increased in the presence of EB proteins in vitro. Specifically, the EB proteins increased the frequencies of growth (rescue) and shortening (catastrophe) of the PTX-treated microtubules (31). Similar to these findings, study by Pagano et al, showed that at low PTX concentration, microtubule dynamics is increased instead of suppressed in the presence of EB protein, EB3 (60).

In summary, we show from our cellular and biochemical results that inhibition of EB1 expression sensitizes breast cancer cells to PTX-induced cytotoxicity and apoptosis. The ability of EB1 to inhibit PTX-induced microtubule polymerization and PTX binding to the microtubules suggests that EB1 confers resistance against the microtubule-stabilizing activity of PTX. The results support the hypothesis that EB1 downregulates PTX sensitivity through a mechanism in which it inhibits PTX binding to the microtubules and suppresses PTX-induced stabilization of microtubule polymerization.

Acknowledgements

We thank S. Murty Srinivasula, IISER-Thiruvananthapuram for critical comments and suggestions, and for providing cell lines. We also thank Stephen Doxsey, UMass Medical (Worcester, MA, USA) for providing pGFP-EB1 plasmid. This study was supported by a CSIR-Govt. of India research grant to T.M.

References

1 

Kamangar F, Dores GM and Anderson WF: Patterns of cancer incidence, mortality, and prevalence across five continents: defining priorities to reduce cancer disparities in different geographical regions of the world. J Clin Oncol. 24:2137–2150. 2006. View Article : Google Scholar

2 

Mamounas EP, Bryant J, Lembersky B, Fehrenbacher L, Sedlacek SM, Fisher B, Wickerham DL, Yothers G, Soran A and Wolmark N: Paclitaxel after doxorubicin and cyclophosphamide as adjuvant chemotherapy for node-positive breast cancer: results from NSABP B-28. J Clin Oncol. 23:3686–3696. 2005. View Article : Google Scholar : PubMed/NCBI

3 

Martin M, Seguí MA, Antón A, Ruiz A, Ramos M, Adrover E, Aranda I, Rodríguez-Lescure A, Grosse R, Calvo L, et al: GEICAM 9805 Investigators: Adjuvant docetaxel for high-risk, node-negative breast cancer. N Engl J Med. 363:2200–2210. 2010. View Article : Google Scholar : PubMed/NCBI

4 

Henderson IC, Berry DA, Demetri GD, Cirrincione CT, Goldstein LJ, Martino S, Ingle JN, Cooper MR, Hayes DF, Tkaczuk KH, et al: Improved outcomes from adding sequential Paclitaxel but not from escalating Doxorubicin dose in an adjuvant chemotherapy regimen for patients with node-positive primary breast cancer. J Clin Oncol. 21:976–983. 2003. View Article : Google Scholar

5 

McGrogan BT, Gilmartin B, Carney DN and McCann A: Taxanes, microtubules and chemoresistant breast cancer. Biochim Biophys Acta. 1785:96–132. 2008.PubMed/NCBI

6 

Jordan MA and Wilson L: Microtubules as a target for anticancer drugs. Nat Rev Cancer. 4:253–265. 2004. View Article : Google Scholar : PubMed/NCBI

7 

Manfredi JJ, Parness J and Horwitz SB: Taxol binds to cellular microtubules. J Cell Biol. 94:688–696. 1982. View Article : Google Scholar : PubMed/NCBI

8 

Giannakakou P, Gussio R, Nogales E, Downing KH, Zaharevitz D, Bollbuck B, Poy G, Sackett D, Nicolaou KC and Fojo T: A common pharmacophore for epothilone and taxanes: molecular basis for drug resistance conferred by tubulin mutations in human cancer cells. Proc Natl Acad Sci USA. 97:2904–2909. 2000. View Article : Google Scholar : PubMed/NCBI

9 

Orr GA, Verdier-Pinard P, McDaid H and Horwitz SB: Mechanisms of Taxol resistance related to microtubules. Oncogene. 22:7280–7295. 2003. View Article : Google Scholar : PubMed/NCBI

10 

Kavallaris M, Kuo DY, Burkhart CA, Regl DL, Norris MD, Haber M and Horwitz SB: Taxol-resistant epithelial ovarian tumors are associated with altered expression of specific beta-tubulin isotypes. J Clin Invest. 100:1282–1293. 1997. View Article : Google Scholar : PubMed/NCBI

11 

Derry WB, Wilson L and Jordan MA: Substoichiometric binding of taxol suppresses microtubule dynamics. Biochemistry. 34:2203–2211. 1995. View Article : Google Scholar : PubMed/NCBI

12 

Jordan MA, Toso RJ, Thrower D and Wilson L: Mechanism of mitotic block and inhibition of cell proliferation by taxol at low concentrations. Proc Natl Acad Sci USA. 90:9552–9556. 1993. View Article : Google Scholar : PubMed/NCBI

13 

Blagosklonny MV, Robey R, Sheikh MS and Fojo T: Paclitaxel-induced FasL-independent apoptosis and slow (non-apoptotic) cell death. Cancer Biol Ther. 1:113–117. 2002. View Article : Google Scholar : PubMed/NCBI

14 

Ganguly A, Yang H, Pedroza M, Bhattacharya R and Cabral F: Mitotic centromere-associated kinesin (MCAK) mediates paclitaxel resistance. J Biol Chem. 286:36378–36384. 2011. View Article : Google Scholar : PubMed/NCBI

15 

Sung M and Giannakakou P: BRCA1 regulates microtubule dynamics and taxanes-induced apoptotic cell signaling. Oncogene. 33:1418–1428. 2014. View Article : Google Scholar : PubMed/NCBI

16 

Wang H, Liu B, Zhang C, Peng G, Liu M, Li D, Gu F, Chen Q, Dong JT, Fu L and Zhou J: Parkin regulates paclitaxel sensitivity in breast cancer via a microtubule-dependent mechanism. J Pathol. 218:76–85. 2009. View Article : Google Scholar : PubMed/NCBI

17 

Alli E, Bash-Babula J, Yang JM and Hait WN: Effect of stathmin on the sensitivity to antimicrotubule drugs in human breast cancer. Cancer Res. 62:6864–6869. 2002.PubMed/NCBI

18 

Sun X, Li D, Yang Y, Ren Y, Li J, Wang Z, Dong B, Liu M and Zhou J: Microtubule-binding protein CLIP-170 is a mediator of paclitaxel sensitivity. J Pathol. 226:666–673. 2012. View Article : Google Scholar : PubMed/NCBI

19 

Lu Q and Luduena RF: Removal of beta III isotype enhances taxol induced microtubule assembly. Cell Struct Funct. 18:173–182. 1993. View Article : Google Scholar : PubMed/NCBI

20 

Kamath K, Wilson L, Cabral F and Jordan MA: Beta-tubulin induces paclitaxel resistance in association with reduced effects on microtubule dynamic instability. J Biol Chem. 280:12902–12907. 2005. View Article : Google Scholar : PubMed/NCBI

21 

Akhmanova A and Steinmetz MO: Tracking the ends: a dynamic protein network controls the fate of microtubule tips. Nat Rev Mol Cell Biol. 9:309–322. 2008. View Article : Google Scholar : PubMed/NCBI

22 

Honnappa S, Okhrimenko O, Jaussi R, Jawhari H, Jelesarov I, Winkler FK and Steinmetz MO: Key interaction modes of dynamic +TIP networks. Mol Cell. 23:663–671. 2006. View Article : Google Scholar : PubMed/NCBI

23 

Komarova Y, Lansbergen G, Galjart N, Grosveld F, Borisy GG and Akhmanova A: EB1 and EB3 control CLIP dissociation from the ends of growing microtubules. Mol Biol Cell. 16:5334–5345. 2005. View Article : Google Scholar : PubMed/NCBI

24 

Askham JM, Vaughan KT, Goodson HV and Morrison EE: Evidence that an interaction between EB1 and p150(Glued) is required for the formation and maintenance of a radial microtubule array anchored at the centrosome. Mol Biol Cell. 13:3627–3645. 2002. View Article : Google Scholar : PubMed/NCBI

25 

Dixit R, Barnett B, Lazarus JE, Tokito M, Goldman YE and Holzbaur EL: Microtubule plus-end tracking by CLIP-170 requires EB1. Proc Natl Acad Sci USA. 106:492–497. 2009. View Article : Google Scholar : PubMed/NCBI

26 

Su LK and Qi Y: Characterization of human MAPRE genes and their proteins. Genomics. 71:142–149. 2001. View Article : Google Scholar : PubMed/NCBI

27 

Tirnauer JS, Grego S, Salmon ED and Mitchison TJ: EB1-microtubule interactions in Xenopus egg extracts: role of EB1 in microtubule stabilization and mechanisms of targeting to microtubules. Mol Biol Cell. 13:3614–3626. 2002. View Article : Google Scholar : PubMed/NCBI

28 

Manna T, Honnappa S, Steinmetz MO and Wilson L: Suppression of microtubule dynamic instability by the +TIP protein EB1 and its modulation by the CAP-Gly domain of p150glued. Biochemistry. 47:779–786. 2008. View Article : Google Scholar : PubMed/NCBI

29 

Vitre B, Coquelle FM, Heichette C, Garnier C, Chrétien D and Arnal I: EB1 regulates microtubule dynamics and tubulin sheet closure in vitro. Nat Cell Biol. 10:415–421. 2008. View Article : Google Scholar : PubMed/NCBI

30 

Komarova Y, De Groot CO, Grigoriev I, Gouveia SM, Munteanu EL, Schober JM, Honnappa S, Buey RM, Hoogenraad CC, Dogterom M, et al: Mammalian end binding proteins control persistent microtubule growth. J Cell Biol. 184:691–706. 2009. View Article : Google Scholar : PubMed/NCBI

31 

Mohan R, Katrukha EA, Doodhi H, Smal I, Meijering E, Kapitein LC, Steinmetz MO and Akhmanova A: End-binding proteins sensitize microtubules to the action of microtubule-targeting agents. Proc Natl Acad Sci USA. 110:8900–8905. 2013. View Article : Google Scholar : PubMed/NCBI

32 

Maurer SP, Cade NI, Bohner G, Gustafsson N, Boutant E and Surrey T: EB1 accelerates two conformational transitions important for microtubule maturation and dynamics. Curr Biol. 24:372–384. 2014. View Article : Google Scholar : PubMed/NCBI

33 

Draviam VM, Shapiro I, Aldridge B and Sorger PK: Misorientation and reduced stretching of aligned sister kinetochores promote chromosome missegregation in EB1- or APC-depleted cells. EMBO J. 25:2814–2827. 2006. View Article : Google Scholar : PubMed/NCBI

34 

Brüning-Richardson A, Langford KJ, Ruane P, Lee T, Askham JM and Morrison EE: EB1 is required for spindle symmetry in mammalian mitosis. PLoS One. 6:e288842011.PubMed/NCBI

35 

Ward T, Wang M, Liu X, Wang Z, Xia P, Chu Y, Wang X, Liu L, Jiang K, Yu H, et al: Regulation of a dynamic interaction between two microtubule-binding proteins, EB1 and TIP150, by the mitotic p300/CBP-associated factor (PCAF) orchestrates kinetochore microtubule plasticity and chromosome stability during mitosis. J Biol Chem. 288:15771–15785. 2013. View Article : Google Scholar

36 

Green RA, Wollman R and Kaplan KB: APC and EB1 function together in mitosis to regulate spindle dynamics and chromosome alignment. Mol Biol Cell. 16:4609–4622. 2005. View Article : Google Scholar : PubMed/NCBI

37 

Sun L, Gao J, Dong X, Liu M, Li D, Shi X, Dong J, Lu X, Liu C and Zhou J: EB1 promotes Aurora-B kinase activity through blocking its inactivation by protein phosphatase 2A. Proc Natl Acad Sci USA. 105:7153–7158. 2008. View Article : Google Scholar : PubMed/NCBI

38 

Dong X, Liu F, Sun L, Liu M, Li D, Su D, Zhu Z, Dong JT, Fu L and Zhou J: Oncogenic function of microtubule end-binding protein 1 in breast cancer. J Pathol. 220:361–369. 2010.PubMed/NCBI

39 

Jaïs P, Sabourin JC, Bombled J, Rougier P, Lasser P, Duvillard P, Bénard J and Bressac-de Paillerets B: Absence of somatic alterations of the EB1 gene adenomatous polyposis coli-associated protein in human sporadic colorectal cancers. Br J Cancer. 78:1356–1360. 1998.PubMed/NCBI

40 

Nishigaki R, Osaki M, Hiratsuka M, Toda T, Murakami K, Jeang KT, Ito H, Inoue T and Oshimura M: Proteomic identification of differentially-expressed genes in human gastric carcinomas. Proteomics. 5:3205–3213. 2005. View Article : Google Scholar : PubMed/NCBI

41 

Wang Y, Zhou X, Zhu H, Liu S, Zhou C, Zhang G, Xue L, Lu N, Quan L, Bai J, et al: Overexpression of EB1 in human esophageal squamous cell carcinoma (ESCC) may promote cellular growth by activating beta-catenin/TCF pathway. Oncogene. 24:6637–6645. 2005. View Article : Google Scholar : PubMed/NCBI

42 

Soule HD, Maloney TM, Wolman SR, Peterson WD Jr, Brenz R, McGrath CM, Russo J, Pauley RJ, Jones RF and Brooks SC: Isolation and characterization of a spontaneously immortalized human breast epithelial cell line, MCF-10. Cancer Res. 50:6075–6086. 1990.PubMed/NCBI

43 

Kittler R, Heninger AK, Franke K, Habermann B and Buchholz F: Production of endoribonuclease-prepared short interfering RNAs for gene silencing in mammalian cells. Nat Methods. 2:779–784. 2005. View Article : Google Scholar : PubMed/NCBI

44 

Calegari F, Haubensak W, Yang D, Huttner WB and Buchholz F: Tissue-specific RNA interference in post-implantation mouse embryos with endoribonuclease-prepared short interfering RNA. Proc Natl Acad Sci USA. 99:14236–14240. 2002. View Article : Google Scholar : PubMed/NCBI

45 

Manna T, Thrower DA, Honnappa S, Steinmetz MO and Wilson L: Regulation of microtubule dynamic instability in vitro by differentially phosphorylated stathmin. J Biol Chem. 284:15640–15649. 2009. View Article : Google Scholar : PubMed/NCBI

46 

Bradford MM: A rapid and sensitive method for the quantitation of microgram quantities of protein utilizing the principle of protein-dye binding. Anal Biochem. 72:248–254. 1976. View Article : Google Scholar : PubMed/NCBI

47 

Zhang L, Lau YK, Xia W, Hortobagyi GN and Hung MC: Tyrosine kinase inhibitor emodin suppresses growth of HER-2/neu-overexpressing breast cancer cells in athylic mice and sensitizes these cells to the inhibitory effects of paclitaxel. Clin Cancer Res. 5:343–353. 1999.

48 

Gireesh KK, Rashid A, Chakraborti S, Panda D and Manna T: CIL-102 binds to tubulin at colchicine binding site and triggers apoptosis in MCF-7 cells by inducing monopolar and multinucleated cells. Biochem Pharmacol. 84:633–645. 2012. View Article : Google Scholar : PubMed/NCBI

49 

Young A, Dictenberg JB, Purohit A, Tuft R and Doxsey SJ: Cytoplasmic dynein-mediated assembly of pericentrin and gamma tubulin onto centrosomes. Mol Biol Cell. 11:2047–2056. 2000. View Article : Google Scholar : PubMed/NCBI

50 

Ciardiello F, Caputo R, Pomatico G, De Laurentiis M, De Placido S, Bianco AR and Tortora G: Resistance to taxanes is induced by c-erbB-2 overexpression in human MCF-10A mammary epithelial cells and is blocked by combined treatment with an antisense oligonucleotide targeting type I protein kinase A. Int J Cancer. 85:710–715. 2000. View Article : Google Scholar

51 

Giannakakou P, Sackett DL, Ward Y, Webster KR, Blagosklonny MV and Fojo T: p53 is associated with cellular microtubules and is transported to the nucleus by dynein. Nat Cell Biol. 2:709–717. 2000. View Article : Google Scholar : PubMed/NCBI

52 

Luo Y, Li D, Ran J, Yan B, Chen J, Dong X, Liu Z, Liu R, Zhou J and Liu M: End-binding protein 1 stimulates paclitaxel sensitivity in breast cancer by promoting its actions toward microtubule assembly and stability. Protein Cell. 5:469–479. 2014. View Article : Google Scholar : PubMed/NCBI

53 

Skube SB, Chaverri JM and Goodson HV: Effect of GFP tags on the localization of EB1 and EB1 fragments in vivo. Cytoskeleton (Hoboken). 67:1–12. 2010. View Article : Google Scholar : PubMed/NCBI

54 

Giannakakou P, Nakano M, Nicolaou KC, O’Brate A, Yu J, Blagosklonny MV, Greber UF and Fojo T: Enhanced microtubule-dependent trafficking and p53 nuclear accumulation by suppression of microtubule dynamics. Proc Natl Acad Sci USA. 99:10855–10860. 2002. View Article : Google Scholar : PubMed/NCBI

55 

Rathinasamy K and Panda D: Kinetic stabilization of microtubule dynamic instability by benomyl increases the nuclear transport of p53. Biochem Pharmacol. 76:1669–1680. 2008. View Article : Google Scholar : PubMed/NCBI

56 

Carney BK, Caruso Silva V and Cassimeris L: The microtubule cytoskeleton is required for a G2 cell cycle delay in cancer cells lacking stathmin and p53. Cytoskeleton (Hoboken). 69:278–289. 2012. View Article : Google Scholar : PubMed/NCBI

57 

Tao W, South VJ, Zhang Y, Davide JP, Farrell L, Kohl NE, Sepp-Lorenzino L and Lobell RB: Induction of apoptosis by an inhibitor of the mitotic kinesin KSP requires both activation of the spindle assembly checkpoint and mitotic slippage. Cancer Cell. 8:49–59. 2005. View Article : Google Scholar : PubMed/NCBI

58 

Maurer SP, Fourniol FJ, Bohner G, Moores CA and Surrey T: EBs recognize a nucleotide dependent structural cap at growing microtubule ends. Cell. 149:371–382. 2012. View Article : Google Scholar : PubMed/NCBI

59 

Sandblad L, Busch KE, Tittmann P, Gross H, Brunner D and Hoenger A: The Schizosaccharomyces pombe EB1 homolog Mal3p binds and stabilizes microtubule lattice seam. Cell. 127:1415–1424. 2006. View Article : Google Scholar

60 

Pagano A, Honoré S, Mohan R, Berges R, Akhmanova A and Braguer D: Epothilone B inhibits migration of glioblastoma cells by inducing microtubule catastrophes and affecting EB1 accumulation at microtubule plus ends. Biochem Pharmacol. 84:432–443. 2012. View Article : Google Scholar : PubMed/NCBI

Related Articles

Journal Cover

January-2015
Volume 46 Issue 1

Print ISSN: 1019-6439
Online ISSN:1791-2423

Sign up for eToc alerts

Recommend to Library

Copy and paste a formatted citation
x
Spandidos Publications style
Thomas GE, Sreeja JS, Gireesh KK, Gupta H and Manna TK: +TIP EB1 downregulates paclitaxel‑induced proliferation inhibition and apoptosis in breast cancer cells through inhibition of paclitaxel binding on microtubules. Int J Oncol 46: 133-146, 2015
APA
Thomas, G.E., Sreeja, J.S., Gireesh, K.K., Gupta, H., & Manna, T.K. (2015). +TIP EB1 downregulates paclitaxel‑induced proliferation inhibition and apoptosis in breast cancer cells through inhibition of paclitaxel binding on microtubules. International Journal of Oncology, 46, 133-146. https://doi.org/10.3892/ijo.2014.2701
MLA
Thomas, G. E., Sreeja, J. S., Gireesh, K. K., Gupta, H., Manna, T. K."+TIP EB1 downregulates paclitaxel‑induced proliferation inhibition and apoptosis in breast cancer cells through inhibition of paclitaxel binding on microtubules". International Journal of Oncology 46.1 (2015): 133-146.
Chicago
Thomas, G. E., Sreeja, J. S., Gireesh, K. K., Gupta, H., Manna, T. K."+TIP EB1 downregulates paclitaxel‑induced proliferation inhibition and apoptosis in breast cancer cells through inhibition of paclitaxel binding on microtubules". International Journal of Oncology 46, no. 1 (2015): 133-146. https://doi.org/10.3892/ijo.2014.2701