Differentially expressed proteins in human breast cancer cells sensitive and resistant to paclitaxel

  • Authors:
    • Nela Pavlikova
    • Irena Bartonova
    • Lucia Dincakova
    • Petr Halada
    • Jan Kovar
  • View Affiliations

  • Published online on: June 3, 2014     https://doi.org/10.3892/ijo.2014.2484
  • Pages: 822-830
Metrics: Total Views: 0 (Spandidos Publications: | PMC Statistics: )
Total PDF Downloads: 0 (Spandidos Publications: | PMC Statistics: )


Abstract

The resistance of cancer cells to chemotherapeutic drugs represents a major problem in cancer treatment. Despite all efforts, mechanisms of resistance have not yet been elucidated. To reveal proteins that could be involved in resistance to taxanes, we compared protein expression in whole cell lysates of SK-BR-3 breast cancer cells sensitive to paclitaxel and in lysates of the same line with acquired resistance to paclitaxel. The resistant SK-BR-3 cell line was established in our lab. Protein separation was achieved using high-resolution 2D-electrophoresis, computer analysis and mass spectro-metry. With these techniques we identified four proteins with different expression in resistant SK-BR-3 cells, i.e., serpin B3, serpin B4, heat shock protein 27 (all three upregulated) and cytokeratin 18 (downregulated). Observed changes were confirmed using western blot analysis. This study suggests new directions worthy of further study in the effort to reveal the mechanism of resistance to paclitaxel in breast cancer cells.

Introduction

Taxanes, diterpenes originally isolated from the bark of Taxus brevifolia, represent a relatively new group of anticancer drugs. Paclitaxel, the first taxane used in chemotherapy, came into clinical use in the early nineties and has become one of the main anticancer drugs, especially for the treatment of ovarian, breast and lung cancer, and Kaposi’s sarcoma. In rapidly dividing cancer cells, paclitaxel and other taxanes bind to β-tubulin subunit and stabilize it. In doing so, they destroy the dynamic instability of microtubules, which is necessary for their proper function. As a result, cells are arrested in M phase of the cell cycle and subsequently undergo apoptosis (1). Nevertheless, cancer cell resistance (either intrinsic or acquired) to these compounds often reverses the benefits of taxane treatment.

The last decade has seen enormous efforts to elucidated the mechanisms of resistance to anticancer drugs (27). Nonetheless, elucidation is far from complete; although some mechanisms of resistance have been described that could provide useful markers of taxane resistance.

Drug efflux pumps often top the list of basic mechanisms of resistance that are common for many anticancer compounds. Upregulation of the ABC transporter P-glycoprotein (Pgp) (encoded by the ABCB1 gene) results in enhanced efflux of anticancer drugs from cells. The mechanism has been well established in resistance to taxanes (811). Other significant ABC transporters connected with taxane resistance include multidrug resistance-associated proteins 1 (ABCC1/MRP1) (12), 2 (ABCC2/MRP2) (13) and 3 (ABCC3/MRP3) (14). Oddly, upregulation of these proteins has not always been detected in taxane resistant cells (1517), which conflicts with expectations.

Enhanced drug metabolism within the cell is considered to be another possible explanation for reduced taxane effectiveness in cancer cells. Two isoforms of cytochrome p450 are mainly responsible for paclitaxel utilization, i.e. cyp3A4 and cyp2C8 (18). In some cases, overexpression of these isoforms have been found in taxane resistant cancer cells (19,20). Nevertheless, individual patient genetic variability could account for the differences (21). Also phase II metabolic enzyme glutathione S-transferase P1 (GSTP1) has been reported to be connected with paclitaxel resistance. This enzyme conjugates glutathione with various drugs causing their deactivation. Localized inside the nucleus, GSTP1 can also protect DNA against damage caused by anticancer drugs (22). Some studies have suggested that expression of GSTP1 could be associated with higher survival rates of cancer cells after taxane treatment (2325). Interestingly, GSTP1 has been shown to co-localize with another possible marker of taxane resistance, Bcl-2, within the nucleus (2628).

Another mechanism of taxane resistance involve various β-tubulin mutations which can weaken the binding of taxane to β-tubulin and change the dynamics of the microtubule system (2931). Mutations of α-tubulin are also believed to be connected with taxane resistance, but to a lesser extent; their importance lies more in influencing the binding of microtubule-associated proteins (MAPs) (32,33).

Molecules directly connected with cell survival and apoptosis can also be involved. Expression of proteins, such as survivin, an inhibitor of apoptosis known to interact with tubulin (34,35), cyclin-dependent kinase inhibitor p21 (35,36) or p53, a factor inducing its expression (37,38), have been reported to influence cell sensitivity to taxanes.

Even though the mechanisms and compounds mentioned here is a heterogeneous group, there have been attempts to find connections between various mechanisms and markers of taxane resistance. De Hoon et al (22) have suggested that some mechanisms of resistance in breast cancer cells might be connected to expression and signaling of human epidermal growth factor receptor 2 (HER2) and subsequent activation of STAT3 as well as other downstream effectors.

Despite the current knowledge concerning resistance to taxanes, more studies are required to elucidate this phenomenon and provide reliable biomarkers to reveal tumors where taxane treatment (with all its side-effects) would have no impact. Indeed, investigation of resistance mechanisms (insights into treatment failure) remains a key task in cancer treatment (7,39).

To study further the mechanisms of resistance to taxanes, we employed a proteomic approach using the SK-BR-3 cell line (human breast adenocarcinoma) with acquired resistance to paclitaxel. We compared protein expression patterns from paclitaxel resistant cells to patterns from non-resistant cells and searched for proteins with different expression.

Materials and methods

Materials

Immobiline DryStrips pH 3–11NL 18 cm, Protein Extraction Buffer-III, 2-D Clean-Up Kit, 2-D Quant Kit, DeStreak Reagent and IPG Buffer 3–11NL were obtained from GE Healthcare (Uppsala, Sweden). Rabbit polyclonal antibody against serpin B4 (anti-SERPINB4 antibody defined by manufacturer also as ‘SCCA2/SCCA1 fusion protein antibody’: ab104338), mouse monoclonal antibody against cytokeratin 18 (anti-cytokeratin 18 antibody: ab55395), mouse monoclonal antibody against heat shock protein 27 [anti-Hsp27 antibody (8A7): ab78436] and rabbit polyclonal antibody against GAPDH (anti-GAPDH antibody: ab9485) were obtained from Abcam (Cambridge, UK). Mouse monoclonal antibody anti-actin (clone AC-40: A3853) against human actin was obtained from Sigma-Aldrich (St. Louis, MO, USA).

Cells and cell culture conditions

Human breast carcinoma cell line SK-BR-3 was obtained from American Type Culture Collection (ATCC, Rockville, MD, USA). Resistant SK-BR-3 cells were derived in our lab from original sensitive SK-BR-3 cells by gradual adaptation to paclitaxel. The concentration of paclitaxel in the culture medium was increased approximately every 20th passage. Resistant SK-BR-3 cells display long-term proliferation in culture medium containing 100 nM paclitaxel. At this taxane concentration original sensitive SK-BR-3 cells die within 72 h. This concentration of paclitaxel approximates concentrations used in clinical practice (26,40).

The culture medium consisted of basic medium supplemented with 10% heat-inactivated fetal bovine serum. The basic medium was RPMI-1640 based medium containing extra L-glutamine (300 μg/ml), sodium pyruvate (110 μg/ml), HEPES (15 mM), penicillin (100 U/ml) and streptomycin (100 μg/ml), as previously described (4143). Cells were maintained at 37°C in a humidified atmosphere of 5% CO2 in air. For maintaining taxane-resistant cells, the medium was supplemented with paclitaxel (in DMSO) to a final concentration of 100 nM (final concentration of DMSO in medium was below 0.1%) just prior to use. For experiments, cells were harvested and seeded (approximately 5×106 in 15 ml of medium per sample). After 24-h preincubation the culture medium was replaced with a fresh medium and after another 24 h the cells were harvested.

Sample preparation for 2D-electrophoresis

Cells were trypsinized, washed with ice-cold PBS and resuspended in Protein Extraction Buffer-III (GE Healthcare) (urea, thiourea, ASB-16, CHAPS) containing 1% Protease-Inhibitor Mix G (SERVA Electrophoresis GmbH, Heidelberg, Germany). A 2-D Clean-Up Kit (GE Healthcare) was used for sample purification, per manufacturer’s instructions. Briefly, the sample was precipitated using Precipitant and Co-precipitant, incubated for 15 min at 4°C and centrifuged. The pellet was then washed with Wash Buffer with Wash Additive, incubated at −20°C for 30 min and centrifuged. After brief air drying, the pellet was dissolved again in Protein Extraction Buffer-III as described above. Protein concentrations were determined using a 2-D Quant Kit (GE Healthcare), which is compatible with both the detergents and thiourea present in Protein Extraction Buffer-III.

2D-electrophoresis: isoelectric focusing

Isoelectric focusing was carried out in an IPGphor focusing unit (GE Healthcare, former Amersham Biosciences) using 3–11NL Immobiline DryStrips 18 cm (GE Healthcare). Each strip was rehydrated for at least 24 h with 340 μl of the diluted protein sample containing 400 μg of protein, 6.8 μl of bromophenol blue solution (0.1%), 4 μl of DeStreak Reagent (GE Healthcare) and 6.8 μl of IPG buffer (pH 3.0–11.0; GE Healthcare) in Protein Extraction Buffer-III as described above. After rehydration, strips were focused at 20°C with current limited to 50 μA/strip using the following conditions: 150 V for 1 h, gradient 150–300 V for 10 min, 300 V for 2 h, gradient 300–1,000 V for 10 min, 1,000 V for 2 h, gradient 1,000–8,000 V for 1 h, 8,000 V for 15 h.

2D-electrophoresis: equilibration

After focusing, strips were equilibrated for 20 min in equilibration buffer containing 6 M urea, 30% glycerol, 2% SDS, 50 mM Tris (stock solution of 1.5 M Tris-HCl, pH 8.8), 2% dithiothreitol and then equilibrated for another 20 min in the same buffer with 2.5% iodoacetamide instead of dithiothreitol. Then the strips were placed on the top of gels and sealed using 0.5% agarose solution containing bromophenol blue.

2D-electrophoresis: SDS-PAGE

The second dimension was performed using an Ettan DaltSix Electrophoresis System (GE Healthcare, former Amersham Biosciences). A total of 10% polyacrylamide gel (pH 8.8) with a 4% stacking gel (pH 8.8) was used for protein separation. Gels were run at a constant current starting with 35 mA/gel for 1 h and then 65 mA/gel till the blue line reached the bottom of the gels (approximately 4 h). After running the second dimension, each gel was washed 3×10 min in distilled water and stained overnight in 500 ml of colloidal Coomassie Blue solution per gel according to (44).

Gel image and analysis

Stained gels were scanned using a calibrated UMAX PowerLook 1120 scanner employing LabScan software (both from GE Healthcare). Gel couples were analyzed using Image MasterTM 2D Platinum 6.0 software (GE Healthcare, former Amersham Biosciences). We analyzed differences in intensity between corresponding spots on each set of gels (each set contained gel with proteins from sensitive SK-BR-3 cells and gel with proteins from resistant SK-BR-3). Spots with at least 2-fold average difference in expression between sensitive and resistant cell lysates were selected. Statistical significance of difference in intensity of these spots was determined using the Student’s t-test.

Enzymatic digestions

CBB-stained protein spots were excised from the gel, cut into small pieces and destained using 50 mM 4-ethylmorpholine acetate (pH 8.1) in 50% acetonitrile (MeCN). After complete destaining, gels were washed with water, reduced in size by dehydration in MeCN and reconstituted again in water. The supernatant was removed and the gel was partly dried in a SpeedVac concentrator. Gel pieces were then incubated overnight at 37°C in a cleavage buffer containing 25 mM 4-ethylmorpholine acetate, 5% MeCN and trypsin (100 ng; Promega). The resulting peptides were extracted with 40% MeCN/0.1% trifluoroacetic acid (TFA).

MALDI mass spectrometry and protein identification

An aqueous 50% MeCN/0.1% TFA solution of α-cyano-4-hydroxycinnamic acid (5 mg/ml; Sigma-Aldrich, St. Louis, MO, USA) was used as a MALDI matrix. Peptide mixture (1 μl) was deposited on a MALDI plate, allowed to air-dry at room temperature and overlaid with 0.4 μl of matrix. Mass spectra were measured using an Ultraflex III MALDI-TOF (Bruker Daltonics, Bremen, Germany); mass range of 700–4,000 Da, calibrated internally using the monoisotopic [M+H]+ ions of trypsin auto-proteolytic fragments (842.5 and 2,211.1 Da). The peak lists created using the flexAnalysis 3.3 program were searched using an in-house MASCOT search engine against SwissProt 2013_12 database subset of human proteins with the following search settings: peptide tolerance of 30 ppm, missed cleavage site value set to one, variable carbamidomethylation of cysteine, oxidation of methionine and protein N-term acetylation. Proteins with MOWSE scores over the threshold, 56 (calculated for the used settings) were considered as identified. The identity of protein candidate was confirmed using MS/MS analysis.

Western blot analysis

Western blot was carried as described previously (41,43,45) with minor modification. Briefly, whole cell lysates were separated by SDS-PAGE using 9% acrylamide gels and blotted into 0.2 μm nitrocellulose membrane for 2 h at 0.25 A using a Mini-Protean II blotting apparatus (Bio-Rad). The membrane was blocked with 5% low fat milk in TBS for 30 min. Tween-20 (0.1%) in TBS was used for washing. The washed membrane was then incubated with the relevant primary antibody. After incubation (overnight, room temperature), the washed membrane was incubated for 1 h with the corresponding horseradish peroxidase-conjugated secondary antibody (Santa Cruz Biotechnology, Santa Cruz, CA, USA). The horseradish peroxidase-conjugated secondary antibody was detected by enhanced chemiluminescence using the SuperSignal reagent from Pierce (Rockford, IL, USA) and a Gel Logic 4000 PRO device. Optical density of bands was analyzed using Carestream Molecular Imaging Software v. 5.2.0 (Carestream Health, Berlin, Germany).

Statistical analysis

Statistics was performed using Sigma Plot, version 11.00. Significant differences in intensity of spots (2-D gels) and bends (western blot) were analyzed by Student’s t-test.

Results

Detection of proteins with changed expression

In order to compare protein expression in the human breast adenocarcinoma cell line (SK-BR-3) sensitive to paclitaxel and in SK-BR-3 cells with acquired resistance to paclitaxel, we prepared whole cell lysates of these cells. Lysates were prepared using Protein Extraction Buffer. Samples were purified using TCA-based precipitation and 400 μg of proteins of each sample were used for two-dimensional electrophoresis (2-DE) (see Materials and methods).

We prepared three pairs of 2-DE gels from three independent sets of samples containing SK-BR-3 cells resistant to paclitaxel and SK-BR-3 cells sensitive to paclitaxel. Gels were stained using colloidal coomassie blue. The stained gels were scanned and differences in intensity of corresponding spots in each pair of gels were analyzed using ImageMaster 2D Platinum 6.0 software (see Materials and methods).

Approximately 700 stable spots were detected on each 2-DE gel. The expression profiles of proteins with isoelectric point within pH range 3.0–11.0 and molecular mass of 20–150 kD were highly reproducible for both sensitive and resistant cells as well as for individual sets of samples. Differences in intensity of corresponding spots were analyzed for each pair of gels and spots with 2-fold higher or lower intensity, when comparing sensitive and resistant cells, and considered as proteins with changed expression. Eight spots with significantly changed expression were detected (Figs. 1 and 2). These spots were cut from the gels and subjected to MS analysis (see Materials and methods).

Identification of proteins with changed expression

The identified proteins (Table I) with changed expression in resistant SK-BR-3 cells were: serpin B3 (spots 1, 2, 3 with intensity increased to 235, 264 and 387%, respectively), serpin B4 (spots 4 and 5 with intensity increased to 190 and 195%, respectively), heat shock protein 27 (HSP27) (spots 6 and 7 with intensity increased to 218 and 340%, respectively) and cytokeratin 18 (spot 8 with intensity decreased to 41%) (Fig. 2).

Table I

Results of MS analysis.

Table I

Results of MS analysis.

No.Protein nameSwissProt no.MUMSC (%)MS (score)MW (kD)pI-value
1Serpin B3SPB3_HUMAN19747FMFDLFQQFR (55)456.4
2Serpin B3SPB3_HUMAN16647FHCNHPFLFFIR (41)456.4
3Serpin B3SPB3_HUMAN9525SVDFANAPEESR (63)456.4
4Serpin B4SPB4_HUMAN11431STDFANAPEESR (66)
TYQFLQEYLDAIKK (59)
455.9
5Serpin B4SPB4_HUMAN8316STDFANAPEESR (55)
TYQFLQEYLDAIKK (57)
455.9
6Heat shock protein beta-1HSPB1_HUMAN8441LFDQAFGLPR (56)236.0
7Heat shock protein beta-1HSPB1_HUMAN8341LFDQAFGLPR (48)
SNEITIPVTFESR (49)
236.0
8Keratin, type I cytoskeletal 18K1C18_HUMAN19444TVQSLEIDLDSMR (51)
SLGSVQAPSYGAR (43)
485.3

[i] Differentially expressed proteins identified from 2-DE experiment. Table includes spot number, protein name, SwissProt database number, number of peptides matched to the identified protein, number of unassigned peaks, sequence coverage, peptide sequences confirmed by MS/MS (MASCOT score of individual peptides is given in parenthesis), MW and pI-values. No, spot number; M, matched peaks; U, unmatched peaks; SC, sequence coverage (%), MS, MS/MS confirmation - AA sequence (MASCOT score); MW, molecular weight of protein (kD).

Western blot analysis of the new sets of samples confirmed the detected changes in expression of all these proteins. Serpin B3 and serpin B4 were detected as one protein. These two proteins have a high homology (92%) and thus cross reactivity of primary antibodies against them is highly probable if not straightly admitted by manufacturer. The expression of serpin B3+B4 in SK-BR-3 cells resistant to paclitaxel detected using western blotting was elevated up to 158% when compared to cells sensitive to paclitaxel (p<0.01) (Fig. 3). The expression of HSP27 in resistant cells was increased to 144% in resistant cells compared to sensitive cells (p<0.01) (Fig. 3). Western blot analysis of cytokeratin 18 showed three bands very close together. We searched for western blots of whole SK-BR-3 lysates using cytokeratin 18 antibody and found that more bands or one very wide band for cytokeratin 18 has been previously published (http://www.scbt.com/datasheet-32722-cytokeratin-18-rck106-antibody.html; accessed June 7, 2013). Thus, for densitometry analysis we took these three bands as one. We detected decreased expression of cytokeratin 18 down to 68% in resistant cells compared to sensitive cells (p<0.01) (Fig. 4).

For serpin B3+B4 and HSP27, actin was used as a loading control. For cytokeratin 18 GAPDH was used. The reason for the different loading controls was that cytokeratin 18 has approximately the same size as actin and both these proteins are very strongly expressed. Therefore stripping off either cytokeratin 18 or actin failed to yield satisfactory results.

Discussion

Using western blot analysis, we found a significant increase (158%) in the expression of serpin B3/B4 in human SK-BR-3 breast cancer cells resistant to paclitaxel compared to sensitive SK-BR-3 cells (Fig. 3). The increase is lower compared to 2-DE gels analysis, but still significant (analyzed by Student’s t-test, p<0.01) (Fig. 2).

Serpin B3 and B4 share the same molecular weight (92% identical sequence of amino acids) but have different pI (serpin B3 represents a neutral form with pI>6.2, serpin B4 represents an acidic form with pI<6.2) (46). This corresponded with position of their spots on our 2-DE gels (Fig. 1). Our gels revealed three spots for serpin B3 and two spots for serpin B4. A higher number of serpin B3 protein species on 2-DE gels was described previously by Ho et al (47). Spots of the same protein with different pI, which we found in our study, could be the result of various levels of phosphorylation of serpins. Because of their almost identical sequence of amino acids, serpin B3 and serpin B4 are often analyzed together as serpin B3+B4.

Serpins (serine protease inhibitors) represent a large group of proteins, most of them capable of inhibiting proteases (serine proteases and in some cases also cysteine proteases) (48). Serpin B3 and B4 are clade B serpins or ovoalbumin serpins (ov-serpins). Serpin B3 inhibits papain-like cysteine proteases (e.g., cathepsin-S, -K and -L), while serpin B4 inhibits both serine proteases (e.g., cathepsin G) and cysteine proteases (e.g., Der p 1 and Der f 1) (49). Both these serpins are often overexpressed in cancer cells of epithelial origin (46,50).

Upregulation of serpin B3 and B4 has previously been described as protective in cells exposed to radiation (51). Suppression of these proteins has been shown to suppress tumor growth (52). Recently, serpin B3 was suggested as a prognostic tool for docetaxel resistance in breast cancer (53) and platinum resistance in epithelial ovarian cancer (54) and lung cancer (55).

The mechanism of pro-survival effect of serpin B3 and B4 on cancer cells is likely to be connected with their ability to inhibit proteases. Cathepsin S, a known target for serpin B3, is essential for proper presentation of antigens to immune cells. Inhibition of its activity could lead to impaired recognition of cancer cells by the immune system. Another cathepsin, cathepsin L, promotes production of endostatin which has an anti-angiogenic effect (56). Therefore, inhibition of cathepsin L by serpin B3 could support angiogenesis. Cathepsin G has been described as an agent opposing tumor cell-cell adhesion (57). Thus, its inhibition would benefit cancer cells. It is worth noting that some of these cathepsins have been reported to have pro-cancer effects, as well (58,59).

More complex explanations of upregulation of serpin B3 and B4 in resistant cancer cells have also been suggested. Serpin B3 is believed to be able to inhibit the lysosomal cell death pathway (LCDP) induced by microtubule-stabilizing agents, including paclitaxel (60), through inhibition of LCDP mediators, e.g. cathepsins. Interestingly, serpin B4 has been described to inhibit also granzyme M-induced cell death, which is the main pathway used by cytotoxic lymphocytes to kill tumor cells (61).

Upstream regulation of serpin B3 and B4 expression has been suggested. Serpins B3+B4 were described as being activated through STAT3 activation (62). De Hoon et al have suggested signaling pathways connected with resistance to paclitaxel (22). They proceed from human epidermal growth factor receptor 2 (HER2) through PI3K/Akt to i) activation of STAT3 and ii) upregulation of P-glycoprotein and survivin, resulting in an overall increase in cancer cell survival (22). Based on these two studies, we suspect that increased expression of serpins B3+B4 is connected to paclitaxel resistance via the mechanism suggested by De Hoon et al (22).

In this study we confirmed that increased expression of serpin B3 and B4 can potentially affect resistance to paclitaxel and we hypothesize that the mechanism of serpin B3 and B4 upregulation involves activation of STAT3.

Another protein found to have significantly higher expression (144%) in resistant SK-BR-3 cells compared to sensitive cells was heat shock protein 27 (HSP27). Again, the increase in HSP27 expression detected using western blot analysis was lower than the increase found using 2D gels analysis (Figs. 2 and 3), however, it was nonetheless statistically significant (Student’s t-test, p<0.01).

Heat shock protein 27 is an ATP-independent chaperon and functions as a protective agent against protein aggregation (63). It is highly expressed even in normal cells under stress conditions like heat, oxidative stress or exposure to chemotherapeutic agents (64). In some studies, HSP27 was thought to protect cancer cells against cell death. Its overexpression was reported to inhibit activation of procaspase-3 and procaspase-9 (65). It antagonizes Bax-mediated mitochondrial changes (66) and it is generally thought to block programmed cell death by directly sequestering intermediates in the caspase-dependent apoptosis pathway (67).

Thus, we hypothesize that HSP27 can positively affect cell survival through inhibition of the inner (mitochondrial) apoptotic pathway. Development of the ability to prevent activation of this pathway could be crucial for survival of SK-BR-3 cells when exposed to paclitaxel and thus could be part of the resistance mechanism to taxanes in certain types of cancer cells.

Cytokeratin 18 was the only protein with reduced expression in resistant SK-BR-3 cells compared to sensitive cells. The cytokeratin 18 molecule (together with cytokeratin 8) is a component of the most common intermediate filaments in cells of epithelial origin. Besides the many functions connected with its role in intracellular scaffolding (e.g., structuring cytoplasm, providing resistance against external stresses) and cellular processes like mitosis or apoptosis, it also has a role in tumor cell behavior (68). Additionally, some studies have suggested the involvement of cytokeratin 18 in certain signaling pathways (68,69).

Cytokeratin 18 was reported to be involved in pro-survival PI3K/Akt pathway (69), which can counteract Fas-mediated apoptosis (70). Fortier et al reported overexpression of cytokeratin 18 (and cytokeratin 8) as a result of overexpression of Akt1 (71). This would suggest that higher expression of cytokeratin 18 could be a marker of reduced susceptibility to cell death (i.e., resistant cells); however, our findings do not support this conclusion.

The tumor necrosis factor receptor 2 (TNFR2) is another signaling pathway with which cytokeratin 18 can interact. Binding of cytokeratin 18 to the cytoplasmic domain of tumor necrosis factor receptor 2 (TNRF2) leads to changes in JNK signaling and NF-κB activation. Cytokeratin 18 is also able to negatively influence TNF-induced apoptosis (72). However, our results showed a different pattern, which could mean that TNF-induced cell death does not play as important role in paclitaxel-induced cell death in SK-BR-3 cells.

Relative to our results, Meng et al (73) found a lower expression of cytokeratin 18 promoting proliferation of breast cancer cells MCF-7; however, they connected this effect to the modulation of estrogen receptor-α pathway. Nevertheless, SK-BR-3 cells do not express estrogen receptor-α. A trend similar to that in our study was found in some primary breast carcinoma screenings where lower expression of cytokeratin 18 as associated with a worse prognosis (74,75).

The function and significance of cytokeratin 18 in cancer cells needs to be elucidated more thoroughly; more precisely we need to determine why upregulation of cytokeratin 18 indicates reduced susceptibility to treatment in some cases, but not in others.

Our study found four proteins (serpin B3, serpin B4, heat shock protein 27, cytokeratin 18) with changed expression in breast cancer cells (the type not expressing estrogen receptor but overexpressing HER2) resistant to paclitaxel. These proteins represent a heterogeneous group and do not seem to be directly connected with each other (with serpin B3 and B4 as an exception). This suggests that processes involved in cancer cells surviving normally lethal doses of paclitaxel are both numerous and complex. Nevertheless, even if revealed proteins interact with different pathways of cell signaling, they still represent a group of potential biomarkers that should be probably seen as a kind of whole and tested together rather than considered and treated as four individual biomarkers.

Acknowledgements

This study was supported by grant NT 13679-4 of the Internal Grant Agency, Ministry of Health of the Czech Republic, and by the Institutional Research Project of the Institute of Microbiology (RVO61388971). We thank Thomas Secrest for the English language revision of the manuscript.

References

1 

Jordan MA: Mechanism of action of antitumor drugs that interact with microtubules and tubulin. Curr Med Chem Anticancer Agents. 2:1–17. 2002. View Article : Google Scholar : PubMed/NCBI

2 

Villeneuve DJ, Hembruff SL, Veitch Z, Cecchetto M, Dew WA and Parissenti AM: cDNA microarray analysis of isogenic paclitaxel- and doxorubicin-resistant breast tumor cell lines reveals distinct drug-specific genetic signatures of resistance. Breast Cancer Res Treat. 96:17–39. 2006. View Article : Google Scholar

3 

Galletti E, Magnani M, Renzulli ML and Botta M: Paclitaxel and docetaxel resistance: Molecular mechanisms and development of new generation taxanes. Chem Med Chem. 2:920–942. 2007. View Article : Google Scholar : PubMed/NCBI

4 

Stordal B and Davey R: A systematic review of genes involved in the inverse resistance relationship between cisplatin and paclitaxel chemotherapy: Role of BRCA1. Curr Cancer Drug Targets. 9:354–365. 2009. View Article : Google Scholar : PubMed/NCBI

5 

Smoter M, Bodnar L, Duchnowska R, Stec R, Grala B and Szczylik C: The role of Tau protein in resistance to paclitaxel. Cancer Chemother Pharmacol. 68:553–557. 2011. View Article : Google Scholar : PubMed/NCBI

6 

Vergara D, Tinelli A, Iannone A and Maffia M: The impact of proteomics in the understanding of the molecular basis of paclitaxel-resistance in ovarian tumors. Curr Cancer Drug Targets. 12:987–997. 2012. View Article : Google Scholar : PubMed/NCBI

7 

Rebucci M and Michiels C: Molecular aspects of cancer cell resistance to chemotherapy. Biochem Pharmacol. 85:1219–1226. 2013. View Article : Google Scholar : PubMed/NCBI

8 

Mechetner E, Kyshtoobayeva A, Zonis S, et al: Levels of multidrug resistance (MDR1) P-glycoprotein expression by human breast cancer correlate with in vitro resistance to taxol and doxorubicin. Clin Cancer Res. 4:389–398. 1998.PubMed/NCBI

9 

Pires MM, Emmert D, Hrycyna CA and Chmielewski J: Inhibition of P-glycoprotein-mediated paclitaxel resistance by reversibly linked quinine homodimers. Mol Pharmacol. 75:92–100. 2009. View Article : Google Scholar : PubMed/NCBI

10 

Thollet A, Vendrell JA, Payen L, et al: ZNF217 confers resistance to the pro-apoptotic signals of paclitaxel and aberrant expression of Aurora-A in breast cancer cells. Mol Cancer. 9:2912010. View Article : Google Scholar : PubMed/NCBI

11 

Wang Y, Chen QF, Jin S, et al: Up-regulation of P-glycoprotein is involved in the increased paclitaxel resistance in human esophageal cancer radioresistant cells. Scand J Gastroenterol. 47:802–808. 2012. View Article : Google Scholar : PubMed/NCBI

12 

Kwak JO, Lee SH, Lee GS, et al: Selective inhibition of MDR1 (ABCB1) by HM30181 increases oral bioavailability and therapeutic efficacy of paclitaxel. Eur J Pharmacol. 627:92–98. 2010. View Article : Google Scholar : PubMed/NCBI

13 

Huisman MT, Chhatta AA, van Tellingen O, Beijnen JH and Schinkel AH: MRP2 (ABCC2) transports taxanes and confers paclitaxel resistance and both processes are stimulated by probenecid. Int J Cancer. 116:824–829. 2005. View Article : Google Scholar : PubMed/NCBI

14 

O’Brien C, Cavet G, Pandita A, et al: Functional genomics identifies ABCC3 as a mediator of taxane resistance in HER2-amplified breast cancer. Cancer Res. 68:5380–5389. 2008.PubMed/NCBI

15 

Violini S, D’Ascenzo S, Bagnoli M, et al: Induction of a multifactorial resistance phenotype by high paclitaxel selective pressure in a human ovarian carcinoma cell line. J Exp Clin Cancer Res. 23:83–91. 2004.PubMed/NCBI

16 

Takano M, Otani Y, Tanda M, Kawami M, Nagai J and Yumoto R: Paclitaxel-resistance conferred by altered expression of efflux and influx transporters for paclitaxel in the human hepatoma cell line, HepG2. Drug Metab Pharmacokinet. 24:418–427. 2009. View Article : Google Scholar : PubMed/NCBI

17 

Kars MD, Iseri OD and Gunduz U: A microarray based expression profiling of paclitaxel and vincristine resistant MCF-7 cells. Eur J Pharmacol. 657:4–9. 2011. View Article : Google Scholar : PubMed/NCBI

18 

Taniguchi R, Kumai T, Matsumoto N, et al: Utilization of human liver microsomes to explain individual differences in paclitaxel metabolism by CYP2C8 and CYP3A4. J Pharmacol Sci. 97:83–90. 2005. View Article : Google Scholar : PubMed/NCBI

19 

Garcia-Martin E, Pizarro RM, Martinez C, et al: Acquired resistance to the anticancer drug paclitaxel is associated with induction of cytochrome P4502C8. Pharmacogenomics. 7:575–585. 2006. View Article : Google Scholar : PubMed/NCBI

20 

Hendrikx J, Lagas JS, Rosing H, Schellens JHM, Beijnen JH and Schinkel AH: P-glycoprotein and cytochrome P450 3A act together in restricting the oral bioavailability of paclitaxel. Int J Cancer. 132:2439–2447. 2013. View Article : Google Scholar : PubMed/NCBI

21 

Henningsson A, Sparreboom A, Sandstrom M, et al: Population pharmacokinetic modelling of unbound and total plasma concentrations of paclitaxel in cancer patients. Eur J Cancer. 39:1105–1114. 2003. View Article : Google Scholar : PubMed/NCBI

22 

De Hoon JPJ, Veeck J, Vriens B, Calon TGA, van Engeland M and Tjan-Heijnen VCG: Taxane resistance in breast cancer: A closed HER2 circuit? Biochim Biophys Acta Rev Cancer. 1825:197–206. 2012.PubMed/NCBI

23 

Arai T, Miyoshi Y, Kim SJ, et al: Association of GSTP1 expression with resistance to docetaxel and paclitaxel in human breast cancers. Eur J Surg Oncol. 34:734–738. 2008. View Article : Google Scholar : PubMed/NCBI

24 

Jardim BV, Moschetta MG, Gelaleti GB, et al: Glutathione transferase pi (GSTpi) expression in breast cancer: An immunohistochemical and molecular study. Acta Histochem. 114:510–517. 2012. View Article : Google Scholar : PubMed/NCBI

25 

Miyake T, Nakayama T, Naoi Y, et al: GSTP1 expression predicts poor pathological complete response to neoadjuvant chemotherapy in ER-negative breast cancer. Cancer Sci. 103:913–920. 2012. View Article : Google Scholar : PubMed/NCBI

26 

Ferlini C, Raspaglio G, Mozzetti S, et al: Bcl-2 down-regulation is a novel mechanism of paclitaxel resistance. Mol Pharmacol. 64:51–58. 2003. View Article : Google Scholar : PubMed/NCBI

27 

Pan Z and Gollahon L: Paclitaxel attenuates Bcl-2 resistance to apoptosis in breast cancer cells through an endoplasmic reticulum-mediated calcium release in a dosage dependent manner. Biochem Biophys Res Commun. 432:431–437. 2013. View Article : Google Scholar

28 

Ali-Osman F, Brunner JM, Kutluk TM and Hess K: Prognostic significance of glutathione S-transferase pi expression and subcellular localization in human gliomas. Clin Cancer Res. 3:2253–2261. 1997.PubMed/NCBI

29 

Cheung CHA, Wu SY, Lee TR, et al: Cancer cells acquire mitotic drug resistance properties through beta I-tubulin mutations and alterations in the expression of beta-tubulin isotypes. PLoS One. 5:e125642010. View Article : Google Scholar

30 

Wang YH, Sparano JA, Fineberg S, et al: High expression of class III beta-tubulin predicts good response to neoadjuvant taxane and doxorubicin/cyclophosphamide-based chemotherapy in estrogen receptor-negative breast cancer. Clin Breast Cancer. 13:103–108. 2013. View Article : Google Scholar

31 

Yin SH, Zeng CQ, Hari M and Cabral F: Random mutagenesis of beta-tubulin defines a set of dispersed mutations that confer paclitaxel resistance. Pharm Res. 29:2994–3006. 2012. View Article : Google Scholar : PubMed/NCBI

32 

Edelman MJ and Shvartsbeyn M: Epothilones in development for non-small-cell lung cancer: Novel anti-tubulin agents with the potential to overcome taxane resistance. Clin Lung Cancer. 13:171–180. 2012. View Article : Google Scholar : PubMed/NCBI

33 

Diaz JF, Barasoain I and Andreu JM: Fast kinetics of taxol binding to microtubules. Effects of solution variables and microtubule-associated proteins. J Biol Chem. 278:8407–8419. 2003. View Article : Google Scholar : PubMed/NCBI

34 

Zhang M, Mukherjee N, Bermudez RS, et al: Adenovirus-mediated inhibition of survivin expression sensitizes human prostate cancer cells to paclitaxel in vitro and in vivo. Prostate. 64:293–302. 2005. View Article : Google Scholar

35 

Wang S, Huang X, Lee CK and Liu B: Elevated expression of erbB3 confers paclitaxel resistance in erbB2-overexpressing breast cancer cells via upregulation of survivin. Oncogene. 29:4225–4236. 2010. View Article : Google Scholar : PubMed/NCBI

36 

Lv KZ, Liu LQ, Wang LB, et al: Lin28 mediates paclitaxel resistance by modulating p21, Rb and Let-7a miRNA in breast cancer cells. PLoS One. 7:e400082012. View Article : Google Scholar : PubMed/NCBI

37 

Lu MS, Xiao L, Hu JL, Deng S and Xu Y: Targeting of p38 mitogen-activated protein kinases to early growth response gene 1 (EGR-1) in the human paclitaxel-resistance ovarian carcinoma cells. J Huazhong Univ Sci Tech-Med. 28:451–455. 2008. View Article : Google Scholar : PubMed/NCBI

38 

Arai K, Matsumoto Y, Nagashima Y and Yagasaki K: Regulation of class II beta-tubulin expression by tumor suppressor p53 protein in mouse melanoma cells in response to vinca alkaloid. Mol Cancer Res. 4:247–255. 2006. View Article : Google Scholar : PubMed/NCBI

39 

Farrell A: A close look at cancer. Nat Med. 17:262–265. 2011. View Article : Google Scholar

40 

Pan Z, Gao YL, Heng LS, et al: Amphiphilic N-(2,3-dihydroxypropyl)-chitosan-cholic acid micelles for paclitaxel delivery. Carbohydr Polym. 94:394–399. 2013. View Article : Google Scholar : PubMed/NCBI

41 

Ehrlichova M, Koc M, Truksa J, Naldova Z, Vaclavikova R and Kovarr J: Cell death induced by taxanes in breast cancer cells: Cytochrome c is released in resistant but not in sensitive cells. Anticancer Res. 25:4215–4224. 2005.PubMed/NCBI

42 

Ehrlichova M, Ojima I, Chen J, et al: Transport, metabolism, cytotoxicity and effects of novel taxanes on the cell cycle in MDA-MB-435 and NCI/ADR-RES cells. Naunyn-Schmiedebergs Arch Pharmacol. 385:1035–1048. 2012. View Article : Google Scholar : PubMed/NCBI

43 

Nemcova-Furstova V, Balusikova K, Sramek J, James RF and Kovar J: Caspase-2 and JNK activated by saturated fatty acids are not involved in apoptosis induction but modulate ER stress in human pancreatic beta-cells. Cell Physiol Biochem. 31:277–289. 2013. View Article : Google Scholar : PubMed/NCBI

44 

Dyballa N and Metzger S: Fast and sensitive colloidal coomassie G-250 staining for proteins in polyacrylamide gels. J Vis Exp. 30:1–5. 2009.PubMed/NCBI

45 

Jelinek M, Balusikova K, Kopperova D, et al: Caspase-2 is involved in cell death induction by taxanes in breast cancer cells. Cancer Cell Int. 13:422013. View Article : Google Scholar

46 

Vidalino L, Doria A, Quarta S, Zen M, Gatta A and Pontisso P: Serpin B3, apoptosis and autoimmunity. Autoimmun Rev. 9:108–112. 2009. View Article : Google Scholar

47 

Ho KY, Huang HH, Hung KF, et al: Cholesteatoma growth and proliferation: Relevance with serpin B3. Laryngoscope. 122:2818–2823. 2012. View Article : Google Scholar : PubMed/NCBI

48 

Gettins PGW: Serpin structure, mechanism, and function. Chem Rev. 102:4751–4803. 2002. View Article : Google Scholar : PubMed/NCBI

49 

Izuhara K, Ohta S, Kanaji S, Shiraishi H and Arima K: Recent progress in understanding the diversity of the human ov-serpin/clade B serpin family. Cell Mol Life Sci. 65:2541–2553. 2008. View Article : Google Scholar : PubMed/NCBI

50 

Cataltepe S, Gornstein ER, Schick C, et al: Co-expression of the squamous cell carcinoma antigens 1 and 2 in normal adult human tissues and squamous cell carcinomas. J Histochem Cytochem. 48:113–122. 2000.PubMed/NCBI

51 

Murakami A, Suminami Y, Hirakawa H, Nawata S, Numa F and Kato H: Squamous cell carcinoma antigen suppresses radiation-induced cell death. Br J Cancer. 84:851–858. 2001. View Article : Google Scholar : PubMed/NCBI

52 

Suminami Y, Nagashima S, Murakami A, et al: Suppression of a squamous cell carcinoma (SCC)-related serpin, SCC antigen, inhibits tumor growth with increased intratumor infiltration of natural killer cells. Cancer Res. 61:1776–1780. 2001.

53 

Collie-Duguid ESR, Sweeney K, Stewart KN, Miller ID, Smyth E and Heys SD: SerpinB3, a new prognostic tool in breast cancer patients treated with neoadjuvant chemotherapy. Breast Cancer Res Treat. 132:807–818. 2011. View Article : Google Scholar

54 

Lim W, Kim HS, Jeong W, et al: Serpin B3 in the chicken model of ovarian cancer: A prognostic factor for platinum resistance and survival in patients with epithelial ovarian cancer. Plos One. 7:e498692012. View Article : Google Scholar : PubMed/NCBI

55 

Kerr KM: Personalized medicine for lung cancer: new challenges for pathology. Histopathology. 60:531–546. 2012. View Article : Google Scholar : PubMed/NCBI

56 

Felbor U, Dreier L, Bryant RAR, Ploegh HL, Olsen BR and Mothes W: Secreted cathepsin L generates endostatin from collagen XVIII. EMBO J. 19:1187–1194. 2000. View Article : Google Scholar : PubMed/NCBI

57 

Kudo T, Kigoshi H, Hagiwara T, Takino T, Yamazaki M and Yui S: Cathepsin G, a neutrophil protease, induces compact cell-cell adhesion in MCF-7 human breast cancer cells. Mediat Inflamm. 2009:8509402009. View Article : Google Scholar : PubMed/NCBI

58 

Lankelma JM, Voorend DM, Barwari T, et al: Cathepsin L, target in cancer treatment? Life Sci. 86:225–233. 2012. View Article : Google Scholar : PubMed/NCBI

59 

Small DM, Burden RE, Jaworski J, et al: Cathepsin S from both tumor and tumor-associated cells promote cancer growth and neovascularization. Int J Cancer. 133:2102–2112. 2013. View Article : Google Scholar : PubMed/NCBI

60 

Broker LE, Huisman C, Span SW, Rodriguez JA, Kruyt FAE and Giaccone G: Cathepsin B mediates caspase-independent cell death induced by microtubule stabilizing agents in non-small cell lung cancer cells. Cancer Res. 64:27–30. 2004. View Article : Google Scholar : PubMed/NCBI

61 

De Koning PJA, Kummer JA, de Poot SAH, et al: Intracellular serine protease inhibitor serpin B4 inhibits granzyme M-induced cell death. PLoS One. 6:e226452011.PubMed/NCBI

62 

Ahmed ST and Darnell JE: Serpin B3/B4, activated by STAT3, promote survival of squamous carcinoma cells. Biochem Biophys Res Commun. 378:821–825. 2009. View Article : Google Scholar : PubMed/NCBI

63 

Ehrnsperger M, Graber S, Gaestel M and Buchner J: Binding of nonnative protein to Hsp25 during heat shock creates a reservoir of folding intermediates for reactivation. EMBO J. 16:221–229. 1997. View Article : Google Scholar : PubMed/NCBI

64 

Schmitt E, Gehrmann M, Brunet M, Multhoff G and Garrido C: Intracellular and extracellular functions of heat shock proteins: repercussions in cancer therapy. J Leukoc Biol. 81:15–27. 2007. View Article : Google Scholar : PubMed/NCBI

65 

Garrido C, Bruey JM, Fromentin A, Hammann A, Arrigo AP and Solary E: HSP27 inhibits cytochrome c-dependent activation of procaspase-9. FASEB J. 13:2061–2070. 1999.PubMed/NCBI

66 

Havasi A, Li ZJ, Wang ZY, et al: Hsp27 inhibits Bax activation and apoptosis via a phosphatidylinositol 3-kinase-dependent mechanism. J Biol Chem. 283:12305–12313. 2008. View Article : Google Scholar : PubMed/NCBI

67 

Ciocca DR, Arrigo AP and Calderwood SK: Heat shock proteins and heat shock factor 1 in carcinogenesis and tumor development: an update. Arch Toxicol. 87:19–48. 2012. View Article : Google Scholar : PubMed/NCBI

68 

Weng YR, Cui Y and Fang JY: Biological functions of cytokeratin 18 in cancer. Mol Cancer Res. 10:485–493. 2012. View Article : Google Scholar : PubMed/NCBI

69 

Galarneau L, Loranger A, Gilbert S and Marceau N: Keratins modulate hepatic cell adhesion, size and G1/S transition. Exp Cell Res. 313:179–194. 2007. View Article : Google Scholar : PubMed/NCBI

70 

Abreu MT, Arnold ET, Chow JYC and Barrett KE: Phosphatidylinositol 3-kinase-dependent pathways oppose Fas-induced apoptosis and limit chloride secretion in human intestinal epithelial cells. Implications for inflammatory diarrheal states. J Biol Chem. 276:47563–47574. 2001. View Article : Google Scholar

71 

Fortier AM, Van Themsche C, Asselin E and Cadrin M: Akt isoforms regulate intermediate filament protein levels in epithelial carcinoma cells. FEBS Lett. 584:984–988. 2010. View Article : Google Scholar : PubMed/NCBI

72 

Inada H, Izawa I, Nishizawa M, et al: Keratin attenuates tumor necrosis factor-induced cytotoxicity through association with TRADD. J Cell Biol. 155:415–425. 2001. View Article : Google Scholar : PubMed/NCBI

73 

Meng YG, Wu ZQ, Yin XY, et al: Keratin 18 attenuates estrogen receptor alpha-mediated signaling by sequestering LRP16 in cytoplasm. BMC Cell Biol. 10:962009. View Article : Google Scholar : PubMed/NCBI

74 

Woelfle U, Sauter G, Santjer S, Brakenhoff R and Pantel K: Down-regulated expression of cytokeratin 18 promotes progression of human breast cancer. Clin Cancer Res. 10:2670–2674. 2004. View Article : Google Scholar : PubMed/NCBI

75 

Schaller G, Fuchs I, Pritze W, et al: Elevated keratin 18 protein expression indicates a favorable prognosis in patients with breast cancer. Clin Cancer Res. 2:1879–1885. 1996.PubMed/NCBI

Related Articles

Journal Cover

August-2014
Volume 45 Issue 2

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
Pavlikova N, Bartonova I, Dincakova L, Halada P and Kovar J: Differentially expressed proteins in human breast cancer cells sensitive and resistant to paclitaxel. Int J Oncol 45: 822-830, 2014
APA
Pavlikova, N., Bartonova, I., Dincakova, L., Halada, P., & Kovar, J. (2014). Differentially expressed proteins in human breast cancer cells sensitive and resistant to paclitaxel. International Journal of Oncology, 45, 822-830. https://doi.org/10.3892/ijo.2014.2484
MLA
Pavlikova, N., Bartonova, I., Dincakova, L., Halada, P., Kovar, J."Differentially expressed proteins in human breast cancer cells sensitive and resistant to paclitaxel". International Journal of Oncology 45.2 (2014): 822-830.
Chicago
Pavlikova, N., Bartonova, I., Dincakova, L., Halada, P., Kovar, J."Differentially expressed proteins in human breast cancer cells sensitive and resistant to paclitaxel". International Journal of Oncology 45, no. 2 (2014): 822-830. https://doi.org/10.3892/ijo.2014.2484