Close correlation between MEK/ERK and Aurora-B signaling pathways in sustaining tumorigenic potential and radioresistance of gynecological cancer cell lines

  • Authors:
    • Francesco Marampon
    • Giovanni Luca Gravina
    • Valdimir M. Popov
    • Luca Scarsella
    • Claudio Festuccia
    • Maria Emilia La Verghetta
    • Silvia Parente
    • Manuela Cerasani
    • Gemma Bruera
    • Corrado Ficorella
    • Enrico Ricevuto
    • Vincenzo Tombolini
    • Ernesto Di Cesare
    • Bianca Maria Zani
  • View Affiliations

  • Published online on: November 5, 2013     https://doi.org/10.3892/ijo.2013.2167
  • Pages: 285-294
Metrics: Total Views: 0 (Spandidos Publications: | PMC Statistics: )
Total PDF Downloads: 0 (Spandidos Publications: | PMC Statistics: )


Abstract

Both Aurora-A and -B kinases have been implicated in tumorigenesis; and as such, they represent an attractive therapeutic target. Recent studies found that Aurora-A is a downstream target of mitogen-activated protein kinase 1/ERK2, while Aurora-B has been found to be a prognostic/predictive therapeutic target for epithelial cancer. In a wide range of human cancers, the Ras/Raf/MEK/ERK/MAP kinase pathway is enhanced and the cellular response to growth signals is known to increase. The purpose of this study was to investigate whether the MEK/ERK cascade regulates tumorigenic signaling and radioresistance via the Aurora-B-mediated pathway in a panel of gynecological cancer cell lines. Exponentially growing human endometrial (Ishikawa), cervical (HeLa), cervical (CASKI) and vulva (SiHa) cancer cells were used in culture treated with either control or MEK/ERK inhibitor or AZD1152 before and after irradiation. Western blotting, ERK1/2 siRNA transfection, growth assay in modified monolayer, Annexin V and migration/invasion assays were performed. The specific MEK/ERK inhibitor U0126 decreased the tumorigenic potential and improved the radiation response in all cellular models. The modulation of radioresponse upon U0126 treatment positively correlated with the inhibition of phospho-ERKs and the reduction of Aurora-B kinase expression. In addition, upon U0126 treatment DNA-PKcs protein expression was found to be downregulated, indicating that the improved radiation response may be caused by decreased DNA double-strand damage repair mechanisms. The knockdown of ERK by siRNA confirmed the MEK/ERK-dependent Aurora-B kinase expression. The use of AZD1152, a selective Aurora-B inhibitor, counteracted tumorigenic potential and radioresistance phenotype by highly increasing apoptotic mechanisms in all gynecological cancer cell lines used. Evidence from our experiments show that tumorigenic potential and radiation response in gynecological cancer cells may ensue from a MEK/ERK or Aurora-B inhibition. Together with the close correlation of MEK/ERK and Aurora-B protein expression, this study underlines the potential role of a MEK/ERK/Aurora-B axis whose interruption recovers the antitumor effects of radiotherapy.

Introduction

Even though cervical, endometrial and ovarian cancers are relatively common, vulvar, vaginal, and fallopian tube cancers, and choriocarcinomas are very rare (1). Treatment of these malignancies requires a multimodal approach combining surgery, radiotherapy (RT) and/or chemotherapy. Although overall statistics have improved considerably, the outcome for patients with high-risk disease remains relatively poor, which points to a clear need for new therapeutic strategies. In situations like these, radiation therapy can be used to prevent local cancer recurrences after surgery (24). It has been clarified that the radiation resistance is associated with the abnormal expression of activated oncogenes, including Ras (5,6) and Aurora-B (7). The Ras/Raf/mitogen-activated protein kinase kinase (MEK)/extracellular signal-regulated kinase (ERK) cascade regulates proliferation, differentiation, survival, motility (818) and when constitutively activated, mediates resistance to ionizing radiation (15). The aurora kinases are a family of oncogenic serine/threonine kinases involved in the mitotic (M) phase of the cell cycle, acting to establish the mitotic spindle, bipolar spindle formation, alignment of centrosomes on mitotic spindle, centrosome separation, cytokinesis and monitoring of the mitotic checkpoint (1922). There are three known aurora kinases (Aurora-A, -B and -C) in human neoplastic and non-neoplastic tissues (19,2226). Inhibition of aurora kinase activity leads to catastrophic errors of mitosis, defective cytokinesis, misaligned centrosomes, mitotic spindle malformation and apoptosis (2630). However, the role of aurora kinases in radioresistance phenomena remains unclear. The Ras/Raf/MEK/ERKs pathway positively regulates Aurora-B expression at the transcriptional level (31). DNA double-strand break is critical in DNA lesions induced by radiation. In mammalian cells the repair of these lesions occurs by non-homologous end joining (NHEJ) requiring Ku70/Ku86 and the recruitment of the catalytic subunit of DNA-dependent PK (DNAPKcs). DNAPKcs is implicated in radioresponsiveness of embryonal rhabdomyosarcomauterine-derived cell lines (3234) and human glioblastoma cell lines (35,36). DNA-PKcs are necessary for genomic stability whereas abnormal levels in cancer cell may contribute to cell proliferation, radioprotection eventually contributing to oncogenic phenotype (3740). Herein we investigated whether MEK/ERK pathway, collaborating with Aurora-B kinase, sustains the tumorigenic potential and radioprotection in a panel of gynecological cancer cell lines.

Materials and methods

Cell culture, treatment and radiation exposure

The human Ishikawa endometrial cancer cell line was kindly provided by Professor Marcello Maggiolini (University of Calabria, Italy). The human HeLa cervical cancer cell line was kindly provided by Professor Roberto Maggio (University of L’Aquila, Italy), while the human CASKI cervical- and SiHa vulva-cancer cell line were kindly provided by Professor P.J.F. Snijders (VU Medisch Centrum, Germany). Tumor cell lines were cultured in the appropriate medium supplemented with 10% fetal calf serum (FCS). Treatments with 10 μmol/l MEK/ERK inhibitor U0126 (1, 4-diamino-2, 3-dicyano-1, 4-bis[2-aminophenylthio] butadiene; Promega) or with AZD1152 (60 nM), an Aurora-B kinase inhibitor, was done as shown in the figures. Radiation was delivered at room temperature using an X-ray LINAC at the dose rate of 2.5 Gy/min. For clonogenic survival assay, exponentially growing cells, diluted to appropriate densities, were plated with complete medium in presence of U0126, AZD1152 or vehicle/control [dimethyl sulfoxide (DMSO) 0.1%] and then irradiated with graded doses (0–2-4–6 Gy). Cells were then cultured in drug-free medium for 14 days, fixed with methanol/acetic acid (10:1, v/v) and stained with crystal violet. Colonies containing >50 cells were counted. The plating efficiency (PE) was calculated as the number of colonies observed/the number of cell plated; the surviving fraction (SF) was calculated as: colonies counted/cells seeded (PE/100).

Cell proliferation assay

Cells from adherent culture were counted using hemocytometer and tested for exclusion of trypan blue. Results represent the average of triplicate experiments, including standard error.

Western blot analysis

Proteins of whole cell lysates were assessed using the Lowry method (40) and equal amounts of proteins were separated on SDS-PAGE. The proteins were transferred to a nitrocellulose membrane (Schleicher & Schell Bioscience GmbH, Germany) by electroblotting. Immunoblottings were performed with the following antibodies directed against c-Myc, ERK1/2, phospho-ERK1/2, p27, cyclin D1, CDK2, Aurora-B, DNAPKcs and α-tubulin (B-7) (all from Santa Cruz Biotechnology, Santa Cruz, CA, USA). Peroxidase-conjugate anti-mouse or anti-rabbit IgG (Amersham-Pharmacia Biotech, UK or Santa Cruz) were used for enhanced chemiluminescence (ECL) detection.

Cell cycle and apoptosis analysis by flow cytometry

After the appropriate treatments, cells (1×106) were fixed for 30 min in 70% ethanol and pelleted by centrifugation (720 g; 5 min). After removal of ethanol, cells were incubated and resuspended in 1 ml of DNA staining solution (PBS containing 200 mg/ml RNase A, 20 mg/ml propidium iodide plus 0.1% Triton X-100) and left at room temperature for 60 min. Ten thousand events per sample were acquired using a FACScan flow cytometer (Becton-Dickinson, San Jose, CA, USA) and the percentage of cells in G1/S, G2/M and Sub-G2/M phases of the cell cycle were determined using CellQuest software (Becton-Dickinson). Apoptosis was analyzed by using Annexin V staining (GenScript, Piscataway, NJ, USA). All cells were then measured on a FACScan flow cytometer with an argon laser at 488 nm for excitation and analyzed using CellQuest software. Apoptotic cells were detected by the percentage of Annexin V stained cells. The results were expressed as the percentage of death by apoptosis induced by a specific treatment.

Soft agar clonogenic assays

Soft agar assays were performed as previously described (34). Briefly, 2×103 cells were suspended in 0.3% Bacto-agar (Life Technologies) containing the appropriate medium supplemented with 10% FCS and/or 10 μM U0126 and seeded in 60-mm cell culture plates. The plates were incubated at 37°C for 14 days. The numbers of colonies containing ≥6 cells were counted.

Small interfering RNA transfection

Tumor cells (1.5×105 cells/well) were plated in 6-well plates and grown in the appropriate medium supplemented with 10% FCS for 2 days. These cells were transfected with ERK1siRNA and ERK2 siRNA (sc-29308 and sc-44224 respectively, Santa Cruz Biotechnology). All siRNA duplexes were transfected using Oligofectamine reagent (Invitrogen) according to the manufacturer’s instructions. After ERK transfection, cells were grown in the appropriate medium for 72 h.

Invasion and migration assays

Transwell membrane (Corning Costar Corp.) was used. Cancer cells were trypsinized, washed and kept suspended in the appropriate medium without FCS. To the lower wells of the chambers, migration-inducing medium (with 10% FCS) was added. Upper wells were filled with serum-free medium with cells (20,000 cells per well) in the absence or in the presence of the appropriate treatments. After 8 h, filters were removed and fixed with methanol and subsequently the cells on the upper side were wiped off using a Q-tip. Filters were stained with 20% Giemsa solution. Evaluation of complete transmigration was performed under the microscope and random fields were scanned (four fields per filter) for the presence of cells at the lower membrane side only. Invasion assays were done in a similar manner as the migration assays described above, unless the inserts were pre-coated with Matrigel (BD Biosciences).

Statistical methods

Continuous variables were summarized as the mean and standard deviation (SD) and the statistical comparisons between control and treatments were established by carrying out the ANOVA test or the t-test when appropriate. Dichotomous variables were summarized by absolute and/or relative frequencies and statistical comparisons between control and treated groups were established by carrying out the Fisher’s exact test. For multiple comparisons the level of significance was corrected according to Bonferroni correction. All tests were two-sided and were determined by Monte Carlo significance. P<0.05 was considered statistically significant.

Results

Persistent ERK inhibition induces growth arrest in G1, inhibition of migration/invasion and promotes anchorage-independent growth

In order to verify the effects of MEK-inhibitor, U0126, a time course experiment with or without U0126 treatment (10 μM) was performed. Cells were treated with U0126 either for 3 h or for 4 days and subsequently processed for immunoblotting, cell count and FACS analysis. As shown in Fig. 1A and B, U0126 induced a rapid (3 h) and persistent (4 days) decrease in phospho-active ERKs (Fig. 1A) concomitant with a decrease in the proliferation rate ranging from 65.5 to 74.9% (Fig. 1B). As shown in Fig. 1C, treatment with U0126 resulted in preferential accumulation of tumor cells in the G1-S phase of cell cycle, with the percentage of Ishikawa, SiHa, Caski and HeLa cells in thiS phase of 61, 69, 74 and 77%, respectively. This phenomenon was consistent with cyclin D1 and CDK2 downregulation and p27Cip/Kip upregulation (Fig. 1D). c-Myc, one of the most tumorigenic transcription factors and downstream target of ERKs, while increasing in the expression levels comparing 3 h and 4 days untreated cultured cells, was early (3 h) and persistently (≤4 days) inhibited by the U0126 treatment as shown in Fig. 1D.

MEK/ERK inhibition is followed by downregulation of growth signal and tumorigenic molecules

To verify whether this inhibition affects anchorage-independent growth of cancer cells, we performed soft agar and cell growth assay in modified layer. Soft agar assay showed that tumor cells cultured with vehicle only formed cell aggregates of different sizes depending on the cell line used (Fig. 2A–D, upper panel). U0126 (10 μM) greatly affected the propensity of tumor cells to grow in modified layer (polyHEMA coated dishes) compared to controls (Fig. 2A–D, middle panel) with a decrease in the proliferation rate of 86, 84, 93 and 89% in Ishikawa, SiHa, Caski and HeLa cell lines, respectively (ANOVA test; P<0.01). The decrease of phospho-active-ERK levels in tumor cells cultured on poly-HEMA-coated dishes was still present after U0126 treatments (Fig. 2A–D, lower panel). Finally, U0126 (10 μM) significantly inhibited Ishikawa, SiHa, Caski and HeLa invasion and migration both at 3 h and 4 days after treatment, with the most evident effect at the longest time (Fig. 3). All together, these results indicate that U0126 induces growth arrest by blocking the molecular mechanism responsible of G1/S cell cycle phase progression and reduces the signals enabling tumorigenic and metastatic potential of tumor cells.

U0126 increases the radiosensitivity of gynecological cancer cell lines by delaying DNA repair machinery and enhancing apoptotic signaling

Tumor cells were treated with U0126 (10 μM) 24 h before the delivery of increasing doses of ionizing radiation (0–600 cGy) (Fig. 4). All cell lines were basically radioresistant at all doses tested (Fig. 4) and U0126 treatments increased the radiosensitivity with effects already evident at lowest radiation doses (Fig. 4). We further investigated whether suppressing ERKs function would influence the repair machinery of DNA double-strand breaks (DSBs) induced by irradiation. Tumor cells treated with U0126 were irradiated and the number of DNA foci representing the amount of unrepaired DSBs was counted in 4 different cell lines: i) untreated, ii) U0126-treated, iii) U0126-pretreated and iv) treated with RT (Fig. 5A–D, upper panel). For these experiments, a single dose of 4-Gy radiation was used. We have found that this dose is the most suitable to effectively distinguish the individual from combined treatments. Radiation-induced γ-H2AX foci were readily detectable at 1 h of irradiation in all treated cells (Fig. 5A–D, upper panel). Interestingly, adding U0126 together with radiation led to a substantially increased number of cells retaining these foci for ≥14 h with respect to cells treated with RT alone. The difference in the percentage of cells retaining DNA foci between the two groups was significant (P<0.01). Due to the fact that DNA-PKcs is usually activated in malignant tumor cells and its inactivation impairs DNA repair following ionizing irradiation we examined whether U0126 would affect DNA-PKcs expression. Of note, DNA-PKcs expression measured by western blotting at 24 h after treatments significantly decreased in all cell lines tested in the present study (Fig. 5A–D, lower panel).

Functional correlation between MEKs/ERKs and Aurora-B supports tumorigenic and radioresistant phenotype

We tested if the U0126-mediated effect on radiosesitization involved Aurora-B, known to play a central role in the control of DNA repair machinery. To this purpose cancer cells were cultured with U0126 24 h before the delivery of ionizing radiation (4 Gy) (Fig. 6) and expression levels of Aurora-B kinases was evaluated in these cell lines. As shown in Fig. 6A, MEK/ERK inhibition reduced Aurora-B expression in all cell lines used (Fig. 6A). To verify whether in these cell lines the Aurora-B played a role of increased radioresponse, experiments with AZD1152, a selective Aurora-B inhibitor, were performed. The functional inhibition of Aurora-B was assessed by quantifying p-H3, the active phosphorylated form of histone H3 required for normal chromosomal condensation. As shown in Fig. 6B, 24 h after AZD1152 (60 nM) treatment a dramatic decrease in p-H3 expression, consistent with inhibition of Aurora-B H3-phosphorylating activity, was observed in all cell lines. The functional inactivation of Aurora-B by AZD1152 increased the radiosensitivity of all cellular models (Fig. 6C) with a kinetic of radiosensitization similar to that observed during U0126 treatment (compare Fig. 4, with Fig. 6C). The radiosensitizing effect of AZD1152 was partially due to increased apoptotic stimulus that was greatly potentiated in association with radiotherapy (Fig. 6D) as measured by Annexin V assay. This evidence suggests a possible role of Aurora-B in the induction of pro-apoptotic stimulus upon radiation treatment.

In order to verify the existence of a direct functional link between ERK1/2 and Aurora-B, ERK1/2 RNA interference experiments were performed. Upon 72 h of treatment with ERK1/ERK2 siRNA, downregulation of total ERKs and Aurora-B protein levels was observed (Fig. 7A). Since MEK/ERK silencing caused downregulation of Aurora-B expression we further studied AZD1152 effects in cellular growth, growth in modified monolayer and in migration/invasion assays. The Aurora-B inactivation by AZ1152 significantly decreased growth rate and growth in modified monolayer (Fig. 7B, left and middle panels) as well as migration/invasion (Fig. 7B, right panels) potential of all tumor cell models. This body of evidence suggests that Aurora-B mediates tumorigenic potential of gynecological cancer cell lines controlling their invasion and metastatic potential.

Discussion

Ras/MEK/ERK signaling inhibition is considered a potential novel strategic approach for therapeutic use (10). In this study, we addressed the issue of whether MEK/ERK inhibition, by interfering with Aurora-B kinase, affects the tumorigenic phenotype and radiation response of a panel of gynecological cancer cell lines. The decrease in tumorigenic signaling induced by the MEK inhibitor, U0126, clearly demonstrated that the MEK/ERK pathway is a pre-requisite for the aberrant growth in in vitro and in vivo system of these cells. Indeed, the permanent phospho-active ERK inhibition in all tumor cell lines used was followed by growth arrest, loss of both anchorage-independent growth and migration/invasion potential. Importantly, MEK/ERK inhibition had significant effects on cell cycle distribution causing an increased accumulation of cells in G1 phase, ranging from 65-80% depending on cell line in 24-h treatment. This anti-growth response is countersigned by a decreased levels of a number of proteins that are globally correlated with cell cycle progression (CDK2, Myc and cyclin D1) or with cell cycle inhibition (p27).

Moreover, the effects of Ras/MEKs/ERKs inhibition and the downstream target pathways on radiation response have not yet been studied in detail in gynecological tumors. With respect to intrinsic cellular radiation sensitivity, it is known that cells are more responsive to the ionizing radiation effects in G2/M than in G1 phase. In these lines the MEK/ERK inhibitor led a significant percent of cells (65-80%) into G1 phase of the cell cycle within 24-h treatment, while radiation, combined with the MEK inhibitor, in just one hour gained the main effects on DNA foci. From this we can conclude that MEK inhibitor does not lower RT-mediated DNA lesions but rather helps radiosensitization as we previously reported in RMS in vivo and in vitro models (32,34). Effects of combined MEK/ERK inhibitor and radiation are particularly evident in the persistence of DNA foci, which decays within 14 h after radiation alone. Our data point to a possible loss of DNA repair mechanisms, resulting in MEK/ERK inhibitor-induced DNA-PKcs downregulation. Our study for the first time, demonstrated a close correlation between MEK/ERK and Aurora-B in a functional axis where the loss of ERK causes Aurora-B downregulation enhancing the response to DNA damage by radiation. The idea that MEK/ERK is a therapeutical target which enhances radiosensitivity was corroborated by the MEK inhibitor-mediated loss of DNA-PKcs, a member of the DNA repair machinery.

The close correlation between MEK/ERK signaling pathway and Aurora-B is of particular significance in light of recent findings reporting that MAPKs and Aurora-B cooperate in promoting invasive growth (4145), tumorigenic potential (46,47) and radioresistance phenomena (4244). We successfully demonstrated that MEK/ERK kinases regulate Aurora-B expression, suggesting that MAPK pathway and Aurora-B kinase rely on a functional collaboration, which is most likely required to sustain the tumorigenic and radioresistant phenotype of the gynecological cancer cells. This evidence was supported by the findings that selective Aurora-B inhibition by AZD1152 recovered radioprotection, induced apoptotic events and elicited anti-growth responses, similarly to that seen after MEK/ERK inhibition. Aurora-B expression dependence on active ERKs is also demonstrated by the siRNA-mediated ERK silencing, which induced downregulation of Aurora-B protein expression. Collectively, our findings suggest that the modulation of MEK/ERK signaling pathway has a strong impact on Aurora-B kinase resulting in reduced tumorigenic radioresistant phenotype in all gynecological cancer cell lines.

In conclusion, results from our studies on MEK/ERK/Aurora-B axis inhibition by MEK/ERK inihibitor can be considered of high relevance in molding the new therapeutic approaches in treating tumorigenesis such as endometrial, cervical and vaginal cancers.

References

1. 

Weiderpass E and Labrèche F: Malignant tumors of the female reproductive system. Saf Health Work. 3:166–180. 2012. View Article : Google Scholar : PubMed/NCBI

2. 

Sharma DN: Radiation in vulvar cancer. Curr Opin Obstet Gynecol. 24:24–30. 2012. View Article : Google Scholar

3. 

Wright JD, Barrena Medel NI, Sehouli J, Fujiwara K and Herzog TJ: Contemporary management of endometrial cancer. Lancet. 379:1352–1360. 2012. View Article : Google Scholar : PubMed/NCBI

4. 

Willmott LJ and Monk BJ: Cervical cancer therapy: current, future and anti-angiogensis targeted treatment. Expert Rev Anticancer Ther. 9:895–903. 2009. View Article : Google Scholar : PubMed/NCBI

5. 

McKenna WG, Weiss MC, Bakanauskas VJ, et al: The role of the H-ras oncogene in radiation resistance and metastasis. Int J Radiat Oncol Biol Phys. 18:849–859. 1990. View Article : Google Scholar : PubMed/NCBI

6. 

Jones HA, Hahn SM, Bernhard E and McKenna WG: Ras inhibitors and radiation therapy. Semin Radiat Oncol. 11:328–337. 2001. View Article : Google Scholar

7. 

Dar AA, Goff LW, Majid S, Berlin J and El-Rifai W: Aurora kinase inhibitors-rising stars in cancer therapeutics? Mol Cancer Ther. 9:268–278. 2010. View Article : Google Scholar : PubMed/NCBI

8. 

O’Neill E and Kolch W: Conferring specificity on the ubiquitous Raf/MEK signaling pathway. Br J Cancer. 90:283–288. 2004.PubMed/NCBI

9. 

Murphy LO and Blenis J: MAPK signal specificity: the right place at the right time. Trends Biochem Sci. 31:268–275. 2006. View Article : Google Scholar : PubMed/NCBI

10. 

Kohno M and Pouyssegur J: Pharmacological inhibitors of the ERK signaling pathway: application as anticancer drugs. Prog Cell Cycle Res. 5:219–224. 2003.PubMed/NCBI

11. 

Faivre S, Djelloul S and Raymond E: New paradigms in anti-cancer therapy: targeting multiple signaling pathways with kinase inhibitors. Semin Oncol. 33:407–420. 2006. View Article : Google Scholar : PubMed/NCBI

12. 

Liu D, Liu Z, Condouris S and Xing M: BRAF V600E maintains proliferation, transformation, and tumorigenicity of BRAF-mutant papillary thyroid cancer cells. J Clin Endocrinol Metab. 92:2264–2271. 2007. View Article : Google Scholar : PubMed/NCBI

13. 

Martinelli S, McDowell HP, Vigne SD, Kokai G, Uccini S and Tartaglia M: RAS signaling dysregulation in human embryonal rhabdomyosarcoma. Genes Chromosomes Cancer. 48:975–982. 2009. View Article : Google Scholar : PubMed/NCBI

14. 

Marshall CJ: The ras oncogenes. J Cell Sci. 10:157–169. 1998.

15. 

Gupta AK, Bakanauskas VJ, Cerniglia GJ, et al: The Ras radiation resistance pathway. Cancer Res. 61:4278–4282. 2001.PubMed/NCBI

16. 

Toulany M, Baumann M and Rodemann HP: Stimulated PI3K-AKT signaling mediated through ligand or radiation-induced EGFR depends indirectly, but not directly, on constitutive K-Ras activity. Mol Cancer Res. 5:863–872. 2007. View Article : Google Scholar : PubMed/NCBI

17. 

Adhikary S and Eilers M: Transcriptional regulation and transformation by Myc proteins. Nat Rev Mol Cell Biol. 6:635–645. 2005. View Article : Google Scholar : PubMed/NCBI

18. 

Mammas IN, Zafiropoulos A and Spandidos DA: Involvement of the ras genes in female genital tract cancer. Int J Oncol. 26:1241–1255. 2005.PubMed/NCBI

19. 

Carmena M and Earnshaw WC: The cellular geography of aurora kinases. Nat Rev Mol Cell Biol. 4:842–854. 2003. View Article : Google Scholar : PubMed/NCBI

20. 

Ducat D and Zheng Y: Aurora kinases in spindle assembly and chromosome segregation. Exp Cell Res. 301:60–67. 2004. View Article : Google Scholar : PubMed/NCBI

21. 

Marumoto T, Zhang D and Saya H: Aurora A, a guardian of poles. Nat Rev Cancer. 5:42–50. 2005. View Article : Google Scholar : PubMed/NCBI

22. 

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

23. 

Kobayashi M, Nakamura S, Ono T, et al: Analysis of aurora kinase expressions and cell cycle regulation by Aurora C in leukemia cells. Blood (ASH Annual Meeting Abstracts). 108:13662006.

24. 

Slattery SD, Mancini MA, Brinkley BR and Hall RM: Aurora-C kinase supports mitotic progression in the absence of Aurora-B. Cell Cycle. 8:2986–2997. 2009. View Article : Google Scholar : PubMed/NCBI

25. 

Zhang X: Aurora kinases. Curr Biol. 18:R146–R148. 2008. View Article : Google Scholar : PubMed/NCBI

26. 

Carvajal RD, Tse A and Schwartz GK: Aurora kinases: new targets for cancer therapy. Clin Cancer Res. 12:6869–6875. 2006. View Article : Google Scholar : PubMed/NCBI

27. 

Girdler F, Gascoigne KE, Eyers PA, et al: Validating Aurora B as an anti-cancer drug target. J Cell Sci. 119:3664–3675. 2006. View Article : Google Scholar : PubMed/NCBI

28. 

Smith SL, Bowers NL, Betticher DC, et al: Overexpression of Aurora B kinase (AURBK) in primary non-small cell lung carcinoma is frequent, generally driven from one allele, and correlates with the level of genetic instability. Br J Cancer. 93:719–729. 2005. View Article : Google Scholar : PubMed/NCBI

29. 

Gautschi O, Heighway J, Mack PC, Purnell PR, Lara PN Jr and Gandara DR: Aurora kinases as anticancer drug targets. Clin Cancer Res. 14:1639–1648. 2008. View Article : Google Scholar : PubMed/NCBI

30. 

Keen M and Taylor S: Mitotic drivers - inhibitors of the Aurora B kinase. Cancer Metastasis Rev. 28:185–195. 2009. View Article : Google Scholar

31. 

Bonet C, Giuliano S, Ohanna M, et al: Aurora B is regulated by the mitogen-activated protein kinase/extracellular signal-regulated kinase (MAPK/ERK) signaling pathway and is a valuable potential target in melanoma cells. J Biol Chem. 287:29887–29898. 2012. View Article : Google Scholar

32. 

Marampon F, Gravina GL, Di Rocco A, et al: MEK/ERK inhibitor U0126 increases the radiosensitivity of rhabdomyosarcoma cells in vitro and in vivo by downregulating growth and DNA repair signals. Mol Cancer Ther. 10:159–168. 2011. View Article : Google Scholar : PubMed/NCBI

33. 

Marampon F, Bossi G, Ciccarelli C, et al: MEK/ERK inhibitor U0126 affects in vitro and in vivo growth of embryonal rhabdomyosarcoma. Mol Cancer Ther. 8:543–551. 2009. View Article : Google Scholar : PubMed/NCBI

34. 

Marampon F, Ciccarelli C and Zani BM: Down-regulation of c-Myc following MEK/ERK inhibition halts the expression of malignant phenotype in rhabdomyosarcoma and in non muscle-derived human tumors. Mol Cancer. 5:312009. View Article : Google Scholar : PubMed/NCBI

35. 

Allalunis-Turner MJ, Zia PK, Barron GM, Mirzayans R and Day RS III: Radiation-induced DNA damage and repair in cells of a radiosensitive human malignant glioma cell line. Radiat Res. 144:288–293. 1995. View Article : Google Scholar : PubMed/NCBI

36. 

Lees-Miller SP, Godbout R, Chan DW, Weinfeld M, Day RS III, Barron GM and Allalunis-Turner J: Absence of p350 subunit of DNA-activated protein kinase from a radiosensitive human cell line. Science. 267:1183–1185. 1995. View Article : Google Scholar : PubMed/NCBI

37. 

Tian X, Chen G, Xing H, Weng D, Guo Y and Ma D: The relationship between the down-regulation of DNA-PKcs or Ku70 and the chemosensitization in human cervical carcinoma cell line HeLa. Oncol Rep. 18:927–932. 2007.PubMed/NCBI

38. 

Gerweck LE, Vijayappa S, Kurimasa A, Ogawa K and Chen DJ: Tumor cell radiosensitivity is a major determinant of tumor response to radiation. Cancer Res. 66:8352–8355. 2006. View Article : Google Scholar : PubMed/NCBI

39. 

van Gent DC, Hoeijmakers JH and Kanaar R: Chromosomal stability and the DNA double-stranded break connection. Nat Rev Genet. 2:196–206. 2001.

40. 

Lowry OH, Rosebrough NJ, Farr AL and Randall RJ: Protein measurement with the Folin phenol reagent. Biol Chem. 193:265–275. 1951.PubMed/NCBI

41. 

Mori S, Chang JT, Andrechek ER, et al: Anchorage-independent cell growth signature identifies tumors with metastatic potential. Oncogene. 28:2796–2805. 2009. View Article : Google Scholar : PubMed/NCBI

42. 

Sak A, Stuschke M, Groneberg M, Kübler D, Pöttgen C and Eberhardt WE: Inhibiting the Aurora B kinase potently suppresses repopulation during fractionated irradiation of human lung cancer cell lines. Int J Radiat Oncol Biol Phys. 84:492–499. 2012. View Article : Google Scholar : PubMed/NCBI

43. 

Borges KS, Castro-Gamero AM, Moreno DA, et al: Inhibition of Aurora kinases enhances chemosensitivity to temozolomide and causes radiosensitization in glioblastoma cells. J Cancer Res Clin Oncol. 138:405–414. 2012. View Article : Google Scholar : PubMed/NCBI

44. 

Niermann KJ, Moretti L, Giacalone NJ, et al: Enhanced radiosensitivity of androgen-resistant prostate cancer: AZD1152-mediated Aurora kinase B inhibition. Radiat Res. 175:444–451. 2011. View Article : Google Scholar : PubMed/NCBI

45. 

Kosik A, Bekier ME, Katusin JD, et al: Investigating the role of Aurora kinases in RAS signaling. J Cell Biochem. 106:33–41. 2009. View Article : Google Scholar : PubMed/NCBI

46. 

Ma Y, Weimer J, Fredrik R, et al: Aurora kinase inhibitor AZD1152 has an additional effect of platinum on a sequential application at the human ovarian cancer cell line SKOV3. Arch Gynecol Obstet. 288:173–182. 2013. View Article : Google Scholar : PubMed/NCBI

47. 

Baldini E, Sorrenti S, D’Armiento E, et al: Aurora kinases: new molecular targets in thyroid cancer therapy. Clin Ter. 163:e457–462. 2012.PubMed/NCBI

Related Articles

Journal Cover

2014-January
Volume 44 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
Marampon F, Gravina GL, Popov VM, Scarsella L, Festuccia C, La Verghetta ME, Parente S, Cerasani M, Bruera G, Ficorella C, Ficorella C, et al: Close correlation between MEK/ERK and Aurora-B signaling pathways in sustaining tumorigenic potential and radioresistance of gynecological cancer cell lines. Int J Oncol 44: 285-294, 2014
APA
Marampon, F., Gravina, G.L., Popov, V.M., Scarsella, L., Festuccia, C., La Verghetta, M.E. ... Zani, B.M. (2014). Close correlation between MEK/ERK and Aurora-B signaling pathways in sustaining tumorigenic potential and radioresistance of gynecological cancer cell lines. International Journal of Oncology, 44, 285-294. https://doi.org/10.3892/ijo.2013.2167
MLA
Marampon, F., Gravina, G. L., Popov, V. M., Scarsella, L., Festuccia, C., La Verghetta, M. E., Parente, S., Cerasani, M., Bruera, G., Ficorella, C., Ricevuto, E., Tombolini, V., Di Cesare, E., Zani, B. M."Close correlation between MEK/ERK and Aurora-B signaling pathways in sustaining tumorigenic potential and radioresistance of gynecological cancer cell lines". International Journal of Oncology 44.1 (2014): 285-294.
Chicago
Marampon, F., Gravina, G. L., Popov, V. M., Scarsella, L., Festuccia, C., La Verghetta, M. E., Parente, S., Cerasani, M., Bruera, G., Ficorella, C., Ricevuto, E., Tombolini, V., Di Cesare, E., Zani, B. M."Close correlation between MEK/ERK and Aurora-B signaling pathways in sustaining tumorigenic potential and radioresistance of gynecological cancer cell lines". International Journal of Oncology 44, no. 1 (2014): 285-294. https://doi.org/10.3892/ijo.2013.2167