Skip to main content
Top
Published in: Molecular Cancer 1/2015

Open Access 01-12-2015 | Research

miR-634 restores drug sensitivity in resistant ovarian cancer cells by targeting the Ras-MAPK pathway

Authors: Marijn T. M. van Jaarsveld, Patricia F. van Kuijk, Antonius W. M. Boersma, Jozien Helleman, Wilfred F. van IJcken, Ron H. J. Mathijssen, Joris Pothof, Els M. J. J. Berns, Jaap Verweij, Erik A. C. Wiemer

Published in: Molecular Cancer | Issue 1/2015

Login to get access

Abstract

Background

Drug resistance hampers the efficient treatment of malignancies, including advanced stage ovarian cancer, which has a 5-year survival rate of only 30 %. The molecular processes underlying resistance have been extensively studied, however, not much is known about the involvement of microRNAs.

Methods

Differentially expressed microRNAs between cisplatin sensitive and resistant cancer cell line pairs were determined using microarrays. Mimics were used to study the role of microRNAs in drug sensitivity of ovarian cancer cell lines and patient derived tumor cells. Luciferase reporter constructs were used to establish regulation of target genes by microRNAs.

Results

MiR-634 downregulation was associated with cisplatin resistance. Overexpression of miR-634 affected cell cycle progression and enhanced apoptosis in ovarian cancer cells. miR-634 resensitized resistant ovarian cancer cell lines and patient derived drug resistant tumor cells to cisplatin. Similarly, miR-634 enhanced the response to carboplatin and doxorubicin, but not to paclitaxel. The cell cycle regulator CCND1, and Ras-MAPK pathway components GRB2, ERK2 and RSK2 were directly repressed by miR-634 overexpression. Repression of the Ras-MAPK pathway using a MEK inhibitor phenocopied the miR-634 effects on viability and chemosensitivity.

Conclusion

miR-634 levels determine chemosensitivity in ovarian cancer cells. We identify miR-634 as a therapeutic candidate to resensitize chemotherapy resistant ovarian tumors.
Appendix
Available only for authorised users
Literature
1.
go back to reference Berns EM, Bowtell DD. The changing view of high-grade serous ovarian cancer. Cancer Res. 2012;72(11):2701–4.CrossRefPubMed Berns EM, Bowtell DD. The changing view of high-grade serous ovarian cancer. Cancer Res. 2012;72(11):2701–4.CrossRefPubMed
2.
go back to reference Ledermann JA, Marth C, Carey MS, Birrer M, Bowtell DD, Kaye S, et al. Role of molecular agents and targeted therapy in clinical trials for women with ovarian cancer. Int J Gynecol Cancer. 2011;21(4):763–70.CrossRefPubMed Ledermann JA, Marth C, Carey MS, Birrer M, Bowtell DD, Kaye S, et al. Role of molecular agents and targeted therapy in clinical trials for women with ovarian cancer. Int J Gynecol Cancer. 2011;21(4):763–70.CrossRefPubMed
3.
go back to reference Vergote I, Trope CG, Amant F, Kristensen GB, Ehlen T, Johnson N, et al. Neoadjuvant chemotherapy or primary surgery in stage IIIC or IV ovarian cancer. N Engl J Med. 2010;363(10):943–53.CrossRefPubMed Vergote I, Trope CG, Amant F, Kristensen GB, Ehlen T, Johnson N, et al. Neoadjuvant chemotherapy or primary surgery in stage IIIC or IV ovarian cancer. N Engl J Med. 2010;363(10):943–53.CrossRefPubMed
4.
5.
go back to reference Ozols RF. Epithelial Ovarian Cancer. In: Hoskins WJYR, Markman M, Perez CA, Barakat R, Randall M, editors. Principles and Practice of Gynaecologic Oncology. Philadelphia, PA: Lippincott Williams & Wilkins; 2005. p. 895–985. Ozols RF. Epithelial Ovarian Cancer. In: Hoskins WJYR, Markman M, Perez CA, Barakat R, Randall M, editors. Principles and Practice of Gynaecologic Oncology. Philadelphia, PA: Lippincott Williams & Wilkins; 2005. p. 895–985.
6.
go back to reference Zhang W, Liu HT. MAPK signal pathways in the regulation of cell proliferation in mammalian cells. Cell Res. 2002;12(1):9–18.CrossRefPubMed Zhang W, Liu HT. MAPK signal pathways in the regulation of cell proliferation in mammalian cells. Cell Res. 2002;12(1):9–18.CrossRefPubMed
7.
go back to reference Li W, Melton DW. Cisplatin regulates the MAPK kinase pathway to induce increased expression of DNA repair gene ERCC1 and increase melanoma chemoresistance. Oncogene. 2012;31(19):2412–22.CrossRefPubMed Li W, Melton DW. Cisplatin regulates the MAPK kinase pathway to induce increased expression of DNA repair gene ERCC1 and increase melanoma chemoresistance. Oncogene. 2012;31(19):2412–22.CrossRefPubMed
8.
go back to reference Dhillon AS, Hagan S, Rath O, Kolch W. MAP kinase signalling pathways in cancer. Oncogene. 2007;26(22):3279–90.CrossRefPubMed Dhillon AS, Hagan S, Rath O, Kolch W. MAP kinase signalling pathways in cancer. Oncogene. 2007;26(22):3279–90.CrossRefPubMed
9.
go back to reference MacCorkle RA, Tan TH. Mitogen-activated protein kinases in cell-cycle control. Cell Biochem Biophys. 2005;43(3):451–61.CrossRefPubMed MacCorkle RA, Tan TH. Mitogen-activated protein kinases in cell-cycle control. Cell Biochem Biophys. 2005;43(3):451–61.CrossRefPubMed
10.
go back to reference Cagnol S, Chambard JC. ERK and cell death: mechanisms of ERK-induced cell death--apoptosis, autophagy and senescence. Febs J. 2010;277(1):2–21.CrossRefPubMed Cagnol S, Chambard JC. ERK and cell death: mechanisms of ERK-induced cell death--apoptosis, autophagy and senescence. Febs J. 2010;277(1):2–21.CrossRefPubMed
12.
go back to reference van Jaarsveld MT, Helleman J, Berns EM, Wiemer EA. MicroRNAs in ovarian cancer biology and therapy resistance. Int J Biochem Cell Biol. 2010;42(8):1282–90.CrossRefPubMed van Jaarsveld MT, Helleman J, Berns EM, Wiemer EA. MicroRNAs in ovarian cancer biology and therapy resistance. Int J Biochem Cell Biol. 2010;42(8):1282–90.CrossRefPubMed
13.
go back to reference van Jaarsveld MT, Helleman J, Boersma AW, van Kuijk PF, van Ijcken WF, Despierre E, et al. miR-141 regulates KEAP1 and modulates cisplatin sensitivity in ovarian cancer cells. Oncogene. 2013;32(36):4284–93.CrossRefPubMed van Jaarsveld MT, Helleman J, Boersma AW, van Kuijk PF, van Ijcken WF, Despierre E, et al. miR-141 regulates KEAP1 and modulates cisplatin sensitivity in ovarian cancer cells. Oncogene. 2013;32(36):4284–93.CrossRefPubMed
14.
go back to reference Kool M, de Haas M, Scheffer GL, Scheper RJ, van Eijk MJ, Juijn JA, et al. Analysis of expression of cMOAT (MRP2), MRP3, MRP4, and MRP5, homologues of the multidrug resistance-associated protein gene (MRP1), in human cancer cell lines. Cancer Res. 1997;57(16):3537–47.PubMed Kool M, de Haas M, Scheffer GL, Scheper RJ, van Eijk MJ, Juijn JA, et al. Analysis of expression of cMOAT (MRP2), MRP3, MRP4, and MRP5, homologues of the multidrug resistance-associated protein gene (MRP1), in human cancer cell lines. Cancer Res. 1997;57(16):3537–47.PubMed
15.
go back to reference Schmidt W, Chaney SG. Role of carrier ligand in platinum resistance of human carcinoma cell lines. Cancer Res. 1993;53(4):799–805.PubMed Schmidt W, Chaney SG. Role of carrier ligand in platinum resistance of human carcinoma cell lines. Cancer Res. 1993;53(4):799–805.PubMed
17.
go back to reference Shepherd TG, Theriault BL, Campbell EJ, Nachtigal MW. Primary culture of ovarian surface epithelial cells and ascites-derived ovarian cancer cells from patients. Nat Protoc. 2006;1(6):2643–9.CrossRefPubMed Shepherd TG, Theriault BL, Campbell EJ, Nachtigal MW. Primary culture of ovarian surface epithelial cells and ascites-derived ovarian cancer cells from patients. Nat Protoc. 2006;1(6):2643–9.CrossRefPubMed
18.
go back to reference Ayhan A, Kurman RJ, Yemelyanova A, Vang R, Logani S, Seidman JD, et al. Defining the cut point between low-grade and high-grade ovarian serous carcinomas: a clinicopathologic and molecular genetic analysis. Am J Surg Pathol. 2009;33(8):1220–4.CrossRefPubMedCentralPubMed Ayhan A, Kurman RJ, Yemelyanova A, Vang R, Logani S, Seidman JD, et al. Defining the cut point between low-grade and high-grade ovarian serous carcinomas: a clinicopathologic and molecular genetic analysis. Am J Surg Pathol. 2009;33(8):1220–4.CrossRefPubMedCentralPubMed
19.
go back to reference Brozovic A, Osmak M. Activation of mitogen-activated protein kinases by cisplatin and their role in cisplatin-resistance. Cancer Lett. 2007;251(1):1–16.CrossRefPubMed Brozovic A, Osmak M. Activation of mitogen-activated protein kinases by cisplatin and their role in cisplatin-resistance. Cancer Lett. 2007;251(1):1–16.CrossRefPubMed
20.
go back to reference van Jaarsveld MT, Blijdorp IC, Boersma AW, Pothof J, Mathijssen RH, Verweij J, et al. The kinase RSK2 modulates the sensitivity of ovarian cancer cells to cisplatin. Eur J Cancer. 2013;49(2):345–51.CrossRefPubMed van Jaarsveld MT, Blijdorp IC, Boersma AW, Pothof J, Mathijssen RH, Verweij J, et al. The kinase RSK2 modulates the sensitivity of ovarian cancer cells to cisplatin. Eur J Cancer. 2013;49(2):345–51.CrossRefPubMed
21.
22.
go back to reference Ostling P, Leivonen SK, Aakula A, Kohonen P, Makela R, Hagman Z, et al. Systematic analysis of microRNAs targeting the androgen receptor in prostate cancer cells. Cancer Res. 2011;71(5):1956–67.CrossRefPubMed Ostling P, Leivonen SK, Aakula A, Kohonen P, Makela R, Hagman Z, et al. Systematic analysis of microRNAs targeting the androgen receptor in prostate cancer cells. Cancer Res. 2011;71(5):1956–67.CrossRefPubMed
23.
go back to reference Li Y, Choi PS, Casey SC, Dill DL, Felsher DW. MYC through miR-17-92 suppresses specific target genes to maintain survival, autonomous proliferation, and a neoplastic state. Cancer Cell. 2014;26(2):262–72.CrossRefPubMedCentralPubMed Li Y, Choi PS, Casey SC, Dill DL, Felsher DW. MYC through miR-17-92 suppresses specific target genes to maintain survival, autonomous proliferation, and a neoplastic state. Cancer Cell. 2014;26(2):262–72.CrossRefPubMedCentralPubMed
24.
go back to reference Johnson SM, Grosshans H, Shingara J, Byrom M, Jarvis R, Cheng A, et al. RAS is regulated by the let-7 microRNA family. Cell. 2005;120(5):635–47.CrossRefPubMed Johnson SM, Grosshans H, Shingara J, Byrom M, Jarvis R, Cheng A, et al. RAS is regulated by the let-7 microRNA family. Cell. 2005;120(5):635–47.CrossRefPubMed
26.
go back to reference Gregory PA, Bert AG, Paterson EL, Barry SC, Tsykin A, Farshid G, et al. The miR-200 family and miR-205 regulate epithelial to mesenchymal transition by targeting ZEB1 and SIP1. Nat Cell Biol. 2008;10(5):593–601.CrossRefPubMed Gregory PA, Bert AG, Paterson EL, Barry SC, Tsykin A, Farshid G, et al. The miR-200 family and miR-205 regulate epithelial to mesenchymal transition by targeting ZEB1 and SIP1. Nat Cell Biol. 2008;10(5):593–601.CrossRefPubMed
27.
go back to reference Park SM, Gaur AB, Lengyel E, Peter ME. The miR-200 family determines the epithelial phenotype of cancer cells by targeting the E-cadherin repressors ZEB1 and ZEB2. Genes Dev. 2008;22(7):894–907.CrossRefPubMedCentralPubMed Park SM, Gaur AB, Lengyel E, Peter ME. The miR-200 family determines the epithelial phenotype of cancer cells by targeting the E-cadherin repressors ZEB1 and ZEB2. Genes Dev. 2008;22(7):894–907.CrossRefPubMedCentralPubMed
28.
go back to reference Burk U, Schubert J, Wellner U, Schmalhofer O, Vincan E, Spaderna S, et al. A reciprocal repression between ZEB1 and members of the miR-200 family promotes EMT and invasion in cancer cells. EMBO Rep. 2008;9(6):582–9.CrossRefPubMedCentralPubMed Burk U, Schubert J, Wellner U, Schmalhofer O, Vincan E, Spaderna S, et al. A reciprocal repression between ZEB1 and members of the miR-200 family promotes EMT and invasion in cancer cells. EMBO Rep. 2008;9(6):582–9.CrossRefPubMedCentralPubMed
29.
go back to reference Liu YN, Yin JJ, Abou-Kheir W, Hynes PG, Casey OM, Fang L, et al. MiR-1 and miR-200 inhibit EMT via Slug-dependent and tumorigenesis via Slug-independent mechanisms. Oncogene. 2013;32(3):296–306.CrossRefPubMed Liu YN, Yin JJ, Abou-Kheir W, Hynes PG, Casey OM, Fang L, et al. MiR-1 and miR-200 inhibit EMT via Slug-dependent and tumorigenesis via Slug-independent mechanisms. Oncogene. 2013;32(3):296–306.CrossRefPubMed
31.
go back to reference Ewings KE, Wiggins CM, Cook SJ. Bim and the pro-survival Bcl-2 proteins: opposites attract. ERK repels Cell Cycle. 2007;6(18):2236–40.CrossRefPubMed Ewings KE, Wiggins CM, Cook SJ. Bim and the pro-survival Bcl-2 proteins: opposites attract. ERK repels Cell Cycle. 2007;6(18):2236–40.CrossRefPubMed
32.
go back to reference Romeo Y, Zhang X, Roux PP. Regulation and function of the RSK family of protein kinases. Biochem J. 2012;441(2):553–69.CrossRefPubMed Romeo Y, Zhang X, Roux PP. Regulation and function of the RSK family of protein kinases. Biochem J. 2012;441(2):553–69.CrossRefPubMed
33.
go back to reference Ray-David H, Romeo Y, Lavoie G, Deleris P, Tcherkezian J, Galan JA, et al. RSK promotes G2 DNA damage checkpoint silencing and participates in melanoma chemoresistance. Oncogene. 2013;32(38):4480–9.CrossRefPubMed Ray-David H, Romeo Y, Lavoie G, Deleris P, Tcherkezian J, Galan JA, et al. RSK promotes G2 DNA damage checkpoint silencing and participates in melanoma chemoresistance. Oncogene. 2013;32(38):4480–9.CrossRefPubMed
35.
go back to reference Chen C, Zhang L, Huang NJ, Huang B, Kornbluth S. Suppression of DNA-damage checkpoint signaling by Rsk-mediated phosphorylation of Mre11. Proc Natl Acad Sci U S A. 2013;110(51):20605–10.CrossRefPubMedCentralPubMed Chen C, Zhang L, Huang NJ, Huang B, Kornbluth S. Suppression of DNA-damage checkpoint signaling by Rsk-mediated phosphorylation of Mre11. Proc Natl Acad Sci U S A. 2013;110(51):20605–10.CrossRefPubMedCentralPubMed
36.
37.
go back to reference Pritchard AL, Hayward NK. Molecular pathways: mitogen-activated protein kinase pathway mutations and drug resistance. Clin Cancer Res. 2013;19(9):2301–9.CrossRefPubMed Pritchard AL, Hayward NK. Molecular pathways: mitogen-activated protein kinase pathway mutations and drug resistance. Clin Cancer Res. 2013;19(9):2301–9.CrossRefPubMed
38.
go back to reference Seton-Rogers S. Therapeutics: Delving deeper into resistance. Nat Rev Cancer. 2013;14(1):7.CrossRef Seton-Rogers S. Therapeutics: Delving deeper into resistance. Nat Rev Cancer. 2013;14(1):7.CrossRef
39.
go back to reference Shan W, Liu J. Epithelial ovarian cancer: focus on genetics and animal models. Cell Cycle. 2009;8(5):731–5.CrossRefPubMed Shan W, Liu J. Epithelial ovarian cancer: focus on genetics and animal models. Cell Cycle. 2009;8(5):731–5.CrossRefPubMed
41.
go back to reference Leivonen SK, Sahlberg KK, Makela R, Due EU, Kallioniemi O, Borresen-Dale AL, et al. High-throughput screens identify microRNAs essential for HER2 positive breast cancer cell growth. Mol Oncol. 2014;8(1):93–104.CrossRefPubMed Leivonen SK, Sahlberg KK, Makela R, Due EU, Kallioniemi O, Borresen-Dale AL, et al. High-throughput screens identify microRNAs essential for HER2 positive breast cancer cell growth. Mol Oncol. 2014;8(1):93–104.CrossRefPubMed
42.
go back to reference Rainer J, Ploner C, Jesacher S, Ploner A, Eduardoff M, Mansha M, et al. Glucocorticoid-regulated microRNAs and mirtrons in acute lymphoblastic leukemia. Leukemia. 2009;23(4):746–52.CrossRefPubMed Rainer J, Ploner C, Jesacher S, Ploner A, Eduardoff M, Mansha M, et al. Glucocorticoid-regulated microRNAs and mirtrons in acute lymphoblastic leukemia. Leukemia. 2009;23(4):746–52.CrossRefPubMed
43.
go back to reference Bach D, Fuereder J, Karbiener M, Scheideler M, Ress AL, Neureiter D, et al. Comprehensive analysis of alterations in the miRNome in response to photodynamic treatment. J Photochem Photobiol B. 2013;120:74–81.CrossRefPubMed Bach D, Fuereder J, Karbiener M, Scheideler M, Ress AL, Neureiter D, et al. Comprehensive analysis of alterations in the miRNome in response to photodynamic treatment. J Photochem Photobiol B. 2013;120:74–81.CrossRefPubMed
44.
go back to reference Peng X, Cao P, He D, Han S, Zhou J, Tan G, et al. MiR-634 sensitizes nasopharyngeal carcinoma cells to paclitaxel and inhibits cell growth both in vitro and in vivo. Int J Clin Exp Pathol. 2014;7(10):6784–91.PubMedCentralPubMed Peng X, Cao P, He D, Han S, Zhou J, Tan G, et al. MiR-634 sensitizes nasopharyngeal carcinoma cells to paclitaxel and inhibits cell growth both in vitro and in vivo. Int J Clin Exp Pathol. 2014;7(10):6784–91.PubMedCentralPubMed
45.
go back to reference Yamamoto S, Inoue J, Kawano T, Kozaki K, Omura K, Inazawa J. The impact of miRNA-based molecular diagnostics and treatment of NRF2-stabilized tumors. Mol Cancer Res. 2014;12(1):58–68.CrossRefPubMed Yamamoto S, Inoue J, Kawano T, Kozaki K, Omura K, Inazawa J. The impact of miRNA-based molecular diagnostics and treatment of NRF2-stabilized tumors. Mol Cancer Res. 2014;12(1):58–68.CrossRefPubMed
46.
47.
go back to reference Wyman SK, Knouf EC, Parkin RK, Fritz BR, Lin DW, Dennis LM, et al. Post-transcriptional generation of miRNA variants by multiple nucleotidyl transferases contributes to miRNA transcriptome complexity. Genome Res. 2011;21(9):1450–61.CrossRefPubMedCentralPubMed Wyman SK, Knouf EC, Parkin RK, Fritz BR, Lin DW, Dennis LM, et al. Post-transcriptional generation of miRNA variants by multiple nucleotidyl transferases contributes to miRNA transcriptome complexity. Genome Res. 2011;21(9):1450–61.CrossRefPubMedCentralPubMed
48.
go back to reference Kotoh S, Naito S, Yokomizo A, Kumazawa J, Asakuno K, Kohno K, et al. Increased expression of DNA topoisomerase I gene and collateral sensitivity to camptothecin in human cisplatin-resistant bladder cancer cells. Cancer Res. 1994;54(12):3248–52.PubMed Kotoh S, Naito S, Yokomizo A, Kumazawa J, Asakuno K, Kohno K, et al. Increased expression of DNA topoisomerase I gene and collateral sensitivity to camptothecin in human cisplatin-resistant bladder cancer cells. Cancer Res. 1994;54(12):3248–52.PubMed
49.
go back to reference Goto S, Yoshida K, Morikawa T, Urata Y, Suzuki K, Kondo T. Augmentation of transport for cisplatin-glutathione adduct in cisplatin-resistant cancer cells. Cancer Res. 1995;55(19):4297–301.PubMed Goto S, Yoshida K, Morikawa T, Urata Y, Suzuki K, Kondo T. Augmentation of transport for cisplatin-glutathione adduct in cisplatin-resistant cancer cells. Cancer Res. 1995;55(19):4297–301.PubMed
50.
go back to reference Beaufort CM, Helmijr JC, Piskorz AM, Hoogstraat M, Ruigrok-Ritstier K, Besselink N, et al. Ovarian cancer cell line panel (OCCP): clinical importance of in vitro morphological subtypes. PLoS One. 2014;9(9):e103988.CrossRefPubMedCentralPubMed Beaufort CM, Helmijr JC, Piskorz AM, Hoogstraat M, Ruigrok-Ritstier K, Besselink N, et al. Ovarian cancer cell line panel (OCCP): clinical importance of in vitro morphological subtypes. PLoS One. 2014;9(9):e103988.CrossRefPubMedCentralPubMed
51.
go back to reference Schmittgen TD, Livak KJ. Analyzing real-time PCR data by the comparative C(T) method. Nat Protoc. 2008;3(6):1101–8.CrossRefPubMed Schmittgen TD, Livak KJ. Analyzing real-time PCR data by the comparative C(T) method. Nat Protoc. 2008;3(6):1101–8.CrossRefPubMed
52.
go back to reference Carmichael J, DeGraff WG, Gazdar AF, Minna JD, Mitchell JB. Evaluation of a tetrazolium-based semiautomated colorimetric assay: assessment of chemosensitivity testing. Cancer Res. 1987;47(4):936–42.PubMed Carmichael J, DeGraff WG, Gazdar AF, Minna JD, Mitchell JB. Evaluation of a tetrazolium-based semiautomated colorimetric assay: assessment of chemosensitivity testing. Cancer Res. 1987;47(4):936–42.PubMed
53.
go back to reference Burger H, Zoumaro-Djayoon A, Boersma AW, Helleman J, Berns EM, Mathijssen RH, et al. Differential transport of platinum compounds by the human organic cation transporter hOCT2 (hSLC22A2). Br J Pharmacol. 2010;159(4):898–908.CrossRefPubMedCentralPubMed Burger H, Zoumaro-Djayoon A, Boersma AW, Helleman J, Berns EM, Mathijssen RH, et al. Differential transport of platinum compounds by the human organic cation transporter hOCT2 (hSLC22A2). Br J Pharmacol. 2010;159(4):898–908.CrossRefPubMedCentralPubMed
54.
go back to reference Tusher VG, Tibshirani R, Chu G. Significance analysis of microarrays applied to the ionizing radiation response. Proc Natl Acad Sci U S A. 2001;98(9):5116–21.CrossRefPubMedCentralPubMed Tusher VG, Tibshirani R, Chu G. Significance analysis of microarrays applied to the ionizing radiation response. Proc Natl Acad Sci U S A. 2001;98(9):5116–21.CrossRefPubMedCentralPubMed
55.
go back to reference Bonni A, Brunet A, West AE, Datta SR, Takasu MA, Greenberg ME. Cell survival promoted by the Ras-MAPK signaling pathway by transcription-dependent and -independent mechanisms. Science. 1999;286(5443):1358–62.CrossRefPubMed Bonni A, Brunet A, West AE, Datta SR, Takasu MA, Greenberg ME. Cell survival promoted by the Ras-MAPK signaling pathway by transcription-dependent and -independent mechanisms. Science. 1999;286(5443):1358–62.CrossRefPubMed
56.
go back to reference Ewings KE, Hadfield-Moorhouse K, Wiggins CM, Wickenden JA, Balmanno K, Gilley R, et al. ERK1/2-dependent phosphorylation of BimEL promotes its rapid dissociation from Mcl-1 and Bcl-xL. Embo J. 2007;26(12):2856–67.CrossRefPubMedCentralPubMed Ewings KE, Hadfield-Moorhouse K, Wiggins CM, Wickenden JA, Balmanno K, Gilley R, et al. ERK1/2-dependent phosphorylation of BimEL promotes its rapid dissociation from Mcl-1 and Bcl-xL. Embo J. 2007;26(12):2856–67.CrossRefPubMedCentralPubMed
57.
go back to reference Dehan E, Bassermann F, Guardavaccaro D, Vasiliver-Shamis G, Cohen M, Lowes KN, et al. betaTrCP- and Rsk1/2-mediated degradation of BimEL inhibits apoptosis. Mol Cell. 2009;33(1):109–16.CrossRefPubMedCentralPubMed Dehan E, Bassermann F, Guardavaccaro D, Vasiliver-Shamis G, Cohen M, Lowes KN, et al. betaTrCP- and Rsk1/2-mediated degradation of BimEL inhibits apoptosis. Mol Cell. 2009;33(1):109–16.CrossRefPubMedCentralPubMed
Metadata
Title
miR-634 restores drug sensitivity in resistant ovarian cancer cells by targeting the Ras-MAPK pathway
Authors
Marijn T. M. van Jaarsveld
Patricia F. van Kuijk
Antonius W. M. Boersma
Jozien Helleman
Wilfred F. van IJcken
Ron H. J. Mathijssen
Joris Pothof
Els M. J. J. Berns
Jaap Verweij
Erik A. C. Wiemer
Publication date
01-12-2015
Publisher
BioMed Central
Published in
Molecular Cancer / Issue 1/2015
Electronic ISSN: 1476-4598
DOI
https://doi.org/10.1186/s12943-015-0464-4

Other articles of this Issue 1/2015

Molecular Cancer 1/2015 Go to the issue
Webinar | 19-02-2024 | 17:30 (CET)

Keynote webinar | Spotlight on antibody–drug conjugates in cancer

Antibody–drug conjugates (ADCs) are novel agents that have shown promise across multiple tumor types. Explore the current landscape of ADCs in breast and lung cancer with our experts, and gain insights into the mechanism of action, key clinical trials data, existing challenges, and future directions.

Dr. Véronique Diéras
Prof. Fabrice Barlesi
Developed by: Springer Medicine