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

Open Access 01-12-2015 | Research article

Role of the splicing factor SRSF4 in cisplatin-induced modifications of pre-mRNA splicing and apoptosis

Authors: Maude Gabriel, Yves Delforge, Adeline Deward, Yvette Habraken, Benoit Hennuy, Jacques Piette, Roscoe Klinck, Benoit Chabot, Alain Colige, Charles Lambert

Published in: BMC Cancer | Issue 1/2015

Login to get access

Abstract

Background

Modification of splicing by chemotherapeutic drugs has usually been evaluated on a limited number of pre-mRNAs selected for their recognized or potential importance in cell proliferation or apoptosis. However, the pathways linking splicing alterations to the efficiency of cancer therapy remain unclear.

Methods

Next-generation sequencing was used to analyse the transcriptome of breast carcinoma cells treated by cisplatin. Pharmacological inhibitors, RNA interference, cells deficient in specific signalling pathways, RT-PCR and FACS analysis were used to investigate how the anti-cancer drug cisplatin affected alternative splicing and the cell death pathway.

Results

We identified 717 splicing events affected by cisplatin, including 245 events involving cassette exons. Gene ontology analysis indicates that cell cycle, mRNA processing and pre-mRNA splicing were the main pathways affected. Importantly, the cisplatin–induced splicing alterations required class I PI3Ks P110β but not components such as ATM, ATR and p53 that are involved in the DNA damage response. The siRNA-mediated depletion of the splicing regulator SRSF4, but not SRSF6, expression abrogated many of the splicing alterations as well as cell death induced by cisplatin.

Conclusion

Many of the splicing alterations induced by cisplatin are caused by SRSF4 and they contribute to apoptosis in a process requires class I PI3K.
Appendix
Available only for authorised users
Literature
1.
go back to reference Wang D, Lippard SJ. Cellular processing of platinum anticancer drugs. Nat Rev Drug Discov. 2005;4(4):307–20. doi:10.1038/nrd1691.CrossRefPubMed Wang D, Lippard SJ. Cellular processing of platinum anticancer drugs. Nat Rev Drug Discov. 2005;4(4):307–20. doi:10.1038/nrd1691.CrossRefPubMed
2.
go back to reference Kartalou M, Essigmann JM. Mechanisms of resistance to cisplatin. Mutat Res. 2001;478(1–2):23–43.CrossRefPubMed Kartalou M, Essigmann JM. Mechanisms of resistance to cisplatin. Mutat Res. 2001;478(1–2):23–43.CrossRefPubMed
3.
go back to reference Yang J, Yu Y, Hamrick HE, Duerksen-Hughes PJ. ATM, ATR and DNA-PK: initiators of the cellular genotoxic stress responses. Carcinogenesis. 2003;24(10):1571–80. doi:10.1093/carcin/bgg137.CrossRefPubMed Yang J, Yu Y, Hamrick HE, Duerksen-Hughes PJ. ATM, ATR and DNA-PK: initiators of the cellular genotoxic stress responses. Carcinogenesis. 2003;24(10):1571–80. doi:10.1093/carcin/bgg137.CrossRefPubMed
4.
go back to reference Blencowe BJ. Alternative splicing: new insights from global analyses. Cell. 2006;126(1):37–47. doi:10.1016/j.cell.2006.06.023.CrossRefPubMed Blencowe BJ. Alternative splicing: new insights from global analyses. Cell. 2006;126(1):37–47. doi:10.1016/j.cell.2006.06.023.CrossRefPubMed
5.
go back to reference Burge C, Karlin S. Prediction of complete gene structures in human genomic DNA. J Mol Biol. 1997;268(1):78–94. doi:10.1006/jmbi.1997.0951.CrossRefPubMed Burge C, Karlin S. Prediction of complete gene structures in human genomic DNA. J Mol Biol. 1997;268(1):78–94. doi:10.1006/jmbi.1997.0951.CrossRefPubMed
6.
go back to reference Shkreta L, Froehlich U, Paquet ER, Toutant J, Elela SA, Chabot B. Anticancer drugs affect the alternative splicing of Bcl-x and other human apoptotic genes. Mol Cancer Ther. 2008;7(6):1398–409. doi:10.1158/1535-7163.mct-08-0192.CrossRefPubMed Shkreta L, Froehlich U, Paquet ER, Toutant J, Elela SA, Chabot B. Anticancer drugs affect the alternative splicing of Bcl-x and other human apoptotic genes. Mol Cancer Ther. 2008;7(6):1398–409. doi:10.1158/1535-7163.mct-08-0192.CrossRefPubMed
7.
go back to reference McGlincy NJ, Smith CW. Alternative splicing resulting in nonsense-mediated mRNA decay: what is the meaning of nonsense? Trends Biochem Sci. 2008;33(8):385–93. doi:10.1016/j.tibs.2008.06.001.CrossRefPubMed McGlincy NJ, Smith CW. Alternative splicing resulting in nonsense-mediated mRNA decay: what is the meaning of nonsense? Trends Biochem Sci. 2008;33(8):385–93. doi:10.1016/j.tibs.2008.06.001.CrossRefPubMed
8.
go back to reference Zhou Z, Fu XD. Regulation of splicing by SR proteins and SR protein-specific kinases. Chromosoma. 2013;122(3):191–207. doi:10.1007/s00412-013-0407-z.CrossRefPubMedPubMedCentral Zhou Z, Fu XD. Regulation of splicing by SR proteins and SR protein-specific kinases. Chromosoma. 2013;122(3):191–207. doi:10.1007/s00412-013-0407-z.CrossRefPubMedPubMedCentral
9.
go back to reference Tang JY, Lee JC, Hou MF, Wang CL, Chen CC, Huang HW, et al. Alternative splicing for diseases, cancers, drugs, and databases. Sci World J. 2013;2013:703568. doi:10.1155/2013/703568. Tang JY, Lee JC, Hou MF, Wang CL, Chen CC, Huang HW, et al. Alternative splicing for diseases, cancers, drugs, and databases. Sci World J. 2013;2013:703568. doi:10.1155/2013/703568.
10.
go back to reference Venables JP. Unbalanced alternative splicing and its significance in cancer. Bioessays. 2006;28(4):378–86. doi:10.1002/bies.20390.CrossRefPubMed Venables JP. Unbalanced alternative splicing and its significance in cancer. Bioessays. 2006;28(4):378–86. doi:10.1002/bies.20390.CrossRefPubMed
11.
go back to reference Shkreta L, Bell B, Revil T, Venables JP, Prinos P, Elela SA, et al. Cancer-associated perturbations in alternative Pre-messenger RNA splicing. Cancer Treat Res. 2013;158:41–94. doi:10.1007/978-3-642-31659-3_3.CrossRefPubMed Shkreta L, Bell B, Revil T, Venables JP, Prinos P, Elela SA, et al. Cancer-associated perturbations in alternative Pre-messenger RNA splicing. Cancer Treat Res. 2013;158:41–94. doi:10.1007/978-3-642-31659-3_3.CrossRefPubMed
12.
go back to reference Venables JP, Klinck R, Koh C, Gervais-Bird J, Bramard A, Inkel L, et al. Cancer-associated regulation of alternative splicing. Nat Struct Mol Biol. 2009;16(6):670–6. doi:10.1038/nsmb.1608.CrossRefPubMed Venables JP, Klinck R, Koh C, Gervais-Bird J, Bramard A, Inkel L, et al. Cancer-associated regulation of alternative splicing. Nat Struct Mol Biol. 2009;16(6):670–6. doi:10.1038/nsmb.1608.CrossRefPubMed
13.
go back to reference Solier S, Barb J, Zeeberg BR, Varma S, Ryan MC, Kohn KW, et al. Genome-wide analysis of novel splice variants induced by topoisomerase I poisoning shows preferential occurrence in genes encoding splicing factors. Cancer Res. 2010;70(20):8055–65. doi:10.1158/0008-5472.can-10-2491.CrossRefPubMedPubMedCentral Solier S, Barb J, Zeeberg BR, Varma S, Ryan MC, Kohn KW, et al. Genome-wide analysis of novel splice variants induced by topoisomerase I poisoning shows preferential occurrence in genes encoding splicing factors. Cancer Res. 2010;70(20):8055–65. doi:10.1158/0008-5472.can-10-2491.CrossRefPubMedPubMedCentral
14.
go back to reference Mineur P, Colige AC, Deroanne CF, Dubail J, Kesteloot F, Habraken Y, et al. Newly identified biologically active and proteolysis-resistant VEGF-A isoform VEGF111 is induced by genotoxic agents. J Cell Biol. 2007;179(6):1261–73. doi:10.1083/jcb.200703052.CrossRefPubMedPubMedCentral Mineur P, Colige AC, Deroanne CF, Dubail J, Kesteloot F, Habraken Y, et al. Newly identified biologically active and proteolysis-resistant VEGF-A isoform VEGF111 is induced by genotoxic agents. J Cell Biol. 2007;179(6):1261–73. doi:10.1083/jcb.200703052.CrossRefPubMedPubMedCentral
15.
go back to reference Chandler DS, Singh RK, Caldwell LC, Bitler JL, Lozano G. Genotoxic stress induces coordinately regulated alternative splicing of the p53 modulators MDM2 and MDM4. Cancer Res. 2006;66(19):9502–8. doi:10.1158/0008-5472.can-05-4271.CrossRefPubMed Chandler DS, Singh RK, Caldwell LC, Bitler JL, Lozano G. Genotoxic stress induces coordinately regulated alternative splicing of the p53 modulators MDM2 and MDM4. Cancer Res. 2006;66(19):9502–8. doi:10.1158/0008-5472.can-05-4271.CrossRefPubMed
16.
go back to reference Dutertre M, Sanchez G, Barbier J, Corcos L, Auboeuf D. The emerging role of pre-messenger RNA splicing in stress responses: sending alternative messages and silent messengers. RNA Biol. 2011;8(5):740–7. doi:10.4161/rna.8.5.16016.CrossRefPubMed Dutertre M, Sanchez G, Barbier J, Corcos L, Auboeuf D. The emerging role of pre-messenger RNA splicing in stress responses: sending alternative messages and silent messengers. RNA Biol. 2011;8(5):740–7. doi:10.4161/rna.8.5.16016.CrossRefPubMed
17.
go back to reference Neutelings T, Lambert CA, Nusgens BV, Colige AC. Effects of mild cold shock (25 degrees C) followed by warming up at 37 degrees C on the cellular stress response. PLoS One. 2013;8(7):e69687. doi:10.1371/journal.pone.0069687.CrossRefPubMedPubMedCentral Neutelings T, Lambert CA, Nusgens BV, Colige AC. Effects of mild cold shock (25 degrees C) followed by warming up at 37 degrees C on the cellular stress response. PLoS One. 2013;8(7):e69687. doi:10.1371/journal.pone.0069687.CrossRefPubMedPubMedCentral
18.
go back to reference Bustin SA, Benes V, Garson JA, Hellemans J, Huggett J, Kubista M, et al. The MIQE guidelines: minimum information for publication of quantitative real-time PCR experiments. Clin Chem. 2009;55(4):611–22. doi:10.1373/clinchem.2008.112797.CrossRefPubMed Bustin SA, Benes V, Garson JA, Hellemans J, Huggett J, Kubista M, et al. The MIQE guidelines: minimum information for publication of quantitative real-time PCR experiments. Clin Chem. 2009;55(4):611–22. doi:10.1373/clinchem.2008.112797.CrossRefPubMed
19.
go back to reference Langmead B, Trapnell C, Pop M, Salzberg SL. Ultrafast and memory-efficient alignment of short DNA sequences to the human genome. Genome Biol. 2009;10(3):R25. doi:10.1186/gb-2009-10-3-r25.CrossRefPubMedPubMedCentral Langmead B, Trapnell C, Pop M, Salzberg SL. Ultrafast and memory-efficient alignment of short DNA sequences to the human genome. Genome Biol. 2009;10(3):R25. doi:10.1186/gb-2009-10-3-r25.CrossRefPubMedPubMedCentral
20.
go back to reference Ryan MC, Cleland J, Kim R, Wong WC, Weinstein JN. SpliceSeq: a resource for analysis and visualization of RNA-Seq data on alternative splicing and its functional impacts. Bioinformatics (Oxford, England). 2012;28(18):2385–7. doi:10.1093/bioinformatics/bts452.CrossRef Ryan MC, Cleland J, Kim R, Wong WC, Weinstein JN. SpliceSeq: a resource for analysis and visualization of RNA-Seq data on alternative splicing and its functional impacts. Bioinformatics (Oxford, England). 2012;28(18):2385–7. doi:10.1093/bioinformatics/bts452.CrossRef
21.
go back to reference Chen J, Bardes EE, Aronow BJ, Jegga AG. ToppGene Suite for gene list enrichment analysis and candidate gene prioritization. Nucleic Acids Res. 2009;37(Web Server issue):W305–11. doi:10.1093/nar/gkp427.CrossRefPubMedPubMedCentral Chen J, Bardes EE, Aronow BJ, Jegga AG. ToppGene Suite for gene list enrichment analysis and candidate gene prioritization. Nucleic Acids Res. 2009;37(Web Server issue):W305–11. doi:10.1093/nar/gkp427.CrossRefPubMedPubMedCentral
22.
go back to reference Ho TT, Merajver SD, Lapiere CM, Nusgens BV, Deroanne CF. RhoA-GDP regulates RhoB protein stability. Potential involvement of RhoGDIalpha. J Biol Chem. 2008;283(31):21588–98. doi:10.1074/jbc.M710033200.CrossRefPubMed Ho TT, Merajver SD, Lapiere CM, Nusgens BV, Deroanne CF. RhoA-GDP regulates RhoB protein stability. Potential involvement of RhoGDIalpha. J Biol Chem. 2008;283(31):21588–98. doi:10.1074/jbc.M710033200.CrossRefPubMed
23.
go back to reference Deroanne C, Vouret-Craviari V, Wang B, Pouyssegur J. EphrinA1 inactivates integrin-mediated vascular smooth muscle cell spreading via the Rac/PAK pathway. J Cell Sci. 2003;116(Pt 7):1367–76.CrossRefPubMed Deroanne C, Vouret-Craviari V, Wang B, Pouyssegur J. EphrinA1 inactivates integrin-mediated vascular smooth muscle cell spreading via the Rac/PAK pathway. J Cell Sci. 2003;116(Pt 7):1367–76.CrossRefPubMed
24.
go back to reference Avery-Kiejda KA, Zhang XD, Adams LJ, Scott RJ, Vojtesek B, Lane DP, et al. Small molecular weight variants of p53 are expressed in human melanoma cells and are induced by the DNA-damaging agent cisplatin. Clin Cancer Res. 2008;14(6):1659–68. doi:10.1158/1078-0432.ccr-07-1422.CrossRefPubMed Avery-Kiejda KA, Zhang XD, Adams LJ, Scott RJ, Vojtesek B, Lane DP, et al. Small molecular weight variants of p53 are expressed in human melanoma cells and are induced by the DNA-damaging agent cisplatin. Clin Cancer Res. 2008;14(6):1659–68. doi:10.1158/1078-0432.ccr-07-1422.CrossRefPubMed
25.
go back to reference Edmond V, Moysan E, Khochbin S, Matthias P, Brambilla C, Brambilla E, et al. Acetylation and phosphorylation of SRSF2 control cell fate decision in response to cisplatin. EMBO J. 2011;30(3):510–23. doi:10.1038/emboj.2010.333.CrossRefPubMed Edmond V, Moysan E, Khochbin S, Matthias P, Brambilla C, Brambilla E, et al. Acetylation and phosphorylation of SRSF2 control cell fate decision in response to cisplatin. EMBO J. 2011;30(3):510–23. doi:10.1038/emboj.2010.333.CrossRefPubMed
26.
go back to reference Muscella A, Urso L, Calabriso N, Ciccarese A, Migoni D, Fanizzi FP, et al. Differential response of normal, dedifferentiated and transformed thyroid cell lines to cisplatin treatment. Biochem Pharmacol. 2005;71(1–2):50–60. doi:10.1016/j.bcp.2005.10.022.CrossRefPubMed Muscella A, Urso L, Calabriso N, Ciccarese A, Migoni D, Fanizzi FP, et al. Differential response of normal, dedifferentiated and transformed thyroid cell lines to cisplatin treatment. Biochem Pharmacol. 2005;71(1–2):50–60. doi:10.1016/j.bcp.2005.10.022.CrossRefPubMed
27.
go back to reference Lu HP, Chao CC. Cancer cells acquire resistance to anticancer drugs: an update. Biomed J. 2012;35(6):464–72. doi:10.4103/2319-4170.104411.CrossRefPubMed Lu HP, Chao CC. Cancer cells acquire resistance to anticancer drugs: an update. Biomed J. 2012;35(6):464–72. doi:10.4103/2319-4170.104411.CrossRefPubMed
28.
go back to reference Boulikas T, Vougiouka M. Recent clinical trials using cisplatin, carboplatin and their combination chemotherapy drugs (review). Oncol Rep. 2004;11(3):559–95.PubMed Boulikas T, Vougiouka M. Recent clinical trials using cisplatin, carboplatin and their combination chemotherapy drugs (review). Oncol Rep. 2004;11(3):559–95.PubMed
29.
go back to reference Cobleigh MA. Other options in the treatment of advanced breast cancer. Semin Oncol. 2011;38 Suppl 2:S11–6. doi:10.1053/j.seminoncol.2011.04.005.CrossRefPubMed Cobleigh MA. Other options in the treatment of advanced breast cancer. Semin Oncol. 2011;38 Suppl 2:S11–6. doi:10.1053/j.seminoncol.2011.04.005.CrossRefPubMed
30.
go back to reference Shkreta L, Michelle L, Toutant J, Tremblay ML, Chabot B. The DNA damage response pathway regulates the alternative splicing of the apoptotic mediator Bcl-x. J Biol Chem. 2011;286(1):331–40. doi:10.1074/jbc.M110.162644.CrossRefPubMed Shkreta L, Michelle L, Toutant J, Tremblay ML, Chabot B. The DNA damage response pathway regulates the alternative splicing of the apoptotic mediator Bcl-x. J Biol Chem. 2011;286(1):331–40. doi:10.1074/jbc.M110.162644.CrossRefPubMed
31.
go back to reference Blaustein M, Pelisch F, Srebrow A. Signals, pathways and splicing regulation. Int J Biochem Cell Biol. 2007;39(11):2031–48. doi:10.1016/j.biocel.2007.04.004.CrossRefPubMed Blaustein M, Pelisch F, Srebrow A. Signals, pathways and splicing regulation. Int J Biochem Cell Biol. 2007;39(11):2031–48. doi:10.1016/j.biocel.2007.04.004.CrossRefPubMed
32.
go back to reference Eisenreich A, Malz R, Pepke W, Ayral Y, Poller W, Schultheiss HP, et al. Role of the phosphatidylinositol 3-kinase/protein kinase B pathway in regulating alternative splicing of tissue factor mRNA in human endothelial cells. Circ J. 2009;73(9):1746–52.CrossRefPubMed Eisenreich A, Malz R, Pepke W, Ayral Y, Poller W, Schultheiss HP, et al. Role of the phosphatidylinositol 3-kinase/protein kinase B pathway in regulating alternative splicing of tissue factor mRNA in human endothelial cells. Circ J. 2009;73(9):1746–52.CrossRefPubMed
33.
go back to reference Zhou Z, Qiu J, Liu W, Zhou Y, Plocinik RM, Li H, et al. The Akt-SRPK-SR axis constitutes a major pathway in transducing EGF signaling to regulate alternative splicing in the nucleus. Mol Cell. 2012;47(3):422–33. doi:10.1016/j.molcel.2012.05.014.CrossRefPubMedPubMedCentral Zhou Z, Qiu J, Liu W, Zhou Y, Plocinik RM, Li H, et al. The Akt-SRPK-SR axis constitutes a major pathway in transducing EGF signaling to regulate alternative splicing in the nucleus. Mol Cell. 2012;47(3):422–33. doi:10.1016/j.molcel.2012.05.014.CrossRefPubMedPubMedCentral
35.
go back to reference Martelli AM, Ognibene A, Buontempo F, Fini M, Bressanin D, Goto K, et al. Nuclear phosphoinositides and their roles in cell biology and disease. Crit Rev Biochem Mol Biol. 2011;46(5):436–57. doi:10.3109/10409238.2011.609530.CrossRefPubMed Martelli AM, Ognibene A, Buontempo F, Fini M, Bressanin D, Goto K, et al. Nuclear phosphoinositides and their roles in cell biology and disease. Crit Rev Biochem Mol Biol. 2011;46(5):436–57. doi:10.3109/10409238.2011.609530.CrossRefPubMed
36.
go back to reference Chen R, Kang VH, Chen J, Shope JC, Torabinejad J, DeWald DB, et al. A monoclonal antibody to visualize PtdIns(3,4,5)P(3) in cells. J Histochem Cytochem. 2002;50(5):697–708.CrossRefPubMed Chen R, Kang VH, Chen J, Shope JC, Torabinejad J, DeWald DB, et al. A monoclonal antibody to visualize PtdIns(3,4,5)P(3) in cells. J Histochem Cytochem. 2002;50(5):697–708.CrossRefPubMed
37.
go back to reference Anko ML, Muller-McNicoll M, Brandl H, Curk T, Gorup C, Henry I, et al. The RNA-binding landscapes of two SR proteins reveal unique functions and binding to diverse RNA classes. Genome Biol. 2012;13(3):R17. doi:10.1186/gb-2012-13-3-r17.CrossRefPubMedPubMedCentral Anko ML, Muller-McNicoll M, Brandl H, Curk T, Gorup C, Henry I, et al. The RNA-binding landscapes of two SR proteins reveal unique functions and binding to diverse RNA classes. Genome Biol. 2012;13(3):R17. doi:10.1186/gb-2012-13-3-r17.CrossRefPubMedPubMedCentral
38.
go back to reference Landau DA, Carter SL, Stojanov P, McKenna A, Stevenson K, Lawrence MS, et al. Evolution and impact of subclonal mutations in chronic lymphocytic leukemia. Cell. 2013;152(4):714–26. doi:10.1016/j.cell.2013.01.019.CrossRefPubMedPubMedCentral Landau DA, Carter SL, Stojanov P, McKenna A, Stevenson K, Lawrence MS, et al. Evolution and impact of subclonal mutations in chronic lymphocytic leukemia. Cell. 2013;152(4):714–26. doi:10.1016/j.cell.2013.01.019.CrossRefPubMedPubMedCentral
39.
go back to reference Rozovski U, Keating M, Estrov Z. The significance of spliceosome mutations in chronic lymphocytic leukemia. Leuk Lymphoma. 2013;54(7):1364–6. doi:10.3109/10428194.2012.742528.CrossRefPubMedPubMedCentral Rozovski U, Keating M, Estrov Z. The significance of spliceosome mutations in chronic lymphocytic leukemia. Leuk Lymphoma. 2013;54(7):1364–6. doi:10.3109/10428194.2012.742528.CrossRefPubMedPubMedCentral
40.
go back to reference Shultz JC, Goehe RW, Murudkar CS, Wijesinghe DS, Mayton EK, Massiello A, et al. SRSF1 regulates the alternative splicing of caspase 9 via a novel intronic splicing enhancer affecting the chemotherapeutic sensitivity of non-small cell lung cancer cells. Mol Cancer Res. 2011;9(7):889–900. doi:10.1158/1541-7786.mcr-11-0061.CrossRefPubMedPubMedCentral Shultz JC, Goehe RW, Murudkar CS, Wijesinghe DS, Mayton EK, Massiello A, et al. SRSF1 regulates the alternative splicing of caspase 9 via a novel intronic splicing enhancer affecting the chemotherapeutic sensitivity of non-small cell lung cancer cells. Mol Cancer Res. 2011;9(7):889–900. doi:10.1158/1541-7786.mcr-11-0061.CrossRefPubMedPubMedCentral
41.
go back to reference Kamihira S, Yamada Y, Tomonaga M, Sugahara K, Tsuruda K. Discrepant expression of membrane and soluble isoforms of Fas (CD95/APO-1) in adult T-cell leukaemia: soluble Fas isoform is an independent risk factor for prognosis. Br J Haematol. 1999;107(4):851–60.CrossRefPubMed Kamihira S, Yamada Y, Tomonaga M, Sugahara K, Tsuruda K. Discrepant expression of membrane and soluble isoforms of Fas (CD95/APO-1) in adult T-cell leukaemia: soluble Fas isoform is an independent risk factor for prognosis. Br J Haematol. 1999;107(4):851–60.CrossRefPubMed
42.
go back to reference Konno R, Takano T, Sato S, Yajima A. Serum soluble fas level as a prognostic factor in patients with gynecological malignancies. Clin Cancer Res. 2000;6(9):3576–80.PubMed Konno R, Takano T, Sato S, Yajima A. Serum soluble fas level as a prognostic factor in patients with gynecological malignancies. Clin Cancer Res. 2000;6(9):3576–80.PubMed
43.
go back to reference Eblen ST. Regulation of chemoresistance via alternative messenger RNA splicing. Biochem Pharmacol. 2012;83(8):1063–72. doi:10.1016/j.bcp.2011.12.041.CrossRefPubMedPubMedCentral Eblen ST. Regulation of chemoresistance via alternative messenger RNA splicing. Biochem Pharmacol. 2012;83(8):1063–72. doi:10.1016/j.bcp.2011.12.041.CrossRefPubMedPubMedCentral
44.
go back to reference Subramanian A, Tamayo P, Mootha VK, Mukherjee S, Ebert BL, Gillette MA, et al. Gene set enrichment analysis: a knowledge-based approach for interpreting genome-wide expression profiles. Proc Natl Acad Sci U S A. 2005;102(43):15545–50. doi:10.1073/pnas.0506580102.CrossRefPubMedPubMedCentral Subramanian A, Tamayo P, Mootha VK, Mukherjee S, Ebert BL, Gillette MA, et al. Gene set enrichment analysis: a knowledge-based approach for interpreting genome-wide expression profiles. Proc Natl Acad Sci U S A. 2005;102(43):15545–50. doi:10.1073/pnas.0506580102.CrossRefPubMedPubMedCentral
45.
go back to reference Mootha VK, Lindgren CM, Eriksson KF, Subramanian A, Sihag S, Lehar J, et al. PGC-1alpha-responsive genes involved in oxidative phosphorylation are coordinately downregulated in human diabetes. Nat Genet. 2003;34(3):267–73. doi:10.1038/ng1180.CrossRefPubMed Mootha VK, Lindgren CM, Eriksson KF, Subramanian A, Sihag S, Lehar J, et al. PGC-1alpha-responsive genes involved in oxidative phosphorylation are coordinately downregulated in human diabetes. Nat Genet. 2003;34(3):267–73. doi:10.1038/ng1180.CrossRefPubMed
46.
go back to reference Bonnal S, Vigevani L, Valcarcel J. The spliceosome as a target of novel antitumour drugs. Nat Rev Drug Discov. 2012;11(11):847–59. doi:10.1038/nrd3823.CrossRefPubMed Bonnal S, Vigevani L, Valcarcel J. The spliceosome as a target of novel antitumour drugs. Nat Rev Drug Discov. 2012;11(11):847–59. doi:10.1038/nrd3823.CrossRefPubMed
47.
go back to reference Webb TR, Joyner AS, Potter PM. The development and application of small molecule modulators of SF3b as therapeutic agents for cancer. Drug Discov Today. 2013;18(1–2):43–9. doi:10.1016/j.drudis.2012.07.013.CrossRefPubMed Webb TR, Joyner AS, Potter PM. The development and application of small molecule modulators of SF3b as therapeutic agents for cancer. Drug Discov Today. 2013;18(1–2):43–9. doi:10.1016/j.drudis.2012.07.013.CrossRefPubMed
48.
go back to reference Butler MS. Remediating cancer via splicing modulation. J Med Chem. 2013;56(17):6573–5. doi:10.1021/jm401289z.CrossRefPubMed Butler MS. Remediating cancer via splicing modulation. J Med Chem. 2013;56(17):6573–5. doi:10.1021/jm401289z.CrossRefPubMed
49.
go back to reference Gao Y, Vogt A, Forsyth CJ, Koide K. Comparison of splicing factor 3b inhibitors in human cells. Chembiochem. 2013;14(1):49–52. doi:10.1002/cbic.201200558.CrossRefPubMed Gao Y, Vogt A, Forsyth CJ, Koide K. Comparison of splicing factor 3b inhibitors in human cells. Chembiochem. 2013;14(1):49–52. doi:10.1002/cbic.201200558.CrossRefPubMed
50.
go back to reference Yokoi A, Kotake Y, Takahashi K, Kadowaki T, Matsumoto Y, Minoshima Y, et al. Biological validation that SF3b is a target of the antitumor macrolide pladienolide. FEBS J. 2011;278(24):4870–80. doi:10.1111/j.1742-4658.2011.08387.x.CrossRefPubMed Yokoi A, Kotake Y, Takahashi K, Kadowaki T, Matsumoto Y, Minoshima Y, et al. Biological validation that SF3b is a target of the antitumor macrolide pladienolide. FEBS J. 2011;278(24):4870–80. doi:10.1111/j.1742-4658.2011.08387.x.CrossRefPubMed
51.
go back to reference Chang WH, Liu TC, Yang WK, Lee CC, Lin YH, Chen TY, et al. Amiloride modulates alternative splicing in leukemic cells and resensitizes Bcr-AblT315I mutant cells to imatinib. Cancer Res. 2011;71(2):383–92. doi:10.1158/0008-5472.can-10-1037.CrossRefPubMed Chang WH, Liu TC, Yang WK, Lee CC, Lin YH, Chen TY, et al. Amiloride modulates alternative splicing in leukemic cells and resensitizes Bcr-AblT315I mutant cells to imatinib. Cancer Res. 2011;71(2):383–92. doi:10.1158/0008-5472.can-10-1037.CrossRefPubMed
52.
go back to reference Fang HY, Chen SB, Guo DJ, Pan SY, Yu ZL. Proteomic identification of differentially expressed proteins in curcumin-treated MCF-7 cells. Phytomedicine. 2011;18(8–9):697–703. doi:10.1016/j.phymed.2010.11.012.CrossRefPubMed Fang HY, Chen SB, Guo DJ, Pan SY, Yu ZL. Proteomic identification of differentially expressed proteins in curcumin-treated MCF-7 cells. Phytomedicine. 2011;18(8–9):697–703. doi:10.1016/j.phymed.2010.11.012.CrossRefPubMed
53.
go back to reference Markus MA, Marques FZ, Morris BJ. Resveratrol, by modulating RNA processing factor levels, can influence the alternative splicing of pre-mRNAs. PLoS One. 2011;6(12):e28926. doi:10.1371/journal.pone.0028926.CrossRefPubMedPubMedCentral Markus MA, Marques FZ, Morris BJ. Resveratrol, by modulating RNA processing factor levels, can influence the alternative splicing of pre-mRNAs. PLoS One. 2011;6(12):e28926. doi:10.1371/journal.pone.0028926.CrossRefPubMedPubMedCentral
54.
go back to reference Kim MH. Protein phosphatase 1 activation and alternative splicing of Bcl-X and Mcl-1 by EGCG + ibuprofen. J Cell Biochem. 2008;104(4):1491–9. doi:10.1002/jcb.21725.CrossRefPubMed Kim MH. Protein phosphatase 1 activation and alternative splicing of Bcl-X and Mcl-1 by EGCG + ibuprofen. J Cell Biochem. 2008;104(4):1491–9. doi:10.1002/jcb.21725.CrossRefPubMed
55.
go back to reference Jiang M, Huang O, Zhang X, Xie Z, Shen A, Liu H, et al. Curcumin induces cell death and restores tamoxifen sensitivity in the antiestrogen-resistant breast cancer cell lines MCF-7/LCC2 and MCF-7/LCC9. Molecules (Basel, Switzerland). 2013;18(1):701–20. doi:10.3390/molecules18010701.CrossRef Jiang M, Huang O, Zhang X, Xie Z, Shen A, Liu H, et al. Curcumin induces cell death and restores tamoxifen sensitivity in the antiestrogen-resistant breast cancer cell lines MCF-7/LCC2 and MCF-7/LCC9. Molecules (Basel, Switzerland). 2013;18(1):701–20. doi:10.3390/molecules18010701.CrossRef
Metadata
Title
Role of the splicing factor SRSF4 in cisplatin-induced modifications of pre-mRNA splicing and apoptosis
Authors
Maude Gabriel
Yves Delforge
Adeline Deward
Yvette Habraken
Benoit Hennuy
Jacques Piette
Roscoe Klinck
Benoit Chabot
Alain Colige
Charles Lambert
Publication date
01-12-2015
Publisher
BioMed Central
Published in
BMC Cancer / Issue 1/2015
Electronic ISSN: 1471-2407
DOI
https://doi.org/10.1186/s12885-015-1259-0

Other articles of this Issue 1/2015

BMC 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