How Driver Oncogenes Shape and Are Shaped by Alternative Splicing Mechanisms in Tumors
Abstract
:Simple Summary
Abstract
1. Introduction
2. Alternative Splicing under the Control of Driver Oncogenes
2.1. CMYC
2.2. RAS and Downstream Pathways
2.3. Mutant p53
3. Driver Oncogenes Regulated by Alternative Splicing
3.1. TP53 Regulation by Splicing
3.2. CMYC
3.3. KRAS and Downstream Pathways
3.4. EGFR and FGFR
4. SR and hnRNP Families of Splicing Factors: The Oncogenes That Regulate and Are Regulated by Alternative Splicing
5. Therapeutic Implications
5.1. Splicing Inhibitors
5.2. Targeting of Specific Isoforms
5.3. Resistance to Targeted Therapies
6. Conclusions
Author Contributions
Funding
Acknowledgments
Conflicts of Interest
References
- Matlin, A.J.; Clark, F.; Smith, C.W. Understanding alternative splicing: Towards a cellular code. Nat. Rev. Mol. Cell. Biol. 2005, 6, 386–398. [Google Scholar] [CrossRef]
- Blencowe, B.J. Alternative splicing: New insights from global analyses. Cell 2006, 126, 37–47. [Google Scholar] [CrossRef]
- Pan, Q.; Shai, O.; Lee, L.J.; Frey, B.J.; Blencowe, B.J. Deep surveying of alternative splicing complexity in the human transcriptome by high-throughput sequencing. Nat. Genet. 2008, 40, 1413–1415. [Google Scholar] [CrossRef]
- Baralle, F.E.; Giudice, J. Alternative splicing as a regulator of development and tissue identity. Nat. Rev. Mol. Cell. Biol. 2017, 18, 437–451. [Google Scholar] [CrossRef]
- Singh, A.; Rajeevan, A.; Gopalan, V.; Agrawal, P.; Day, C.P.; Hannenhalli, S. Broad misappropriation of developmental splicing profile by cancer in multiple organs. Nat. Commun. 2022, 13, 7664. [Google Scholar] [CrossRef]
- Xiong, W.; Gao, D.; Li, Y.; Liu, X.; Dai, P.; Qin, J.; Wang, G.; Li, K.; Bai, H.; Li, W. Genome-wide profiling of chemoradiationinduced changes in alternative splicing in colon cancer cells. Oncol. Rep. 2016, 36, 2142–2150. [Google Scholar] [CrossRef]
- Yang, C.; Wu, Q.; Huang, K.; Wang, X.; Yu, T.; Liao, X.; Huang, J.; Zhu, G.; Gong, Y.; Han, C.; et al. Genome-Wide Profiling Reveals the Landscape of Prognostic Alternative Splicing Signatures in Pancreatic Ductal Adenocarcinoma. Front. Oncol. 2019, 9, 511. [Google Scholar] [CrossRef] [PubMed]
- Danan-Gotthold, M.; Golan-Gerstl, R.; Eisenberg, E.; Meir, K.; Karni, R.; Levanon, E.Y. Identification of recurrent regulated alternative splicing events across human solid tumors. Nucleic Acids Res. 2015, 43, 5130–5144. [Google Scholar] [CrossRef] [PubMed]
- Oltean, S.; Bates, D.O. Hallmarks of alternative splicing in cancer. Oncogene 2014, 33, 5311–5318. [Google Scholar] [CrossRef] [PubMed]
- Ladomery, M. Aberrant alternative splicing is another hallmark of cancer. Int. J. Cell Biol. 2013, 2013, 463786. [Google Scholar] [CrossRef] [PubMed]
- Anczukow, O.; Krainer, A.R. Splicing-factor alterations in cancers. RNA 2016, 22, 1285–1301. [Google Scholar] [CrossRef] [PubMed]
- Urbanski, L.M.; Leclair, N.; Anczuków, O. Alternative-splicing defects in cancer: Splicing regulators and their downstream targets, guiding the way to novel cancer therapeutics. Wiley Interdiscip. Rev. RNA 2018, 9, e1476. [Google Scholar] [CrossRef] [PubMed]
- de la Mata, M.; Alonso, C.R.; Kadener, S.; Fededa, J.P.; Blaustein, M.; Pelisch, F.; Cramer, P.; Bentley, D.; Kornblihtt, A.R. A slow RNA polymerase II affects alternative splicing in vivo. Mol. Cell 2003, 12, 525–532. [Google Scholar] [CrossRef] [PubMed]
- Shukla, S.; Oberdoerffer, S. Co-transcriptional regulation of alternative pre-mRNA splicing. Biochim. Biophys. Acta 2012, 1819, 673–683. [Google Scholar] [CrossRef] [PubMed]
- Lareau, L.F.; Inada, M.; Green, R.E.; Wengrod, J.C.; Brenner, S.E. Unproductive splicing of SR genes associated with highly conserved and ultraconserved DNA elements. Nature 2007, 446, 926–929. [Google Scholar] [CrossRef] [PubMed]
- Corkery, D.P.; Holly, A.C.; Lahsaee, S.; Dellaire, G. Connecting the speckles: Splicing kinases and their role in tumorigenesis and treatment response. Nucleus 2015, 6, 279–288. [Google Scholar] [CrossRef]
- Aubol, B.E.; Plocinik, R.M.; Hagopian, J.C.; Ma, C.T.; McGlone, M.L.; Bandyopadhyay, R.; Fu, X.D.; Adams, J.A. Partitioning RS domain phosphorylation in an SR protein through the CLK and SRPK protein kinases. J. Mol. Biol. 2013, 425, 2894–2909. [Google Scholar] [CrossRef]
- Dvinge, H.; Guenthoer, J.; Porter, P.L.; Bradley, R.K. RNA components of the spliceosome regulate tissue- and cancer-specific alternative splicing. Genome Res. 2019, 29, 1591–1604. [Google Scholar] [CrossRef]
- Duffy, M.J.; Tang, M.; Rajaram, S.; O’Grady, S.; Crown, J. Targeting Mutant p53 for Cancer Treatment: Moving Closer to Clinical Use? Cancers 2022, 14, 4499. [Google Scholar] [CrossRef]
- Nagasaka, M.; Potugari, B.; Nguyen, A.; Sukari, A.; Azmi, A.S.; Ou, S.I. KRAS Inhibitors- yes but what next? Direct targeting of KRAS- vaccines, adoptive T cell therapy and beyond. Cancer Treat. Rev. 2021, 101, 102309. [Google Scholar] [CrossRef]
- Garralda, E.; Moreno, V.; Alonso, G.; Corral, E.; Hernandez-Guerrero, T.; Ramon, J.; de Spéville, B.D.; Martinez, E.; Soucek, L.; Niewel, M.; et al. Dose escalation study of OMO-103, a first in class Pan-MYC-Inhibitor in patients (pts) with advanced solid tumors. Eur. J. Cancer 2022, 174, S5–S6. [Google Scholar] [CrossRef]
- Jin, H.; Wang, L.; Bernards, R. Rational combinations of targeted cancer therapies: Background, advances and challenges. Nat. Rev. Drug Discov. 2022, 22, 213–234. [Google Scholar] [CrossRef]
- Kress, T.R.; Sabo, A.; Amati, B. MYC: Connecting selective transcriptional control to global RNA production. Nat. Rev. Cancer 2015, 15, 593–607. [Google Scholar] [CrossRef]
- Spencer, C.A.; Groudine, M. Control of c-myc regulation in normal and neoplastic cells. Adv. Cancer Res. 1991, 56, 1–48. [Google Scholar] [CrossRef]
- Kalkat, M.; De Melo, J.; Hickman, K.A.; Lourenco, C.; Redel, C.; Resetca, D.; Tamachi, A.; Tu, W.B.; Penn, L.Z. MYC Deregulation in Primary Human Cancers. Genes 2017, 8, 151. [Google Scholar] [CrossRef]
- Gabay, M.; Li, Y.; Felsher, D.W. MYC activation is a hallmark of cancer initiation and maintenance. Cold Spring Harb. Perspect. Med. 2014, 4, a014241. [Google Scholar] [CrossRef]
- Grzes, M.; Oron, M.; Staszczak, Z.; Jaiswar, A.; Nowak-Niezgoda, M.; Walerych, D. A Driver Never Works Alone-Interplay Networks of Mutant p53, MYC, RAS, and Other Universal Oncogenic Drivers in Human Cancer. Cancers 2020, 12, 1532. [Google Scholar] [CrossRef]
- Van Dang, C.; McMahon, S.B. Emerging Concepts in the Analysis of Transcriptional Targets of the MYC Oncoprotein: Are the Targets Targetable? Genes Cancer 2010, 1, 560–567. [Google Scholar] [CrossRef]
- Hsu, T.Y.; Simon, L.M.; Neill, N.J.; Marcotte, R.; Sayad, A.; Bland, C.S.; Echeverria, G.V.; Sun, T.; Kurley, S.J.; Tyagi, S.; et al. The spliceosome is a therapeutic vulnerability in MYC-driven cancer. Nature 2015, 525, 384–388. [Google Scholar] [CrossRef]
- Koh, C.M.; Bezzi, M.; Low, D.H.; Ang, W.X.; Teo, S.X.; Gay, F.P.; Al-Haddawi, M.; Tan, S.Y.; Osato, M.; Sabo, A.; et al. MYC regulates the core pre-mRNA splicing machinery as an essential step in lymphomagenesis. Nature 2015, 523, 96–100. [Google Scholar] [CrossRef]
- Ciesla, M.; Ngoc, P.C.T.; Cordero, E.; Martinez, A.S.; Morsing, M.; Muthukumar, S.; Beneventi, G.; Madej, M.; Munita, R.; Jonsson, T.; et al. Oncogenic translation directs spliceosome dynamics revealing an integral role for SF3A3 in breast cancer. Mol. Cell 2021, 81, 1453–1468. [Google Scholar] [CrossRef]
- Meister, G.; Eggert, C.; Buhler, D.; Brahms, H.; Kambach, C.; Fischer, U. Methylation of Sm proteins by a complex containing PRMT5 and the putative U snRNP assembly factor pICln. Curr. Biol. 2001, 11, 1990–1994. [Google Scholar] [CrossRef] [PubMed]
- Das, S.; Anczukow, O.; Akerman, M.; Krainer, A.R. Oncogenic splicing factor SRSF1 is a critical transcriptional target of MYC. Cell Rep. 2012, 1, 110–117. [Google Scholar] [CrossRef]
- Urbanski, L.; Brugiolo, M.; Park, S.; Angarola, B.L.; Leclair, N.K.; Yurieva, M.; Palmer, P.; Sahu, S.K.; Anczukow, O. MYC regulates a pan-cancer network of co-expressed oncogenic splicing factors. Cell Rep. 2022, 41, 111704. [Google Scholar] [CrossRef]
- Rauch, J.; Moran-Jones, K.; Albrecht, V.; Schwarzl, T.; Hunter, K.; Gires, O.; Kolch, W. c-Myc regulates RNA splicing of the A-Raf kinase and its activation of the ERK pathway. Cancer Res. 2011, 71, 4664–4674. [Google Scholar] [CrossRef]
- David, C.J.; Chen, M.; Assanah, M.; Canoll, P.; Manley, J.L. HnRNP proteins controlled by c-Myc deregulate pyruvate kinase mRNA splicing in cancer. Nature 2010, 463, 364–368. [Google Scholar] [CrossRef]
- Zhang, S.; Wei, J.S.; Li, S.Q.; Badgett, T.C.; Song, Y.K.; Agarwal, S.; Coarfa, C.; Tolman, C.; Hurd, L.; Liao, H.; et al. MYCN controls an alternative RNA splicing program in high-risk metastatic neuroblastoma. Cancer Lett. 2016, 371, 214–224. [Google Scholar] [CrossRef]
- Baluapuri, A.; Hofstetter, J.; Dudvarski Stankovic, N.; Endres, T.; Bhandare, P.; Vos, S.M.; Adhikari, B.; Schwarz, J.D.; Narain, A.; Vogt, M.; et al. MYC Recruits SPT5 to RNA Polymerase II to Promote Processive Transcription Elongation. Mol. Cell 2019, 74, 674–687. [Google Scholar] [CrossRef]
- Girard, C.; Will, C.L.; Peng, J.; Makarov, E.M.; Kastner, B.; Lemm, I.; Urlaub, H.; Hartmuth, K.; Luhrmann, R. Post-transcriptional spliceosomes are retained in nuclear speckles until splicing completion. Nat. Commun. 2012, 3, 994. [Google Scholar] [CrossRef]
- Caggiano, C.; Pieraccioli, M.; Panzeri, V.; Sette, C.; Bielli, P. c-MYC empowers transcription and productive splicing of the oncogenic splicing factor Sam68 in cancer. Nucleic Acids Res. 2019, 47, 6160–6171. [Google Scholar] [CrossRef]
- Phillips, J.W.; Pan, Y.; Tsai, B.L.; Xie, Z.; Demirdjian, L.; Xiao, W.; Yang, H.T.; Zhang, Y.; Lin, C.H.; Cheng, D.; et al. Pathway-guided analysis identifies Myc-dependent alternative pre-mRNA splicing in aggressive prostate cancers. Proc. Natl. Acad. Sci. USA 2020, 117, 5269–5279. [Google Scholar] [CrossRef] [PubMed]
- Prior, I.A.; Hood, F.E.; Hartley, J.L. The Frequency of Ras Mutations in Cancer. Cancer Res. 2020, 80, 2969–2974. [Google Scholar] [CrossRef] [PubMed]
- Prior, I.A.; Lewis, P.D.; Mattos, C. A comprehensive survey of Ras mutations in cancer. Cancer Res. 2012, 72, 2457–2467. [Google Scholar] [CrossRef]
- Gimple, R.C.; Wang, X. RAS: Striking at the Core of the Oncogenic Circuitry. Front. Oncol. 2019, 9, 965. [Google Scholar] [CrossRef] [PubMed]
- Lo, A.; McSharry, M.; Berger, A.H. Oncogenic KRAS alters splicing factor phosphorylation and alternative splicing in lung cancer. BMC Cancer 2022, 22, 1315. [Google Scholar] [CrossRef]
- Naro, C.; Sette, C. Phosphorylation-mediated regulation of alternative splicing in cancer. Int. J. Cell Biol. 2013, 2013, 151839. [Google Scholar] [CrossRef]
- Zhou, Z.; Qiu, J.; Liu, W.; Zhou, Y.; Plocinik, R.M.; Li, H.; Hu, Q.; Ghosh, G.; Adams, J.A.; Rosenfeld, M.G.; 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, 422–433. [Google Scholar] [CrossRef]
- Yea, S.; Narla, G.; Zhao, X.; Garg, R.; Tal-Kremer, S.; Hod, E.; Villanueva, A.; Loke, J.; Tarocchi, M.; Akita, K.; et al. Ras promotes growth by alternative splicing-mediated inactivation of the KLF6 tumor suppressor in hepatocellular carcinoma. Gastroenterology 2008, 134, 1521–1531. [Google Scholar] [CrossRef]
- Hu, K.; Zheng, Q.K.; Ma, R.J.; Ma, C.; Sun, Z.G.; Zhang, N. Kruppel-Like Factor 6 Splice Variant 1: An Oncogenic Transcription Factor Involved in the Progression of Multiple Malignant Tumors. Front. Cell Dev. Biol. 2021, 9, 661731. [Google Scholar] [CrossRef]
- Munoz, U.; Puche, J.E.; Hannivoort, R.; Lang, U.E.; Cohen-Naftaly, M.; Friedman, S.L. Hepatocyte growth factor enhances alternative splicing of the Kruppel-like factor 6 (KLF6) tumor suppressor to promote growth through SRSF1. Mol. Cancer Res. 2012, 10, 1216–1227. [Google Scholar] [CrossRef]
- Cheng, C.; Yaffe, M.B.; Sharp, P.A. A positive feedback loop couples Ras activation and CD44 alternative splicing. Genes Dev. 2006, 20, 1715–1720. [Google Scholar] [CrossRef] [PubMed]
- Matter, N.; Herrlich, P.; Konig, H. Signal-dependent regulation of splicing via phosphorylation of Sam68. Nature 2002, 420, 691–695. [Google Scholar] [CrossRef] [PubMed]
- Frisone, P.; Pradella, D.; Di Matteo, A.; Belloni, E.; Ghigna, C.; Paronetto, M.P. SAM68: Signal Transduction and RNA Metabolism in Human Cancer. BioMed Res. Int. 2015, 2015, 528954. [Google Scholar] [CrossRef] [PubMed]
- Walerych, D.; Napoli, M.; Collavin, L.; Del Sal, G. The rebel angel: Mutant p53 as the driving oncogene in breast cancer. Carcinogenesis 2012, 33, 2007–2017. [Google Scholar] [CrossRef]
- Oren, M.; Rotter, V. Mutant p53 gain-of-function in cancer. Cold Spring Harb. Perspect. Biol. 2010, 2, a001107. [Google Scholar] [CrossRef]
- Schilling, T.; Kairat, A.; Melino, G.; Krammer, P.H.; Stremmel, W.; Oren, M.; Muller, M. Interference with the p53 family network contributes to the gain of oncogenic function of mutant p53 in hepatocellular carcinoma. Biochem. Biophys. Res. Commun. 2010, 394, 817–823. [Google Scholar] [CrossRef]
- Fontemaggi, G.; Dell’Orso, S.; Trisciuoglio, D.; Shay, T.; Melucci, E.; Fazi, F.; Terrenato, I.; Mottolese, M.; Muti, P.; Domany, E.; et al. The execution of the transcriptional axis mutant p53, E2F1 and ID4 promotes tumor neo-angiogenesis. Nat. Struct. Mol. Biol. 2009, 16, 1086–1093. [Google Scholar] [CrossRef]
- Pruszko, M.; Milano, E.; Forcato, M.; Donzelli, S.; Ganci, F.; Di Agostino, S.; De Panfilis, S.; Fazi, F.; Bates, D.O.; Bicciato, S.; et al. The mutant p53-ID4 complex controls VEGFA isoforms by recruiting lncRNA MALAT1. EMBO Rep. 2017, 18, 1331–1351. [Google Scholar] [CrossRef]
- Escobar-Hoyos, L.F.; Penson, A.; Kannan, R.; Cho, H.; Pan, C.H.; Singh, R.K.; Apken, L.H.; Hobbs, G.A.; Luo, R.; Lecomte, N.; et al. Altered RNA Splicing by Mutant p53 Activates Oncogenic RAS Signaling in Pancreatic Cancer. Cancer Cell 2020, 38, 198–211. [Google Scholar] [CrossRef]
- Pruszko, M.; Milano, E.; Zylicz, A.; Zylicz, M.; Blandino, G.; Fontemaggi, G. Zebrafish as experimental model to establish the contribution of mutant p53 and ID4 to breast cancer angiogenesis in vivo. J. Thorac. Dis. 2018, 10, E231–E233. [Google Scholar] [CrossRef]
- Surget, S.; Khoury, M.P.; Bourdon, J.C. Uncovering the role of p53 splice variants in human malignancy: A clinical perspective. OncoTargets Ther. 2013, 7, 57–68. [Google Scholar] [CrossRef]
- Tang, Y.; Horikawa, I.; Ajiro, M.; Robles, A.I.; Fujita, K.; Mondal, A.M.; Stauffer, J.K.; Zheng, Z.M.; Harris, C.C. Downregulation of splicing factor SRSF3 induces p53beta, an alternatively spliced isoform of p53 that promotes cellular senescence. Oncogene 2013, 32, 2792–2798. [Google Scholar] [CrossRef]
- Marcel, V.; Fernandes, K.; Terrier, O.; Lane, D.P.; Bourdon, J.C. Modulation of p53beta and p53gamma expression by regulating the alternative splicing of TP53 gene modifies cellular response. Cell Death Differ. 2014, 21, 1377–1387. [Google Scholar] [CrossRef] [PubMed]
- Chen, J.; Crutchley, J.; Zhang, D.; Owzar, K.; Kastan, M.B. Identification of a DNA Damage-Induced Alternative Splicing Pathway That Regulates p53 and Cellular Senescence Markers. Cancer Discov. 2017, 7, 766–781. [Google Scholar] [CrossRef] [PubMed]
- Steffens Reinhardt, L.; Zhang, X.; Wawruszak, A.; Groen, K.; De Iuliis, G.N.; Avery-Kiejda, K.A. Good Cop, Bad Cop: Defining the Roles of Delta40p53 in Cancer and Aging. Cancers 2020, 12, 1659. [Google Scholar] [CrossRef]
- Joruiz, S.M.; Beck, J.A.; Horikawa, I.; Harris, C.C. The Delta133p53 Isoforms, Tuners of the p53 Pathway. Cancers 2020, 12, 3422. [Google Scholar] [CrossRef]
- Solomon, H.; Sharon, M.; Rotter, V. Modulation of alternative splicing contributes to cancer development: Focusing on p53 isoforms, p53beta and p53gamma. Cell Death Differ. 2014, 21, 1347–1349. [Google Scholar] [CrossRef]
- Bourdon, J.C.; Khoury, M.P.; Diot, A.; Baker, L.; Fernandes, K.; Aoubala, M.; Quinlan, P.; Purdie, C.A.; Jordan, L.B.; Prats, A.C.; et al. p53 mutant breast cancer patients expressing p53gamma have as good a prognosis as wild-type p53 breast cancer patients. Breast Cancer Res. 2011, 13, R7. [Google Scholar] [CrossRef]
- Avery-Kiejda, K.A.; Morten, B.; Wong-Brown, M.W.; Mathe, A.; Scott, R.J. The relative mRNA expression of p53 isoforms in breast cancer is associated with clinical features and outcome. Carcinogenesis 2014, 35, 586–596. [Google Scholar] [CrossRef]
- Oh, L.; Hainaut, P.; Blanchet, S.; Ariffin, H. Expression of p53 N-terminal isoforms in B-cell precursor acute lymphoblastic leukemia and its correlation with clinicopathological profiles. BMC Cancer 2020, 20, 110. [Google Scholar] [CrossRef]
- Hofstetter, G.; Berger, A.; Berger, R.; Zoric, A.; Braicu, E.I.; Reimer, D.; Fiegl, H.; Marth, C.; Zeimet, A.G.; Ulmer, H.; et al. The N-terminally truncated p53 isoform Delta40p53 influences prognosis in mucinous ovarian cancer. Int. J. Gynecol. Cancer 2012, 22, 372–379. [Google Scholar] [CrossRef] [PubMed]
- Avery-Kiejda, K.A.; Zhang, X.D.; Adams, L.J.; Scott, R.J.; Vojtesek, B.; Lane, D.P.; Hersey, P. 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, 1659–1668. [Google Scholar] [CrossRef]
- Hafsi, H.; Santos-Silva, D.; Courtois-Cox, S.; Hainaut, P. Effects of Delta40p53, an isoform of p53 lacking the N-terminus, on transactivation capacity of the tumor suppressor protein p53. BMC Cancer 2013, 13, 134. [Google Scholar] [CrossRef]
- Kitayner, M.; Rozenberg, H.; Kessler, N.; Rabinovich, D.; Shaulov, L.; Haran, T.E.; Shakked, Z. Structural basis of DNA recognition by p53 tetramers. Mol. Cell 2006, 22, 741–753. [Google Scholar] [CrossRef] [PubMed]
- Sun, Y.; Manceau, A.; Frydman, L.; Cappuccio, L.; Neves, D.; Basso, V.; Wang, H.; Fombonne, J.; Maisse, C.; Mehlen, P.; et al. Delta40p53 isoform up-regulates netrin-1/UNC5B expression and potentiates netrin-1 pro-oncogenic activity. Proc. Natl. Acad. Sci. USA 2021, 118, e2103319118. [Google Scholar] [CrossRef] [PubMed]
- Ungewitter, E.; Scrable, H. Delta40p53 controls the switch from pluripotency to differentiation by regulating IGF signaling in ESCs. Genes Dev. 2010, 24, 2408–2419. [Google Scholar] [CrossRef]
- Gong, L.; Gong, H.; Pan, X.; Chang, C.; Ou, Z.; Ye, S.; Yin, L.; Yang, L.; Tao, T.; Zhang, Z.; et al. p53 isoform Delta113p53/Delta133p53 promotes DNA double-strand break repair to protect cell from death and senescence in response to DNA damage. Cell Res. 2015, 25, 351–369. [Google Scholar] [CrossRef]
- Kazantseva, M.; Mehta, S.; Eiholzer, R.A.; Gimenez, G.; Bowie, S.; Campbell, H.; Reily-Bell, A.L.; Roth, I.; Ray, S.; Drummond, C.J.; et al. The Delta133p53beta isoform promotes an immunosuppressive environment leading to aggressive prostate cancer. Cell Death Dis. 2019, 10, 631. [Google Scholar] [CrossRef]
- Ozretic, P.; Hanzic, N.; Proust, B.; Sabol, M.; Trnski, D.; Radic, M.; Musani, V.; Ciribilli, Y.; Milas, I.; Puljiz, Z.; et al. Expression profiles of p53/p73, NME and GLI families in metastatic melanoma tissue and cell lines. Sci. Rep. 2019, 9, 12470. [Google Scholar] [CrossRef]
- Fujita, K.; Mondal, A.M.; Horikawa, I.; Nguyen, G.H.; Kumamoto, K.; Sohn, J.J.; Bowman, E.D.; Mathe, E.A.; Schetter, A.J.; Pine, S.R.; et al. p53 isoforms Delta133p53 and p53beta are endogenous regulators of replicative cellular senescence. Nat. Cell Biol. 2009, 11, 1135–1142. [Google Scholar] [CrossRef]
- Kazantseva, M.; Eiholzer, R.A.; Mehta, S.; Taha, A.; Bowie, S.; Roth, I.; Zhou, J.; Joruiz, S.M.; Royds, J.A.; Hung, N.A.; et al. Elevation of the TP53 isoform Delta133p53beta in glioblastomas: An alternative to mutant p53 in promoting tumor development. J. Pathol. 2018, 246, 77–88. [Google Scholar] [CrossRef] [PubMed]
- Gadea, G.; Arsic, N.; Fernandes, K.; Diot, A.; Joruiz, S.M.; Abdallah, S.; Meuray, V.; Vinot, S.; Anguille, C.; Remenyi, J.; et al. TP53 drives invasion through expression of its Delta133p53beta variant. Elife 2016, 5, e14734. [Google Scholar] [CrossRef]
- Campbell, H.; Fleming, N.; Roth, I.; Mehta, S.; Wiles, A.; Williams, G.; Vennin, C.; Arsic, N.; Parkin, A.; Pajic, M.; et al. ∆133p53 isoform promotes tumour invasion and metastasis via interleukin-6 activation of JAK-STAT and RhoA-ROCK signalling. Nat. Commun. 2018, 9, 254. [Google Scholar] [CrossRef] [PubMed]
- Bernard, H.; Garmy-Susini, B.; Ainaoui, N.; Van Den Berghe, L.; Peurichard, A.; Javerzat, S.; Bikfalvi, A.; Lane, D.P.; Bourdon, J.C.; Prats, A.C. The p53 isoform, Delta133p53alpha, stimulates angiogenesis and tumour progression. Oncogene 2013, 32, 2150–2160. [Google Scholar] [CrossRef]
- Aoubala, M.; Murray-Zmijewski, F.; Khoury, M.P.; Fernandes, K.; Perrier, S.; Bernard, H.; Prats, A.C.; Lane, D.P.; Bourdon, J.C. p53 directly transactivates Delta133p53alpha, regulating cell fate outcome in response to DNA damage. Cell Death Differ. 2011, 18, 248–258. [Google Scholar] [CrossRef] [PubMed]
- Candeias, M.M.; Hagiwara, M.; Matsuda, M. Cancer-specific mutations in p53 induce the translation of Delta160p53 promoting tumorigenesis. EMBO Rep. 2016, 17, 1542–1551. [Google Scholar] [CrossRef]
- Marcel, V.; Perrier, S.; Aoubala, M.; Ageorges, S.; Groves, M.J.; Diot, A.; Fernandes, K.; Tauro, S.; Bourdon, J.C. Delta160p53 is a novel N-terminal p53 isoform encoded by Delta133p53 transcript. FEBS Lett. 2010, 584, 4463–4468. [Google Scholar] [CrossRef]
- Tadijan, A.; Precazzini, F.; Hanzic, N.; Radic, M.; Gavioli, N.; Vlasic, I.; Ozretic, P.; Pinto, L.; Skreblin, L.; Barban, G.; et al. Altered Expression of Shorter p53 Family Isoforms Can Impact Melanoma Aggressiveness. Cancers 2021, 13, 5231. [Google Scholar] [CrossRef] [PubMed]
- Lai, M.Y.; Chang, H.C.; Li, H.P.; Ku, C.K.; Chen, P.J.; Sheu, J.C.; Huang, G.T.; Lee, P.H.; Chen, D.S. Splicing mutations of the p53 gene in human hepatocellular carcinoma. Cancer Res. 1993, 53, 1653–1656. [Google Scholar]
- Austin, F.; Oyarbide, U.; Massey, G.; Grimes, M.; Corey, S.J. Synonymous mutation in TP53 results in a cryptic splice site affecting its DNA-binding site in an adolescent with two primary sarcomas. Pediatr. Blood Cancer 2017, 64, e26584. [Google Scholar] [CrossRef]
- Eicheler, W.; Zips, D.; Dorfler, A.; Grenman, R.; Baumann, M. Splicing mutations in TP53 in human squamous cell carcinoma lines influence immunohistochemical detection. J. Histochem. Cytochem. 2002, 50, 197–204. [Google Scholar] [CrossRef] [PubMed]
- Smeby, J.; Sveen, A.; Eilertsen, I.A.; Danielsen, S.A.; Hoff, A.M.; Eide, P.W.; Johannessen, B.; Hektoen, M.; Skotheim, R.I.; Guren, M.G.; et al. Transcriptional and functional consequences of TP53 splice mutations in colorectal cancer. Oncogenesis 2019, 8, 35. [Google Scholar] [CrossRef] [PubMed]
- Holmila, R.; Fouquet, C.; Cadranel, J.; Zalcman, G.; Soussi, T. Splice mutations in the p53 gene: Case report and review of the literature. Hum. Mutat. 2003, 21, 101–102. [Google Scholar] [CrossRef] [PubMed]
- Bartel, F.; Pinkert, D.; Fiedler, W.; Kappler, M.; Wurl, P.; Schmidt, H.; Taubert, H. Expression of alternatively and aberrantly spliced transcripts of the MDM2 mRNA is not tumor-specific. Int. J. Oncol. 2004, 24, 143–151. [Google Scholar] [CrossRef]
- Inoue, K.; Fry, E.A. Aberrant splicing of the DMP1-ARF-MDM2-p53 pathway in cancer. Int. J. Cancer 2016, 139, 33–41. [Google Scholar] [CrossRef]
- Lukashchuk, N.; Vousden, K.H. Ubiquitination and degradation of mutant p53. Mol. Cell. Biol. 2007, 27, 8284–8295. [Google Scholar] [CrossRef]
- Traweek, R.S.; Cope, B.M.; Roland, C.L.; Keung, E.Z.; Nassif, E.F.; Erstad, D.J. Targeting the MDM2-p53 pathway in dedifferentiated liposarcoma. Front. Oncol. 2022, 12, 1006959. [Google Scholar] [CrossRef]
- Momand, J.; Jung, D.; Wilczynski, S.; Niland, J. The MDM2 gene amplification database. Nucleic Acids Res. 1998, 26, 3453–3459. [Google Scholar] [CrossRef]
- Zheng, T.; Wang, J.; Zhao, Y.; Zhang, C.; Lin, M.; Wang, X.; Yu, H.; Liu, L.; Feng, Z.; Hu, W. Spliced MDM2 isoforms promote mutant p53 accumulation and gain-of-function in tumorigenesis. Nat. Commun. 2013, 4, 2996. [Google Scholar] [CrossRef]
- Wiech, M.; Olszewski, M.B.; Tracz-Gaszewska, Z.; Wawrzynow, B.; Zylicz, M.; Zylicz, A. Molecular mechanism of mutant p53 stabilization: The role of HSP70 and MDM2. PLoS ONE 2012, 7, e51426. [Google Scholar] [CrossRef]
- Comiskey, D.F., Jr.; Montes, M.; Khurshid, S.; Singh, R.K.; Chandler, D.S. SRSF2 Regulation of MDM2 Reveals Splicing as a Therapeutic Vulnerability of the p53 Pathway. Mol. Cancer Res. 2020, 18, 194–203. [Google Scholar] [CrossRef] [PubMed]
- Comiskey, D.F., Jr.; Jacob, A.G.; Singh, R.K.; Tapia-Santos, A.S.; Chandler, D.S. Splicing factor SRSF1 negatively regulates alternative splicing of MDM2 under damage. Nucleic Acids Res. 2015, 43, 4202–4218. [Google Scholar] [CrossRef] [PubMed]
- Aptullahoglu, E.; Ciardullo, C.; Wallis, J.P.; Marr, H.; Marshall, S.; Bown, N.; Willmore, E.; Lunec, J. Splicing Modulation Results in Aberrant Isoforms and Protein Products of p53 Pathway Genes and the Sensitization of B Cells to Non-Genotoxic MDM2 Inhibition. Int. J. Mol. Sci. 2023, 24, 2410. [Google Scholar] [CrossRef]
- Matsushita, K.; Tomonaga, T.; Shimada, H.; Shioya, A.; Higashi, M.; Matsubara, H.; Harigaya, K.; Nomura, F.; Libutti, D.; Levens, D.; et al. An essential role of alternative splicing of c-myc suppressor FUSE-binding protein-interacting repressor in carcinogenesis. Cancer Res. 2006, 66, 1409–1417. [Google Scholar] [CrossRef]
- Liu, Z.; Yoshimi, A.; Wang, J.; Cho, H.; Chun-Wei Lee, S.; Ki, M.; Bitner, L.; Chu, T.; Shah, H.; Liu, B.; et al. Mutations in the RNA Splicing Factor SF3B1 Promote Tumorigenesis through MYC Stabilization. Cancer Discov. 2020, 10, 806–821. [Google Scholar] [CrossRef]
- Qu, S.; Jiao, Z.; Lu, G.; Yao, B.; Wang, T.; Rong, W.; Xu, J.; Fan, T.; Sun, X.; Yang, R.; et al. PD-L1 lncRNA splice isoform promotes lung adenocarcinoma progression via enhancing c-Myc activity. Genome Biol. 2021, 22, 104. [Google Scholar] [CrossRef] [PubMed]
- Heimberger, A.B.; Suki, D.; Yang, D.; Shi, W.; Aldape, K. The natural history of EGFR and EGFRvIII in glioblastoma patients. J. Transl. Med. 2005, 3, 38. [Google Scholar] [CrossRef]
- Babic, I.; Anderson, E.S.; Tanaka, K.; Guo, D.; Masui, K.; Li, B.; Zhu, S.; Gu, Y.; Villa, G.R.; Akhavan, D.; et al. EGFR mutation-induced alternative splicing of Max contributes to growth of glycolytic tumors in brain cancer. Cell Metab. 2013, 17, 1000–1008. [Google Scholar] [CrossRef]
- Voice, J.K.; Klemke, R.L.; Le, A.; Jackson, J.H. Four human ras homologs differ in their abilities to activate Raf-1, induce transformation, and stimulate cell motility. J. Biol. Chem. 1999, 274, 17164–17170. [Google Scholar] [CrossRef]
- Nuevo-Tapioles, C.; Philips, M.R. The role of KRAS splice variants in cancer biology. Front. Cell Dev. Biol. 2022, 10, 1033348. [Google Scholar] [CrossRef]
- Miller, S.P.; Maio, G.; Zhang, X.; Badillo Soto, F.S.; Zhu, J.; Ramirez, S.Z.; Lin, H. A Proteomic Approach Identifies Isoform-Specific and Nucleotide-Dependent RAS Interactions. Mol. Cell. Proteom. 2022, 21, 100268. [Google Scholar] [CrossRef] [PubMed]
- Zhang, X.; Cao, J.; Miller, S.P.; Jing, H.; Lin, H. Comparative Nucleotide-Dependent Interactome Analysis Reveals Shared and Differential Properties of KRas4a and KRas4b. ACS Cent. Sci. 2018, 4, 71–80. [Google Scholar] [CrossRef] [PubMed]
- Amendola, C.R.; Mahaffey, J.P.; Parker, S.J.; Ahearn, I.M.; Chen, W.C.; Zhou, M.; Court, H.; Shi, J.; Mendoza, S.L.; Morten, M.J.; et al. KRAS4A directly regulates hexokinase 1. Nature 2019, 576, 482–486. [Google Scholar] [CrossRef]
- Hall, A.E.; Pohl, S.O.; Cammareri, P.; Aitken, S.; Younger, N.T.; Raponi, M.; Billard, C.V.; Carrancio, A.B.; Bastem, A.; Freile, P.; et al. RNA splicing is a key mediator of tumour cell plasticity and a therapeutic vulnerability in colorectal cancer. Nat. Commun. 2022, 13, 2791. [Google Scholar] [CrossRef]
- Chen, W.C.; To, M.D.; Westcott, P.M.K.; Delrosario, R.; Kim, I.J.; Philips, M.; Tran, Q.; Bollam, S.R.; Goodarzi, H.; Bayani, N.; et al. Targeting KRAS4A splicing through the RBM39/DCAF15 pathway inhibits cancer stem cells. Nat. Commun. 2021, 12, 4288. [Google Scholar] [CrossRef]
- Siegfried, Z.; Bonomi, S.; Ghigna, C.; Karni, R. Regulation of the Ras-MAPK and PI3K-mTOR Signalling Pathways by Alternative Splicing in Cancer. Int. J. Cell Biol. 2013, 2013, 568931. [Google Scholar] [CrossRef] [PubMed]
- Shilo, A.; Ben Hur, V.; Denichenko, P.; Stein, I.; Pikarsky, E.; Rauch, J.; Kolch, W.; Zender, L.; Karni, R. Splicing factor hnRNP A2 activates the Ras-MAPK-ERK pathway by controlling A-Raf splicing in hepatocellular carcinoma development. RNA 2014, 20, 505–515. [Google Scholar] [CrossRef]
- Mogilevsky, M.; Shimshon, O.; Kumar, S.; Mogilevsky, A.; Keshet, E.; Yavin, E.; Heyd, F.; Karni, R. Modulation of MKNK2 alternative splicing by splice-switching oligonucleotides as a novel approach for glioblastoma treatment. Nucleic Acids Res. 2018, 46, 11396–11404. [Google Scholar] [CrossRef]
- Maimon, A.; Mogilevsky, M.; Shilo, A.; Golan-Gerstl, R.; Obiedat, A.; Ben-Hur, V.; Lebenthal-Loinger, I.; Stein, I.; Reich, R.; Beenstock, J.; et al. Mnk2 alternative splicing modulates the p38-MAPK pathway and impacts Ras-induced transformation. Cell Rep. 2014, 7, 501–513. [Google Scholar] [CrossRef]
- Thorpe, L.M.; Yuzugullu, H.; Zhao, J.J. PI3K in cancer: Divergent roles of isoforms, modes of activation and therapeutic targeting. Nat. Rev. Cancer 2015, 15, 7–24. [Google Scholar] [CrossRef]
- Panasyuk, G.; Nemazanyy, I.; Zhyvoloup, A.; Filonenko, V.; Davies, D.; Robson, M.; Pedley, R.B.; Waterfield, M.; Gout, I. mTORbeta splicing isoform promotes cell proliferation and tumorigenesis. J. Biol. Chem. 2009, 284, 30807–30814. [Google Scholar] [CrossRef] [PubMed]
- Boch, T.; Kohler, J.; Janning, M.; Loges, S. Targeting the EGF receptor family in non-small cell lung cancer-increased complexity and future perspectives. Cancer Biol. Med. 2022, 19, 1543–1564. [Google Scholar] [CrossRef] [PubMed]
- Wieduwilt, M.J.; Moasser, M.M. The epidermal growth factor receptor family: Biology driving targeted therapeutics. Cell. Mol. Life Sci. 2008, 65, 1566–1584. [Google Scholar] [CrossRef] [PubMed]
- Abou-Faycal, C.; Hatat, A.S.; Gazzeri, S.; Eymin, B. Splice Variants of the RTK Family: Their Role in Tumour Progression and Response to Targeted Therapy. Int. J. Mol. Sci. 2017, 18, 383. [Google Scholar] [CrossRef]
- Sanderson, M.P.; Keller, S.; Alonso, A.; Riedle, S.; Dempsey, P.J.; Altevogt, P. Generation of novel, secreted epidermal growth factor receptor (EGFR/ErbB1) isoforms via metalloprotease-dependent ectodomain shedding and exosome secretion. J. Cell. Biochem. 2008, 103, 1783–1797. [Google Scholar] [CrossRef]
- Reiter, J.L.; Threadgill, D.W.; Eley, G.D.; Strunk, K.E.; Danielsen, A.J.; Sinclair, C.S.; Pearsall, R.S.; Green, P.J.; Yee, D.; Lampland, A.L.; et al. Comparative genomic sequence analysis and isolation of human and mouse alternative EGFR transcripts encoding truncated receptor isoforms. Genomics 2001, 71, 1–20. [Google Scholar] [CrossRef]
- Maramotti, S.; Paci, M.; Manzotti, G.; Rapicetta, C.; Gugnoni, M.; Galeone, C.; Cesario, A.; Lococo, F. Soluble Epidermal Growth Factor Receptors (sEGFRs) in Cancer: Biological Aspects and Clinical Relevance. Int. J. Mol. Sci. 2016, 17, 593. [Google Scholar] [CrossRef]
- Asgeirsson, K.S.; Agrawal, A.; Allen, C.; Hitch, A.; Ellis, I.O.; Chapman, C.; Cheung, K.L.; Robertson, J.F. Serum epidermal growth factor receptor and HER2 expression in primary and metastatic breast cancer patients. Breast Cancer Res. 2007, 9, R75. [Google Scholar] [CrossRef]
- Baron, A.T.; Cora, E.M.; Lafky, J.M.; Boardman, C.H.; Buenafe, M.C.; Rademaker, A.; Liu, D.; Fishman, D.A.; Podratz, K.C.; Maihle, N.J. Soluble epidermal growth factor receptor (sEGFR/sErbB1) as a potential risk, screening, and diagnostic serum biomarker of epithelial ovarian cancer. Cancer Epidemiol. Biomark. Prev. 2003, 12, 103–113. [Google Scholar]
- Choi, J.H.; Oh, J.Y.; Ryu, S.K.; Kim, S.J.; Lee, N.Y.; Kim, Y.S.; Yi, S.Y.; Shim, K.S.; Han, W.S. Detection of epidermal growth factor receptor in the serum of gastric carcinoma patients. Cancer 1997, 79, 1879–1883. [Google Scholar] [CrossRef]
- Piccione, E.C.; Lieu, T.J.; Gentile, C.F.; Williams, T.R.; Connolly, A.J.; Godwin, A.K.; Koong, A.C.; Wong, A.J. A novel epidermal growth factor receptor variant lacking multiple domains directly activates transcription and is overexpressed in tumors. Oncogene 2012, 31, 2953–2967. [Google Scholar] [CrossRef] [PubMed]
- Hart, V.; Gautrey, H.; Kirby, J.; Tyson-Capper, A. HER2 splice variants in breast cancer: Investigating their impact on diagnosis and treatment outcomes. Oncotarget 2020, 11, 4338–4357. [Google Scholar] [CrossRef] [PubMed]
- Shamieh, L.S.; Evans, A.J.; Denton, M.C.; Clinton, G.M. Receptor binding specificities of Herstatin and its intron 8-encoded domain. FEBS Lett. 2004, 568, 163–166. [Google Scholar] [CrossRef] [PubMed]
- Scott, G.K.; Robles, R.; Park, J.W.; Montgomery, P.A.; Daniel, J.; Holmes, W.E.; Lee, J.; Keller, G.A.; Li, W.L.; Fendly, B.M.; et al. A truncated intracellular HER2/neu receptor produced by alternative RNA processing affects growth of human carcinoma cells. Mol. Cell. Biol. 1993, 13, 2247–2257. [Google Scholar] [CrossRef] [PubMed]
- Castagnoli, L.; Iorio, E.; Dugo, M.; Koschorke, A.; Faraci, S.; Canese, R.; Casalini, P.; Nanni, P.; Vernieri, C.; Di Nicola, M.; et al. Intratumor lactate levels reflect HER2 addiction status in HER2-positive breast cancer. J. Cell. Physiol. 2019, 234, 1768–1779. [Google Scholar] [CrossRef] [PubMed]
- Palladini, A.; Nicoletti, G.; Lamolinara, A.; Dall’Ora, M.; Balboni, T.; Ianzano, M.L.; Laranga, R.; Landuzzi, L.; Giusti, V.; Ceccarelli, C.; et al. HER2 isoforms co-expression differently tunes mammary tumor phenotypes affecting onset, vasculature and therapeutic response. Oncotarget 2017, 8, 54444–54458. [Google Scholar] [CrossRef]
- Turpin, J.; Ling, C.; Crosby, E.J.; Hartman, Z.C.; Simond, A.M.; Chodosh, L.A.; Rennhack, J.P.; Andrechek, E.R.; Ozcelik, J.; Hallett, M.; et al. The ErbB2DeltaEx16 splice variant is a major oncogenic driver in breast cancer that promotes a pro-metastatic tumor microenvironment. Oncogene 2016, 35, 6053–6064. [Google Scholar] [CrossRef]
- Marchini, C.; Gabrielli, F.; Iezzi, M.; Zenobi, S.; Montani, M.; Pietrella, L.; Kalogris, C.; Rossini, A.; Ciravolo, V.; Castagnoli, L.; et al. The human splice variant Delta16HER2 induces rapid tumor onset in a reporter transgenic mouse. PLoS ONE 2011, 6, e18727. [Google Scholar] [CrossRef]
- Castagnoli, L.; Iezzi, M.; Ghedini, G.C.; Ciravolo, V.; Marzano, G.; Lamolinara, A.; Zappasodi, R.; Gasparini, P.; Campiglio, M.; Amici, A.; et al. Activated d16HER2 homodimers and SRC kinase mediate optimal efficacy for trastuzumab. Cancer Res. 2014, 74, 6248–6259. [Google Scholar] [CrossRef]
- Babina, I.S.; Turner, N.C. Advances and challenges in targeting FGFR signalling in cancer. Nat. Rev. Cancer 2017, 17, 318–332. [Google Scholar] [CrossRef]
- Eswarakumar, V.P.; Lax, I.; Schlessinger, J. Cellular signaling by fibroblast growth factor receptors. Cytokine Growth Factor Rev. 2005, 16, 139–149. [Google Scholar] [CrossRef]
- Holzmann, K.; Grunt, T.; Heinzle, C.; Sampl, S.; Steinhoff, H.; Reichmann, N.; Kleiter, M.; Hauck, M.; Marian, B. Alternative Splicing of Fibroblast Growth Factor Receptor IgIII Loops in Cancer. J. Nucleic Acids 2012, 2012, 950508. [Google Scholar] [CrossRef] [PubMed]
- De Moerlooze, L.; Spencer-Dene, B.; Revest, J.M.; Hajihosseini, M.; Rosewell, I.; Dickson, C. An important role for the IIIb isoform of fibroblast growth factor receptor 2 (FGFR2) in mesenchymal-epithelial signalling during mouse organogenesis. Development 2000, 127, 483–492. [Google Scholar] [CrossRef] [PubMed]
- Thiery, J.P.; Sleeman, J.P. Complex networks orchestrate epithelial-mesenchymal transitions. Nat. Rev. Mol. Cell Biol. 2006, 7, 131–142. [Google Scholar] [CrossRef]
- Matsuda, Y.; Hagio, M.; Seya, T.; Ishiwata, T. Fibroblast growth factor receptor 2 IIIc as a therapeutic target for colorectal cancer cells. Mol. Cancer Ther. 2012, 11, 2010–2020. [Google Scholar] [CrossRef] [PubMed]
- Ricol, D.; Cappellen, D.; El Marjou, A.; Gil-Diez-de-Medina, S.; Girault, J.M.; Yoshida, T.; Ferry, G.; Tucker, G.; Poupon, M.F.; Chopin, D.; et al. Tumour suppressive properties of fibroblast growth factor receptor 2-IIIb in human bladder cancer. Oncogene 1999, 18, 7234–7243. [Google Scholar] [CrossRef] [PubMed]
- Allerstorfer, S.; Sonvilla, G.; Fischer, H.; Spiegl-Kreinecker, S.; Gauglhofer, C.; Setinek, U.; Czech, T.; Marosi, C.; Buchroithner, J.; Pichler, J.; et al. FGF5 as an oncogenic factor in human glioblastoma multiforme: Autocrine and paracrine activities. Oncogene 2008, 27, 4180–4190. [Google Scholar] [CrossRef] [PubMed]
- Fischer, H.; Taylor, N.; Allerstorfer, S.; Grusch, M.; Sonvilla, G.; Holzmann, K.; Setinek, U.; Elbling, L.; Cantonati, H.; Grasl-Kraupp, B.; et al. Fibroblast growth factor receptor-mediated signals contribute to the malignant phenotype of non-small cell lung cancer cells: Therapeutic implications and synergism with epidermal growth factor receptor inhibition. Mol. Cancer Ther. 2008, 7, 3408–3419. [Google Scholar] [CrossRef]
- Sonvilla, G.; Allerstorfer, S.; Heinzle, C.; Stattner, S.; Karner, J.; Klimpfinger, M.; Wrba, F.; Fischer, H.; Gauglhofer, C.; Spiegl-Kreinecker, S.; et al. Fibroblast growth factor receptor 3-IIIc mediates colorectal cancer growth and migration. Br. J. Cancer 2010, 102, 1145–1156. [Google Scholar] [CrossRef]
- Lafitte, M.; Moranvillier, I.; Garcia, S.; Peuchant, E.; Iovanna, J.; Rousseau, B.; Dubus, P.; Guyonnet-Duperat, V.; Belleannee, G.; Ramos, J.; et al. FGFR3 has tumor suppressor properties in cells with epithelial phenotype. Mol. Cancer 2013, 12, 83. [Google Scholar] [CrossRef]
- Yan, G.; Fukabori, Y.; McBride, G.; Nikolaropolous, S.; McKeehan, W.L. Exon switching and activation of stromal and embryonic fibroblast growth factor (FGF)-FGF receptor genes in prostate epithelial cells accompany stromal independence and malignancy. Mol. Cell. Biol. 1993, 13, 4513–4522. [Google Scholar] [CrossRef] [PubMed]
- Scotet, E.; Houssaint, E. Exon III splicing switch of fibroblast growth factor (FGF) receptor-2 and -3 can be induced by FGF-1 or FGF-2. Oncogene 1998, 17, 67–76. [Google Scholar] [CrossRef] [PubMed]
- Mauger, D.M.; Lin, C.; Garcia-Blanco, M.A. hnRNP H and hnRNP F complex with Fox2 to silence fibroblast growth factor receptor 2 exon IIIc. Mol. Cell. Biol. 2008, 28, 5403–5419. [Google Scholar] [CrossRef] [PubMed]
- Warzecha, C.C.; Sato, T.K.; Nabet, B.; Hogenesch, J.B.; Carstens, R.P. ESRP1 and ESRP2 are epithelial cell-type-specific regulators of FGFR2 splicing. Mol. Cell 2009, 33, 591–601. [Google Scholar] [CrossRef]
- Warzecha, C.C.; Jiang, P.; Amirikian, K.; Dittmar, K.A.; Lu, H.; Shen, S.; Guo, W.; Xing, Y.; Carstens, R.P. An ESRP-regulated splicing programme is abrogated during the epithelial-mesenchymal transition. EMBO J. 2010, 29, 3286–3300. [Google Scholar] [CrossRef]
- Jin, W.; McCutcheon, I.E.; Fuller, G.N.; Huang, E.S.; Cote, G.J. Fibroblast growth factor receptor-1 alpha-exon exclusion and polypyrimidine tract-binding protein in glioblastoma multiforme tumors. Cancer Res. 2000, 60, 1221–1224. [Google Scholar]
- Tomlinson, D.C.; Knowles, M.A. Altered splicing of FGFR1 is associated with high tumor grade and stage and leads to increased sensitivity to FGF1 in bladder cancer. Am. J. Pathol. 2010, 177, 2379–2386. [Google Scholar] [CrossRef]
- Zhao, M.; Zhuo, M.L.; Zheng, X.; Su, X.; Meric-Bernstam, F. FGFR1beta is a driver isoform of FGFR1 alternative splicing in breast cancer cells. Oncotarget 2019, 10, 30–44. [Google Scholar] [CrossRef]
- Roy, R.; Huang, Y.; Seckl, M.J.; Pardo, O.E. Emerging roles of hnRNPA1 in modulating malignant transformation. Wiley Interdiscip. Rev. RNA 2017, 8, e1431. [Google Scholar] [CrossRef]
- Yang, X.; Zhan, P.; Feng, S.; Ji, H.; Tian, W.; Wang, M.; Cheng, C.; Song, B. SRSF6 regulates alternative splicing of genes involved in DNA damage response and DNA repair in HeLa cells. Oncol. Rep. 2020, 44, 1851–1862. [Google Scholar] [CrossRef]
- Zheng, X.; Peng, Q.; Wang, L.; Zhang, X.; Huang, L.; Wang, J.; Qin, Z. Serine/arginine-rich splicing factors: The bridge linking alternative splicing and cancer. Int. J. Biol. Sci. 2020, 16, 2442–2453. [Google Scholar] [CrossRef] [PubMed]
- Cheng, R.; Xiao, L.; Zhou, W.; Jin, X.; Xu, Z.; Xu, C.; Wang, P.; Luo, M.; Wang, M.; Ma, K.; et al. A pan-cancer analysis of alternative splicing of splicing factors in 6904 patients. Oncogene 2021, 40, 5441–5450. [Google Scholar] [CrossRef]
- Chan, J.J.; Zhang, B.; Chew, X.H.; Salhi, A.; Kwok, Z.H.; Lim, C.Y.; Desi, N.; Subramaniam, N.; Siemens, A.; Kinanti, T.; et al. Pan-cancer pervasive upregulation of 3′ UTR splicing drives tumourigenesis. Nat. Cell. Biol. 2022, 24, 928–939. [Google Scholar] [CrossRef] [PubMed]
- Leclair, N.K.; Brugiolo, M.; Urbanski, L.; Lawson, S.C.; Thakar, K.; Yurieva, M.; George, J.; Hinson, J.T.; Cheng, A.; Graveley, B.R.; et al. Poison Exon Splicing Regulates a Coordinated Network of SR Protein Expression during Differentiation and Tumorigenesis. Mol. Cell 2020, 80, 648–665.E9. [Google Scholar] [CrossRef] [PubMed]
- She, W.; Shao, J.; Jia, R. Targeting Splicing Factor SRSF6 for Cancer Therapy. Front. Cell Dev. Biol. 2021, 9, 780023. [Google Scholar] [CrossRef]
- Park, S.; Brugiolo, M.; Akerman, M.; Das, S.; Urbanski, L.; Geier, A.; Kesarwani, A.K.; Fan, M.; Leclair, N.; Lin, K.T.; et al. Differential Functions of Splicing Factors in Mammary Transformation and Breast Cancer Metastasis. Cell Rep. 2019, 29, 2672–2688. [Google Scholar] [CrossRef] [PubMed]
- Hara, H.; Takeda, T.; Yamamoto, N.; Furuya, K.; Hirose, K.; Kamiya, T.; Adachi, T. Zinc-induced modulation of SRSF6 activity alters Bim splicing to promote generation of the most potent apoptotic isoform BimS. FEBS J. 2013, 280, 3313–3327. [Google Scholar] [CrossRef] [PubMed]
- Jiang, C.C.; Lai, F.; Tay, K.H.; Croft, A.; Rizos, H.; Becker, T.M.; Yang, F.; Liu, H.; Thorne, R.F.; Hersey, P.; et al. Apoptosis of human melanoma cells induced by inhibition of B-RAFV600E involves preferential splicing of bimS. Cell Death Dis. 2010, 1, e69. [Google Scholar] [CrossRef]
- Cohen-Eliav, M.; Golan-Gerstl, R.; Siegfried, Z.; Andersen, C.L.; Thorsen, K.; Orntoft, T.F.; Mu, D.; Karni, R. The splicing factor SRSF6 is amplified and is an oncoprotein in lung and colon cancers. J. Pathol. 2013, 229, 630–639. [Google Scholar] [CrossRef]
- Wan, L.; Yu, W.; Shen, E.; Sun, W.; Liu, Y.; Kong, J.; Wu, Y.; Han, F.; Zhang, L.; Yu, T.; et al. SRSF6-regulated alternative splicing that promotes tumour progression offers a therapy target for colorectal cancer. Gut 2019, 68, 118–129. [Google Scholar] [CrossRef]
- Kong, J.; Sun, W.; Li, C.; Wan, L.; Wang, S.; Wu, Y.; Xu, E.; Zhang, H.; Lai, M. Long non-coding RNA LINC01133 inhibits epithelial-mesenchymal transition and metastasis in colorectal cancer by interacting with SRSF6. Cancer Lett. 2016, 380, 476–484. [Google Scholar] [CrossRef] [PubMed]
- Li, M.; Wu, P.; Yang, Z.; Deng, S.; Ni, L.; Zhang, Y.; Jin, L.; Pan, Y. miR-193a-5p promotes pancreatic cancer cell metastasis through SRSF6-mediated alternative splicing of OGDHL and ECM1. Am. J. Cancer Res. 2020, 10, 38–59. [Google Scholar] [PubMed]
- Zhang, F.; Wang, H.; Yu, J.; Yao, X.; Yang, S.; Li, W.; Xu, L.; Zhao, L. LncRNA CRNDE attenuates chemoresistance in gastric cancer via SRSF6-regulated alternative splicing of PICALM. Mol. Cancer 2021, 20, 6. [Google Scholar] [CrossRef] [PubMed]
- Nowak, D.G.; Woolard, J.; Amin, E.M.; Konopatskaya, O.; Saleem, M.A.; Churchill, A.J.; Ladomery, M.R.; Harper, S.J.; Bates, D.O. Expression of pro- and anti-angiogenic isoforms of VEGF is differentially regulated by splicing and growth factors. J. Cell Sci. 2008, 121, 3487–3495. [Google Scholar] [CrossRef]
- Muehlbauer, L.K.; Wei, T.; Shishkova, E.; Coon, J.J.; Lambert, P.F. IQGAP1 and RNA Splicing in the Context of Head and Neck via Phosphoproteomics. J. Proteome Res. 2022, 21, 2211–2223. [Google Scholar] [CrossRef]
- Jia, Q.; Nie, H.; Yu, P.; Xie, B.; Wang, C.; Yang, F.; Wei, G.; Ni, T. HNRNPA1-mediated 3′ UTR length changes of HN1 contributes to cancer- and senescence-associated phenotypes. Aging 2019, 11, 4407–4437. [Google Scholar] [CrossRef]
- Chu, P.C.; Yang, M.C.; Kulp, S.K.; Salunke, S.B.; Himmel, L.E.; Fang, C.S.; Jadhav, A.M.; Shan, Y.S.; Lee, C.T.; Lai, M.D.; et al. Regulation of oncogenic KRAS signaling via a novel KRAS-integrin-linked kinase-hnRNPA1 regulatory loop in human pancreatic cancer cells. Oncogene 2016, 35, 3897–3908. [Google Scholar] [CrossRef]
- Cogoi, S.; Rapozzi, V.; Cauci, S.; Xodo, L.E. Critical role of hnRNP A1 in activating KRAS transcription in pancreatic cancer cells: A molecular mechanism involving G4 DNA. Biochim. Biophys. Acta Gen. Subj. 2017, 1861, 1389–1398. [Google Scholar] [CrossRef]
- Nadiminty, N.; Tummala, R.; Liu, C.; Lou, W.; Evans, C.P.; Gao, A.C. NF-kappaB2/p52:c-Myc: hnRNPA1 Pathway Regulates Expression of Androgen Receptor Splice Variants and Enzalutamide Sensitivity in Prostate Cancer. Mol. Cancer Ther. 2015, 14, 1884–1895. [Google Scholar] [CrossRef]
- Zou, Y.; Tang, H.; Miao, Y.; Zhu, H.; Wang, L.; Fan, L.; Fu, J.; Xu, W.; Li, J.; Xia, Y. Overexpression of c-Myc-dependent heterogeneous nuclear ribonucleoprotein A1 promotes proliferation and inhibits apoptosis in NOTCH1-mutated chronic lymphocytic leukemia cells. Chin. Med. J. 2022, 135, 920–929. [Google Scholar] [CrossRef]
- Clower, C.V.; Chatterjee, D.; Wang, Z.; Cantley, L.C.; Vander Heiden, M.G.; Krainer, A.R. The alternative splicing repressors hnRNP A1/A2 and PTB influence pyruvate kinase isoform expression and cell metabolism. Proc. Natl. Acad. Sci. USA 2010, 107, 1894–1899. [Google Scholar] [CrossRef] [PubMed]
- Li, Z.; Zheng, W.; Li, H.; Li, C.; Gong, Z. Synergistic Induction of Potential Warburg Effect in Zebrafish Hepatocellular Carcinoma by Co-Transgenic Expression of Myc and xmrk Oncogenes. PLoS ONE 2015, 10, e0132319. [Google Scholar] [CrossRef] [PubMed]
- Bykov, V.J.; Issaeva, N.; Shilov, A.; Hultcrantz, M.; Pugacheva, E.; Chumakov, P.; Bergman, J.; Wiman, K.G.; Selivanova, G. Restoration of the tumor suppressor function to mutant p53 by a low-molecular-weight compound. Nat. Med. 2002, 8, 282–288. [Google Scholar] [CrossRef] [PubMed]
- Lehmann, S.; Bykov, V.J.; Ali, D.; Andren, O.; Cherif, H.; Tidefelt, U.; Uggla, B.; Yachnin, J.; Juliusson, G.; Moshfegh, A.; et al. Targeting p53 in vivo: A first-in-human study with p53-targeting compound APR-246 in refractory hematologic malignancies and prostate cancer. J. Clin. Oncol. 2012, 30, 3633–3639. [Google Scholar] [CrossRef]
- Guiley, K.Z.; Shokat, K.M. A small molecule reacts with the p53 somatic mutant Y220C to rescue wild-type thermal stability. Cancer Discov. 2022, 13, 56–69. [Google Scholar] [CrossRef]
- Wang, X.; Allen, S.; Blake, J.F.; Bowcut, V.; Briere, D.M.; Calinisan, A.; Dahlke, J.R.; Fell, J.B.; Fischer, J.P.; Gunn, R.J.; et al. Identification of MRTX1133, a Noncovalent, Potent, and Selective KRAS(G12D) Inhibitor. J. Med. Chem. 2022, 65, 3123–3133. [Google Scholar] [CrossRef]
- Lanman, B.A.; Allen, J.R.; Allen, J.G.; Amegadzie, A.K.; Ashton, K.S.; Booker, S.K.; Chen, J.J.; Chen, N.; Frohn, M.J.; Goodman, G.; et al. Discovery of a Covalent Inhibitor of KRAS(G12C) (AMG 510) for the Treatment of Solid Tumors. J. Med. Chem. 2020, 63, 52–65. [Google Scholar] [CrossRef]
- FDA Approves First KRAS Inhibitor: Sotorasib. Cancer Discov. 2021, 11, OF4. [CrossRef]
- Liu, J.; Kang, R.; Tang, D. The KRAS-G12C inhibitor: Activity and resistance. Cancer Gene Ther. 2022, 29, 875–878. [Google Scholar] [CrossRef]
- Savino, M.; Annibali, D.; Carucci, N.; Favuzzi, E.; Cole, M.D.; Evan, G.I.; Soucek, L.; Nasi, S. The action mechanism of the Myc inhibitor termed Omomyc may give clues on how to target Myc for cancer therapy. PLoS ONE 2011, 6, e22284. [Google Scholar] [CrossRef]
- Kaida, D.; Motoyoshi, H.; Tashiro, E.; Nojima, T.; Hagiwara, M.; Ishigami, K.; Watanabe, H.; Kitahara, T.; Yoshida, T.; Nakajima, H.; et al. Spliceostatin A targets SF3b and inhibits both splicing and nuclear retention of pre-mRNA. Nat. Chem. Biol. 2007, 3, 576–583. [Google Scholar] [CrossRef]
- Kumar, D.; Kashyap, M.K.; Yu, Z.; Spaanderman, I.; Villa, R.; Kipps, T.J.; La Clair, J.J.; Burkart, M.D.; Castro, J.E. Modulation of RNA splicing associated with Wnt signaling pathway using FD-895 and pladienolide B. Aging 2022, 14, 2081–2100. [Google Scholar] [CrossRef]
- Convertini, P.; Shen, M.; Potter, P.M.; Palacios, G.; Lagisetti, C.; de la Grange, P.; Horbinski, C.; Fondufe-Mittendorf, Y.N.; Webb, T.R.; Stamm, S. Sudemycin E influences alternative splicing and changes chromatin modifications. Nucleic Acids Res. 2014, 42, 4947–4961. [Google Scholar] [CrossRef]
- Corrionero, A.; Minana, B.; Valcarcel, J. Reduced fidelity of branch point recognition and alternative splicing induced by the anti-tumor drug spliceostatin A. Genes Dev. 2011, 25, 445–459. [Google Scholar] [CrossRef]
- Darman, R.B.; Seiler, M.; Agrawal, A.A.; Lim, K.H.; Peng, S.; Aird, D.; Bailey, S.L.; Bhavsar, E.B.; Chan, B.; Colla, S.; et al. Cancer-Associated SF3B1 Hotspot Mutations Induce Cryptic 3′ Splice Site Selection through Use of a Different Branch Point. Cell Rep. 2015, 13, 1033–1045. [Google Scholar] [CrossRef]
- Obeng, E.A.; Chappell, R.J.; Seiler, M.; Chen, M.C.; Campagna, D.R.; Schmidt, P.J.; Schneider, R.K.; Lord, A.M.; Wang, L.; Gambe, R.G.; et al. Physiologic Expression of Sf3b1(K700E) Causes Impaired Erythropoiesis, Aberrant Splicing, and Sensitivity to Therapeutic Spliceosome Modulation. Cancer Cell 2016, 30, 404–417. [Google Scholar] [CrossRef]
- Seiler, M.; Yoshimi, A.; Darman, R.; Chan, B.; Keaney, G.; Thomas, M.; Agrawal, A.A.; Caleb, B.; Csibi, A.; Sean, E.; et al. H3B-8800, an orally available small-molecule splicing modulator, induces lethality in spliceosome-mutant cancers. Nat. Med. 2018, 24, 497–504. [Google Scholar] [CrossRef]
- Eskens, F.A.; Ramos, F.J.; Burger, H.; O’Brien, J.P.; Piera, A.; de Jonge, M.J.; Mizui, Y.; Wiemer, E.A.; Carreras, M.J.; Baselga, J.; et al. Phase I pharmacokinetic and pharmacodynamic study of the first-in-class spliceosome inhibitor E7107 in patients with advanced solid tumors. Clin. Cancer Res. 2013, 19, 6296–6304. [Google Scholar] [CrossRef]
- Alors-Perez, E.; Blazquez-Encinas, R.; Alcala, S.; Viyuela-Garcia, C.; Pedraza-Arevalo, S.; Herrero-Aguayo, V.; Jimenez-Vacas, J.M.; Mafficini, A.; Sanchez-Frias, M.E.; Cano, M.T.; et al. Dysregulated splicing factor SF3B1 unveils a dual therapeutic vulnerability to target pancreatic cancer cells and cancer stem cells with an anti-splicing drug. J. Exp. Clin. Cancer Res. 2021, 40, 382. [Google Scholar] [CrossRef]
- Fuentes-Fayos, A.C.; Perez-Gomez, J.M.; ME, G.G.; Jimenez-Vacas, J.M.; Blanco-Acevedo, C.; Sanchez-Sanchez, R.; Solivera, J.; Breunig, J.J.; Gahete, M.D.; Castano, J.P.; et al. SF3B1 inhibition disrupts malignancy and prolongs survival in glioblastoma patients through BCL2L1 splicing and mTOR/ss-catenin pathways imbalances. J. Exp. Clin. Cancer Res. 2022, 41, 39. [Google Scholar] [CrossRef]
- Han, C.; Khodadadi-Jamayran, A.; Lorch, A.H.; Jin, Q.; Serafin, V.; Zhu, P.; Politanska, Y.; Sun, L.; Gutierrez-Diaz, B.T.; Pryzhkova, M.V.; et al. SF3B1 homeostasis is critical for survival and therapeutic response in T cell leukemia. Sci. Adv. 2022, 8, eabj8357. [Google Scholar] [CrossRef]
- O’Brien, K.; Matlin, A.J.; Lowell, A.M.; Moore, M.J. The biflavonoid isoginkgetin is a general inhibitor of Pre-mRNA splicing. J. Biol. Chem. 2008, 283, 33147–33154. [Google Scholar] [CrossRef]
- Feustel, K.; Falchook, G.S. Protein Arginine Methyltransferase 5 (PRMT5) Inhibitors in Oncology Clinical Trials: A review. J. Immunother. Precis. Oncol. 2022, 5, 58–67. [Google Scholar] [CrossRef]
- Chen, Y.; Shao, X.; Zhao, X.; Ji, Y.; Liu, X.; Li, P.; Zhang, M.; Wang, Q. Targeting protein arginine methyltransferase 5 in cancers: Roles, inhibitors and mechanisms. Biomed. Pharmacother. 2021, 144, 112252. [Google Scholar] [CrossRef]
- Fong, J.Y.; Pignata, L.; Goy, P.A.; Kawabata, K.C.; Lee, S.C.; Koh, C.M.; Musiani, D.; Massignani, E.; Kotini, A.G.; Penson, A.; et al. Therapeutic Targeting of RNA Splicing Catalysis through Inhibition of Protein Arginine Methylation. Cancer Cell 2019, 36, 194–209. [Google Scholar] [CrossRef]
- Hamard, P.J.; Santiago, G.E.; Liu, F.; Karl, D.L.; Martinez, C.; Man, N.; Mookhtiar, A.K.; Duffort, S.; Greenblatt, S.; Verdun, R.E.; et al. PRMT5 Regulates DNA Repair by Controlling the Alternative Splicing of Histone-Modifying Enzymes. Cell Rep. 2018, 24, 2643–2657. [Google Scholar] [CrossRef]
- Che, Y.; Liu, Y.; Yao, Y.; Hill, H.A.; Li, Y.; Cai, Q.; Yan, F.; Jain, P.; Wang, W.; Rui, L.; et al. Exploiting PRMT5 as a target for combination therapy in mantle cell lymphoma characterized by frequent ATM and TP53 mutations. Blood Cancer J. 2023, 13, 27. [Google Scholar] [CrossRef]
- Iwai, K.; Yaguchi, M.; Nishimura, K.; Yamamoto, Y.; Tamura, T.; Nakata, D.; Dairiki, R.; Kawakita, Y.; Mizojiri, R.; Ito, Y.; et al. Anti-tumor efficacy of a novel CLK inhibitor via targeting RNA splicing and MYC-dependent vulnerability. EMBO Mol. Med. 2018, 10, e8289. [Google Scholar] [CrossRef]
- Denichenko, P.; Mogilevsky, M.; Clery, A.; Welte, T.; Biran, J.; Shimshon, O.; Barnabas, G.D.; Danan-Gotthold, M.; Kumar, S.; Yavin, E.; et al. Specific inhibition of splicing factor activity by decoy RNA oligonucleotides. Nat. Commun. 2019, 10, 1590. [Google Scholar] [CrossRef]
- Patel, M.; Sachidanandan, M.; Adnan, M. Serine arginine protein kinase 1 (SRPK1): A moonlighting protein with theranostic ability in cancer prevention. Mol. Biol. Rep. 2019, 46, 1487–1497. [Google Scholar] [CrossRef]
- Siqueira, R.P.; Caetano, M.M.M.; de Souza, L.A.; Dos Passos, P.M.S.; Simaroli, N.B.; Barros, M.V.A.; de Souza, A.P.M.; de Oliveira, L.L.; Silva-Junior, A.; Fietto, J.L.R.; et al. Combined SRPK and AKT pharmacological inhibition is synergistic in T-cell acute lymphoblastic leukemia cells. Toxicol. Vitr. 2020, 65, 104777. [Google Scholar] [CrossRef]
- ElHady, A.K.; El-Gamil, D.S.; Abadi, A.H.; Abdel-Halim, M.; Engel, M. An overview of cdc2-like kinase 1 (Clk1) inhibitors and their therapeutic indications. Med. Res. Rev. 2023, 43, 343–398. [Google Scholar] [CrossRef]
- Oltean, S.; Gammons, M.; Hulse, R.; Hamdollah-Zadeh, M.; Mavrou, A.; Donaldson, L.; Salmon, A.H.; Harper, S.J.; Ladomery, M.R.; Bates, D.O. SRPK1 inhibition in vivo: Modulation of VEGF splicing and potential treatment for multiple diseases. Biochem. Soc. Trans. 2012, 40, 831–835. [Google Scholar] [CrossRef]
- He, C.; Liu, B.; Wang, H.Y.; Wu, L.; Zhao, G.; Huang, C.; Liu, Y.; Shan, B.; Liu, L. Inhibition of SRPK1, a key splicing regulator, exhibits antitumor and chemotherapeutic-sensitizing effects on extranodal NK/T-cell lymphoma cells. BMC Cancer 2022, 22, 1100. [Google Scholar] [CrossRef]
- Tummala, R.; Lou, W.; Gao, A.C.; Nadiminty, N. Quercetin Targets hnRNPA1 to Overcome Enzalutamide Resistance in Prostate Cancer Cells. Mol. Cancer Ther. 2017, 16, 2770–2779. [Google Scholar] [CrossRef]
- Ma, W.K.; Voss, D.M.; Scharner, J.; Costa, A.S.H.; Lin, K.T.; Jeon, H.Y.; Wilkinson, J.E.; Jackson, M.; Rigo, F.; Bennett, C.F.; et al. ASO-Based PKM Splice-Switching Therapy Inhibits Hepatocellular Carcinoma Growth. Cancer Res. 2022, 82, 900–915. [Google Scholar] [CrossRef]
- Li, L.; Hobson, L.; Perry, L.; Clark, B.; Heavey, S.; Haider, A.; Sridhar, A.; Shaw, G.; Kelly, J.; Freeman, A.; et al. Targeting the ERG oncogene with splice-switching oligonucleotides as a novel therapeutic strategy in prostate cancer. Br. J. Cancer 2020, 123, 1024–1032. [Google Scholar] [CrossRef]
- Meijboom, K.E.; Wood, M.J.A.; McClorey, G. Splice-Switching Therapy for Spinal Muscular Atrophy. Genes 2017, 8, 161. [Google Scholar] [CrossRef]
- Aartsma-Rus, A.; Krieg, A.M. FDA Approves Eteplirsen for Duchenne Muscular Dystrophy: The Next Chapter in the Eteplirsen Saga. Nucleic Acid Ther. 2017, 27, 1–3. [Google Scholar] [CrossRef]
- Jbara, A.; Siegfried, Z.; Karni, R. Splice-switching as cancer therapy. Curr. Opin. Pharmacol. 2021, 59, 140–148. [Google Scholar] [CrossRef]
- Havens, M.A.; Hastings, M.L. Splice-switching antisense oligonucleotides as therapeutic drugs. Nucleic Acids Res. 2016, 44, 6549–6563. [Google Scholar] [CrossRef]
- Godfrey, C.; Desviat, L.R.; Smedsrod, B.; Pietri-Rouxel, F.; Denti, M.A.; Disterer, P.; Lorain, S.; Nogales-Gadea, G.; Sardone, V.; Anwar, R.; et al. Delivery is key: Lessons learnt from developing splice-switching antisense therapies. EMBO Mol. Med. 2017, 9, 545–557. [Google Scholar] [CrossRef]
- Soudah, T.; Mogilevsky, M.; Karni, R.; Yavin, E. CLIP6-PNA-Peptide Conjugates: Non-Endosomal Delivery of Splice Switching Oligonucleotides. Bioconjugate Chem. 2017, 28, 3036–3042. [Google Scholar] [CrossRef]
- Scharner, J.; Qi, S.; Rigo, F.; Bennett, C.F.; Krainer, A.R. Delivery of GalNAc-Conjugated Splice-Switching ASOs to Non-hepatic Cells through Ectopic Expression of Asialoglycoprotein Receptor. Mol. Therapy. Nucleic Acids 2019, 16, 313–325. [Google Scholar] [CrossRef]
- Li, L.; Zheng, J.; Stevens, M.; Oltean, S. A repositioning screen using an FGFR2 splicing reporter reveals compounds that regulate epithelial-mesenchymal transitions and inhibit growth of prostate cancer xenografts. Mol. Ther. Methods Clin. Dev. 2022, 25, 147–157. [Google Scholar] [CrossRef]
- Star, E.; Stevens, M.; Gooding, C.; Smith, C.W.J.; Li, L.; Ayine, M.L.; Harper, S.J.; Bates, D.O.; Oltean, S. A drug-repositioning screen using splicing-sensitive fluorescent reporters identifies novel modulators of VEGF-A splicing with anti-angiogenic properties. Oncogenesis 2021, 10, 36. [Google Scholar] [CrossRef]
- Bokharaie, H.; Kolch, W.; Krstic, A. Analysis of Alternative mRNA Splicing in Vemurafenib-Resistant Melanoma Cells. Biomolecules 2022, 12, 993. [Google Scholar] [CrossRef]
- Poulikakos, P.I.; Persaud, Y.; Janakiraman, M.; Kong, X.; Ng, C.; Moriceau, G.; Shi, H.; Atefi, M.; Titz, B.; Gabay, M.T.; et al. RAF inhibitor resistance is mediated by dimerization of aberrantly spliced BRAF(V600E). Nature 2011, 480, 387–390. [Google Scholar] [CrossRef]
- Dehm, S.M.; Schmidt, L.J.; Heemers, H.V.; Vessella, R.L.; Tindall, D.J. Splicing of a novel androgen receptor exon generates a constitutively active androgen receptor that mediates prostate cancer therapy resistance. Cancer Res. 2008, 68, 5469–5477. [Google Scholar] [CrossRef]
- Antonarakis, E.S.; Lu, C.; Wang, H.; Luber, B.; Nakazawa, M.; Roeser, J.C.; Chen, Y.; Mohammad, T.A.; Chen, Y.; Fedor, H.L.; et al. AR-V7 and resistance to enzalutamide and abiraterone in prostate cancer. N. Engl. J. Med. 2014, 371, 1028–1038. [Google Scholar] [CrossRef]
- Thiebaut, C.; Konan, H.P.; Guerquin, M.J.; Chesnel, A.; Livera, G.; Le Romancer, M.; Dumond, H. The Role of ERalpha36 in Development and Tumor Malignancy. Int. J. Mol. Sci. 2020, 21, 4116. [Google Scholar] [CrossRef] [PubMed]
- Wang, Y.; Bernhardy, A.J.; Cruz, C.; Krais, J.J.; Nacson, J.; Nicolas, E.; Peri, S.; van der Gulden, H.; van der Heijden, I.; O’Brien, S.W.; et al. The BRCA1-Delta11q Alternative Splice Isoform Bypasses Germline Mutations and Promotes Therapeutic Resistance to PARP Inhibition and Cisplatin. Cancer Res. 2016, 76, 2778–2790. [Google Scholar] [CrossRef] [PubMed]
- Liu, J.; Bhadra, M.; Sinnakannu, J.R.; Yue, W.L.; Tan, C.W.; Rigo, F.; Ong, S.T.; Roca, X. Overcoming imatinib resistance conferred by the BIM deletion polymorphism in chronic myeloid leukemia with splice-switching antisense oligonucleotides. Oncotarget 2017, 8, 77567–77585. [Google Scholar] [CrossRef] [PubMed]
- Ng, K.P.; Hillmer, A.M.; Chuah, C.T.; Juan, W.C.; Ko, T.K.; Teo, A.S.; Ariyaratne, P.N.; Takahashi, N.; Sawada, K.; Fei, Y.; et al. A common BIM deletion polymorphism mediates intrinsic resistance and inferior responses to tyrosine kinase inhibitors in cancer. Nat. Med. 2012, 18, 521–528. [Google Scholar] [CrossRef]
- Asnani, M.; Hayer, K.E.; Naqvi, A.S.; Zheng, S.; Yang, S.Y.; Oldridge, D.; Ibrahim, F.; Maragkakis, M.; Gazzara, M.R.; Black, K.L.; et al. Retention of CD19 intron 2 contributes to CART-19 resistance in leukemias with subclonal frameshift mutations in CD19. Leukemia 2020, 34, 1202–1207. [Google Scholar] [CrossRef]
- Sotillo, E.; Barrett, D.M.; Black, K.L.; Bagashev, A.; Oldridge, D.; Wu, G.; Sussman, R.; Lanauze, C.; Ruella, M.; Gazzara, M.R.; et al. Convergence of Acquired Mutations and Alternative Splicing of CD19 Enables Resistance to CART-19 Immunotherapy. Cancer Discov. 2015, 5, 1282–1295. [Google Scholar] [CrossRef]
- Mitra, D.; Brumlik, M.J.; Okamgba, S.U.; Zhu, Y.; Duplessis, T.T.; Parvani, J.G.; Lesko, S.M.; Brogi, E.; Jones, F.E. An oncogenic isoform of HER2 associated with locally disseminated breast cancer and trastuzumab resistance. Mol. Cancer Ther. 2009, 8, 2152–2162. [Google Scholar] [CrossRef]
- Martinez-Martin, S.; Soucek, L. MYC inhibitors in multiple myeloma. Cancer Drug Resist. 2021, 4, 842–865. [Google Scholar] [CrossRef]
Disclaimer/Publisher’s Note: The statements, opinions and data contained in all publications are solely those of the individual author(s) and contributor(s) and not of MDPI and/or the editor(s). MDPI and/or the editor(s) disclaim responsibility for any injury to people or property resulting from any ideas, methods, instructions or products referred to in the content. |
© 2023 by the authors. Licensee MDPI, Basel, Switzerland. This article is an open access article distributed under the terms and conditions of the Creative Commons Attribution (CC BY) license (https://creativecommons.org/licenses/by/4.0/).
Share and Cite
Wojtyś, W.; Oroń, M. How Driver Oncogenes Shape and Are Shaped by Alternative Splicing Mechanisms in Tumors. Cancers 2023, 15, 2918. https://doi.org/10.3390/cancers15112918
Wojtyś W, Oroń M. How Driver Oncogenes Shape and Are Shaped by Alternative Splicing Mechanisms in Tumors. Cancers. 2023; 15(11):2918. https://doi.org/10.3390/cancers15112918
Chicago/Turabian StyleWojtyś, Weronika, and Magdalena Oroń. 2023. "How Driver Oncogenes Shape and Are Shaped by Alternative Splicing Mechanisms in Tumors" Cancers 15, no. 11: 2918. https://doi.org/10.3390/cancers15112918
APA StyleWojtyś, W., & Oroń, M. (2023). How Driver Oncogenes Shape and Are Shaped by Alternative Splicing Mechanisms in Tumors. Cancers, 15(11), 2918. https://doi.org/10.3390/cancers15112918