Skip to main content
Top
Published in: Molecular Cancer 1/2017

Open Access 01-12-2017 | Review

EMT and stemness: flexible processes tuned by alternative splicing in development and cancer progression

Authors: Davide Pradella, Chiara Naro, Claudio Sette, Claudia Ghigna

Published in: Molecular Cancer | Issue 1/2017

Login to get access

Abstract

Epithelial-to-mesenchymal transition (EMT) is associated with metastasis formation as well as with generation and maintenance of cancer stem cells. In this way, EMT contributes to tumor invasion, heterogeneity and chemoresistance. Morphological and functional changes involved in these processes require robust reprogramming of gene expression, which is only partially accomplished at the transcriptional level. Alternative splicing is another essential layer of gene expression regulation that expands the cell proteome. This step in post-transcriptional regulation of gene expression tightly controls cell identity between epithelial and mesenchymal states and during stem cell differentiation. Importantly, dysregulation of splicing factor function and cancer-specific splicing isoform expression frequently occurs in human tumors, suggesting the importance of alternative splicing regulation for cancer biology.
In this review, we briefly discuss the role of EMT programs in development, stem cell differentiation and cancer progression. Next, we focus on selected examples of key factors involved in EMT and stem cell differentiation that are regulated post-transcriptionally through alternative splicing mechanisms. Lastly, we describe relevant oncogenic splice-variants that directly orchestrate cancer stem cell biology and tumor EMT, which may be envisioned as novel targets for therapeutic intervention.
Literature
2.
go back to reference Gavert N, Ben-Ze’ev A. Epithelial–mesenchymal transition and the invasive potential of tumors. Trends Mol Med. 2008;14:199–209.PubMedCrossRef Gavert N, Ben-Ze’ev A. Epithelial–mesenchymal transition and the invasive potential of tumors. Trends Mol Med. 2008;14:199–209.PubMedCrossRef
3.
go back to reference Thiery J-P, Acloque H, Huang RYJ, Nieto MA. Epithelial-mesenchymal transitions in development and disease. Cell. 2009;139:871–90.PubMedCrossRef Thiery J-P, Acloque H, Huang RYJ, Nieto MA. Epithelial-mesenchymal transitions in development and disease. Cell. 2009;139:871–90.PubMedCrossRef
5.
go back to reference Scheel C, Weinberg RA. Cancer stem cells and epithelial-mesenchymal transition: concepts and molecular links. Semin Cancer Biol. 2012;22:396–403.PubMedCrossRef Scheel C, Weinberg RA. Cancer stem cells and epithelial-mesenchymal transition: concepts and molecular links. Semin Cancer Biol. 2012;22:396–403.PubMedCrossRef
6.
go back to reference Visvader JE, Lindeman GJ. Cancer stem cells: current status and evolving complexities. Cell Stem Cell. 2012;10:717–28.PubMedCrossRef Visvader JE, Lindeman GJ. Cancer stem cells: current status and evolving complexities. Cell Stem Cell. 2012;10:717–28.PubMedCrossRef
8.
go back to reference Sveen A, Kilpinen S, Ruusulehto A, Lothe RA, Skotheim RI. Aberrant RNA splicing in cancer; expression changes and driver mutations of splicing factor genes. Oncogene. 2016;35:2413–27.PubMedCrossRef Sveen A, Kilpinen S, Ruusulehto A, Lothe RA, Skotheim RI. Aberrant RNA splicing in cancer; expression changes and driver mutations of splicing factor genes. Oncogene. 2016;35:2413–27.PubMedCrossRef
10.
go back to reference Bonomi S, Gallo S, Catillo M, Pignataro D, Biamonti G, Ghigna C. Oncogenic alternative splicing switches: role in cancer progression and prospects for therapy. Int J Cell Biol. 2013;2013:962038.PubMedPubMedCentralCrossRef Bonomi S, Gallo S, Catillo M, Pignataro D, Biamonti G, Ghigna C. Oncogenic alternative splicing switches: role in cancer progression and prospects for therapy. Int J Cell Biol. 2013;2013:962038.PubMedPubMedCentralCrossRef
11.
go back to reference Paronetto MP, Passacantilli I, Sette C. Alternative splicing and cell survival: from tissue homeostasis to disease. Cell Death Differ. 2016;23:1919–29.PubMedCrossRef Paronetto MP, Passacantilli I, Sette C. Alternative splicing and cell survival: from tissue homeostasis to disease. Cell Death Differ. 2016;23:1919–29.PubMedCrossRef
12.
go back to reference Sebestyén E, Singh B, Miñana B, Pagès A, Mateo F, Pujana MA, et al. Large-scale analysis of genome and transcriptome alterations in multiple tumors unveils novel cancer-relevant splicing networks. Genome Res. 2016;26:732–44.PubMedPubMedCentralCrossRef Sebestyén E, Singh B, Miñana B, Pagès A, Mateo F, Pujana MA, et al. Large-scale analysis of genome and transcriptome alterations in multiple tumors unveils novel cancer-relevant splicing networks. Genome Res. 2016;26:732–44.PubMedPubMedCentralCrossRef
13.
go back to reference Biamonti G, Bonomi S, Gallo S, Ghigna C. Making alternative splicing decisions during epithelial-to-mesenchymal transition (EMT). Cell Mol Life Sci. 2012;69:2515–26.PubMedCrossRef Biamonti G, Bonomi S, Gallo S, Ghigna C. Making alternative splicing decisions during epithelial-to-mesenchymal transition (EMT). Cell Mol Life Sci. 2012;69:2515–26.PubMedCrossRef
14.
go back to reference Yang Y, Park JW, Bebee TW, Warzecha CC, Guo Y, Shang X, et al. Determination of a comprehensive alternative splicing regulatory network and combinatorial regulation by key factors during the epithelial-to-mesenchymal transition. Mol Cell Biol. 2016;36:1704–19.PubMedPubMedCentralCrossRef Yang Y, Park JW, Bebee TW, Warzecha CC, Guo Y, Shang X, et al. Determination of a comprehensive alternative splicing regulatory network and combinatorial regulation by key factors during the epithelial-to-mesenchymal transition. Mol Cell Biol. 2016;36:1704–19.PubMedPubMedCentralCrossRef
15.
go back to reference Le K, Mitsouras K, Roy M, Wang Q, Xu Q, Nelson SF, et al. Detecting tissue-specific regulation of alternative splicing as a qualitative change in microarray data. Nucleic Acids Res. 2004;32:e180.PubMedPubMedCentralCrossRef Le K, Mitsouras K, Roy M, Wang Q, Xu Q, Nelson SF, et al. Detecting tissue-specific regulation of alternative splicing as a qualitative change in microarray data. Nucleic Acids Res. 2004;32:e180.PubMedPubMedCentralCrossRef
16.
go back to reference Pan Q, Shai O, Misquitta C, Zhang W, Saltzman AL, Mohammad N, et al. Revealing global regulatory features of mammalian alternative splicing using a quantitative microarray platform. Mol Cell. 2004;16:929–41.PubMedCrossRef Pan Q, Shai O, Misquitta C, Zhang W, Saltzman AL, Mohammad N, et al. Revealing global regulatory features of mammalian alternative splicing using a quantitative microarray platform. Mol Cell. 2004;16:929–41.PubMedCrossRef
17.
go back to reference Nakaya Y, Sheng G. Epithelial to mesenchymal transition during gastrulation: an embryological view. Develop Growth Differ. 2008;50:755–66.CrossRef Nakaya Y, Sheng G. Epithelial to mesenchymal transition during gastrulation: an embryological view. Develop Growth Differ. 2008;50:755–66.CrossRef
18.
go back to reference Kalcheim C. Epithelial-mesenchymal transitions during neural crest and somite development. J Clin Med. 2015;5. Kalcheim C. Epithelial-mesenchymal transitions during neural crest and somite development. J Clin Med. 2015;5.
20.
go back to reference Lim J, Thiery JP. Epithelial-mesenchymal transitions: insights from development. Development. 2012;139:3471–86.PubMedCrossRef Lim J, Thiery JP. Epithelial-mesenchymal transitions: insights from development. Development. 2012;139:3471–86.PubMedCrossRef
21.
go back to reference Skromne I, Stern CD. Interactions between Wnt and Vg1 signalling pathways initiate primitive streak formation in the chick embryo. Development. 2001;128:2915–27.PubMed Skromne I, Stern CD. Interactions between Wnt and Vg1 signalling pathways initiate primitive streak formation in the chick embryo. Development. 2001;128:2915–27.PubMed
22.
go back to reference Groves AK, LaBonne C. Setting appropriate boundaries: fate, patterning and competence at the neural plate border. Dev Biol. 2014;389:2–12.PubMedCrossRef Groves AK, LaBonne C. Setting appropriate boundaries: fate, patterning and competence at the neural plate border. Dev Biol. 2014;389:2–12.PubMedCrossRef
23.
go back to reference Carver EA, Jiang R, Lan Y, Oram KF, Gridley T. The mouse snail gene encodes a key regulator of the epithelial-mesenchymal transition. Mol Cell Biol. 2001;21:8184–8.PubMedPubMedCentralCrossRef Carver EA, Jiang R, Lan Y, Oram KF, Gridley T. The mouse snail gene encodes a key regulator of the epithelial-mesenchymal transition. Mol Cell Biol. 2001;21:8184–8.PubMedPubMedCentralCrossRef
24.
go back to reference Tien CL, Jones A, Wang H, Gerigk M, Nozell S, Chang C. Snail2/Slug cooperates with Polycomb repressive complex 2 (PRC2) to regulate neural crest development. Development. 2015;142:722–31.PubMedPubMedCentralCrossRef Tien CL, Jones A, Wang H, Gerigk M, Nozell S, Chang C. Snail2/Slug cooperates with Polycomb repressive complex 2 (PRC2) to regulate neural crest development. Development. 2015;142:722–31.PubMedPubMedCentralCrossRef
25.
go back to reference Nakagawa S, Takeichi M. Neural crest emigration from the neural tube depends on regulated cadherin expression. Development. 1998;125:2963–71.PubMed Nakagawa S, Takeichi M. Neural crest emigration from the neural tube depends on regulated cadherin expression. Development. 1998;125:2963–71.PubMed
26.
go back to reference Vallin J, Girault JM, Thiery JP, Broders F. Xenopus cadherin-11 is expressed in different populations of migrating neural crest cells. Mech Dev. 1998;75:171–4.PubMedCrossRef Vallin J, Girault JM, Thiery JP, Broders F. Xenopus cadherin-11 is expressed in different populations of migrating neural crest cells. Mech Dev. 1998;75:171–4.PubMedCrossRef
27.
28.
go back to reference Scarpa E, Szabó A, Bibonne A, Theveneau E, Parsons M, Mayor R. Cadherin switch during EMT in neural crest cells leads to contact inhibition of locomotion via repolarization of forces. Dev Cell. 2015;34:421–34.PubMedPubMedCentralCrossRef Scarpa E, Szabó A, Bibonne A, Theveneau E, Parsons M, Mayor R. Cadherin switch during EMT in neural crest cells leads to contact inhibition of locomotion via repolarization of forces. Dev Cell. 2015;34:421–34.PubMedPubMedCentralCrossRef
29.
go back to reference Nistico P, Bissell MJ, Radisky DC. Epithelial-mesenchymal transition: general principles and pathological relevance with special emphasis on the role of matrix metalloproteinases. Cold Spring Harb Perspect Biol. 2012;4:a011908–8.PubMedPubMedCentralCrossRef Nistico P, Bissell MJ, Radisky DC. Epithelial-mesenchymal transition: general principles and pathological relevance with special emphasis on the role of matrix metalloproteinases. Cold Spring Harb Perspect Biol. 2012;4:a011908–8.PubMedPubMedCentralCrossRef
30.
go back to reference Duong TD, Erickson CA. MMP-2 plays an essential role in producing epithelial-mesenchymal transformations in the avian embryo. Dev Dyn. 2004;229:42–53.PubMedCrossRef Duong TD, Erickson CA. MMP-2 plays an essential role in producing epithelial-mesenchymal transformations in the avian embryo. Dev Dyn. 2004;229:42–53.PubMedCrossRef
31.
go back to reference Niessen CM. Tight junctions/adherens junctions: basic structure and function. J Invest Dermatol. 2007;127:2525–32.PubMedCrossRef Niessen CM. Tight junctions/adherens junctions: basic structure and function. J Invest Dermatol. 2007;127:2525–32.PubMedCrossRef
32.
go back to reference Shin K, Fogg VC, Margolis B. Tight junctions and cell polarity. Annu Rev Cell Dev Biol. 2006;22:207–35.PubMedCrossRef Shin K, Fogg VC, Margolis B. Tight junctions and cell polarity. Annu Rev Cell Dev Biol. 2006;22:207–35.PubMedCrossRef
33.
go back to reference Zihni C, Mills C, Matter K, Balda MS. Tight junctions: from simple barriersto multifunctional molecular gates. Nat Rev Mol Cell Biol. 2016;17:564–80.PubMedCrossRef Zihni C, Mills C, Matter K, Balda MS. Tight junctions: from simple barriersto multifunctional molecular gates. Nat Rev Mol Cell Biol. 2016;17:564–80.PubMedCrossRef
34.
go back to reference Niessen CM, Gottardi CJ. Molecular components of the adherens junction. Biochim Biophys Acta. 1778;2008:562–71. Niessen CM, Gottardi CJ. Molecular components of the adherens junction. Biochim Biophys Acta. 1778;2008:562–71.
36.
37.
go back to reference Oas RG, Nanes BA, Esimai CC, Vincent PA, García AJ, Kowalczyk AP. p120-catenin and β-catenin differentially regulate cadherin adhesive function. Mol Biol Cell. 2013;24:704–14.PubMedPubMedCentralCrossRef Oas RG, Nanes BA, Esimai CC, Vincent PA, García AJ, Kowalczyk AP. p120-catenin and β-catenin differentially regulate cadherin adhesive function. Mol Biol Cell. 2013;24:704–14.PubMedPubMedCentralCrossRef
38.
go back to reference Harris TJC, Tepass U. Adherens junctions: from moleculesto morphogenesis. Nat Rev Mol Cell Biol. 2010;11:502–14.PubMedCrossRef Harris TJC, Tepass U. Adherens junctions: from moleculesto morphogenesis. Nat Rev Mol Cell Biol. 2010;11:502–14.PubMedCrossRef
39.
go back to reference Ikenouchi J, Matsuda M, Furuse M, Tsukita S. Regulation of tight junctions during the epithelium-mesenchyme transition: direct repression of the gene expression of claudins/occludin by Snail. J Cell Sci. 2003;116:1959–67.PubMedCrossRef Ikenouchi J, Matsuda M, Furuse M, Tsukita S. Regulation of tight junctions during the epithelium-mesenchyme transition: direct repression of the gene expression of claudins/occludin by Snail. J Cell Sci. 2003;116:1959–67.PubMedCrossRef
40.
go back to reference Huang RYJ, Guilford P, Thiery JP. Early events in cell adhesion and polarity during epithelial-mesenchymal transition. J Cell Sci. 2012;125:4417–22.PubMedCrossRef Huang RYJ, Guilford P, Thiery JP. Early events in cell adhesion and polarity during epithelial-mesenchymal transition. J Cell Sci. 2012;125:4417–22.PubMedCrossRef
41.
go back to reference Cano A, Pérez-Moreno MA, Rodrigo I, Locascio A, Blanco MJ, del Barrio MG, et al. The transcription factor snail controls epithelial-mesenchymal transitions by repressing E-cadherin expression. Nat Cell Biol. 2000;2:76–83.PubMedCrossRef Cano A, Pérez-Moreno MA, Rodrigo I, Locascio A, Blanco MJ, del Barrio MG, et al. The transcription factor snail controls epithelial-mesenchymal transitions by repressing E-cadherin expression. Nat Cell Biol. 2000;2:76–83.PubMedCrossRef
42.
go back to reference Grieve AG, Rabouille C. Extracellular cleavage of E-cadherin promotes epithelial cell extrusion. J Cell Sci. 2014;127:3331–46.PubMedCrossRef Grieve AG, Rabouille C. Extracellular cleavage of E-cadherin promotes epithelial cell extrusion. J Cell Sci. 2014;127:3331–46.PubMedCrossRef
43.
go back to reference Palacios F, Tushir JS, Fujita Y, D'Souza-Schorey C. Lysosomal targeting of E-cadherin: a unique mechanism for the down-regulation of cell-cell adhesion during epithelial to mesenchymal transitions. Mol Cell Biol. 2005;25:389–402.PubMedPubMedCentralCrossRef Palacios F, Tushir JS, Fujita Y, D'Souza-Schorey C. Lysosomal targeting of E-cadherin: a unique mechanism for the down-regulation of cell-cell adhesion during epithelial to mesenchymal transitions. Mol Cell Biol. 2005;25:389–402.PubMedPubMedCentralCrossRef
44.
go back to reference Yang J, Mani SA, Donaher JL, Ramaswamy S, Itzykson RA, Come C, et al. Twist, a master regulator of morphogenesis, plays an essential role in tumor metastasis. Cell. 2004;117:927–39.PubMedCrossRef Yang J, Mani SA, Donaher JL, Ramaswamy S, Itzykson RA, Come C, et al. Twist, a master regulator of morphogenesis, plays an essential role in tumor metastasis. Cell. 2004;117:927–39.PubMedCrossRef
46.
go back to reference Moreno-Bueno G, Portillo F, Cano A. Transcriptional regulation of cell polarity in EMT and cancer. Oncogene. 2008;27:6958–69.PubMedCrossRef Moreno-Bueno G, Portillo F, Cano A. Transcriptional regulation of cell polarity in EMT and cancer. Oncogene. 2008;27:6958–69.PubMedCrossRef
47.
go back to reference Nelson WJ. Remodeling epithelial cell organization: transitions between front-rear and apical-basal polarity. Cold Spring Harb Perspect Biol. 2009;1:a000513.PubMedPubMedCentralCrossRef Nelson WJ. Remodeling epithelial cell organization: transitions between front-rear and apical-basal polarity. Cold Spring Harb Perspect Biol. 2009;1:a000513.PubMedPubMedCentralCrossRef
49.
go back to reference Jordan NV, Johnson GL, Abell AN. Tracking the intermediate stages of epithelial-mesenchymal transition in epithelial stem cells and cancer. Cell Cycle. 2011;10:2865–73.PubMedPubMedCentralCrossRef Jordan NV, Johnson GL, Abell AN. Tracking the intermediate stages of epithelial-mesenchymal transition in epithelial stem cells and cancer. Cell Cycle. 2011;10:2865–73.PubMedPubMedCentralCrossRef
50.
go back to reference Lee JM, Dedhar S, Kalluri R, Thompson EW. The epithelial-mesenchymal transition: new insights in signaling, development, and disease. J Cell Biol. 2006;172:973–81.PubMedPubMedCentralCrossRef Lee JM, Dedhar S, Kalluri R, Thompson EW. The epithelial-mesenchymal transition: new insights in signaling, development, and disease. J Cell Biol. 2006;172:973–81.PubMedPubMedCentralCrossRef
51.
go back to reference Jolly MK, Boareto M, Huang B, Jia D, Lu M, Ben-Jacob E, et al. Implications of the hybrid epithelial/mesenchymal phenotype in metastasis. Front Oncol. 2015;5:155.PubMedPubMedCentralCrossRef Jolly MK, Boareto M, Huang B, Jia D, Lu M, Ben-Jacob E, et al. Implications of the hybrid epithelial/mesenchymal phenotype in metastasis. Front Oncol. 2015;5:155.PubMedPubMedCentralCrossRef
52.
54.
55.
go back to reference Pantel K, Woelfle U. Micrometastasis in breast cancer and other solid tumors. J Biol Regul Homeost Agents. 2004;18:120–5.PubMed Pantel K, Woelfle U. Micrometastasis in breast cancer and other solid tumors. J Biol Regul Homeost Agents. 2004;18:120–5.PubMed
56.
go back to reference Rhim AD, Mirek ET, Aiello NM, Maitra A, Bailey JM, McAllister F, et al. EMT and dissemination precede pancreatic tumor formation. Cell. 2012;148:349–61.PubMedPubMedCentralCrossRef Rhim AD, Mirek ET, Aiello NM, Maitra A, Bailey JM, McAllister F, et al. EMT and dissemination precede pancreatic tumor formation. Cell. 2012;148:349–61.PubMedPubMedCentralCrossRef
57.
go back to reference Hugo H, Ackland ML, Blick T, Lawrence MG, Clements JA, Williams ED, et al. Epithelial--mesenchymal and mesenchymal--epithelial transitions in carcinoma progression. J Cell Physiol. 2007;213:374–83.PubMedCrossRef Hugo H, Ackland ML, Blick T, Lawrence MG, Clements JA, Williams ED, et al. Epithelial--mesenchymal and mesenchymal--epithelial transitions in carcinoma progression. J Cell Physiol. 2007;213:374–83.PubMedCrossRef
58.
go back to reference Yang J, Weinberg RA. Epithelial-mesenchymal transition: at the crossroads of development and tumor metastasis. Dev Cell. 2008;14:818–29.PubMedCrossRef Yang J, Weinberg RA. Epithelial-mesenchymal transition: at the crossroads of development and tumor metastasis. Dev Cell. 2008;14:818–29.PubMedCrossRef
59.
go back to reference Balkwill FR, Capasso M, Hagemann T. The tumor microenvironment at a glance. J Cell Sci. 2012;125:5591–6.PubMedCrossRef Balkwill FR, Capasso M, Hagemann T. The tumor microenvironment at a glance. J Cell Sci. 2012;125:5591–6.PubMedCrossRef
61.
go back to reference Davies KJ. The complex interaction of matrix metalloproteinases in the migration of cancer cells through breast tissue stroma. Int J Breast Cancer. 2014;2014:839094.PubMedPubMedCentral Davies KJ. The complex interaction of matrix metalloproteinases in the migration of cancer cells through breast tissue stroma. Int J Breast Cancer. 2014;2014:839094.PubMedPubMedCentral
62.
go back to reference Chatterjee S, Seifried L, Feigin ME, Gibbons DL, Scuoppo C, Lin W, et al. Dysregulation of cell polarity proteins synergize with oncogenes or the microenvironment to induce invasive behavior in epithelial cells. Plos ONE. 2012;7:e34343.PubMedPubMedCentralCrossRef Chatterjee S, Seifried L, Feigin ME, Gibbons DL, Scuoppo C, Lin W, et al. Dysregulation of cell polarity proteins synergize with oncogenes or the microenvironment to induce invasive behavior in epithelial cells. Plos ONE. 2012;7:e34343.PubMedPubMedCentralCrossRef
63.
go back to reference Lee M, Vasioukhin V. Cell polarity and cancer – cell and tissue polarity as a non-canonical tumor suppressor. J Cell Sci. 2008;121:1141–50.PubMedCrossRef Lee M, Vasioukhin V. Cell polarity and cancer – cell and tissue polarity as a non-canonical tumor suppressor. J Cell Sci. 2008;121:1141–50.PubMedCrossRef
64.
go back to reference Radisky ES, Radisky DC. Matrix metalloproteinase-induced epithelial-mesenchymal transition in breast cancer. J Mammary Gland Biol Neoplasia. 2010;15:201–12.PubMedPubMedCentralCrossRef Radisky ES, Radisky DC. Matrix metalloproteinase-induced epithelial-mesenchymal transition in breast cancer. J Mammary Gland Biol Neoplasia. 2010;15:201–12.PubMedPubMedCentralCrossRef
65.
go back to reference Finak G, Bertos N, Pepin F, Sadekova S, Souleimanova M, Zhao H, et al. Stromal gene expression predicts clinical outcome in breast cancer. Nat Med. 2008;14:518–27.PubMedCrossRef Finak G, Bertos N, Pepin F, Sadekova S, Souleimanova M, Zhao H, et al. Stromal gene expression predicts clinical outcome in breast cancer. Nat Med. 2008;14:518–27.PubMedCrossRef
66.
go back to reference Noë V, Fingleton B, Jacobs K, Crawford HC, Vermeulen S, Steelant W, et al. Release of an invasion promoter E-cadherin fragment by matrilysin and stromelysin-1. J Cell Sci. 2001;114:111–8.PubMed Noë V, Fingleton B, Jacobs K, Crawford HC, Vermeulen S, Steelant W, et al. Release of an invasion promoter E-cadherin fragment by matrilysin and stromelysin-1. J Cell Sci. 2001;114:111–8.PubMed
67.
go back to reference Hu X, Li D, Zhang W, Zhou J, Tang B, Li L. Matrix metalloproteinase-9 expression correlates with prognosis and involved in ovarian cancer cell invasion. Arch Gynecol Obstet. 2012;286:1537–43.PubMedCrossRef Hu X, Li D, Zhang W, Zhou J, Tang B, Li L. Matrix metalloproteinase-9 expression correlates with prognosis and involved in ovarian cancer cell invasion. Arch Gynecol Obstet. 2012;286:1537–43.PubMedCrossRef
68.
go back to reference Li HC, Cao DC, Liu Y, Hou YF, Wu J, Lu JS, et al. Prognostic value of matrix metalloproteinases (MMP-2 and MMP-9) in patients with lymph node-negative breast carcinoma. Breast Cancer Res Treat. 2004;88:75–85.PubMedCrossRef Li HC, Cao DC, Liu Y, Hou YF, Wu J, Lu JS, et al. Prognostic value of matrix metalloproteinases (MMP-2 and MMP-9) in patients with lymph node-negative breast carcinoma. Breast Cancer Res Treat. 2004;88:75–85.PubMedCrossRef
69.
go back to reference Inoue T, Yashiro M, Nishimura S, Maeda K, Sawada T, Ogawa Y, et al. Matrix metalloproteinase-1 expression is a prognostic factor for patients with advanced gastric cancer. Int J Mol Med. 1999;4:73–7.PubMed Inoue T, Yashiro M, Nishimura S, Maeda K, Sawada T, Ogawa Y, et al. Matrix metalloproteinase-1 expression is a prognostic factor for patients with advanced gastric cancer. Int J Mol Med. 1999;4:73–7.PubMed
70.
go back to reference Zucker S, Vacirca J. Role of matrix metalloproteinases (MMPs) in colorectal cancer. Cancer Metastasis Rev. 2004;23:101–17.PubMedCrossRef Zucker S, Vacirca J. Role of matrix metalloproteinases (MMPs) in colorectal cancer. Cancer Metastasis Rev. 2004;23:101–17.PubMedCrossRef
71.
go back to reference Smit MA, Peeper DS. Epithelial-mesenchymal transition and senescence: two cancer-related processes are crossing paths. Aging (Albany NY). 2010;2:735–41.CrossRef Smit MA, Peeper DS. Epithelial-mesenchymal transition and senescence: two cancer-related processes are crossing paths. Aging (Albany NY). 2010;2:735–41.CrossRef
72.
go back to reference Fischer KR, Durrans A, Lee S, Sheng J, Li F, Wong STC, et al. Epithelial-to-mesenchymal transitionis not required for lung metastasis butcontributes to chemoresistance. Nature. 2015;527:472–6.PubMedPubMedCentralCrossRef Fischer KR, Durrans A, Lee S, Sheng J, Li F, Wong STC, et al. Epithelial-to-mesenchymal transitionis not required for lung metastasis butcontributes to chemoresistance. Nature. 2015;527:472–6.PubMedPubMedCentralCrossRef
73.
go back to reference Zheng X, Carstens JL, Kim J, Scheible M, Kaye J, Sugimoto H, et al. Epithelial-to-mesenchymal transitionis dispensable for metastasis but induceschemoresistance in pancreatic cancer. Nature. 2015;527:525–30.PubMedPubMedCentralCrossRef Zheng X, Carstens JL, Kim J, Scheible M, Kaye J, Sugimoto H, et al. Epithelial-to-mesenchymal transitionis dispensable for metastasis but induceschemoresistance in pancreatic cancer. Nature. 2015;527:525–30.PubMedPubMedCentralCrossRef
74.
go back to reference Ohashi S, Natsuizaka M, Wong GS, Michaylira CZ, Grugan KD, Stairs DB, et al. Epidermal growth factor receptor and mutant p53 expand an esophageal cellular subpopulation capable of epithelial-to-mesenchymal transition through ZEB transcription factors. Cancer Res. 2010;70:4174–84.PubMedPubMedCentralCrossRef Ohashi S, Natsuizaka M, Wong GS, Michaylira CZ, Grugan KD, Stairs DB, et al. Epidermal growth factor receptor and mutant p53 expand an esophageal cellular subpopulation capable of epithelial-to-mesenchymal transition through ZEB transcription factors. Cancer Res. 2010;70:4174–84.PubMedPubMedCentralCrossRef
75.
go back to reference Ansieau S, Bastid J, Doreau A, Morel AP, Bouchet BP, Thomas C, et al. Induction of EMT by twist proteins as a collateral effect of tumor-promoting inactivation of premature senescence. Cancer Cell. 2008;14:79–89.PubMedCrossRef Ansieau S, Bastid J, Doreau A, Morel AP, Bouchet BP, Thomas C, et al. Induction of EMT by twist proteins as a collateral effect of tumor-promoting inactivation of premature senescence. Cancer Cell. 2008;14:79–89.PubMedCrossRef
76.
go back to reference Robson EJD, Khaled WT, Abell K, Watson CJ. Epithelial-to-mesenchymal transition confers resistance to apoptosis in three murine mammary epithelial cell lines. Differentiation. 2006;74:254–64.PubMedCrossRef Robson EJD, Khaled WT, Abell K, Watson CJ. Epithelial-to-mesenchymal transition confers resistance to apoptosis in three murine mammary epithelial cell lines. Differentiation. 2006;74:254–64.PubMedCrossRef
77.
go back to reference Franco DL, Mainez J, Vega S, Sancho P, Murillo MM, de Frutos CA, et al. Snail1 suppresses TGF-beta-induced apoptosis and is sufficient to trigger EMT in hepatocytes. J Cell Sci. 2010;123:3467–77.PubMedCrossRef Franco DL, Mainez J, Vega S, Sancho P, Murillo MM, de Frutos CA, et al. Snail1 suppresses TGF-beta-induced apoptosis and is sufficient to trigger EMT in hepatocytes. J Cell Sci. 2010;123:3467–77.PubMedCrossRef
78.
go back to reference Creighton CJ, Li X, Landis M, Dixon JM, Neumeister VM, Sjolund A, et al. Residual breast cancers after conventional therapy display mesenchymal as well as tumor-initiating features. Proc Natl Acad Sci U S A. 2009;106:13820–5.PubMedPubMedCentralCrossRef Creighton CJ, Li X, Landis M, Dixon JM, Neumeister VM, Sjolund A, et al. Residual breast cancers after conventional therapy display mesenchymal as well as tumor-initiating features. Proc Natl Acad Sci U S A. 2009;106:13820–5.PubMedPubMedCentralCrossRef
79.
go back to reference Kawamoto A, Yokoe T, Tanaka K, Saigusa S, Toiyama Y, Yasuda H, et al. Radiation induces epithelial-mesenchymal transition in colorectal cancer cells. Oncol Rep. 2012;27:51–7.PubMed Kawamoto A, Yokoe T, Tanaka K, Saigusa S, Toiyama Y, Yasuda H, et al. Radiation induces epithelial-mesenchymal transition in colorectal cancer cells. Oncol Rep. 2012;27:51–7.PubMed
80.
go back to reference Shintani Y, Okimura A, Sato K, Nakagiri T, Kadota Y, Inoue M, et al. Epithelial to mesenchymal transition is a determinant of sensitivity to chemoradiotherapy in non-small cell lung cancer. Ann Thorac Surg. 2011;92:1794–804.PubMedCrossRef Shintani Y, Okimura A, Sato K, Nakagiri T, Kadota Y, Inoue M, et al. Epithelial to mesenchymal transition is a determinant of sensitivity to chemoradiotherapy in non-small cell lung cancer. Ann Thorac Surg. 2011;92:1794–804.PubMedCrossRef
81.
go back to reference Wellner U, Brabletz T, Keck T. ZEB1 in pancreatic cancer. Cancers (Basel). 2010;2:1617–28.CrossRef Wellner U, Brabletz T, Keck T. ZEB1 in pancreatic cancer. Cancers (Basel). 2010;2:1617–28.CrossRef
82.
go back to reference Kurrey NK, Jalgaonkar SP, Joglekar AV, Ghanate AD, Chaskar PD, Doiphode RY, et al. Snail and slug mediate radioresistance and chemoresistance by antagonizing p53-mediated apoptosis and acquiring a stem-like phenotype in ovarian cancer cells. Stem Cells. 2009;27:2059–68.PubMedCrossRef Kurrey NK, Jalgaonkar SP, Joglekar AV, Ghanate AD, Chaskar PD, Doiphode RY, et al. Snail and slug mediate radioresistance and chemoresistance by antagonizing p53-mediated apoptosis and acquiring a stem-like phenotype in ovarian cancer cells. Stem Cells. 2009;27:2059–68.PubMedCrossRef
83.
go back to reference Brabletz T, Jung A, Reu S, Porzner M, Hlubek F, Kunz-Schughart LA, et al. Variable beta-catenin expression in colorectal cancers indicates tumor progression driven by the tumor environment. Proc Natl Acad Sci U S A. 2001;98:10356–61.PubMedPubMedCentralCrossRef Brabletz T, Jung A, Reu S, Porzner M, Hlubek F, Kunz-Schughart LA, et al. Variable beta-catenin expression in colorectal cancers indicates tumor progression driven by the tumor environment. Proc Natl Acad Sci U S A. 2001;98:10356–61.PubMedPubMedCentralCrossRef
84.
go back to reference Yu M, Bardia A, Wittner BS, Stott SL, Smas ME, Ting DT, et al. Circulating breast tumor cells exhibit dynamic changes in epithelial and mesenchymal composition. Science. 2013;339:580–4.PubMedPubMedCentralCrossRef Yu M, Bardia A, Wittner BS, Stott SL, Smas ME, Ting DT, et al. Circulating breast tumor cells exhibit dynamic changes in epithelial and mesenchymal composition. Science. 2013;339:580–4.PubMedPubMedCentralCrossRef
85.
go back to reference Strauss R, Sova P, Liu Y, Li ZY, Tuve S, Pritchard D, et al. Epithelial phenotype confers resistance of ovarian cancer cells to oncolytic adenoviruses. Cancer Res. 2009;69:5115–25.PubMedPubMedCentralCrossRef Strauss R, Sova P, Liu Y, Li ZY, Tuve S, Pritchard D, et al. Epithelial phenotype confers resistance of ovarian cancer cells to oncolytic adenoviruses. Cancer Res. 2009;69:5115–25.PubMedPubMedCentralCrossRef
86.
go back to reference Lecharpentier A, Vielh P, Perez-Moreno P, Planchard D, Soria JC, Farace F. Detection of circulating tumour cells with a hybrid (epithelial/mesenchymal) phenotype in patients with metastatic non-small cell lung cancer. Br J Cancer. 2011;105:1338–41.PubMedPubMedCentralCrossRef Lecharpentier A, Vielh P, Perez-Moreno P, Planchard D, Soria JC, Farace F. Detection of circulating tumour cells with a hybrid (epithelial/mesenchymal) phenotype in patients with metastatic non-small cell lung cancer. Br J Cancer. 2011;105:1338–41.PubMedPubMedCentralCrossRef
87.
go back to reference Ruscetti M, Quach B, Dadashian EL, Mulholland DJ, Wu H. Tracking and functional characterization of epithelial-mesenchymal transition and mesenchymal tumor cells during prostate cancer metastasis. Cancer Res. 2015;75:2749–59.PubMedPubMedCentralCrossRef Ruscetti M, Quach B, Dadashian EL, Mulholland DJ, Wu H. Tracking and functional characterization of epithelial-mesenchymal transition and mesenchymal tumor cells during prostate cancer metastasis. Cancer Res. 2015;75:2749–59.PubMedPubMedCentralCrossRef
88.
go back to reference Aceto N, Bardia A, Miyamoto DT, Donaldson MC, Wittner BS, Spencer JA, et al. Circulating tumor cell clusters are oligoclonal precursors of breast cancer metastasis. Cell. 2014;158:1110–22.PubMedPubMedCentralCrossRef Aceto N, Bardia A, Miyamoto DT, Donaldson MC, Wittner BS, Spencer JA, et al. Circulating tumor cell clusters are oligoclonal precursors of breast cancer metastasis. Cell. 2014;158:1110–22.PubMedPubMedCentralCrossRef
89.
go back to reference Podsypanina K, Du YCN, Jechlinger M, Beverly LJ, Hambardzumyan D, Varmus H. Seeding and propagation of untransformed mouse mammary cells in the lung. Science. 2008;321:1841–4.PubMedPubMedCentralCrossRef Podsypanina K, Du YCN, Jechlinger M, Beverly LJ, Hambardzumyan D, Varmus H. Seeding and propagation of untransformed mouse mammary cells in the lung. Science. 2008;321:1841–4.PubMedPubMedCentralCrossRef
90.
go back to reference Liu L, Salnikov AV, Bauer N, Aleksandrowicz E, Labsch S, Nwaeburu C, et al. Triptolide reverses hypoxia-induced epithelial-mesenchymal transition and stem-like features in pancreatic cancer by NF-κB downregulation. Int J Cancer. 2014;134:2489–503.PubMedCrossRef Liu L, Salnikov AV, Bauer N, Aleksandrowicz E, Labsch S, Nwaeburu C, et al. Triptolide reverses hypoxia-induced epithelial-mesenchymal transition and stem-like features in pancreatic cancer by NF-κB downregulation. Int J Cancer. 2014;134:2489–503.PubMedCrossRef
91.
go back to reference Celià-Terrassa T, Meca-Cortés O, Mateo F, Martinez de Paz A, Rubio N, Arnal-Estapé A, et al. Epithelial-mesenchymal transition can suppress major attributes of human epithelial tumor-initiating cells. J Clin Invest. 2012;122:1849–68.PubMedPubMedCentralCrossRef Celià-Terrassa T, Meca-Cortés O, Mateo F, Martinez de Paz A, Rubio N, Arnal-Estapé A, et al. Epithelial-mesenchymal transition can suppress major attributes of human epithelial tumor-initiating cells. J Clin Invest. 2012;122:1849–68.PubMedPubMedCentralCrossRef
93.
go back to reference Ocaña OH, Córcoles R, Fabra A, Moreno-Bueno G, Acloque H, Vega S, et al. Metastatic colonization requires the repression of the epithelial-mesenchymal transition inducer Prrx1. Cancer Cell. 2012;22:709–24.PubMedCrossRef Ocaña OH, Córcoles R, Fabra A, Moreno-Bueno G, Acloque H, Vega S, et al. Metastatic colonization requires the repression of the epithelial-mesenchymal transition inducer Prrx1. Cancer Cell. 2012;22:709–24.PubMedCrossRef
94.
go back to reference Ohkubo T, Ozawa M. The transcription factor Snail downregulates the tight junction components independently of E-cadherin downregulation. J Cell Sci. 2004;117:1675–85.PubMedCrossRef Ohkubo T, Ozawa M. The transcription factor Snail downregulates the tight junction components independently of E-cadherin downregulation. J Cell Sci. 2004;117:1675–85.PubMedCrossRef
95.
go back to reference Herranz N, Pasini D, Díaz VM, Francí C, Gutierrez A, Dave N, et al. Polycomb complex 2 is required for E-cadherin repression by the Snail1 transcription factor. Mol Cell Biol. 2008;28:4772–81.PubMedPubMedCentralCrossRef Herranz N, Pasini D, Díaz VM, Francí C, Gutierrez A, Dave N, et al. Polycomb complex 2 is required for E-cadherin repression by the Snail1 transcription factor. Mol Cell Biol. 2008;28:4772–81.PubMedPubMedCentralCrossRef
96.
go back to reference Tong ZT, Cai MY, Wang XG, Kong LL, Mai SJ, Liu YH, et al. EZH2 supports nasopharyngeal carcinoma cell aggressiveness by forming a co-repressor complex with HDAC1/HDAC2 and snail to inhibit E-cadherin. Oncogene. 2012;31:583–94.PubMed Tong ZT, Cai MY, Wang XG, Kong LL, Mai SJ, Liu YH, et al. EZH2 supports nasopharyngeal carcinoma cell aggressiveness by forming a co-repressor complex with HDAC1/HDAC2 and snail to inhibit E-cadherin. Oncogene. 2012;31:583–94.PubMed
97.
go back to reference Lin T, Ponn A, Hu X, Law BK, Lu J. Requirement of the histone demethylase LSD1 in Snai1-mediated transcriptional repression during epithelial-mesenchymal transition. Oncogene. 2010;29:4896–904.PubMedPubMedCentralCrossRef Lin T, Ponn A, Hu X, Law BK, Lu J. Requirement of the histone demethylase LSD1 in Snai1-mediated transcriptional repression during epithelial-mesenchymal transition. Oncogene. 2010;29:4896–904.PubMedPubMedCentralCrossRef
98.
go back to reference Dong C, Wu Y, Yao J, Wang Y, Yu Y, Rychahou PG, et al. G9a interacts with snail and is critical for snail-mediated E-cadherin repression in human breast cancer. J Clin Invest. 2012;122:1469–86.PubMedPubMedCentralCrossRef Dong C, Wu Y, Yao J, Wang Y, Yu Y, Rychahou PG, et al. G9a interacts with snail and is critical for snail-mediated E-cadherin repression in human breast cancer. J Clin Invest. 2012;122:1469–86.PubMedPubMedCentralCrossRef
99.
go back to reference Dong C, Wu Y, Wang Y, Wang C, Kang T, Rychahou PG, et al. Interaction with Suv39H1 is critical for Snail-mediated E-cadherin repression in breast cancer. Oncogene. 2013;32:1351–62.PubMedCrossRef Dong C, Wu Y, Wang Y, Wang C, Kang T, Rychahou PG, et al. Interaction with Suv39H1 is critical for Snail-mediated E-cadherin repression in breast cancer. Oncogene. 2013;32:1351–62.PubMedCrossRef
100.
go back to reference Serrano-Gomez SJ, Maziveyi M, Alahari SK. Regulation of epithelial-mesenchymal transition through epigenetic and post-translational modifications. Mol Cancer. 2016;15:18.PubMedPubMedCentralCrossRef Serrano-Gomez SJ, Maziveyi M, Alahari SK. Regulation of epithelial-mesenchymal transition through epigenetic and post-translational modifications. Mol Cancer. 2016;15:18.PubMedPubMedCentralCrossRef
101.
go back to reference Kang Y, Massagué J. Epithelial-mesenchymal transitions: twist in development and metastasis. Cell. 2004;118:277–9.PubMedCrossRef Kang Y, Massagué J. Epithelial-mesenchymal transitions: twist in development and metastasis. Cell. 2004;118:277–9.PubMedCrossRef
102.
go back to reference Comijn J, Berx G, Vermassen P, Verschueren K, van Grunsven L, Bruyneel E, et al. The two-handed E box binding zinc finger protein SIP1 downregulates E-cadherin and induces invasion. Mol Cell. 2001;7:1267–78.PubMedCrossRef Comijn J, Berx G, Vermassen P, Verschueren K, van Grunsven L, Bruyneel E, et al. The two-handed E box binding zinc finger protein SIP1 downregulates E-cadherin and induces invasion. Mol Cell. 2001;7:1267–78.PubMedCrossRef
103.
go back to reference Eger A, Aigner K, Sonderegger S, Dampier B, Oehler S, Schreiber M, et al. DeltaEF1 is a transcriptional repressor of E-cadherin and regulates epithelial plasticity in breast cancer cells. Oncogene. 2005;24:2375–85.PubMedCrossRef Eger A, Aigner K, Sonderegger S, Dampier B, Oehler S, Schreiber M, et al. DeltaEF1 is a transcriptional repressor of E-cadherin and regulates epithelial plasticity in breast cancer cells. Oncogene. 2005;24:2375–85.PubMedCrossRef
104.
go back to reference Mathias RA, Simpson RJ. Towards understanding epithelial-mesenchymal transition: a proteomics perspective. Biochim Biophys Acta. 1794;2009:1325–31. Mathias RA, Simpson RJ. Towards understanding epithelial-mesenchymal transition: a proteomics perspective. Biochim Biophys Acta. 1794;2009:1325–31.
105.
go back to reference Black DL. Mechanisms of alternative pre-messenger RNA splicing. Annu Rev Biochem. 2003;72:291–336.PubMedCrossRef Black DL. Mechanisms of alternative pre-messenger RNA splicing. Annu Rev Biochem. 2003;72:291–336.PubMedCrossRef
108.
go back to reference Wu JY, Maniatis T. Specific interactions between proteins implicated in splice site selection and regulated alternative splicing. Cell. 1993;75:1061–70.PubMedCrossRef Wu JY, Maniatis T. Specific interactions between proteins implicated in splice site selection and regulated alternative splicing. Cell. 1993;75:1061–70.PubMedCrossRef
109.
110.
go back to reference Ghigna C, Giordano S, Shen H, Benvenuto F, Castiglioni F, Comoglio PM, et al. Cell motility is controlled by SF2/ASF through alternative splicing of the ron protooncogene. Mol Cell. 2005;20:881–90.PubMedCrossRef Ghigna C, Giordano S, Shen H, Benvenuto F, Castiglioni F, Comoglio PM, et al. Cell motility is controlled by SF2/ASF through alternative splicing of the ron protooncogene. Mol Cell. 2005;20:881–90.PubMedCrossRef
111.
go back to reference Pandit S, Zhou Y, Shiue L, Coutinho-Mansfield G, Li H, Qiu J, et al. Genome-wide analysis reveals SR protein cooperation and competition in regulated splicing. Mol Cell. 2013;50:223–35.PubMedPubMedCentralCrossRef Pandit S, Zhou Y, Shiue L, Coutinho-Mansfield G, Li H, Qiu J, et al. Genome-wide analysis reveals SR protein cooperation and competition in regulated splicing. Mol Cell. 2013;50:223–35.PubMedPubMedCentralCrossRef
113.
go back to reference Ule J, Stefani G, Mele A, Ruggiu M, Wang X, Taneri B, et al. An RNA map predicting Nova-dependent splicing regulation. Nature. 2006;444:580–6.PubMedCrossRef Ule J, Stefani G, Mele A, Ruggiu M, Wang X, Taneri B, et al. An RNA map predicting Nova-dependent splicing regulation. Nature. 2006;444:580–6.PubMedCrossRef
114.
go back to reference Braunschweig U, Gueroussov S, Plocik AM, Graveley BR, Blencowe BJ. Dynamic integration of splicing within gene regulatory pathways. Cell. 2013;152:1252–69.PubMedPubMedCentralCrossRef Braunschweig U, Gueroussov S, Plocik AM, Graveley BR, Blencowe BJ. Dynamic integration of splicing within gene regulatory pathways. Cell. 2013;152:1252–69.PubMedPubMedCentralCrossRef
115.
go back to reference Giampietro C, Deflorian G, Gallo S, Di Matteo A, Pradella D, Bonomi S, et al. The alternative splicing factor Nova2 regulates vascular development and lumen formation. Nat Comms. 2015;6:8479.CrossRef Giampietro C, Deflorian G, Gallo S, Di Matteo A, Pradella D, Bonomi S, et al. The alternative splicing factor Nova2 regulates vascular development and lumen formation. Nat Comms. 2015;6:8479.CrossRef
118.
go back to reference Aparicio LA, Abella V, Valladares M, Figueroa A. Posttranscriptional regulation by RNA-binding proteins during epithelial-to-mesenchymal transition. Cell Mol Life Sci. 2013;70:4463–77.PubMedPubMedCentralCrossRef Aparicio LA, Abella V, Valladares M, Figueroa A. Posttranscriptional regulation by RNA-binding proteins during epithelial-to-mesenchymal transition. Cell Mol Life Sci. 2013;70:4463–77.PubMedPubMedCentralCrossRef
119.
go back to reference Shapiro IM, Cheng AW, Flytzanis NC, Balsamo M, Condeelis JS, Oktay MH, et al. An EMT–driven alternative splicing program occurs in human breast cancer and modulates cellular phenotype. PLoS Genet. 2011;7:e1002218.PubMedPubMedCentralCrossRef Shapiro IM, Cheng AW, Flytzanis NC, Balsamo M, Condeelis JS, Oktay MH, et al. An EMT–driven alternative splicing program occurs in human breast cancer and modulates cellular phenotype. PLoS Genet. 2011;7:e1002218.PubMedPubMedCentralCrossRef
120.
go back to reference Warzecha CC, Sato TK, Nabet B, Hogenesch JB, Carstens RP. ESRP1 and ESRP2 are epithelial cell-type-specific regulators of FGFR2 splicing. Mol Cell. 2009;33:591–601.PubMedPubMedCentralCrossRef Warzecha CC, Sato TK, Nabet B, Hogenesch JB, Carstens RP. ESRP1 and ESRP2 are epithelial cell-type-specific regulators of FGFR2 splicing. Mol Cell. 2009;33:591–601.PubMedPubMedCentralCrossRef
121.
go back to reference Warzecha CC, Shen S, Xing Y, Carstens RP. The epithelial splicing factors ESRP1 and ESRP2 positively and negatively regulate diverse types of alternative splicing events. RNA Biol. 2009;6:546–62.PubMedPubMedCentralCrossRef Warzecha CC, Shen S, Xing Y, Carstens RP. The epithelial splicing factors ESRP1 and ESRP2 positively and negatively regulate diverse types of alternative splicing events. RNA Biol. 2009;6:546–62.PubMedPubMedCentralCrossRef
122.
go back to reference Warzecha CC, Jiang P, Amirikian K, Dittmar KA, Lu H, Shen S, et al. An ESRP-regulated splicing programme is abrogated during the epithelial-mesenchymal transition. EMBO J. 2010;29:3286–300.PubMedPubMedCentralCrossRef Warzecha CC, Jiang P, Amirikian K, Dittmar KA, Lu H, Shen S, et al. An ESRP-regulated splicing programme is abrogated during the epithelial-mesenchymal transition. EMBO J. 2010;29:3286–300.PubMedPubMedCentralCrossRef
123.
go back to reference Bebee TW, Park JW, Sheridan KI, Warzecha CC, Cieply BW, Rohacek AM, et al. The splicing regulators Esrp1 and Esrp2 direct an epithelial splicing program essential for mammalian development. Elife. 2015;4:e08954. Bebee TW, Park JW, Sheridan KI, Warzecha CC, Cieply BW, Rohacek AM, et al. The splicing regulators Esrp1 and Esrp2 direct an epithelial splicing program essential for mammalian development. Elife. 2015;4:e08954.
124.
go back to reference Yeo, Coufal NG, Liang TY, Peng GE, Fu X-D, Gage FH. An RNA code for the FOX2 splicing regulator revealed by mapping RNA-protein interactions in stem cells. Nat Struct Mol Biol. 2009;16:130–7.PubMedPubMedCentralCrossRef Yeo, Coufal NG, Liang TY, Peng GE, Fu X-D, Gage FH. An RNA code for the FOX2 splicing regulator revealed by mapping RNA-protein interactions in stem cells. Nat Struct Mol Biol. 2009;16:130–7.PubMedPubMedCentralCrossRef
125.
go back to reference Zhao Q, Caballero OL, Davis ID, Jonasch E, Tamboli P, Yung WKA, et al. Tumor-specific isoform switch of the fibroblast growth factor receptor 2 underlies the mesenchymal and malignant phenotypes of clear cell renal cell carcinomas. Clin Cancer Res. 2013;19:2460–72.PubMedPubMedCentralCrossRef Zhao Q, Caballero OL, Davis ID, Jonasch E, Tamboli P, Yung WKA, et al. Tumor-specific isoform switch of the fibroblast growth factor receptor 2 underlies the mesenchymal and malignant phenotypes of clear cell renal cell carcinomas. Clin Cancer Res. 2013;19:2460–72.PubMedPubMedCentralCrossRef
126.
go back to reference Hovhannisyan RH, Carstens RP. Heterogeneous ribonucleoprotein m is a splicing regulatory protein that can enhance or silence splicing of alternatively spliced exons. J Biol Chem. 2007;282:36265–74.PubMedCrossRef Hovhannisyan RH, Carstens RP. Heterogeneous ribonucleoprotein m is a splicing regulatory protein that can enhance or silence splicing of alternatively spliced exons. J Biol Chem. 2007;282:36265–74.PubMedCrossRef
127.
go back to reference Mauger DM, Lin C, Garcia-Blanco MA. hnRNP H and hnRNP F complex with Fox2 to silence fibroblast growth factor receptor 2 exon IIIc. Mol Cell Biol. 2008;28:5403–19.PubMedPubMedCentralCrossRef Mauger DM, Lin C, Garcia-Blanco MA. hnRNP H and hnRNP F complex with Fox2 to silence fibroblast growth factor receptor 2 exon IIIc. Mol Cell Biol. 2008;28:5403–19.PubMedPubMedCentralCrossRef
128.
go back to reference Gesnel M-C, Del Gatto-Konczak F, Breathnach R. Combined use of MS2 and PP7 coat fusions shows that TIA-1 dominates hnRNP A1 for K-SAM exon splicing control. J Biomed Biotechnol. 2009;2009:104853.PubMedCrossRef Gesnel M-C, Del Gatto-Konczak F, Breathnach R. Combined use of MS2 and PP7 coat fusions shows that TIA-1 dominates hnRNP A1 for K-SAM exon splicing control. J Biomed Biotechnol. 2009;2009:104853.PubMedCrossRef
129.
go back to reference Carstens RP, Wagner EJ, Garcia-Blanco MA. An intronic splicing silencer causes skipping of the IIIb exon of fibroblast growth factor receptor 2 through involvement of polypyrimidine tract binding protein. Mol Cell Biol. 2000;20:7388–400.PubMedPubMedCentralCrossRef Carstens RP, Wagner EJ, Garcia-Blanco MA. An intronic splicing silencer causes skipping of the IIIb exon of fibroblast growth factor receptor 2 through involvement of polypyrimidine tract binding protein. Mol Cell Biol. 2000;20:7388–400.PubMedPubMedCentralCrossRef
130.
go back to reference Ishiyama N, Lee SH, Liu S, Li GY, Smith MJ, Reichardt LF, et al. Dynamic and static interactions between p120 catenin and E-cadherin regulate the stability of cell-cell adhesion. Cell. 2010;141:117–28.PubMedCrossRef Ishiyama N, Lee SH, Liu S, Li GY, Smith MJ, Reichardt LF, et al. Dynamic and static interactions between p120 catenin and E-cadherin regulate the stability of cell-cell adhesion. Cell. 2010;141:117–28.PubMedCrossRef
131.
go back to reference Yanagisawa M, Huveldt D, Kreinest P, Lohse CM, Cheville JC, Parker AS, et al. A p120 catenin isoform switch affects Rho activity, induces tumor cell invasion, and predicts metastatic disease. J Biol Chem. 2008;283:18344–54.PubMedPubMedCentralCrossRef Yanagisawa M, Huveldt D, Kreinest P, Lohse CM, Cheville JC, Parker AS, et al. A p120 catenin isoform switch affects Rho activity, induces tumor cell invasion, and predicts metastatic disease. J Biol Chem. 2008;283:18344–54.PubMedPubMedCentralCrossRef
133.
go back to reference Wang Z, Sandiford S, Wu C, Li SSC. Numb regulates cell-cell adhesion and polarity in response to tyrosine kinase signalling. EMBO J. 2009;28:2360–73.PubMedPubMedCentralCrossRef Wang Z, Sandiford S, Wu C, Li SSC. Numb regulates cell-cell adhesion and polarity in response to tyrosine kinase signalling. EMBO J. 2009;28:2360–73.PubMedPubMedCentralCrossRef
134.
go back to reference Yamben IF, Rachel RA, Shatadal S, Copeland NG, Jenkins NA, Warming S, et al. Scrib is required for epithelial cell identity and prevents epithelial to mesenchymal transition in the mouse. Dev Biol. 2013;384:41–52.PubMedCrossRef Yamben IF, Rachel RA, Shatadal S, Copeland NG, Jenkins NA, Warming S, et al. Scrib is required for epithelial cell identity and prevents epithelial to mesenchymal transition in the mouse. Dev Biol. 2013;384:41–52.PubMedCrossRef
135.
go back to reference Vaira V, Faversani A, Dohi T, Maggioni M, Nosotti M, Tosi D, et al. Aberrant overexpression of the cell polarity module scribble in human cancer. Am J Pathol. 2011;178:2478–83.PubMedPubMedCentralCrossRef Vaira V, Faversani A, Dohi T, Maggioni M, Nosotti M, Tosi D, et al. Aberrant overexpression of the cell polarity module scribble in human cancer. Am J Pathol. 2011;178:2478–83.PubMedPubMedCentralCrossRef
136.
go back to reference Warzecha CC, Carstens RP. Complex changes in alternative pre-mRNA splicing play a central role in the epithelial-to-mesenchymal transition (EMT). Semin Cancer Biol. 2012;22:417–27.PubMedPubMedCentralCrossRef Warzecha CC, Carstens RP. Complex changes in alternative pre-mRNA splicing play a central role in the epithelial-to-mesenchymal transition (EMT). Semin Cancer Biol. 2012;22:417–27.PubMedPubMedCentralCrossRef
137.
go back to reference Pino MS, Balsamo M, Di Modugno F, Mottolese M, Alessio M, Melucci E, et al. Human Mena + 11a isoform serves as a marker of epithelial phenotype and sensitivity to epidermal growth factor receptor inhibition in human pancreatic cancer cell lines. Clin Cancer Res. 2008;14:4943–50.PubMedPubMedCentralCrossRef Pino MS, Balsamo M, Di Modugno F, Mottolese M, Alessio M, Melucci E, et al. Human Mena + 11a isoform serves as a marker of epithelial phenotype and sensitivity to epidermal growth factor receptor inhibition in human pancreatic cancer cell lines. Clin Cancer Res. 2008;14:4943–50.PubMedPubMedCentralCrossRef
138.
go back to reference Goswami S, Philippar U, Sun D, Patsialou A, Avraham J, Wang W, et al. Identification of invasion specific splice variants of the cytoskeletal protein Mena present in mammary tumor cells during invasion in vivo. Clin Exp Metastasis. 2009;26:153–9.PubMedCrossRef Goswami S, Philippar U, Sun D, Patsialou A, Avraham J, Wang W, et al. Identification of invasion specific splice variants of the cytoskeletal protein Mena present in mammary tumor cells during invasion in vivo. Clin Exp Metastasis. 2009;26:153–9.PubMedCrossRef
139.
go back to reference Di Modugno F, Iapicca P, Boudreau A, Mottolese M, Terrenato I, Perracchio L, et al. Splicing program of human MENA produces a previously undescribed isoform associated with invasive, mesenchymal-like breast tumors. Proc Natl Acad Sci U S A. 2012;109:19280–5.PubMedPubMedCentralCrossRef Di Modugno F, Iapicca P, Boudreau A, Mottolese M, Terrenato I, Perracchio L, et al. Splicing program of human MENA produces a previously undescribed isoform associated with invasive, mesenchymal-like breast tumors. Proc Natl Acad Sci U S A. 2012;109:19280–5.PubMedPubMedCentralCrossRef
140.
go back to reference Tani K, Sato S, Sukezane T, Kojima H, Hirose H, Hanafusa H, et al. Abl interactor 1 promotes tyrosine 296 phosphorylation of mammalian enabled (Mena) by c-Abl kinase. J Biol Chem. 2003;278:21685–92.PubMedCrossRef Tani K, Sato S, Sukezane T, Kojima H, Hirose H, Hanafusa H, et al. Abl interactor 1 promotes tyrosine 296 phosphorylation of mammalian enabled (Mena) by c-Abl kinase. J Biol Chem. 2003;278:21685–92.PubMedCrossRef
141.
go back to reference Weise A, Bruser K, Elfert S, Wallmen B, Wittel Y, Wöhrle S, et al. Alternative splicing of Tcf7l2 transcripts generates protein variants with differential promoter-binding and transcriptional activation properties at Wnt/beta-catenin targets. Nucleic Acids Res. 2010;38:1964–81.PubMedCrossRef Weise A, Bruser K, Elfert S, Wallmen B, Wittel Y, Wöhrle S, et al. Alternative splicing of Tcf7l2 transcripts generates protein variants with differential promoter-binding and transcriptional activation properties at Wnt/beta-catenin targets. Nucleic Acids Res. 2010;38:1964–81.PubMedCrossRef
143.
go back to reference Shirakihara T, Horiguchi K, Miyazawa K, Ehata S, Shibata T, Morita I, et al. TGF-β regulates isoform switching of FGF receptors and epithelial-mesenchymal transition. EMBO J. 2011;30:783–95.PubMedPubMedCentralCrossRef Shirakihara T, Horiguchi K, Miyazawa K, Ehata S, Shibata T, Morita I, et al. TGF-β regulates isoform switching of FGF receptors and epithelial-mesenchymal transition. EMBO J. 2011;30:783–95.PubMedPubMedCentralCrossRef
144.
go back to reference Horiguchi K, Sakamoto K, Koinuma D, Semba K, Inoue A, Inoue S, et al. TGF-β drives epithelial-mesenchymal transition through δEF1-mediated downregulation of ESRP. Oncogene. 2012;31:3190–201.PubMedCrossRef Horiguchi K, Sakamoto K, Koinuma D, Semba K, Inoue A, Inoue S, et al. TGF-β drives epithelial-mesenchymal transition through δEF1-mediated downregulation of ESRP. Oncogene. 2012;31:3190–201.PubMedCrossRef
146.
go back to reference Ivanov I, Lo KC, Hawthorn L, Cowell JK, Ionov Y. Identifying candidate colon cancer tumor suppressor genes using inhibition of nonsense-mediated mRNA decay in colon cancer cells. Oncogene. 2007;26:2873–84.PubMedCrossRef Ivanov I, Lo KC, Hawthorn L, Cowell JK, Ionov Y. Identifying candidate colon cancer tumor suppressor genes using inhibition of nonsense-mediated mRNA decay in colon cancer cells. Oncogene. 2007;26:2873–84.PubMedCrossRef
147.
go back to reference Taube JH, Herschkowitz JI, Komurov K, Zhou AY, Gupta S, Yang J, et al. Core epithelial-to-mesenchymal transition interactome gene-expression signature is associated with claudin-low and metaplastic breast cancer subtypes. Proc Natl Acad Sci U S A. 2010;107:15449–54.PubMedPubMedCentralCrossRef Taube JH, Herschkowitz JI, Komurov K, Zhou AY, Gupta S, Yang J, et al. Core epithelial-to-mesenchymal transition interactome gene-expression signature is associated with claudin-low and metaplastic breast cancer subtypes. Proc Natl Acad Sci U S A. 2010;107:15449–54.PubMedPubMedCentralCrossRef
148.
go back to reference Onder TT, Gupta PB, Mani SA, Yang J, Lander ES, Weinberg RA. Loss of E-cadherin promotes metastasis via multiple downstream transcriptional pathways. Cancer Res. 2008;68:3645–54.PubMedCrossRef Onder TT, Gupta PB, Mani SA, Yang J, Lander ES, Weinberg RA. Loss of E-cadherin promotes metastasis via multiple downstream transcriptional pathways. Cancer Res. 2008;68:3645–54.PubMedCrossRef
149.
go back to reference Braeutigam C, Rago L, Rolke A, Waldmeier L, Christofori G, Winter J. The RNA-binding protein Rbfox2: an essential regulator of EMT-driven alternative splicing and a mediator of cellular invasion. Oncogene. 2014;33:1082–92.PubMedCrossRef Braeutigam C, Rago L, Rolke A, Waldmeier L, Christofori G, Winter J. The RNA-binding protein Rbfox2: an essential regulator of EMT-driven alternative splicing and a mediator of cellular invasion. Oncogene. 2014;33:1082–92.PubMedCrossRef
150.
go back to reference Dohadwala M, Yang S-C, Luo J, Sharma S, Batra RK, Huang M, et al. Cyclooxygenase-2-dependent regulation of E-cadherin: prostaglandin E(2) induces transcriptional repressors ZEB1 and snail in non-small cell lung cancer. Cancer Res. 2006;66:5338–45.PubMedCrossRef Dohadwala M, Yang S-C, Luo J, Sharma S, Batra RK, Huang M, et al. Cyclooxygenase-2-dependent regulation of E-cadherin: prostaglandin E(2) induces transcriptional repressors ZEB1 and snail in non-small cell lung cancer. Cancer Res. 2006;66:5338–45.PubMedCrossRef
151.
go back to reference Zhang J, Lu C, Zhang J, Kang J, Cao C, Li M. Involvement of ZEB1 and E-cadherin in the invasion of lung squamous cell carcinoma. Mol Biol Rep. 2013;40:949–56.PubMedCrossRef Zhang J, Lu C, Zhang J, Kang J, Cao C, Li M. Involvement of ZEB1 and E-cadherin in the invasion of lung squamous cell carcinoma. Mol Biol Rep. 2013;40:949–56.PubMedCrossRef
152.
go back to reference Naor D, Wallach-Dayan SB, Zahalka MA, Sionov RV. Involvement of CD44, a molecule with a thousand faces, in cancer dissemination. Semin Cancer Biol. 2008;18:260–7.PubMedCrossRef Naor D, Wallach-Dayan SB, Zahalka MA, Sionov RV. Involvement of CD44, a molecule with a thousand faces, in cancer dissemination. Semin Cancer Biol. 2008;18:260–7.PubMedCrossRef
153.
go back to reference Larsen JE, Nathan V, Osborne JK, Farrow RK, Deb D, Sullivan JP, et al. ZEB1 drives epithelial-to-mesenchymal transition in lung cancer. J Clin Invest. 2016;126:3219–35.PubMedPubMedCentralCrossRef Larsen JE, Nathan V, Osborne JK, Farrow RK, Deb D, Sullivan JP, et al. ZEB1 drives epithelial-to-mesenchymal transition in lung cancer. J Clin Invest. 2016;126:3219–35.PubMedPubMedCentralCrossRef
154.
go back to reference Brown RL, Reinke LM, Damerow MS, Perez D, Chodosh LA, Yang J, et al. CD44 splice isoform switching in human and mouse epithelium is essential for epithelial-mesenchymal transition and breast cancer progression. J Clin Invest. 2011;121:1064–74.PubMedPubMedCentralCrossRef Brown RL, Reinke LM, Damerow MS, Perez D, Chodosh LA, Yang J, et al. CD44 splice isoform switching in human and mouse epithelium is essential for epithelial-mesenchymal transition and breast cancer progression. J Clin Invest. 2011;121:1064–74.PubMedPubMedCentralCrossRef
155.
go back to reference Deloria AJ, Höflmayer D, Kienzl P, Łopatecka1 J, Sampl S, Klimpfinger M, et al. Epithelial splicing regulatory protein 1 and 2 paralogues correlate with splice signatures and favorable outcome in human colorectal cancer. Oncotarget. 2016;7:73800–16. Deloria AJ, Höflmayer D, Kienzl P, Łopatecka1 J, Sampl S, Klimpfinger M, et al. Epithelial splicing regulatory protein 1 and 2 paralogues correlate with splice signatures and favorable outcome in human colorectal cancer. Oncotarget. 2016;7:73800–16.
156.
go back to reference Ueda J, Matsuda Y, Yamahatsu K, Uchida E, Naito Z, Korc M, et al. Epithelial splicing regulatory protein 1 is a favorable prognostic factor in pancreatic cancer that attenuates pancreatic metastases. Oncogene. 2014;33:4485–95.PubMedCrossRef Ueda J, Matsuda Y, Yamahatsu K, Uchida E, Naito Z, Korc M, et al. Epithelial splicing regulatory protein 1 is a favorable prognostic factor in pancreatic cancer that attenuates pancreatic metastases. Oncogene. 2014;33:4485–95.PubMedCrossRef
157.
go back to reference Ishii H, Saitoh M, Sakamoto K, Kondo T, Katoh R, Tanaka S, et al. Epithelial splicing regulatory proteins 1 (ESRP1) and 2 (ESRP2) suppress cancer cell motility via different mechanisms. J Biol Chem. 2014;289:27386–99.PubMedPubMedCentralCrossRef Ishii H, Saitoh M, Sakamoto K, Kondo T, Katoh R, Tanaka S, et al. Epithelial splicing regulatory proteins 1 (ESRP1) and 2 (ESRP2) suppress cancer cell motility via different mechanisms. J Biol Chem. 2014;289:27386–99.PubMedPubMedCentralCrossRef
158.
go back to reference Vanharanta S, Marney CB, Shu W, Valiente M, Zou Y, Mele A, et al. Loss of the multifunctional RNA-binding protein RBM47 as a source of selectable metastatic traits in breast cancer. eLife. 2014;3:e02734. Vanharanta S, Marney CB, Shu W, Valiente M, Zou Y, Mele A, et al. Loss of the multifunctional RNA-binding protein RBM47 as a source of selectable metastatic traits in breast cancer. eLife. 2014;3:e02734.
159.
go back to reference Fossat N, Tourle K, Radziewic T, Barratt K, Liebhold D, Studdert JB, et al. C to U RNA editing mediated by APOBEC1 requires RNA-binding protein RBM47. EMBO Rep. 2014;15:903–10.PubMedPubMedCentralCrossRef Fossat N, Tourle K, Radziewic T, Barratt K, Liebhold D, Studdert JB, et al. C to U RNA editing mediated by APOBEC1 requires RNA-binding protein RBM47. EMBO Rep. 2014;15:903–10.PubMedPubMedCentralCrossRef
160.
go back to reference Venables JP, Lapasset L, Gadea G, Fort P, Klinck R, Irimia M, et al. MBNL1 and RBFOX2 cooperate to establish a splicing programme involved in pluripotent stem cell differentiation. Nat Comms. 2013;4:2480.CrossRef Venables JP, Lapasset L, Gadea G, Fort P, Klinck R, Irimia M, et al. MBNL1 and RBFOX2 cooperate to establish a splicing programme involved in pluripotent stem cell differentiation. Nat Comms. 2013;4:2480.CrossRef
161.
go back to reference Venables JP, Brosseau JP, Gadea G, Klinck R, Prinos P, Beaulieu JF, et al. RBFOX2 Is an important regulator of mesenchymal tissue-specific splicing in both normal and cancer tissues. Mol Cell Biol. 2012;33:396–405.PubMedCrossRef Venables JP, Brosseau JP, Gadea G, Klinck R, Prinos P, Beaulieu JF, et al. RBFOX2 Is an important regulator of mesenchymal tissue-specific splicing in both normal and cancer tissues. Mol Cell Biol. 2012;33:396–405.PubMedCrossRef
162.
go back to reference Hall MP, Nagel RJ, Fagg WS, Shiue L, Cline MS, Perriman RJ, et al. Quaking and PTB control overlapping splicing regulatory networks during muscle cell differentiation. RNA. 2013;19:627–38.PubMedPubMedCentralCrossRef Hall MP, Nagel RJ, Fagg WS, Shiue L, Cline MS, Perriman RJ, et al. Quaking and PTB control overlapping splicing regulatory networks during muscle cell differentiation. RNA. 2013;19:627–38.PubMedPubMedCentralCrossRef
163.
go back to reference Masuda A, Andersen HS, Doktor TK, Okamoto T, Ito M, Andresen BS, et al. CUGBP1 and MBNL1 preferentially bind to 3′ UTRs and facilitate mRNA decay. Sci Rep. 2012;2:209.PubMedPubMedCentral Masuda A, Andersen HS, Doktor TK, Okamoto T, Ito M, Andresen BS, et al. CUGBP1 and MBNL1 preferentially bind to 3′ UTRs and facilitate mRNA decay. Sci Rep. 2012;2:209.PubMedPubMedCentral
164.
go back to reference Batra R, Charizanis K, Manchanda M, Mohan A, Li M, Finn DJ, et al. Loss of MBNL leads to disruption of developmentally regulated alternative polyadenylation in RNA-mediated disease. Mol Cell. 2014;56:311–22.PubMedPubMedCentralCrossRef Batra R, Charizanis K, Manchanda M, Mohan A, Li M, Finn DJ, et al. Loss of MBNL leads to disruption of developmentally regulated alternative polyadenylation in RNA-mediated disease. Mol Cell. 2014;56:311–22.PubMedPubMedCentralCrossRef
165.
go back to reference Fish L, Pencheva N, Goodarzi H, Tran H, Yoshida M, Tavazoie SF. Muscleblind-like 1 suppresses breast cancer metastatic colonization and stabilizes metastasis suppressor transcripts. Genes Dev. 2016;30:386–98.PubMedPubMedCentralCrossRef Fish L, Pencheva N, Goodarzi H, Tran H, Yoshida M, Tavazoie SF. Muscleblind-like 1 suppresses breast cancer metastatic colonization and stabilizes metastasis suppressor transcripts. Genes Dev. 2016;30:386–98.PubMedPubMedCentralCrossRef
166.
go back to reference Gout S, Brambilla E, Boudria A, Drissi R, Lantuejoul S, Gazzeri S, et al. Abnormal expression of the pre-mRNA splicing regulators SRSF1, SRSF2, SRPK1 and SRPK2 in non small cell lung carcinoma. PLoS ONE. 2012;7:e46539.PubMedPubMedCentralCrossRef Gout S, Brambilla E, Boudria A, Drissi R, Lantuejoul S, Gazzeri S, et al. Abnormal expression of the pre-mRNA splicing regulators SRSF1, SRSF2, SRPK1 and SRPK2 in non small cell lung carcinoma. PLoS ONE. 2012;7:e46539.PubMedPubMedCentralCrossRef
167.
go back to reference Sen S, Langiewicz M, Jumaa H, Webster NJG. Deletion of serine/arginine-rich splicing factor 3 in hepatocytes predisposes to hepatocellular carcinoma in mice. Hepatology. 2015;61:171–83.PubMedCrossRef Sen S, Langiewicz M, Jumaa H, Webster NJG. Deletion of serine/arginine-rich splicing factor 3 in hepatocytes predisposes to hepatocellular carcinoma in mice. Hepatology. 2015;61:171–83.PubMedCrossRef
168.
go back to reference Karni R, de Stanchina E, Lowe SW, Sinha R, Mu D, Krainer AR. The gene encoding the splicing factor SF2/ASF is a proto-oncogene. Nat Struct Mol Biol. 2007;14:185–93.PubMedPubMedCentralCrossRef Karni R, de Stanchina E, Lowe SW, Sinha R, Mu D, Krainer AR. The gene encoding the splicing factor SF2/ASF is a proto-oncogene. Nat Struct Mol Biol. 2007;14:185–93.PubMedPubMedCentralCrossRef
169.
go back to reference Anczuków O, Rosenberg AZ, Akerman M, Das S, Zhan L, Karni R, et al. The splicing factor SRSF1 regulates apoptosis and proliferation to promote mammary epithelial cell transformation. Nat Struct Mol Biol. 2012;19:220–8.PubMedPubMedCentralCrossRef Anczuków O, Rosenberg AZ, Akerman M, Das S, Zhan L, Karni R, et al. The splicing factor SRSF1 regulates apoptosis and proliferation to promote mammary epithelial cell transformation. Nat Struct Mol Biol. 2012;19:220–8.PubMedPubMedCentralCrossRef
170.
171.
go back to reference Valacca C, Bonomi S, Buratti E, Pedrotti S, Baralle FE, Sette C, et al. Sam68 regulates EMT through alternative splicing-activated nonsense-mediated mRNA decay of the SF2/ASF proto-oncogene. J Cell Biol. 2010;191:87–99.PubMedPubMedCentralCrossRef Valacca C, Bonomi S, Buratti E, Pedrotti S, Baralle FE, Sette C, et al. Sam68 regulates EMT through alternative splicing-activated nonsense-mediated mRNA decay of the SF2/ASF proto-oncogene. J Cell Biol. 2010;191:87–99.PubMedPubMedCentralCrossRef
172.
go back to reference Meseguer S, Mudduluru G, Escamilla JM, Allgayer H, Barettino D. MicroRNAs-10a and -10b contribute to retinoic acid-induced differentiation of neuroblastoma cells and target the alternative splicing regulatory factor SFRS1 (SF2/ASF). J Biol Chem. 2011;286:4150–64.PubMedCrossRef Meseguer S, Mudduluru G, Escamilla JM, Allgayer H, Barettino D. MicroRNAs-10a and -10b contribute to retinoic acid-induced differentiation of neuroblastoma cells and target the alternative splicing regulatory factor SFRS1 (SF2/ASF). J Biol Chem. 2011;286:4150–64.PubMedCrossRef
173.
go back to reference Gonçalves V, Henriques AFA, Henriques A, Pereira JFS, Pereira J, Neves Costa A, et al. Phosphorylation of SRSF1 by SRPK1 regulates alternative splicing of tumor-related Rac1b in colorectal cells. RNA. 2014;20:474–82.PubMedPubMedCentralCrossRef Gonçalves V, Henriques AFA, Henriques A, Pereira JFS, Pereira J, Neves Costa A, et al. Phosphorylation of SRSF1 by SRPK1 regulates alternative splicing of tumor-related Rac1b in colorectal cells. RNA. 2014;20:474–82.PubMedPubMedCentralCrossRef
174.
go back to reference Bielli P, Busa R, Paronetto MP, Sette C. The RNA-binding protein Sam68 is a multifunctional player in human cancer. Endocr Relat Cancer. 2011;18:R91–R102.PubMedCrossRef Bielli P, Busa R, Paronetto MP, Sette C. The RNA-binding protein Sam68 is a multifunctional player in human cancer. Endocr Relat Cancer. 2011;18:R91–R102.PubMedCrossRef
175.
go back to reference Frisone P, Pradella D, Di Matteo A, Belloni E, Ghigna C, Paronetto MP. SAM68: signal transduction and RNA metabolism in human cancer. BioMed Res Int. 2015;2015:528954.PubMedPubMedCentralCrossRef Frisone P, Pradella D, Di Matteo A, Belloni E, Ghigna C, Paronetto MP. SAM68: signal transduction and RNA metabolism in human cancer. BioMed Res Int. 2015;2015:528954.PubMedPubMedCentralCrossRef
176.
go back to reference Hayes GM, Carrigan PE, Miller LJ. Serine-arginine protein kinase 1 overexpression is associated with tumorigenic imbalance in mitogen-activated protein kinase pathways in breast, colonic, and pancreatic carcinomas. Cancer Res. 2007;67:2072–80.PubMedCrossRef Hayes GM, Carrigan PE, Miller LJ. Serine-arginine protein kinase 1 overexpression is associated with tumorigenic imbalance in mitogen-activated protein kinase pathways in breast, colonic, and pancreatic carcinomas. Cancer Res. 2007;67:2072–80.PubMedCrossRef
177.
go back to reference Naro C, Barbagallo F, Chieffi P, Bourgeois CF, Paronetto MP, Sette C. The centrosomal kinase NEK2 is a novel splicing factor kinase involved in cell survival. Nucleic Acids Res. 2014;42:3218–27.PubMedCrossRef Naro C, Barbagallo F, Chieffi P, Bourgeois CF, Paronetto MP, Sette C. The centrosomal kinase NEK2 is a novel splicing factor kinase involved in cell survival. Nucleic Acids Res. 2014;42:3218–27.PubMedCrossRef
178.
go back to reference Nowak DG, Amin EM, Rennel ES, Hoareau-Aveilla C, Gammons M, Damodoran G, et al. Regulation of vascular endothelial growth factor (VEGF) splicing from pro-angiogenic to anti-angiogenic isoforms: a novel therapeutic strategy for angiogenesis. J Biol Chem. 2010;285:5532–40.PubMedCrossRef Nowak DG, Amin EM, Rennel ES, Hoareau-Aveilla C, Gammons M, Damodoran G, et al. Regulation of vascular endothelial growth factor (VEGF) splicing from pro-angiogenic to anti-angiogenic isoforms: a novel therapeutic strategy for angiogenesis. J Biol Chem. 2010;285:5532–40.PubMedCrossRef
179.
go back to reference Danilkovitch-Miagkova A. Oncogenic signaling pathways activated by RON receptor tyrosine kinase. Curr Cancer Drug Targets. 2003;3:31–40.PubMedCrossRef Danilkovitch-Miagkova A. Oncogenic signaling pathways activated by RON receptor tyrosine kinase. Curr Cancer Drug Targets. 2003;3:31–40.PubMedCrossRef
180.
go back to reference Collesi C, Santoro MM, Gaudino G, Comoglio PM. A splicing variant of the RON transcript induces constitutive tyrosine kinase activity and an invasive phenotype. Mol Cell Biol. 1996;16:5518–26.PubMedPubMedCentralCrossRef Collesi C, Santoro MM, Gaudino G, Comoglio PM. A splicing variant of the RON transcript induces constitutive tyrosine kinase activity and an invasive phenotype. Mol Cell Biol. 1996;16:5518–26.PubMedPubMedCentralCrossRef
181.
go back to reference Zhou YQ, He C, Chen YQ, Wang D, Wang MH. Altered expression of the RON receptor tyrosine kinase in primary human colorectal adenocarcinomas: generation of different splicing RON variants and their oncogenic potential. Oncogene. 2003;22:186–97.PubMedCrossRef Zhou YQ, He C, Chen YQ, Wang D, Wang MH. Altered expression of the RON receptor tyrosine kinase in primary human colorectal adenocarcinomas: generation of different splicing RON variants and their oncogenic potential. Oncogene. 2003;22:186–97.PubMedCrossRef
182.
go back to reference Golan-Gerstl R, Cohen M, Shilo A, Suh SS, Bakàcs A, Coppola L, et al. Splicing factor hnRNP A2/B1 regulates tumor suppressor gene splicing and is an oncogenic driver in glioblastoma. Cancer Res. 2011;71:4464–72.PubMedCrossRef Golan-Gerstl R, Cohen M, Shilo A, Suh SS, Bakàcs A, Coppola L, et al. Splicing factor hnRNP A2/B1 regulates tumor suppressor gene splicing and is an oncogenic driver in glioblastoma. Cancer Res. 2011;71:4464–72.PubMedCrossRef
183.
go back to reference Tauler J, Zudaire E, Liu H, Shih J, Mulshine JL. hnRNP A2/B1 modulates epithelial-mesenchymal transition in lung cancer cell lines. Cancer Res. 2010;70:7137–47.PubMedCrossRef Tauler J, Zudaire E, Liu H, Shih J, Mulshine JL. hnRNP A2/B1 modulates epithelial-mesenchymal transition in lung cancer cell lines. Cancer Res. 2010;70:7137–47.PubMedCrossRef
184.
go back to reference Bonomi S, di Matteo A, Buratti E, Cabianca DS, Baralle FE, Ghigna C, et al. HnRNP A1 controls a splicing regulatory circuit promoting mesenchymal-to-epithelial transition. Nucleic Acids Res. 2013;41:8665–79.PubMedPubMedCentralCrossRef Bonomi S, di Matteo A, Buratti E, Cabianca DS, Baralle FE, Ghigna C, et al. HnRNP A1 controls a splicing regulatory circuit promoting mesenchymal-to-epithelial transition. Nucleic Acids Res. 2013;41:8665–79.PubMedPubMedCentralCrossRef
185.
go back to reference Ghigna C, Biamonti G, Gallo S. Pro-metastatic splicing of Ron proto-oncogene mRNA can be reversed: therapeutic potential of bifunctional oligonucleotides and indole derivatives. RNA Biol. 2010;7:495–503.PubMedCrossRef Ghigna C, Biamonti G, Gallo S. Pro-metastatic splicing of Ron proto-oncogene mRNA can be reversed: therapeutic potential of bifunctional oligonucleotides and indole derivatives. RNA Biol. 2010;7:495–503.PubMedCrossRef
186.
187.
go back to reference Jordan P RB, Boavida MG, Gespach C, Chastre E. Cloning of a novel human Rac1b splice variant with increased expression in colorectal tumors. Oncogene. 1999;18:6835–9.PubMedCrossRef Jordan P RB, Boavida MG, Gespach C, Chastre E. Cloning of a novel human Rac1b splice variant with increased expression in colorectal tumors. Oncogene. 1999;18:6835–9.PubMedCrossRef
188.
go back to reference Orlichenko L, Geyer R, Yanagisawa M, Khauv D, Radisky ES, Anastasiadis PZ, et al. The 19-amino acid insertion in the tumor-associated splice isoform Rac1b confers specific binding to p120 catenin. J Biol Chem. 2010;285:19153–61.PubMedPubMedCentralCrossRef Orlichenko L, Geyer R, Yanagisawa M, Khauv D, Radisky ES, Anastasiadis PZ, et al. The 19-amino acid insertion in the tumor-associated splice isoform Rac1b confers specific binding to p120 catenin. J Biol Chem. 2010;285:19153–61.PubMedPubMedCentralCrossRef
189.
go back to reference Li G, Ying L, Wang H, Wei SS, Chen J, Chen YH, et al. Rac1b enhances cell survival through activation of the JNK2/c-JUN/Cyclin-D1 and AKT2/MCL1 pathways. Oncotarget. 2016;7:17970–85.PubMedPubMedCentral Li G, Ying L, Wang H, Wei SS, Chen J, Chen YH, et al. Rac1b enhances cell survival through activation of the JNK2/c-JUN/Cyclin-D1 and AKT2/MCL1 pathways. Oncotarget. 2016;7:17970–85.PubMedPubMedCentral
190.
go back to reference Radisky DC, Levy DD, Littlepage LE, Liu H, Nelson CM, Fata JE, et al. Rac1b and reactive oxygen species mediate MMP-3-induced EMT and genomic instability. Nature. 2005;436:123–7.PubMedPubMedCentralCrossRef Radisky DC, Levy DD, Littlepage LE, Liu H, Nelson CM, Fata JE, et al. Rac1b and reactive oxygen species mediate MMP-3-induced EMT and genomic instability. Nature. 2005;436:123–7.PubMedPubMedCentralCrossRef
191.
go back to reference Stallings-Mann ML, Waldmann J, Zhang Y, Miller E, Gauthier ML, Visscher DW, et al. Matrix metalloproteinase induction of Rac1b, a key effector of lung cancer progression. Sci Transl Med. 2012;4:142ra95.PubMedPubMedCentralCrossRef Stallings-Mann ML, Waldmann J, Zhang Y, Miller E, Gauthier ML, Visscher DW, et al. Matrix metalloproteinase induction of Rac1b, a key effector of lung cancer progression. Sci Transl Med. 2012;4:142ra95.PubMedPubMedCentralCrossRef
192.
go back to reference Henriques AFA, Barros P, Moyer MP, Matos P, Jordan P. Expression of tumor-related Rac1b antagonizes B-Raf-induced senescence in colorectal cells. Cancer Lett. 2015;369:368–75.PubMedCrossRef Henriques AFA, Barros P, Moyer MP, Matos P, Jordan P. Expression of tumor-related Rac1b antagonizes B-Raf-induced senescence in colorectal cells. Cancer Lett. 2015;369:368–75.PubMedCrossRef
194.
go back to reference Nelles DA, Yeo A. Alternative splicing in stem cell self-renewal and diferentiation. Adv Exp Med Biol. 2010;695:92–104.PubMedCrossRef Nelles DA, Yeo A. Alternative splicing in stem cell self-renewal and diferentiation. Adv Exp Med Biol. 2010;695:92–104.PubMedCrossRef
195.
go back to reference Cloonan N, Forrest ARR, Kolle G, Gardiner BBA, Faulkner GJ, Brown MK, et al. Stem cell transcriptome profiling via massive-scale mRNA sequencing. Nat Meth. 2008;5:613–9.CrossRef Cloonan N, Forrest ARR, Kolle G, Gardiner BBA, Faulkner GJ, Brown MK, et al. Stem cell transcriptome profiling via massive-scale mRNA sequencing. Nat Meth. 2008;5:613–9.CrossRef
196.
go back to reference Wu JQ, Habegger L, Noisa P, Szekely A, Qiu C, Hutchison S, et al. Dynamic transcriptomes during neural differentiation of human embryonic stem cells revealed by short, long, and paired-end sequencing. Proc Natl Acad Sci U S A. 2010;107:5254–9.PubMedPubMedCentralCrossRef Wu JQ, Habegger L, Noisa P, Szekely A, Qiu C, Hutchison S, et al. Dynamic transcriptomes during neural differentiation of human embryonic stem cells revealed by short, long, and paired-end sequencing. Proc Natl Acad Sci U S A. 2010;107:5254–9.PubMedPubMedCentralCrossRef
197.
go back to reference Gabut M, Samavarchi-Tehrani P, Wang X, Slobodeniuc V, O’Hanlon D, Sung HK, et al. An alternative splicing switch regulates embryonic stem cell pluripotency and reprogramming. Cell. 2011;147:132–46.PubMedCrossRef Gabut M, Samavarchi-Tehrani P, Wang X, Slobodeniuc V, O’Hanlon D, Sung HK, et al. An alternative splicing switch regulates embryonic stem cell pluripotency and reprogramming. Cell. 2011;147:132–46.PubMedCrossRef
199.
go back to reference Chen L, Kostadima M, Martens JHA, Canu G, Garcia SP, Turro E, et al. Transcriptional diversity during lineage commitment of human blood progenitors. Science. 2014;345:1251033.PubMedPubMedCentralCrossRef Chen L, Kostadima M, Martens JHA, Canu G, Garcia SP, Turro E, et al. Transcriptional diversity during lineage commitment of human blood progenitors. Science. 2014;345:1251033.PubMedPubMedCentralCrossRef
200.
go back to reference Bland CS, Wang ET, Vu A, David MP, Castle JC, Johnson JM, et al. Global regulation of alternative splicing during myogenic differentiation. Nucleic Acids Res. 2010;38:7651–64.PubMedPubMedCentralCrossRef Bland CS, Wang ET, Vu A, David MP, Castle JC, Johnson JM, et al. Global regulation of alternative splicing during myogenic differentiation. Nucleic Acids Res. 2010;38:7651–64.PubMedPubMedCentralCrossRef
201.
go back to reference Singh RK, Xia Z, Bland CS, Kalsotra A, Scavuzzo MA, Curk T, et al. Rbfox2-coordinated alternative splicing of Mef2d and Rock2 controls myoblast fusion during myogenesis. Mol Cell. 2014;55:592–603.PubMedPubMedCentralCrossRef Singh RK, Xia Z, Bland CS, Kalsotra A, Scavuzzo MA, Curk T, et al. Rbfox2-coordinated alternative splicing of Mef2d and Rock2 controls myoblast fusion during myogenesis. Mol Cell. 2014;55:592–603.PubMedPubMedCentralCrossRef
202.
go back to reference Tang F, Barbacioru C, Bao S, Lee C, Nordman E, Wang X, et al. Tracing the derivation of embryonic stem cells from the inner cell mass by single-cell RNA-Seq analysis. Cell Stem Cell. 2010;6:468–78.PubMedPubMedCentralCrossRef Tang F, Barbacioru C, Bao S, Lee C, Nordman E, Wang X, et al. Tracing the derivation of embryonic stem cells from the inner cell mass by single-cell RNA-Seq analysis. Cell Stem Cell. 2010;6:468–78.PubMedPubMedCentralCrossRef
203.
go back to reference Ohta S, Nishida E, Yamanaka S, Yamamoto T. Global splicing pattern reversion during somatic cell reprogramming. Cell Rep. 2013;5:357–66.PubMedCrossRef Ohta S, Nishida E, Yamanaka S, Yamamoto T. Global splicing pattern reversion during somatic cell reprogramming. Cell Rep. 2013;5:357–66.PubMedCrossRef
204.
go back to reference Tanaka Y, Hysolli E, Su J, Xiang Y, Kim KY, Zhong M, et al. Transcriptome signature and regulation in human somatic cell reprogramming. Stem Cell Rep. 2015;4:1125–39.CrossRef Tanaka Y, Hysolli E, Su J, Xiang Y, Kim KY, Zhong M, et al. Transcriptome signature and regulation in human somatic cell reprogramming. Stem Cell Rep. 2015;4:1125–39.CrossRef
205.
go back to reference Cieply B, Park JW, Nakauka-Ddamba A, Bebee TW, Guo Y, Shang X, et al. Multiphasic and dynamic changes in alternative splicing during induction of pluripotency are coordinated by numerous RNA-binding proteins. Cell Rep. 2016;15:247–55.PubMedCrossRef Cieply B, Park JW, Nakauka-Ddamba A, Bebee TW, Guo Y, Shang X, et al. Multiphasic and dynamic changes in alternative splicing during induction of pluripotency are coordinated by numerous RNA-binding proteins. Cell Rep. 2016;15:247–55.PubMedCrossRef
206.
go back to reference Han H, Irimia M, Ross PJ, Sung HK, Alipanahi B, David L, et al. MBNL proteins repress ES-cell-specific alternative splicing and reprogramming. Nature. 2013;498:241–5.PubMedPubMedCentralCrossRef Han H, Irimia M, Ross PJ, Sung HK, Alipanahi B, David L, et al. MBNL proteins repress ES-cell-specific alternative splicing and reprogramming. Nature. 2013;498:241–5.PubMedPubMedCentralCrossRef
208.
go back to reference Lu Y, Loh YH, Li H, Cesana M, Ficarro SB, Parikh JR, et al. Alternative splicing of MBD2 supports self-renewal in human pluripotent stem cells. Stem Cell. 2014;15:92–101. Lu Y, Loh YH, Li H, Cesana M, Ficarro SB, Parikh JR, et al. Alternative splicing of MBD2 supports self-renewal in human pluripotent stem cells. Stem Cell. 2014;15:92–101.
209.
go back to reference Atlasi Y, Mowla SJ, Ziaee SAM, Gokhale PJ, Andrews PW. OCT4 spliced variants are differentially expressed in human pluripotent and nonpluripotent cells. Stem Cells. 2008;26:3068–74.PubMedCrossRef Atlasi Y, Mowla SJ, Ziaee SAM, Gokhale PJ, Andrews PW. OCT4 spliced variants are differentially expressed in human pluripotent and nonpluripotent cells. Stem Cells. 2008;26:3068–74.PubMedCrossRef
210.
go back to reference Das S, Jena S, Levasseur DN. Alternative splicing produces Nanog protein variants with different capacities for self-renewal and pluripotency in embryonic stem cells. J Biol Chem. 2011;286:42690–703.PubMedPubMedCentralCrossRef Das S, Jena S, Levasseur DN. Alternative splicing produces Nanog protein variants with different capacities for self-renewal and pluripotency in embryonic stem cells. J Biol Chem. 2011;286:42690–703.PubMedPubMedCentralCrossRef
211.
go back to reference Yap K, Lim ZQ, Khandelia P, Friedman B, Makeyev EV. Coordinated regulation of neuronal mRNA steady-state levels through developmentally controlled intron retention. Genes Dev. 2012;26:1209–23.PubMedPubMedCentralCrossRef Yap K, Lim ZQ, Khandelia P, Friedman B, Makeyev EV. Coordinated regulation of neuronal mRNA steady-state levels through developmentally controlled intron retention. Genes Dev. 2012;26:1209–23.PubMedPubMedCentralCrossRef
212.
go back to reference Edwards CR, Ritchie W, Wong JJ-L, Schmitz U, Middleton R, An X, et al. A dynamic intron retention program in the mammalian megakaryocyte and erythrocyte lineages. Blood. 2016. Edwards CR, Ritchie W, Wong JJ-L, Schmitz U, Middleton R, An X, et al. A dynamic intron retention program in the mammalian megakaryocyte and erythrocyte lineages. Blood. 2016.
213.
go back to reference Neumüller RA, Richter C, Fischer A, Novatchkova M, Neumüller KG, Knoblich JA. Genome-wide analysis of self-renewal in drosophila neural stem cells by transgenic RNAi. Cell Stem Cell. 2011;8:580–93.PubMedPubMedCentralCrossRef Neumüller RA, Richter C, Fischer A, Novatchkova M, Neumüller KG, Knoblich JA. Genome-wide analysis of self-renewal in drosophila neural stem cells by transgenic RNAi. Cell Stem Cell. 2011;8:580–93.PubMedPubMedCentralCrossRef
214.
go back to reference Lu X, Göke J, Sachs F, Jacques PÉ, Liang H, Feng B, et al. SON connects the splicing-regulatory network with pluripotency in human embryonic stem cells. Nat Cell Biol. 2013;15:1141–52.PubMedPubMedCentralCrossRef Lu X, Göke J, Sachs F, Jacques PÉ, Liang H, Feng B, et al. SON connects the splicing-regulatory network with pluripotency in human embryonic stem cells. Nat Cell Biol. 2013;15:1141–52.PubMedPubMedCentralCrossRef
215.
go back to reference Toh CXD, Chan JW, Chong ZS, Wang HF, Guo HC, Satapathy S, et al. RNAi reveals phase-specific global regulators of human somatic cell reprogramming. Cell Rep. 2016;15:2597–607.PubMedCrossRef Toh CXD, Chan JW, Chong ZS, Wang HF, Guo HC, Satapathy S, et al. RNAi reveals phase-specific global regulators of human somatic cell reprogramming. Cell Rep. 2016;15:2597–607.PubMedCrossRef
216.
go back to reference Shibayama M, Ohno S, Osaka T, Sakamoto R, Tokunaga A, Nakatake Y, et al. Polypyrimidine tract-binding protein is essential for early mouse development and embryonic stem cell proliferation. FEBS J. 2009;276:6658–68.PubMedCrossRef Shibayama M, Ohno S, Osaka T, Sakamoto R, Tokunaga A, Nakatake Y, et al. Polypyrimidine tract-binding protein is essential for early mouse development and embryonic stem cell proliferation. FEBS J. 2009;276:6658–68.PubMedCrossRef
217.
go back to reference Suckale J, Wendling O, Masjkur J, Jäger M, Münster C, Anastassiadis K, et al. PTBP1 is required for embryonic development before gastrulation. PLoS ONE. 2011;6:e16992.PubMedPubMedCentralCrossRef Suckale J, Wendling O, Masjkur J, Jäger M, Münster C, Anastassiadis K, et al. PTBP1 is required for embryonic development before gastrulation. PLoS ONE. 2011;6:e16992.PubMedPubMedCentralCrossRef
218.
go back to reference Licatalosi DD, Yano M, Fak JJ, Mele A, Grabinski SE, Zhang C, et al. Ptbp2 represses adult-specific splicing to regulate the generation of neuronal precursors in the embryonic brain. Genes Dev. 2012;26:1626–42.PubMedPubMedCentralCrossRef Licatalosi DD, Yano M, Fak JJ, Mele A, Grabinski SE, Zhang C, et al. Ptbp2 represses adult-specific splicing to regulate the generation of neuronal precursors in the embryonic brain. Genes Dev. 2012;26:1626–42.PubMedPubMedCentralCrossRef
219.
go back to reference Card DAG, Hebbar PB, Li L, Trotter KW, Komatsu Y, Mishina Y, et al. Oct4/Sox2-regulated miR-302 targets cyclin D1 in human embryonic stem cells. Mol Cell Biol. 2008;28:6426–38.PubMedCrossRef Card DAG, Hebbar PB, Li L, Trotter KW, Komatsu Y, Mishina Y, et al. Oct4/Sox2-regulated miR-302 targets cyclin D1 in human embryonic stem cells. Mol Cell Biol. 2008;28:6426–38.PubMedCrossRef
220.
go back to reference Hirsch CL, Coban Akdemir Z, Wang L, Jayakumaran G, Trcka D, Weiss A, et al. Myc and SAGA rewire an alternative splicing network during early somatic cell reprogramming. Genes Dev. 2015;29:803–16.PubMedPubMedCentralCrossRef Hirsch CL, Coban Akdemir Z, Wang L, Jayakumaran G, Trcka D, Weiss A, et al. Myc and SAGA rewire an alternative splicing network during early somatic cell reprogramming. Genes Dev. 2015;29:803–16.PubMedPubMedCentralCrossRef
222.
go back to reference Gerber JM, Gucwa JL, Esopi D, Gurel M, Haffner MC, Vala M, et al. Genome-wide comparison of the transcriptomes of highly enriched normal and chronic myeloid leukemia stem and progenitor cell populations. Oncotarget. 2013;4:715–28.PubMedPubMedCentralCrossRef Gerber JM, Gucwa JL, Esopi D, Gurel M, Haffner MC, Vala M, et al. Genome-wide comparison of the transcriptomes of highly enriched normal and chronic myeloid leukemia stem and progenitor cell populations. Oncotarget. 2013;4:715–28.PubMedPubMedCentralCrossRef
223.
go back to reference Crews LA, Balaian L, Santos NPD, Leu HS, Court AC, Lazzari E, et al. RNA splicing modulation selectively impairs leukemia stem cell maintenance in secondary human AML. Cell Stem Cell. 2016;19:599–612.PubMedCrossRef Crews LA, Balaian L, Santos NPD, Leu HS, Court AC, Lazzari E, et al. RNA splicing modulation selectively impairs leukemia stem cell maintenance in secondary human AML. Cell Stem Cell. 2016;19:599–612.PubMedCrossRef
224.
225.
go back to reference Holm F, Hellqvist E, Mason CN, Ali SA, Delos-Santos N, Barrett CL, et al. Reversion to an embryonic alternative splicing program enhances leukemia stem cell self-renewal. Proc Natl Acad Sci U S A. 2015;112:15444–9.PubMedPubMedCentralCrossRef Holm F, Hellqvist E, Mason CN, Ali SA, Delos-Santos N, Barrett CL, et al. Reversion to an embryonic alternative splicing program enhances leukemia stem cell self-renewal. Proc Natl Acad Sci U S A. 2015;112:15444–9.PubMedPubMedCentralCrossRef
226.
go back to reference Powell DR, Blasky AJ, Britt SG, Artinger KB. Riding the crest of the wave: parallels between the neural crest and cancer in epithelial-to-mesenchymal transition and migration. Wiley Interdiscip Rev Syst Biol Med. 2013;5:511–22.PubMedPubMedCentralCrossRef Powell DR, Blasky AJ, Britt SG, Artinger KB. Riding the crest of the wave: parallels between the neural crest and cancer in epithelial-to-mesenchymal transition and migration. Wiley Interdiscip Rev Syst Biol Med. 2013;5:511–22.PubMedPubMedCentralCrossRef
227.
go back to reference Chaneton B, Gottlieb E. Rocking cell metabolism: revised functions of the key glycolytic regulator PKM2 in cancer. Trends Biochem Sci. 2012;37:309–16.PubMedCrossRef Chaneton B, Gottlieb E. Rocking cell metabolism: revised functions of the key glycolytic regulator PKM2 in cancer. Trends Biochem Sci. 2012;37:309–16.PubMedCrossRef
228.
go back to reference Takahashi K, Yamanaka S. Induction of pluripotent stem cells from mouse embryonic and adult fibroblast cultures by defined factors. Cell. 2006;126:663–76.PubMedCrossRef Takahashi K, Yamanaka S. Induction of pluripotent stem cells from mouse embryonic and adult fibroblast cultures by defined factors. Cell. 2006;126:663–76.PubMedCrossRef
229.
go back to reference David CJ, Chen M, Assanah M, Canoll P, Manley JL. HnRNP proteins controlled by c-Myc deregulate pyruvate kinase mRNA splicing in cancer. Nature. 2010;463:364–8. David CJ, Chen M, Assanah M, Canoll P, Manley JL. HnRNP proteins controlled by c-Myc deregulate pyruvate kinase mRNA splicing in cancer. Nature. 2010;463:364–8.  
230.
go back to reference Calabretta S, Bielli P, Passacantilli I, Pilozzi E, Fendrich V, Capurso G, et al. Modulation of PKM alternative splicing by PTBP1 promotes gemcitabine resistance in pancreatic cancer cells. Oncogene. 2016;35:2031–9.PubMedCrossRef Calabretta S, Bielli P, Passacantilli I, Pilozzi E, Fendrich V, Capurso G, et al. Modulation of PKM alternative splicing by PTBP1 promotes gemcitabine resistance in pancreatic cancer cells. Oncogene. 2016;35:2031–9.PubMedCrossRef
231.
go back to reference Wang Y, Chen D, Qian H, Tsai YS, Shao S, Liu Q, et al. The splicing factor RBM4 controls apoptosis, proliferation, and migration to suppress tumor progression. Cancer Cell. 2014;26:374–89.PubMedPubMedCentralCrossRef Wang Y, Chen D, Qian H, Tsai YS, Shao S, Liu Q, et al. The splicing factor RBM4 controls apoptosis, proliferation, and migration to suppress tumor progression. Cancer Cell. 2014;26:374–89.PubMedPubMedCentralCrossRef
232.
go back to reference Su CH, Hung KY, Hung SC, Tarn WY. RBM4 regulates neuronal differentiation of mesenchymal stem cells by modulating alternative splicing of pyruvate kinase M. Mol Cell Biol. 2016. Su CH, Hung KY, Hung SC, Tarn WY. RBM4 regulates neuronal differentiation of mesenchymal stem cells by modulating alternative splicing of pyruvate kinase M. Mol Cell Biol. 2016.
233.
go back to reference Goff DJ, Court Recart A, Sadarangani A, Chun HJ, Barrett CL, Krajewska M, et al. A Pan-BCL2 inhibitor renders bone-marrow-resident human leukemia stem cells sensitive to tyrosine kinase inhibition. Cell Stem Cell. 2013;12:316–28.PubMedPubMedCentralCrossRef Goff DJ, Court Recart A, Sadarangani A, Chun HJ, Barrett CL, Krajewska M, et al. A Pan-BCL2 inhibitor renders bone-marrow-resident human leukemia stem cells sensitive to tyrosine kinase inhibition. Cell Stem Cell. 2013;12:316–28.PubMedPubMedCentralCrossRef
234.
go back to reference Abrahamsson AE, Geron I, Gotlib J, Dao KHT, Barroga CF, Newton IG, et al. Glycogen synthase kinase 3beta missplicing contributes to leukemia stem cell generation. Proc Natl Acad Sci U S A. 2009;106:3925–9.PubMedPubMedCentralCrossRef Abrahamsson AE, Geron I, Gotlib J, Dao KHT, Barroga CF, Newton IG, et al. Glycogen synthase kinase 3beta missplicing contributes to leukemia stem cell generation. Proc Natl Acad Sci U S A. 2009;106:3925–9.PubMedPubMedCentralCrossRef
235.
go back to reference Ferrarese R, Harsh GR, Yadav AK, Bug E, Maticzka D, Reichardt W, et al. Lineage-specific splicing of a brain-enriched alternative exon promotes glioblastoma progression. J Clin Invest. 2014;124:2861–76.PubMedPubMedCentralCrossRef Ferrarese R, Harsh GR, Yadav AK, Bug E, Maticzka D, Reichardt W, et al. Lineage-specific splicing of a brain-enriched alternative exon promotes glioblastoma progression. J Clin Invest. 2014;124:2861–76.PubMedPubMedCentralCrossRef
236.
go back to reference Todaro M, Gaggianesi M, Catalano V, Benfante A, Iovino F, Biffoni M, et al. CD44v6 is a marker of constitutive and reprogrammed cancer stem cells driving colon cancer metastasis. Cell Stem Cell. 2014;14:342–56.PubMedCrossRef Todaro M, Gaggianesi M, Catalano V, Benfante A, Iovino F, Biffoni M, et al. CD44v6 is a marker of constitutive and reprogrammed cancer stem cells driving colon cancer metastasis. Cell Stem Cell. 2014;14:342–56.PubMedCrossRef
237.
go back to reference Lau WM, Teng E, Chong HS, Lopez KAP, Tay AYL, Salto-Tellez M, et al. CD44v8-10 is a cancer-specific marker for gastric cancer stem cells. Cancer Res. 2014;74:2630–41.PubMedCrossRef Lau WM, Teng E, Chong HS, Lopez KAP, Tay AYL, Salto-Tellez M, et al. CD44v8-10 is a cancer-specific marker for gastric cancer stem cells. Cancer Res. 2014;74:2630–41.PubMedCrossRef
238.
go back to reference Zeng Y, Wodzenski D, Gao D, Shiraishi T, Terada N, Li Y, et al. Stress-response protein RBM3 attenuates the stem-like properties of prostate cancer cells by interfering with CD44 variant splicing. Cancer Res. 2013;73:4123–33.PubMedCrossRef Zeng Y, Wodzenski D, Gao D, Shiraishi T, Terada N, Li Y, et al. Stress-response protein RBM3 attenuates the stem-like properties of prostate cancer cells by interfering with CD44 variant splicing. Cancer Res. 2013;73:4123–33.PubMedCrossRef
239.
go back to reference Muramaki M, Miyake H, Kamidono S, Hara I. Over expression of CD44V8-10 in human bladder cancer cells decreases their interaction with hyaluronic acid and potentiates their malignant progression. J Urol. 2004;171:426–30.PubMedCrossRef Muramaki M, Miyake H, Kamidono S, Hara I. Over expression of CD44V8-10 in human bladder cancer cells decreases their interaction with hyaluronic acid and potentiates their malignant progression. J Urol. 2004;171:426–30.PubMedCrossRef
240.
go back to reference Cappellari M, Bielli P, Paronetto MP, Ciccosanti F, Fimia GM, Saarikettu J, et al. The transcriptional co-activator SND1 is a novelregulator of alternative splicing in prostate cancer cells. Oncogene. 2014;33:3794–802.PubMedCrossRef Cappellari M, Bielli P, Paronetto MP, Ciccosanti F, Fimia GM, Saarikettu J, et al. The transcriptional co-activator SND1 is a novelregulator of alternative splicing in prostate cancer cells. Oncogene. 2014;33:3794–802.PubMedCrossRef
241.
go back to reference Li R, Liang J, Ni S, Zhou T, Qing X, Li H, et al. A mesenchymal-to-epithelial transition initiates and is required for the nuclear reprogramming of mouse fibroblasts. Cell Stem Cell. 2010;7:51–63.PubMedCrossRef Li R, Liang J, Ni S, Zhou T, Qing X, Li H, et al. A mesenchymal-to-epithelial transition initiates and is required for the nuclear reprogramming of mouse fibroblasts. Cell Stem Cell. 2010;7:51–63.PubMedCrossRef
242.
go back to reference Olsson E, Honeth G, Bendahl PO, Saal LH, Gruvberger-Saal S, Ringnér M, et al. CD44 isoforms are heterogeneously expressed in breast cancer and correlate with tumor subtypes and cancer stem cell markers. BMC Cancer. 2011;11:418.PubMedPubMedCentralCrossRef Olsson E, Honeth G, Bendahl PO, Saal LH, Gruvberger-Saal S, Ringnér M, et al. CD44 isoforms are heterogeneously expressed in breast cancer and correlate with tumor subtypes and cancer stem cell markers. BMC Cancer. 2011;11:418.PubMedPubMedCentralCrossRef
243.
go back to reference Grosse-Wilde A, Fouquier d'Hérouël A, McIntosh E, Ertaylan G, Skupin A, Kuestner RE, et al. Stemness of the hybrid epithelial/mesenchymal state in breast cancer and its association with poor survival. PLoS ONE. 2015;10:e0126522.PubMedPubMedCentralCrossRef Grosse-Wilde A, Fouquier d'Hérouël A, McIntosh E, Ertaylan G, Skupin A, Kuestner RE, et al. Stemness of the hybrid epithelial/mesenchymal state in breast cancer and its association with poor survival. PLoS ONE. 2015;10:e0126522.PubMedPubMedCentralCrossRef
244.
go back to reference Kole R, Krainer AR, Altman S. RNA therapeutics: beyond RNA interference and antisense oligonucleotides. Nat Rev Drug Discov. 2012;11:125–40.PubMedPubMedCentral Kole R, Krainer AR, Altman S. RNA therapeutics: beyond RNA interference and antisense oligonucleotides. Nat Rev Drug Discov. 2012;11:125–40.PubMedPubMedCentral
245.
go back to reference Rigo F, Seth PP, Bennett CF. Antisense oligonucleotide-based therapies for diseases caused by pre-mRNA processing defects. Adv Exp Med Biol. 2014;825:303–52.PubMedCrossRef Rigo F, Seth PP, Bennett CF. Antisense oligonucleotide-based therapies for diseases caused by pre-mRNA processing defects. Adv Exp Med Biol. 2014;825:303–52.PubMedCrossRef
246.
go back to reference McClorey G, Wood MJ. An overview of the clinical application of antisense oligonucleotides for RNA-targeting therapies. Curr Opin Pharmacol. 2015;24:52–8.PubMedCrossRef McClorey G, Wood MJ. An overview of the clinical application of antisense oligonucleotides for RNA-targeting therapies. Curr Opin Pharmacol. 2015;24:52–8.PubMedCrossRef
Metadata
Title
EMT and stemness: flexible processes tuned by alternative splicing in development and cancer progression
Authors
Davide Pradella
Chiara Naro
Claudio Sette
Claudia Ghigna
Publication date
01-12-2017
Publisher
BioMed Central
Published in
Molecular Cancer / Issue 1/2017
Electronic ISSN: 1476-4598
DOI
https://doi.org/10.1186/s12943-016-0579-2

Other articles of this Issue 1/2017

Molecular Cancer 1/2017 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