Skip to main content

Thank you for visiting nature.com. You are using a browser version with limited support for CSS. To obtain the best experience, we recommend you use a more up to date browser (or turn off compatibility mode in Internet Explorer). In the meantime, to ensure continued support, we are displaying the site without styles and JavaScript.

  • Review Article
  • Published:

Oncogenic roles of EMT-inducing transcription factors

Abstract

The plasticity of cancer cells underlies their capacity to adapt to the selective pressures they encounter during tumour development. Aberrant reactivation of epithelial–mesenchymal transition (EMT), an essential embryonic process, can promote cancer cell plasticity and fuel both tumour initiation and metastatic spread. Here we discuss the roles of EMT-inducing transcription factors in creating a pro-tumorigenic setting characterized by an intrinsic ability to withstand oncogenic insults through the mitigation of p53-dependent oncosuppressive functions and the gain of stemness-related properties.

This is a preview of subscription content, access via your institution

Access options

Rent or buy this article

Prices vary by article type

from$1.95

to$39.95

Prices may be subject to local taxes which are calculated during checkout

Figure 1: Schematic overview of EMT pathways.
Figure 2: Inhibition of tumour-suppressive programs by EMT-TFs.
Figure 3: Crosstalk between EMT-TFs, p53 and RB pathways and stemness factors in regulating cancer cell plasticity.

Similar content being viewed by others

References

  1. Thiery, J. P., Acloque, H., Huang, R. Y. & Nieto, M. A. Epithelial–mesenchymal transitions in development and disease. Cell 139, 871–890 (2009).

    Article  CAS  PubMed  Google Scholar 

  2. Nieto, M. A. Epithelial plasticity: a common theme in embryonic and cancer cells. Science 342, 1234850 (2013).

    PubMed  Google Scholar 

  3. Tam, W. L. & Weinberg, R. A. The epigenetics of epithelial-mesenchymal plasticity in cancer. Nat. Med. 19, 1438–1449 (2013).

    CAS  PubMed  PubMed Central  Google Scholar 

  4. Rowe, R. G. et al. Mesenchymal cells reactivate Snail1 expression to drive three-dimensional invasion programs. J. Cell Biol. 184, 399–408 (2009).

    CAS  PubMed  PubMed Central  Google Scholar 

  5. Arnoux, V., Nassour, M., L' Helgoualc'h, A., Hipskind, R. A. & Savagner, P. Erk5 controls Slug expression and keratinocyte activation during wound healing. Mol. Biol. Cell 19, 4738–4749 (2008).

    CAS  PubMed  PubMed Central  Google Scholar 

  6. Peinado, H., Olmeda, D. & Cano, A. Snail, Zeb and bHLH factors in tumour progression: an alliance against the epithelial phenotype? Nat. Rev. Cancer 7, 415–428 (2007).

    CAS  PubMed  Google Scholar 

  7. Iwano, M. et al. Evidence that fibroblasts derive from epithelium during tissue fibrosis. J. Clin. Invest. 110, 341–350 (2002).

    CAS  PubMed  PubMed Central  Google Scholar 

  8. Brabletz, T. et al. Variable beta-catenin expression in colorectal cancers indicates tumor progression driven by the tumor environment. Proc. Natl. Acad. Sci. USA 98, 10356–10361 (2001).

    CAS  PubMed  PubMed Central  Google Scholar 

  9. Scheel, C., Onder, T., Karnoub, A. & Weinberg, R. A. Adaptation versus selection: the origins of metastatic behavior. Cancer Res. 67, 11476–11479 (2007).

    CAS  PubMed  Google Scholar 

  10. Brabletz, T. To differentiate or not — routes towards metastasis. Nat. Rev. Cancer 12, 425–436 (2012).

    CAS  PubMed  Google Scholar 

  11. De Craene, B. & Berx, G. Regulatory networks defining EMT during cancer initiation and progression. Nat. Rev. Cancer 13, 97–110 (2013).

    CAS  PubMed  Google Scholar 

  12. Hugo, H. et al. Epithelial–mesenchymal and mesenchymal–epithelial transitions in carcinoma progression. J. Cell Physiol. 213, 374–383 (2007).

    CAS  PubMed  Google Scholar 

  13. Morel, A. P. et al. EMT inducers catalyze malignant transformation of mammary epithelial cells and drive tumorigenesis towards claudin-low tumors in transgenic mice. PLoS. Genet. 8, e1002723 (2012).

    CAS  PubMed  PubMed Central  Google Scholar 

  14. Valastyan, S. & Weinberg, R. A. Tumor metastasis: molecular insights and evolving paradigms. Cell 147, 275–292 (2011).

    CAS  PubMed  PubMed Central  Google Scholar 

  15. Kalluri, R. & Weinberg, R. A. The basics of epithelial–mesenchymal transition. J. Clin. Invest 119, 1420–1428 (2009).

    CAS  PubMed  PubMed Central  Google Scholar 

  16. Aigner, K. et al. The transcription factor ZEB1 (deltaEF1) promotes tumour cell dedifferentiation by repressing master regulators of epithelial polarity. Oncogene 26, 6979–6988 (2007).

    CAS  PubMed  PubMed Central  Google Scholar 

  17. Cordenonsi, M. et al. The Hippo transducer TAZ confers cancer stem cell-related traits on breast cancer cells. Cell 147, 759–772 (2011).

    CAS  PubMed  Google Scholar 

  18. Spaderna, S. et al. The transcriptional repressor ZEB1 promotes metastasis and loss of cell polarity in cancer. Cancer Res. 68, 537–544 (2008).

    CAS  PubMed  Google Scholar 

  19. Tsai, J. H., Donaher, J. L., Murphy, D. A., Chau, S. & Yang, J. Spatiotemporal regulation of epithelial-mesenchymal transition is essential for squamous cell carcinoma metastasis. Cancer Cell 22, 725–736 (2012).

    CAS  PubMed  PubMed Central  Google Scholar 

  20. Spaderna, S. et al. A transient, EMT-linked loss of basement membranes indicates metastasis and poor survival in colorectal cancer. Gastroenterology 131, 830–840 (2006).

    CAS  PubMed  Google Scholar 

  21. Yokobori, T. et al. Plastin3 is a novel marker for circulating tumor cells undergoing the epithelial–mesenchymal transition and is associated with colorectal cancer prognosis. Cancer Res. 73, 2059–2069 (2013).

    CAS  PubMed  Google Scholar 

  22. Yu, M. et al. Circulating breast tumor cells exhibit dynamic changes in epithelial and mesenchymal composition. Science 339, 580–584 (2013).

    CAS  PubMed  PubMed Central  Google Scholar 

  23. Bednarz-Knoll, N., Alix-Panabieres, C. & Pantel, K. Plasticity of disseminating cancer cells in patients with epithelial malignancies. Cancer Metast. Rev. 31, 673–687 (2012).

    CAS  Google Scholar 

  24. Smit, M. A., Geiger, T. R., Song, J. Y., Gitelman, I. & Peeper, D. S. A Twist–Snail axis critical for TrkB-induced epithelial–mesenchymal transition-like transformation, anoikis resistance, and metastasis. Mol. Cell Biol. 29, 3722–3737 (2009).

    CAS  PubMed  PubMed Central  Google Scholar 

  25. Labelle, M., Begum, S. & Hynes, R. O. Direct signaling between platelets and cancer cells induces an epithelial–mesenchymal-like transition and promotes metastasis. Cancer Cell 20, 576–590 (2011).

    CAS  PubMed  PubMed Central  Google Scholar 

  26. Chaffer, C. L. et al. Mesenchymal-to-epithelial transition facilitates bladder cancer metastasis: role of fibroblast growth factor receptor-2. Cancer Res. 66, 11271–11278 (2006).

    CAS  PubMed  Google Scholar 

  27. Korpal, M. et al. Direct targeting of Sec23a by miR-200s influences cancer cell secretome and promotes metastatic colonization. Nat. Med. 17, 1101–1108 (2011).

    CAS  PubMed  PubMed Central  Google Scholar 

  28. Ocana, O. H. et al. Metastatic colonization requires the repression of the epithelial-mesenchymal transition inducer Prrx1. Cancer Cell 22, 709–724 (2012).

    CAS  PubMed  Google Scholar 

  29. Vega, S. et al. Snail blocks the cell cycle and confers resistance to cell death. Genes Dev. 18, 1131–1143 (2004).

    CAS  PubMed  PubMed Central  Google Scholar 

  30. Scheel, C. & Weinberg, R. A. Phenotypic plasticity and epithelial–mesenchymal transitions in cancer and normal stem cells? Int. J. Cancer 129, 2310–2314 (2011).

    CAS  PubMed  PubMed Central  Google Scholar 

  31. Klymkowsky, M. W. & Savagner, P. Epithelial–mesenchymal transition: a cancer researcher's conceptual friend and foe. Am. J. Pathol. 174, 1588–1593 (2009).

    CAS  PubMed  PubMed Central  Google Scholar 

  32. Tam, W. L. et al. Protein kinase C alpha is a central signaling node and therapeutic target for breast cancer stem cells. Cancer Cell 24, 347–364 (2013).

    CAS  PubMed  PubMed Central  Google Scholar 

  33. Husemann, Y. et al. Systemic spread is an early step in breast cancer. Cancer Cell 13, 58–68 (2008).

    PubMed  Google Scholar 

  34. Rhim, A. D. et al. EMT and dissemination precede pancreatic tumor formation. Cell 148, 349–361 (2012).

    CAS  PubMed  PubMed Central  Google Scholar 

  35. Geradts, J. et al. Nuclear Snail1 and nuclear ZEB1 protein expression in invasive and intraductal human breast carcinomas. Hum. Pathol. 42, 1125–1131 (2011).

    CAS  PubMed  PubMed Central  Google Scholar 

  36. Mironchik, Y. et al. Twist overexpression induces in vivo angiogenesis and correlates with chromosomal instability in breast cancer. Cancer Res. 65, 10801–10809 (2005).

    CAS  PubMed  PubMed Central  Google Scholar 

  37. Montserrat, N. et al. Epithelial to mesenchymal transition in early stage endometrioid endometrial carcinoma. Hum. Pathol. 43, 632–643 (2012).

    PubMed  Google Scholar 

  38. Sarrio, D. et al. Epithelial–mesenchymal transition in breast cancer relates to the basal-like phenotype. Cancer Res. 68, 989–997 (2008).

    CAS  PubMed  Google Scholar 

  39. Matsumoto, T. et al. Loss of heterozygosity analysis shows monoclonal evolution with frequent genetic progression and divergence in esophageal carcinosarcoma. Hum. Pathol. 35, 322–327 (2004).

    CAS  PubMed  Google Scholar 

  40. Castilla, M. A. et al. Micro-RNA signature of the epithelial-mesenchymal transition in endometrial carcinosarcoma. J. Pathol. 223, 72–80 (2011).

    CAS  PubMed  Google Scholar 

  41. Spoelstra, N. S. et al. The transcription factor ZEB1 is aberrantly expressed in aggressive uterine cancers. Cancer Res. 66, 3893–3902 (2006).

    CAS  PubMed  Google Scholar 

  42. Trimboli, A. J. et al. Direct evidence for epithelial-mesenchymal transitions in breast cancer. Cancer Res. 68, 937–945 (2008).

    CAS  PubMed  Google Scholar 

  43. Damonte, P., Gregg, J. P., Borowsky, A. D., Keister, B. A. & Cardiff, R. D. EMT tumorigenesis in the mouse mammary gland. Lab. Invest. 87, 1218–1226 (2007).

    CAS  PubMed  PubMed Central  Google Scholar 

  44. Santisteban, M. et al. Immune-induced epithelial to mesenchymal transition in vivo generates breast cancer stem cells. Cancer Res. 69, 2887–2895 (2009).

    CAS  PubMed  PubMed Central  Google Scholar 

  45. Prat, A. et al. Phenotypic and molecular characterization of the claudin-low intrinsic subtype of breast cancer. Breast Cancer Res. 12, R68 (2010).

    PubMed  PubMed Central  Google Scholar 

  46. Prat, A. & Perou, C. M. Deconstructing the molecular portraits of breast cancer. Mol. Oncol. 5, 5–23 (2011).

    CAS  PubMed  Google Scholar 

  47. Hennessy, B. T. et al. Characterization of a naturally occurring breast cancer subset enriched in epithelial-to-mesenchymal transition and stem cell characteristics. Cancer Res. 69, 4116–4124 (2009).

    CAS  PubMed  PubMed Central  Google Scholar 

  48. Mani, S. A. et al. The epithelial–mesenchymal transition generates cells with properties of stem cells. Cell 133, 704–715 (2008).

    CAS  PubMed  PubMed Central  Google Scholar 

  49. Lamouille, S., Xu, J. & Derynck, R. Molecular mechanisms of epithelial–mesenchymal transition. Nat. Rev. Mol. Cell Biol. 15, 178–196 (2014).

    CAS  PubMed  PubMed Central  Google Scholar 

  50. Lopez-Novoa, J. M. & Nieto, M. A. Inflammation and EMT: an alliance towards organ fibrosis and cancer progression. EMBO Mol. Med. 1, 303–314 (2009).

    CAS  PubMed  PubMed Central  Google Scholar 

  51. Desprat, N., Supatto, W., Pouille, P. A., Beaurepaire, E. & Farge, E. Tissue deformation modulates twist expression to determine anterior midgut differentiation in Drosophila embryos. Dev. Cell 15, 470–477 (2008).

    CAS  PubMed  Google Scholar 

  52. Dong, C. et al. Loss of FBP1 by Snail-mediated repression provides metabolic advantages in basal-like breast cancer. Cancer Cell 23, 316–331 (2013).

    CAS  PubMed  PubMed Central  Google Scholar 

  53. Shin, S., Dimitri, C. A., Yoon, S. O., Dowdle, W. & Blenis, J. ERK2 but not ERK1 induces epithelial-to-mesenchymal transformation via DEF motif-dependent signaling events. Mol. Cell 38, 114–127 (2010).

    CAS  PubMed  PubMed Central  Google Scholar 

  54. Ansieau, S., Morel, A. P., Hinkal, G., Bastid, J. & Puisieux, A. TWISTing an embryonic transcription factor into an oncoprotein. Oncogene 29, 3173–3184 (2010).

    CAS  PubMed  Google Scholar 

  55. Ansieau, S. et al. Induction of EMT by twist proteins as a collateral effect of tumor-promoting inactivation of premature senescence. Cancer Cell 14, 79–89 (2008).

    CAS  PubMed  Google Scholar 

  56. Maestro, R. et al. Twist is a potential oncogene that inhibits apoptosis. Genes Dev. 13, 2207–2217 (1999).

    CAS  PubMed  PubMed Central  Google Scholar 

  57. Valsesia-Wittmann, S. et al. Oncogenic cooperation between H-Twist and N-Myc overrides failsafe programs in cancer cells. Cancer Cell 6, 625–630 (2004).

    CAS  PubMed  Google Scholar 

  58. Serrano, M., Lin, A. W., McCurrach, M. E., Beach, D. & Lowe, S. W. Oncogenic ras provokes premature cell senescence associated with accumulation of p53 and p16INK4a. Cell 88, 593–602 (1997).

    CAS  PubMed  Google Scholar 

  59. Bartkova, J. et al. DNA damage response as a candidate anti-cancer barrier in early human tumorigenesis. Nature 434, 864–870 (2005).

    CAS  PubMed  Google Scholar 

  60. Chen, Z. et al. Crucial role of p53-dependent cellular senescence in suppression of Pten-deficient tumorigenesis. Nature 436, 725–730 (2005).

    CAS  PubMed  PubMed Central  Google Scholar 

  61. Gorgoulis, V. G. et al. Activation of the DNA damage checkpoint and genomic instability in human precancerous lesions. Nature 434, 907–913 (2005).

    CAS  PubMed  Google Scholar 

  62. Collado, M. et al. Tumour biology: senescence in premalignant tumours. Nature 436, 642 (2005).

    CAS  PubMed  Google Scholar 

  63. Piccinin, S. et al. A “twist box” code of p53 inactivation: twist box: p53 interaction promotes p53 degradation. Cancer Cell 22, 404–415 (2012).

    CAS  PubMed  Google Scholar 

  64. Liu, Y., El-Naggar, S., Darling, D. S., Higashi, Y. & Dean, D. C. Zeb1 links epithelial-mesenchymal transition and cellular senescence. Development 135, 579–588 (2008).

    CAS  PubMed  Google Scholar 

  65. Ohashi, S. 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. 70, 4174–4184 (2010).

    CAS  PubMed  PubMed Central  Google Scholar 

  66. Swarbrick, A., Roy, E., Allen, T. & Bishop, J. M. Id1 cooperates with oncogenic Ras to induce metastatic mammary carcinoma by subversion of the cellular senescence response. Proc. Natl. Acad. Sci. USA 105, 5402–5407 (2008).

    CAS  PubMed  PubMed Central  Google Scholar 

  67. Wu, W. S. et al. Slug antagonizes p53-mediated apoptosis of hematopoietic progenitors by repressing puma. Cell 123, 641–653 (2005).

    CAS  PubMed  Google Scholar 

  68. Lee, S. H. et al. Blocking of p53-Snail binding, promoted by oncogenic K-Ras, recovers p53 expression and function. Neoplasia 11, 22–31 (2009).

    CAS  PubMed  PubMed Central  Google Scholar 

  69. Kajita, M., McClinic, K. N. & Wade, P. A. Aberrant expression of the transcription factors snail and slug alters the response to genotoxic stress. Mol. Cell Biol. 24, 7559–7566 (2004).

    CAS  PubMed  PubMed Central  Google Scholar 

  70. Pallier, K. et al. TWIST1 a new determinant of epithelial to mesenchymal transition in EGFR mutated lung adenocarcinoma. PLoS One 7, e29954 (2012).

    CAS  PubMed  PubMed Central  Google Scholar 

  71. Tran, P. T. et al. Twist1 suppresses senescence programs and thereby accelerates and maintains mutant Kras-induced lung tumorigenesis. PLoS Genet. 8, e1002650 (2012).

    CAS  PubMed  PubMed Central  Google Scholar 

  72. Olmeda, D. et al. SNAI1 is required for tumor growth and lymph node metastasis of human breast carcinoma MDA-MB-231 cells. Cancer Res. 67, 11721–11731 (2007).

    CAS  PubMed  Google Scholar 

  73. Olmeda, D. et al. Snai1 and Snai2 collaborate on tumor growth and metastasis properties of mouse skin carcinoma cell lines. Oncogene 27, 4690–4701 (2008).

    CAS  PubMed  Google Scholar 

  74. De, C. B. et al. Epidermal Snail expression drives skin cancer initiation and progression through enhanced cytoprotection, epidermal stem/progenitor cell expansion and enhanced metastatic potential. Cell Death. Differ. 21, 310–320 (2013).

    Google Scholar 

  75. Knudson, A. G. Stem cell regulation, tissue ontogeny, and oncogenic events. Semin. Cancer Biol. 3, 99–106 (1992).

    CAS  PubMed  Google Scholar 

  76. Scotting, P. J., Walker, D. A. & Perilongo, G. Childhood solid tumours: a developmental disorder. Nat. Rev. Cancer 5, 481–488 (2005).

    CAS  PubMed  Google Scholar 

  77. Dick, J. E. Stem cell concepts renew cancer research. Blood 112, 4793–4807 (2008).

    CAS  PubMed  Google Scholar 

  78. Lapidot, T. et al. A cell initiating human acute myeloid leukaemia after transplantation into SCID mice. Nature 367, 645–648 (1994).

    CAS  PubMed  Google Scholar 

  79. Morel, A. P. et al. Generation of breast cancer stem cells through epithelial-mesenchymal transition. PLoS One 3, e2888 (2008).

    PubMed  PubMed Central  Google Scholar 

  80. Vesuna, F., Lisok, A., Kimble, B. & Raman, V. Twist modulates breast cancer stem cells by transcriptional regulation of CD24 expression. Neoplasia 11, 1318–1328 (2009).

    CAS  PubMed  PubMed Central  Google Scholar 

  81. Taube, J. H. 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. USA 107, 15449–15454 (2010).

    CAS  PubMed  PubMed Central  Google Scholar 

  82. McCoy, E. L. et al. Six1 expands the mouse mammary epithelial stem/progenitor cell pool and induces mammary tumors that undergo epithelial–mesenchymal transition. J. Clin. Invest 119, 2663–2677 (2009).

    CAS  PubMed  PubMed Central  Google Scholar 

  83. Micalizzi, D. S. et al. The Six1 homeoprotein induces human mammary carcinoma cells to undergo epithelial-mesenchymal transition and metastasis in mice through increasing TGF-beta signaling. J. Clin. Invest 119, 2678–2690 (2009).

    CAS  PubMed  PubMed Central  Google Scholar 

  84. Yang, M. H. et al. Bmi1 is essential in Twist1-induced epithelial-mesenchymal transition. Nat. Cell Biol. 12, 982–992 (2010).

    PubMed  Google Scholar 

  85. Wellner, U. et al. The EMT-activator ZEB1 promotes tumorigenicity by repressing stemness-inhibiting microRNAs. Nat. Cell Biol. 11, 1487–1495 (2009).

    CAS  PubMed  Google Scholar 

  86. Shimono, Y. et al. Downregulation of miRNA-200c links breast cancer stem cells with normal stem cells. Cell 138, 592–603 (2009).

    CAS  PubMed  PubMed Central  Google Scholar 

  87. Guo, W. et al. Slug and Sox9 cooperatively determine the mammary stem cell state. Cell 148, 1015–1028 (2012).

    CAS  PubMed  PubMed Central  Google Scholar 

  88. Chaffer, C. L. et al. Poised chromatin at the ZEB1 promoter enables breast cancer cell plasticity and enhances tumorigenicity. Cell 154, 61–74 (2013).

    CAS  PubMed  PubMed Central  Google Scholar 

  89. Brabletz, T. EMT and MET in metastasis: where are the cancer stem cells? Cancer Cell 22, 699–701 (2012).

    CAS  PubMed  Google Scholar 

  90. Spike, B. T. & Wahl, G. M. p53, stem cells, and reprogramming: tumor suppression beyond guarding the genome. Genes Cancer 2, 404–419 (2011).

    CAS  PubMed  PubMed Central  Google Scholar 

  91. Donehower, L. A. & Lozano, G. 20 years studying p53 functions in genetically engineered mice. Nat. Rev. Cancer 9, 831–841 (2009).

    CAS  PubMed  Google Scholar 

  92. Jones, S. N., Roe, A. E., Donehower, L. A. & Bradley, A. Rescue of embryonic lethality in Mdm2-deficient mice by absence of p53. Nature 378, 206–208 (1995).

    CAS  PubMed  Google Scholar 

  93. Montes de Oca, L. R., Wagner, D. S. & Lozano, G. Rescue of early embryonic lethality in mdm2-deficient mice by deletion of p53. Nature 378, 203–206 (1995).

    Google Scholar 

  94. Cicalese, A. et al. The tumor suppressor p53 regulates polarity of self-renewing divisions in mammary stem cells. Cell 138, 1083–1095 (2009).

    CAS  PubMed  Google Scholar 

  95. Lin, T. et al. p53 induces differentiation of mouse embryonic stem cells by suppressing Nanog expression. Nat. Cell Biol. 7, 165–171 (2005).

    CAS  PubMed  Google Scholar 

  96. Pereira, L., Yi, F. & Merrill, B. J. Repression of Nanog gene transcription by Tcf3 limits embryonic stem cell self-renewal. Mol. Cell Biol. 26, 7479–7491 (2006).

    CAS  PubMed  PubMed Central  Google Scholar 

  97. Ruiz, S. et al. A high proliferation rate is required for cell reprogramming and maintenance of human embryonic stem cell identity. Curr. Biol. 21, 45–52 (2011).

    CAS  PubMed  Google Scholar 

  98. Li, H. et al. The Ink4/Arf locus is a barrier for iPS cell reprogramming. Nature 460, 1136–1139 (2009).

    CAS  PubMed  PubMed Central  Google Scholar 

  99. Hong, H. et al. Suppression of induced pluripotent stem cell generation by the p53–p21 pathway. Nature 460, 1132–1135 (2009).

    CAS  PubMed  PubMed Central  Google Scholar 

  100. Kawamura, T. et al. Linking the p53 tumour suppressor pathway to somatic cell reprogramming. Nature 460, 1140–1144 (2009).

    CAS  PubMed  PubMed Central  Google Scholar 

  101. Qin, H. et al. Regulation of apoptosis and differentiation by p53 in human embryonic stem cells. J. Biol. Chem. 282, 5842–5852 (2007).

    CAS  PubMed  Google Scholar 

  102. Yang, Y. L. et al. Amplification of PRKCI, located in 3q26, is associated with lymph node metastasis in esophageal squamous cell carcinoma. Genes Chromosome Canc. 47, 127–136 (2008).

    CAS  Google Scholar 

  103. Blum, B. & Benvenisty, N. The tumorigenicity of diploid and aneuploid human pluripotent stem cells. Cell Cycle 8, 3822–3830 (2009).

    CAS  PubMed  Google Scholar 

  104. Mizuno, H., Spike, B. T., Wahl, G. M. & Levine, A. J. Inactivation of p53 in breast cancers correlates with stem cell transcriptional signatures. Proc. Natl. Acad. Sci. USA 107, 22745–22750 (2010).

    CAS  PubMed  PubMed Central  Google Scholar 

  105. Jiang, Z. et al. RB1 and p53 at the crossroad of EMT and triple-negative breast cancer. Cell Cycle 10, 1563–1570 (2011).

    CAS  PubMed  Google Scholar 

  106. Kogan-Sakin, I. et al. Mutant p53(R175H) upregulates Twist1 expression and promotes epithelial–mesenchymal transition in immortalized prostate cells. Cell Death. Differ. 18, 271–281 (2011).

    CAS  PubMed  Google Scholar 

  107. Wang, S. P. et al. p53 controls cancer cell invasion by inducing the MDM2-mediated degradation of Slug. Nat. Cell Biol. 11, 694–704 (2009).

    CAS  PubMed  Google Scholar 

  108. Senoo, M., Pinto, F., Crum, C. P. & McKeon, F. p63 is essential for the proliferative potential of stem cells in stratified epithelia. Cell 129, 523–536 (2007).

    CAS  PubMed  Google Scholar 

  109. Chang, C. J. et al. p53 regulates epithelial-mesenchymal transition and stem cell properties through modulating miRNAs. Nat. Cell Biol. 13, 317–323 (2011).

    CAS  PubMed  PubMed Central  Google Scholar 

  110. He, L. et al. A microRNA component of the p53 tumour suppressor network. Nature 447, 1130–1134 (2007).

    CAS  PubMed  PubMed Central  Google Scholar 

  111. Hermeking, H. The miR-34 family in cancer and apoptosis. Cell Death. Differ. 17, 193–199 (2010).

    CAS  PubMed  Google Scholar 

  112. Kim, T. et al. p53 regulates epithelial–mesenchymal transition through microRNAs targeting ZEB1 and ZEB2. J. Exp. Med. 208, 875–883 (2011).

    CAS  PubMed  PubMed Central  Google Scholar 

  113. Brabletz, S. & Brabletz, T. The ZEB/miR-200 feedback loop — a motor of cellular plasticity in development and cancer? EMBO Rep. 11, 670–677 (2010).

    CAS  PubMed  PubMed Central  Google Scholar 

  114. Bracken, C. P. et al. A double-negative feedback loop between ZEB1-SIP1 and the microRNA-200 family regulates epithelial–mesenchymal transition. Cancer Res. 68, 7846–7854 (2008).

    CAS  PubMed  Google Scholar 

  115. Burk, U. et al. A reciprocal repression between ZEB1 and members of the miR-200 family promotes EMT and invasion in cancer cells. EMBO Rep. 9, 582–589 (2008).

    CAS  PubMed  PubMed Central  Google Scholar 

  116. Siemens, H. et al. miR-34 and SNAIL form a double-negative feedback loop to regulate epithelial-mesenchymal transitions. Cell Cycle 10, 4256–4271 (2011).

    CAS  PubMed  Google Scholar 

  117. Shi, L. et al. p53-induced miR-15a/16–11 and AP4 form a double-negative feedback loop to regulate epithelial–mesenchymal transition and metastasis in colorectal cancer. Cancer Res. 74, 532–542 (2014).

    CAS  PubMed  Google Scholar 

  118. Isenmann, S. et al. TWIST family of basic helix-loop-helix transcription factors mediate human mesenchymal stem cell growth and commitment. Stem Cells 27, 2457–2468 (2009).

    CAS  PubMed  Google Scholar 

  119. Chng, Z., Teo, A., Pedersen, R. A. & Vallier, L. SIP1 mediates cell-fate decisions between neuroectoderm and mesendoderm in human pluripotent stem cells. Cell Stem Cell 6, 59–70 (2010).

    CAS  PubMed  Google Scholar 

  120. Goossens, S. et al. The EMT regulator Zeb2/Sip1 is essential for murine embryonic hematopoietic stem/progenitor cell differentiation and mobilization. Blood 117, 5620–5630 (2011).

    CAS  PubMed  Google Scholar 

  121. Proia, T. A. et al. Genetic predisposition directs breast cancer phenotype by dictating progenitor cell fate. Cell Stem Cell 8, 149–163 (2011).

    CAS  PubMed  PubMed Central  Google Scholar 

  122. Caramel, J. et al. A switch in the expression of embryonic EMT-inducers drives the development of malignant melanoma. Cancer Cell 24, 466–480 (2013).

    CAS  PubMed  Google Scholar 

  123. Hoek, K. S. & Goding, C. R. Cancer stem cells versus phenotype-switching in melanoma. Pigm. Cell Melanoma Res. 23, 746–759 (2010).

    CAS  Google Scholar 

  124. Quintana, E. et al. Phenotypic heterogeneity among tumorigenic melanoma cells from patients that is reversible and not hierarchically organized. Cancer Cell 18, 510–523 (2010).

    CAS  PubMed  PubMed Central  Google Scholar 

  125. Shirley, S. H. et al. Slug expression during melanoma progression. Am. J. Pathol. 180, 2479–2489 (2012).

    CAS  PubMed  PubMed Central  Google Scholar 

  126. Sanchez-Martin, M. et al. SLUG (SNAI2) deletions in patients with Waardenburg disease. Hum. Mol. Genet. 11, 3231–3236 (2002).

    CAS  PubMed  Google Scholar 

  127. Sanchez-Martin, M. et al. Deletion of the SLUG (SNAI2) gene results in human piebaldism. Am. J. Med. Genet. A 122A, 125–132 (2003).

    PubMed  Google Scholar 

  128. Steingrimsson, E., Copeland, N. G. & Jenkins, N. A. Melanocytes and the microphthalmia transcription factor network. Annu. Rev. Genet. 38, 365–411 (2004).

    CAS  PubMed  Google Scholar 

  129. Van de Putte, T. et al. Mice lacking ZFHX1B, the gene that codes for Smad-interacting protein-1, reveal a role for multiple neural crest cell defects in the etiology of Hirschsprung disease-mental retardation syndrome. Am. J. Hum. Genet. 72, 465–470 (2003).

    CAS  PubMed  PubMed Central  Google Scholar 

  130. Dastot-Le, M. F. et al. ZFHX1B mutations in patients with Mowat-Wilson syndrome. Hum. Mutat. 28, 313–321 (2007).

    Google Scholar 

  131. Wakamatsu, N. et al. Mutations in SIP1, encoding Smad interacting protein-1, cause a form of Hirschsprung disease. Nat. Genet. 27, 369–370 (2001).

    CAS  PubMed  Google Scholar 

  132. Karreth, F. A. et al. In vivo identification of tumor-suppressive PTEN ceRNAs in an oncogenic BRAF-induced mouse model of melanoma. Cell 147, 382–395 (2011).

    CAS  PubMed  PubMed Central  Google Scholar 

  133. Mejlvang, J. et al. Direct repression of cyclin D1 by SIP1 attenuates cell cycle progression in cells undergoing an epithelial mesenchymal transition. Mol. Biol. Cell 18, 4615–4624 (2007).

    CAS  PubMed  PubMed Central  Google Scholar 

  134. Ozturk, N. et al. Reprogramming of replicative senescence in hepatocellular carcinoma-derived cells. Proc. Natl. Acad. Sci. USA 103, 2178–2183 (2006).

    CAS  PubMed  PubMed Central  Google Scholar 

  135. Liu, Y. et al. Zeb1 represses Mitf and regulates pigment synthesis, cell proliferation, and epithelial morphology. Invest Ophthalmol. Vis. Sci. 50, 5080–5088 (2009).

    PubMed  Google Scholar 

  136. Gupta, P. B. et al. The melanocyte differentiation program predisposes to metastasis after neoplastic transformation. Nat. Genet. 37, 1047–1054 (2005).

    CAS  PubMed  PubMed Central  Google Scholar 

  137. Klein, C. A. Cancer. The metastasis cascade. Science 321, 1785–1787 (2008).

    CAS  PubMed  Google Scholar 

Download references

Acknowledgements

We thank members of the Puisieux and Brabletz laboratories for helpful discussions and Sarah Kabani for critical reading of the manuscript. A.P. is supported by grants from the Ligue Nationale contre le Cancer, the Association pour la Recherche contre le Cancer (ARC), the Institut National du Cancer (PLBI009), the Lyon integrated Research Institute in Cancer (LYRIC) and the LabEx DEVweCAN (ANR-10-LABX-0061) of Lyon University, within the program 'Investissements d'Avenir' (ANR-11-IDEX-0007) operated by the French National Research Agency (ANR). A.P. is a member of the Institut Universitaire de France. T.B. is supported by the DFG (BR1399/6-1; SFB850,B2 and SFB992,C06) and the Deutsche Krebshilfe (grant no. 109430).

Author information

Authors and Affiliations

Authors

Corresponding author

Correspondence to Alain Puisieux.

Ethics declarations

Competing interests

The authors declare no competing financial interests.

Rights and permissions

Reprints and permissions

About this article

Check for updates. Verify currency and authenticity via CrossMark

Cite this article

Puisieux, A., Brabletz, T. & Caramel, J. Oncogenic roles of EMT-inducing transcription factors. Nat Cell Biol 16, 488–494 (2014). https://doi.org/10.1038/ncb2976

Download citation

  • Published:

  • Issue Date:

  • DOI: https://doi.org/10.1038/ncb2976

This article is cited by

Search

Quick links

Nature Briefing: Cancer

Sign up for the Nature Briefing: Cancer newsletter — what matters in cancer research, free to your inbox weekly.

Get what matters in cancer research, free to your inbox weekly. Sign up for Nature Briefing: Cancer