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.

The EMT-activator Zeb1 is a key factor for cell plasticity and promotes metastasis in pancreatic cancer

Subjects

Abstract

Metastasis is the major cause of cancer-associated death. Partial activation of the epithelial-to-mesenchymal transition program (partial EMT) was considered a major driver of tumour progression from initiation to metastasis. However, the role of EMT in promoting metastasis has recently been challenged, in particular concerning effects of the Snail and Twist EMT transcription factors (EMT-TFs) in pancreatic cancer. In contrast, we show here that in the same pancreatic cancer model, driven by Pdx1-cre-mediated activation of mutant Kras and p53 (KPC model), the EMT-TF Zeb1 is a key factor for the formation of precursor lesions, invasion and notably metastasis. Depletion of Zeb1 suppresses stemness, colonization capacity and in particular phenotypic/metabolic plasticity of tumour cells, probably causing the observed in vivo effects. Accordingly, we conclude that different EMT-TFs have complementary subfunctions in driving pancreatic tumour metastasis. Therapeutic strategies should consider these potential specificities of EMT-TFs to target these factors simultaneously.

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: Zeb1 depletion reduces invasion and metastasis in pancreatic cancer.
Figure 2: Depletion of Zeb1 affects phenotypic variability of tumour cells.
Figure 3: Depletion of Zeb1 affects stemness, tumorigenic and colonization capacities.
Figure 4: Depletion of Zeb1 reduces ADM- and PanIN-precursor lesions.
Figure 5: Depletion of Zeb1 reduces phenotypic variability.
Figure 6: Depletion of Zeb1 reduces TGFβ-induced cellular plasticity.
Figure 7: Depletion of Zeb1 reduces metabolic and phenotypic plasticity.

Similar content being viewed by others

Accession codes

Primary accessions

Gene Expression Omnibus

References

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

    Article  CAS  PubMed  Google Scholar 

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

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  3. Nieto, M. A., Huang, R. Y.-J., Jackson, R. A. & Thiery, J. P. EMT: 2016. Cell 166, 21–45 (2016).

    Article  CAS  PubMed  Google Scholar 

  4. 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).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  5. Vandewalle, C., Van Roy, F. & Berx, G. The role of the ZEB family of transcription factors in development and disease. Cell. Mol. Life Sci. 66, 773–787 (2009).

    Article  CAS  PubMed  Google Scholar 

  6. Zhang, P., Sun, Y. & Ma, L. ZEB1: at the crossroads of epithelial-mesenchymal transition, metastasis and therapy resistance. Cell Cycle 14, 481–487 (2015).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  7. Fischer, K. R. et al. Epithelial-to-mesenchymal transition is not required for lung metastasis but contributes to chemoresistance. Nature 527, 472–476 (2015).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  8. Zheng, X. et al. Epithelial-to-mesenchymal transition is dispensable for metastasis but induces chemoresistance in pancreatic cancer. Nature 527, 525–530 (2015).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  9. Hingorani, S. R. et al. Trp53R172H and KrasG12D cooperate to promote chromosomal instability and widely metastatic pancreatic ductal adenocarcinoma in mice. Cancer Cell 7, 469–483 (2005).

    Article  CAS  PubMed  Google Scholar 

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

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  11. Higashi, Y. et al. Impairment of T Cell Development in deltaEF1 Mutant Mice. J. Exp. Med. 185, 1467–1480 (1997).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  12. Brabletz, S. et al. Generation and characterization of mice for conditional inactivation of Zeb1. Genesis http://dx.doi.org/10.1002/dvg.23024 (2017).

  13. Martinelli, P. et al. GATA6 regulates EMT and tumour dissemination, and is a marker of response to adjuvant chemotherapy in pancreatic cancer. Gut http://dx.doi.org/10.1136/gutjnl-2015-311256 (2016).

  14. Laklai, H. et al. Genotype tunes pancreatic ductal adenocarcinoma tissue tension to induce matricellular fibrosis and tumor progression. Nat. Med. 22, 497–505 (2016).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  15. Erkan, M. et al. The activated stroma index is a novel and independent prognostic marker in pancreatic ductal adenocarcinoma. Clin. Gastroenterol. Hepatol. 6, 1155–1161 (2008).

    Article  PubMed  Google Scholar 

  16. Özdemir, B. C. et al. Depletion of carcinoma-associated fibroblasts and fibrosis induces immunosuppression and accelerates pancreas cancer with reduced survival. Cancer Cell 25, 719–734 (2014).

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  17. Rhim, A. D. et al. Stromal elements act to restrain, rather than support, pancreatic ductal adenocarcinoma. Cancer Cell 25, 735–747 (2014).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  18. 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).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

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

    Article  CAS  PubMed  Google Scholar 

  20. 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).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  21. Raj, D., Aicher, A. & Heeschen, C. Concise review: stem cells in pancreatic cancer: from concept to translation. Stem Cells 33, 2893–2902 (2015).

    Article  PubMed  Google Scholar 

  22. Vannier, C., Mock, K., Brabletz, T. & Driever, W. Zeb1 regulates E-cadherin and Epcam (epithelial cell adhesion molecule) expression to control cell behavior in early zebrafish development. J. Biol. Chem. 288, 18643–18659 (2013).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  23. Dosch, J. S., Ziemke, E. K., Shettigar, A., Rehemtulla, A. & Sebolt-Leopold, J. S. Cancer stem cell marker phenotypes are reversible and functionally homogeneous in a preclinical model of pancreatic cancer. Cancer Res. 75, 4582–4592 (2015).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

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

    Article  CAS  PubMed  Google Scholar 

  25. Herreros-Villanueva, M. et al. SOX2 promotes dedifferentiation and imparts stem cell-like features to pancreatic cancer cells. Oncogenesis 2, e61 (2013).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  26. Sanada, Y. et al. Histopathologic evaluation of stepwise progression of pancreatic carcinoma with immunohistochemical analysis of gastric epithelial transcription factor SOX2: comparison of expression patterns between invasive components and cancerous or nonneoplastic intraductal components. Pancreas 32, 164–170 (2006).

    Article  CAS  PubMed  Google Scholar 

  27. Singh, S. K. et al. Antithetical NFATc1–Sox2 and p53–miR200 signaling networks govern pancreatic cancer cell plasticity. EMBO J. 34, 517–530 (2015).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  28. Muller, P. A. J. & Vousden, K. H. Mutant p53 in cancer: new functions and therapeutic opportunities. Cancer Cell 25, 304–317 (2014).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  29. Rivlin, N., Koifman, G. & Rotter, V. p53 orchestrates between normal differentiation and cancer. Semin. Cancer Biol. 32, 10–17 (2015).

    Article  CAS  PubMed  Google Scholar 

  30. Morton, J. P. et al. Mutant p53 drives metastasis and overcomes growth arrest/senescence in pancreatic cancer. Proc. Natl Acad. Sci. USA 107, 246–251 (2010).

    Article  CAS  PubMed  Google Scholar 

  31. Lehmann, W. et al. ZEB1 turns into a transcriptional activator by interacting with YAP1 in aggressive cancer types. Nat. Commun. 7, 10498 (2016).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  32. Mock, K. et al. The EMT-activator ZEB1 induces bone metastasis associated genes including BMP-inhibitors. Oncotarget 6, 14399–14412 (2015).

    PubMed  PubMed Central  Google Scholar 

  33. Brabletz, S. et al. The ZEB1/miR-200 feedback loop controls Notch signalling in cancer cells. EMBO J. 30, 770–782 (2011).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  34. Singh, A. et al. A gene expression signature associated with “K-Ras addiction” reveals regulators of EMT and tumor cell survival. Cancer Cell 15, 489–500 (2009).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  35. Nakamura, T., Fidler, I. J. & Coombes, K. R. Gene expression profile of metastatic human pancreatic cancer cells depends on the organ microenvironment. Cancer Res. 67, 139–148 (2007).

    Article  CAS  PubMed  Google Scholar 

  36. Collisson, E. A. et al. Subtypes of pancreatic ductal adenocarcinoma and their differing responses to therapy. Nat. Med. 17, 500–503 (2011).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  37. Weissmueller, S. et al. Mutant p53 drives pancreatic cancer metastasis through cell-autonomous PDGF receptor β signaling. Cell 157, 382–394 (2014).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  38. Diepenbruck, M. & Christofori, G. Epithelial–mesenchymal transition (EMT) and metastasis: yes, no, maybe? Curr. Opin. Cell Biol. 43, 7–13 (2016).

    Article  CAS  PubMed  Google Scholar 

  39. Ye, X. & Weinberg, R. A. Epithelial–mesenchymal plasticity: a central regulator of cancer progression. Trends Cell Biol. 25, 675–686 (2015).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  40. Bellomo, C., Caja, L. & Moustakas, A. Transforming growth factor [beta] as regulator of cancer stemness and metastasis. Br. J. Cancer 115, 761–769 (2016).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  41. Korpal, M. & Kang, Y. Targeting the transforming growth factor-β signalling pathway in metastatic cancer. Eur. J. Cancer 46, 1232–1240 (2010).

    Article  CAS  PubMed  Google Scholar 

  42. Lehuédé, C., Dupuy, F., Rabinovitch, R., Jones, R. G. & Siegel, P. M. Metabolic plasticity as a determinant of tumor growth and metastasis. Cancer Res. 76, 5201–5208 (2016).

    Article  PubMed  CAS  Google Scholar 

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

    Article  PubMed  CAS  Google Scholar 

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

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  45. Chaffer, C. L. et al. Normal and neoplastic nonstem cells can spontaneously convert to a stem-like state. Proc. Natl Acad. Sci. USA 108, 7950–7955 (2011).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  46. Puisieux, A., Brabletz, T. & Caramel, J. Oncogenic roles of EMT-inducing transcription factors. Nat. Cell Biol. 16, 488–494 (2014).

    Article  CAS  PubMed  Google Scholar 

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

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  48. Gruber, R. et al. YAP1 and TAZ control pancreatic cancer initiation in mice by direct up-regulation of JAK–STAT3 signaling. Gastroenterology 151, 526–539 (2016).

    Article  CAS  PubMed  Google Scholar 

  49. Zhang, W. et al. Downstream of mutant KRAS, the transcription regulator YAP Is essential for neoplastic progression to pancreatic ductal adenocarcinoma. Sci. Signal. 7, ra42-ra42 (2014).

    Google Scholar 

  50. Moffitt, R. A. et al. Virtual microdissection identifies distinct tumor- and stroma-specific subtypes of pancreatic ductal adenocarcinoma. Nat. Genet. 47, 1168–1178 (2015).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  51. Galvan, J. A. et al. Expression of E-cadherin repressors SNAIL, ZEB1 and ZEB2 by tumour and stromal cells influences tumour-budding phenotype and suggests heterogeneity of stromal cells in pancreatic cancer. Br. J. Cancer 112, 1944–1950 (2015).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  52. Bronsert, P. et al. Prognostic significance of Zinc finger E-box binding homeobox 1 (ZEB1) expression in cancer cells and cancer-associated fibroblasts in pancreatic head cancer. Surgery 156, 97–108 (2014).

    Article  PubMed  Google Scholar 

  53. Singh, A. & Settleman, J. EMT, cancer stem cells and drug resistance: an emerging axis of evil in the war on cancer. Oncogene 29, 4741–4751 (2010).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  54. Eser, S., Schnieke, A., Schneider, G. & Saur, D. Oncogenic KRAS signalling in pancreatic cancer. Br. J. Cancer 111, 817–822 (2014).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  55. Ni, T. et al. Snail1-dependent p53 repression regulates expansion and activity of tumour-initiating cells in breast cancer. Nat. Cell Biol. 18, 1221–1232 (2016).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  56. Turajlic, S. & Swanton, C. Metastasis as an evolutionary process. Science 352, 169–175 (2016).

    Article  CAS  PubMed  Google Scholar 

  57. 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).

    Article  CAS  PubMed  Google Scholar 

  58. Denecker, G. et al. Identification of a ZEB2-MITF-ZEB1 transcriptional network that controls melanogenesis and melanoma progression. Cell Death Differ. 21, 1250–1261 (2014).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  59. Ye, X. et al. Distinct EMT programs control normal mammary stem cells and tumour-initiating cells. Nature 525, 256–260 (2015).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  60. Tiwari, N. et al. Sox4 is a master regulator of epithelial-mesenchymal transition by controlling Ezh2 expression and epigenetic reprogramming. Cancer Cell 23, 768–783 (2013).

    Article  CAS  PubMed  Google Scholar 

  61. Tran, H. D. et al. Transient SNAIL1 expression is necessary for metastatic competence in breast cancer. Cancer Res. 74, 6330–6340 (2014).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  62. Olive, K. P. et al. Mutant p53 gain of function in two mouse models of Li-Fraumeni syndrome. Cell 119, 847–860 (2004).

    Article  CAS  PubMed  Google Scholar 

  63. Jackson, E. L. et al. Analysis of lung tumor initiation and progression using conditional expression of oncogenic K-ras. Genes Dev. 15, 3243–3248 (2001).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  64. Hingorani, S. R. et al. Preinvasive and invasive ductal pancreatic cancer and its early detection in the mouse. Cancer Cell 4, 437–450 (2003).

    Article  CAS  PubMed  Google Scholar 

  65. Chan, I. T. et al. Conditional expression of oncogenic K-ras from its endogenous promoter induces a myeloproliferative disease. J. Clin. Invest. 113, 528–538 (2004).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  66. Novak, A., Guo, C., Yang, W., Nagy, A. & Lobe, C. G. Z/EG, a double reporter mouse line that expresses enhanced green fluorescent protein upon Cre-mediated excision. Genesis 28, 147–155 (2000).

    Article  CAS  PubMed  Google Scholar 

  67. Murray, S. A., Carver, E. A. & Gridley, T. Generation of a Snail1 (Snai1) conditional null allele. Genesis 44, 7–11 (2006).

    Article  CAS  PubMed  Google Scholar 

  68. Meidhof, S. et al. ZEB1-associated drug resistance in cancer cells is reversed by the class I HDAC inhibitor mocetinostat. EMBO Mol. Med. 7, 831–847 (2015).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  69. Dunning, M. J., Smith, M. L., Ritchie, M. E. & Tavare, S. beadarray: R classes and methods for Illumina bead-based data. Bioinformatics 23, 2183–2184 (2007).

    Article  CAS  PubMed  Google Scholar 

  70. Johnson, W. E., Li, C. & Rabinovic, A. Adjusting batch effects in microarray expression data using empirical Bayes methods. Biostatistics 8, 118–127 (2007).

    Article  PubMed  Google Scholar 

Download references

Acknowledgements

We thank B. Schlund, E. Bauer and J. Pfannstiel, as well as U. Appelt and M. Mroz (Core Unit Cell Sorting and Immunomonitoring, FAU Erlangen, Germany) for technical assistance and R. Eccles for critical reading of the manuscript. We are grateful to D. Saur (Department of Internal Medicine, TU Munich, Germany) for providing the KPCS cell lines. We thank J. C. Wu, from Stanford University, for the MSCV-LUC_EF1-GFP-T2A-Puro plasmid. This work was supported by grants to T.B., S.B., M.B. and M.P.S. from the German Research Foundation (SFB850/A4, B2, Z1 and DFG BR 1399/9-1, DFG 1399/10-1, DFG BR4145/1-1) and from the German Consortium for Translational Cancer Research (DKTK).

Author information

Authors and Affiliations

Authors

Contributions

A.M.K. planned and carried out experiments and wrote the manuscript. J.M. carried out mouse experiments. M.L.L. carried out drug studies. O.S. generated the floxed Zeb1 allele. M.B. and H.B. carried out bioinformatics analyses. M.B. and D.M. carried out metabolic tests. W.R. carried out MRI analyses. P.B. carried out histological analyses. V.G.B. established mouse models. C.P. generated cell lines. T.H.W. carried out mouse experiments. S.B. generated the floxed Zeb1 allele, and planned and carried out experiments. M.P.S. generated the floxed Zeb1 allele, planned and carried out mouse experiments, was involved in coordination and wrote the manuscript. T.B. planned and coordinated the project, analysed data and wrote the manuscript. M.P.S. and T.B. contributed equally and share senior authorship.

Corresponding authors

Correspondence to Marc P. Stemmler or Thomas Brabletz.

Ethics declarations

Competing interests

The authors declare no competing financial interests.

Integrated supplementary information

Supplementary Figure 1 Characterisation of KPC, heterozygously and homozygously Zeb1 depleted KPC tumours.

(a) Representative Zeb1-immunolabeling of a GFP lineage-traced primary tumour showing Zeb1/GFP double-positive tumour cells (arrows). n = 5 independent tumors. Scale bar, 50 μm. (b) Representative consecutive sections of HE and indicated immunohistochemical stainings of four Zeb1 expressing KPC tumours demonstrating the heterogeneity in phenotype, grading and marker expression. A representative differentiated Zeb1-negative KPCZ tumour is shown for comparison. Arrows indicate Zeb1 positive tumour cells in the differentiated KPC tumour. n = 15 KPC, 13 KPCZ independent tumours. Scale bar, 100 μm. (c) Tumour-free survival of KPC mice vs. KPC mice with a heterozygous deletion of Zeb1 (KPCz) (n = 15 KPC, 16 KPCz independent tumours); log-rank (Mantel-Cox) test); tumour volume (0 = start of MRI measurements, n = 12 KPC, 14 KPCz independent tumours); error bars show mean ± S.E.M.; multiple t-tests with correction for multiple comparison using the Holm-Sidak method; grading, local invasion and relative ECM deposition of the respective tumours (n = 31 KPC, 17 KPCz; Mann-Whitney test (two-tailed); percentage of metastasized tumours (n = 35 KPC, 17 KPCz independent tumours; Chi-square test (two-tailed); n.s. = not significant.

Supplementary Figure 2 Characterisation of KPC vs. KPCZ tumours.

Representative images of immunohistochemical and histological stainings of KPC and KPCZ tumours and quantifications of the indicated markers are given. Asterisks label Zeb1-expressing stroma cells in KPCZ tumours. Specific blue MTS staining labels collagen fibres. Scale bars, 100 μm, for lower left image 50 μm. n = 48 KPC, 29 KPCZ independent tumours for Zeb1 and MTS; n = 15 independent tumours for KPC, 13 independent tumours for KPCZ for all other markers, error bars show mean ± S.D.; p < 0.0001, n.s. = not significant, Chi-square test (two-tailed) for Zeb1, E-cadherin and Sox2, unpaired Student’s t-test (two-tailed) for Ki67 and Casp3 (with Welch’s correction), Mann-Whitney test (two-tailed) for ECM and CD31.

Supplementary Figure 3 Characterisation of differentiaton markers in KPC vs. KPCZ tumours.

(a) Representative images of positive and negative immunohistochemical stainings and statistical analysis for the indicated EMT-TFs. Scale bar, 150 μm. n = 14 independent tumours for KPC, 13 independent tumours for KPCZ, Chi-square test (two-tailed); n.s. = not significant. (b) Representative images of immunohistochemical stainings and statistical analysis for expression of Gata6. Scale bar, 150 μm. n = 14 independent tumours for KPC, 13 independent tumours for KPCZ; error bars show mean ± S.D.; Mann-Whitney test (two-tailed), p < 0.001. (c) Representative images of differentiated KPCZ and undifferentiated KPC primary tumours (PT) and corresponding metastases (Met) with the same phenotype. Immunohistochemical labelling of Zeb1 expressing tumour cells in the KPC PT and Met (arrows). L = liver or lung tissue. n = 19 KPC, 4 KPCZ independent tumours and corresponding metastases. Scale bar, 100 μm.

Supplementary Figure 4 Characterisation of KPC vs. KPCZ tumour derived cell lines.

(a) Bright field image of primary cell lines from KPC and KPCZ tumours as well as HE stainings of the respective tumours after grafting in syngeneic mice and of the respective primary tumours are shown. Scale bars, 100 μm for bright field, 75 μm for HE stainings. (b) MTT viability assay for the isolated tumour cell lines after treatment with the indicated doses of gemcitabine and erlotinib. The calculated IC50 values for gemcitabine are shown. n = 3 biologically independent experiments, error bars show mean ± S.E.M. (c) Tumour onset after subcutaneous injection of 1 × 105 KPC and KPCZ cells into syngeneic mice. n = 4 mice per cell line, error bars show mean ± S.E.M. (d) Tumour grading, grading at invasive regions and relative ECM deposition of one representative tumour/cell line analysed in c) (n = 6 tumours for KPC, n = 5 tumours for KPCZ); error bars show mean ± S.D.;p < 0.05, p < 0.01, Mann-Whitney test (two-tailed).

Supplementary Figure 5 Depletion of Zeb1 affects tumour promoting capacities.

(a) Representative images of one visual field (n = 6 fields/cell line) showing GFP + cells/cell clusters in the lungs (green dots) 2 h after i.v. injection of KPC and KPCZ tumour cells and control lungs. Scale bar, 500 μm. (b) No. of tumours after subcutaneous injection of the indicated cell numbers for the KPC and KPCZ tumour cell lines and calculated fraction of tumourigenic cells. inf = infinite, Chi-square test. (c) Representative images showing spheres of KPC and KPCZ tumour cells. Scale bar, 500 μm and 50 μm for higher magnifications. (d) Percentage of cells in KPC and KPCZ lines positive for the indicated markers or marker combinations; n = 2 biologically independent experiments, error bars show ± S.D. Source data see Supplementary Table 5, Statistics Source Data. Relative mRNA expression levels (qRT-PCR) of indicated genes, mRNA levels of KPC661 was set to 1; n = 3 biologically independent experiments, Mann-Whitney test (two-tailed), p < 0.05, p < 0.01, error bars show mean ± S.E.M.

Supplementary Figure 6 Depletion of Zeb1 reduces early PanIN lesions.

(a) Consecutive sections showing representative HE and PAS stainings of precancerous PanIN lesions in the pancreas of two different 6 month old KC and of one KCZ mice. Specific dark blue PAS staining indicates the mucin-rich PanIN lesions. Scale bars, 2.5 mm and 150 μm for higher magnifications. Quantification of the PanIN area (% of pancreas area). n = 12 KC and 7 KCZ independent mice, error bars show mean ± S.D.; p < 0.01, unpaired Student’s t-test (two tailed) with Welch’s correction. (b) Gene set enrichment analyses (GSEA) of transcriptome data from KPCZ vs. KPC cells reveals reduction of gene signatures associated with cancer mesenchymal transition and Zeb1 targets in KPCZ vs. KPC cell lines. NES = normalized enrichment score; FDR = false discovery rate.

Supplementary Figure 7 Depletion of Zeb1 reduces tumour cell plasticity.

(a) Relative mRNA expression levels (qRT-PCR) of indicated genes in KPC and KPCZ cell lines treated for different times with TGFβ (time points: 0, 6 h, 1, 3, 7, 14, 21 days). mRNA levels of cell line 661 at day 0 were set to 1. n = 3 biologically independent experiments, error bars show mean ± S.E.M. Statistical analysis is shown for the comparison of TGFβ treated to untreated samples (grey bars) of each individual cell line p < 0.05, p < 0.01, p < 0.001, p < 0.0001, unpaired Student’s t-test (one-tailed) Source data see Supplementary Table 5, Statistics Source Data. (b) Table showing log2FC in mRNA expression levels (microarray) of genes previously determined as common ZEB1/YAP targets in KPC and KPCZ cell lines upon TGFβ treatment for 14 days. (cut-off: adj. p-value < 0.05 and log2FC > 0.5). (c) Representative images of consecutive sections of immunohistochemistry for Ck19 and Zeb1 comparing the plasticity of Zeb1 expression in central and invasive tumour regions. Tumours derived from one KPC and one KPCZ cell line are shown. Asterisks label Zeb1 expression in stroma cells, arrows indicate Zeb1 expression in tumour cells at the invasive front. Ck19 expression is shown to identify cancer cells. n = 15 KPC, 13 KPCZ independent tumours, Scale bars, 50 μm and 150 μm for higher magnifications.

Supplementary information

Supplementary Information

Supplementary Information (PDF 9368 kb)

Supplementary Table 1

Supplementary Information (XLSX 14 kb)

Supplementary Table 2

Supplementary Information (XLS 376 kb)

Supplementary Table 3

Supplementary Information (XLSX 11 kb)

Supplementary Table 4

Supplementary Information (XLSX 12 kb)

Supplementary Table 5

Supplementary Information (XLSX 29 kb)

Rights and permissions

Reprints and permissions

About this article

Check for updates. Verify currency and authenticity via CrossMark

Cite this article

Krebs, A., Mitschke, J., Lasierra Losada, M. et al. The EMT-activator Zeb1 is a key factor for cell plasticity and promotes metastasis in pancreatic cancer. Nat Cell Biol 19, 518–529 (2017). https://doi.org/10.1038/ncb3513

Download citation

  • Received:

  • Accepted:

  • Published:

  • Issue Date:

  • DOI: https://doi.org/10.1038/ncb3513

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