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.

  • Article
  • Published:

Identification of the tumour transition states occurring during EMT

Abstract

In cancer, the epithelial-to-mesenchymal transition (EMT) is associated with tumour stemness, metastasis and resistance to therapy. It has recently been proposed that, rather than being a binary process, EMT occurs through distinct intermediate states. However, there is no direct in vivo evidence for this idea. Here we screen a large panel of cell surface markers in skin and mammary primary tumours, and identify the existence of multiple tumour subpopulations associated with different EMT stages: from epithelial to completely mesenchymal states, passing through intermediate hybrid states. Although all EMT subpopulations presented similar tumour-propagating cell capacity, they displayed differences in cellular plasticity, invasiveness and metastatic potential. Their transcriptional and epigenetic landscapes identify the underlying gene regulatory networks, transcription factors and signalling pathways that control these different EMT transition states. Finally, these tumour subpopulations are localized in different niches that differentially regulate EMT transition states.

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

Fig. 1: Identification of the tumour transition states occurring during EMT in vivo.
Fig. 2: EMT transition states present similar TPC capacity but exhibit differing plasticity.
Fig. 3: Different EMT transition states present distinct metastatic potential.
Fig. 4: Transcriptional and chromatin landscape of EMT transition states.
Fig. 5: Different EMT transition states are localized in different niches.

Similar content being viewed by others

References

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

    Article  CAS  PubMed  Google Scholar 

  2. 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 

  3. Huang, R. Y. et al. An EMT spectrum defines an anoikis-resistant and spheroidogenic intermediate mesenchymal state that is sensitive to e-cadherin restoration by a src-kinase inhibitor, saracatinib (AZD0530). Cell Death Dis. 4, e915 (2013).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  4. Zhang, J. et al. TGF-β-induced epithelial-to-mesenchymal transition proceeds through stepwise activation of multiple feedback loops. Sci. Signal. 7, ra91 (2014).

    Article  PubMed  Google Scholar 

  5. Hong, T. et al. An Ovol2-Zeb1 mutual inhibitory circuit governs bidirectional and multi-step transition between epithelial and mesenchymal states. PLoS Comput. Biol. 11, e1004569 (2015).

    Article  PubMed  PubMed Central  Google Scholar 

  6. Jolly, M. K. et al. Stability of the hybrid epithelial/mesenchymal phenotype. Oncotarget 7, 27067–27084 (2016).

    Article  PubMed  PubMed Central  Google Scholar 

  7. Bierie, B. et al. Integrin-β4 identifies cancer stem cell-enriched populations of partially mesenchymal carcinoma cells. Proc. Natl Acad. Sci. USA 114, E2337–E2346 (2017).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  8. Latil, M. et al. Cell-type-specific chromatin states differentially prime squamous cell carcinoma tumor-initiating cells for epithelial to mesenchymal transition. Cell Stem Cell 20, 191–204 (2017).

    Article  CAS  PubMed  Google Scholar 

  9. Chen, Q., Zhang, X. H. & Massagué, J. Macrophage binding to receptor VCAM-1 transmits survival signals in breast cancer cells that invade the lungs. Cancer Cell 20, 538–549 (2011).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  10. Seguin, L. et al. An integrin β3–KRAS–RalB complex drives tumour stemness and resistance to EGFR inhibition. Nat. Cell Biol. 16, 457–468 (2014).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  11. Slack-Davis, J. K., Atkins, K. A., Harrer, C., Hershey, E. D. & Conaway, M. Vascular cell adhesion molecule-1 is a regulator of ovarian cancer peritoneal metastasis. Cancer Res. 69, 1469–1476 (2009).

    Article  CAS  PubMed  Google Scholar 

  12. Wang, J. et al. CD51 correlates with the TGF-beta pathway and is a functional marker for colorectal cancer stem cells. Oncogene 36, 1351–1363 (2017).

    Article  PubMed  Google Scholar 

  13. Van Keymeulen, A. et al. Reactivation of multipotency by oncogenic PIK3CA induces breast tumour heterogeneity. Nature 525, 119–123 (2015).

    Article  ADS  PubMed  Google Scholar 

  14. Ye, X. et al. Upholding a role for EMT in breast cancer metastasis. Nature 547, E1–E3 (2017).

    Article  CAS  PubMed  Google Scholar 

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

    Article  ADS  CAS  PubMed  PubMed Central  Google Scholar 

  16. DelPozo Martin, Y. et al. Mesenchymal cancer cell-stroma crosstalk promotes niche activation, epithelial reversion, and metastatic colonization. Cell Reports 13, 2456–2469 (2015).

    Article  Google Scholar 

  17. Hidalgo, M. et al. Patient-derived xenograft models: an emerging platform for translational cancer research. Cancer Discov. 4, 998–1013 (2014).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  18. Puram, S. V. et al. Single-cell transcriptomic analysis of primary and metastatic tumor ecosystems in head and neck cancer. Cell 171, 1611–1624 (2017).

    Article  CAS  PubMed  Google Scholar 

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

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  20. 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 

  21. Meacham, C. E. & Morrison, S. J. Tumour heterogeneity and cancer cell plasticity. Nature 501, 328–337 (2013).

    Article  ADS  CAS  PubMed  PubMed Central  Google Scholar 

  22. 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 

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

    Article  ADS  CAS  PubMed  PubMed Central  Google Scholar 

  24. 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  ADS  CAS  PubMed  PubMed Central  Google Scholar 

  25. Aiello, N. M. et al. Upholding a role for EMT in pancreatic cancer metastasis. Nature 547, E7–E8 (2017).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  26. 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 

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

    Article  PubMed  Google Scholar 

  28. Siegel, P. M. & Massagué, J. Cytostatic and apoptotic actions of TGF-β in homeostasis and cancer. Nat. Rev. Cancer 3, 807–821 (2003).

    Article  CAS  PubMed  Google Scholar 

  29. Srinivas, S. et al. Cre reporter strains produced by targeted insertion of EYFP and ECFP into the ROSA26 locus. BMC Dev. Biol. 1, 4 (2001).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  30. Barker, N. et al. Identification of stem cells in small intestine and colon by marker gene Lgr5. Nature 449, 1003–1007 (2007).

    Article  ADS  CAS  PubMed  Google Scholar 

  31. Tuveson, D. A. et al. Endogenous oncogenic K-ras(G12D) stimulates proliferation and widespread neoplastic and developmental defects. Cancer Cell 5, 375–387 (2004).

    Article  CAS  PubMed  Google Scholar 

  32. Jonkers, J. et al. Synergistic tumor suppressor activity of BRCA2 and p53 in a conditional mouse model for breast cancer. Nat. Genet. 29, 418–425 (2001).

    Article  CAS  PubMed  Google Scholar 

  33. Van Keymeulen, A. et al. Distinct stem cells contribute to mammary gland development and maintenance. Nature 479, 189–193 (2011).

    Article  ADS  PubMed  Google Scholar 

  34. Lapouge, G. et al. Skin squamous cell carcinoma propagating cells increase with tumour progression and invasiveness. EMBO J. 31, 4563–4575 (2012).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  35. Dobin, A. et al. STAR: ultrafast universal RNA-seq aligner. Bioinformatics 29, 15–21 (2013).

    Article  CAS  PubMed  Google Scholar 

  36. Kiselev, V. Y. et al. SC3: consensus clustering of single-cell RNA-seq data. Nat. Methods 14, 483–486 (2017).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  37. Trapnell, C. et al. The dynamics and regulators of cell fate decisions are revealed by pseudotemporal ordering of single cells. Nat. Biotechnol. 32, 381–386 (2014).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  38. Langmead, B. & Salzberg, S. L. Fast gapped-read alignment with Bowtie 2. Nat. Methods 9, 357–359 (2012).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  39. Li, H. et al. The Sequence Alignment/Map format and SAMtools. Bioinformatics 25, 2078–2079 (2009).

    Article  PubMed  PubMed Central  Google Scholar 

  40. Zhang, Y. et al. Model-based analysis of ChIP-Seq (MACS). Genome Biol. 9, R137 (2008).

    Article  PubMed  PubMed Central  Google Scholar 

  41. Heinz, S. et al. Simple combinations of lineage-determining transcription factors prime cis-regulatory elements required for macrophage and B cell identities. Mol. Cell 38, 576–589 (2010).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  42. McLean, C. Y. et al. GREAT improves functional interpretation of cis-regulatory regions. Nat. Biotechnol. 28, 495–501 (2010).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  43. Anders, S., Pyl, P. T. & Huber, W. HTSeq—a Python framework to work with high-throughput sequencing data. Bioinformatics 31, 166–169 (2015).

    Article  CAS  PubMed  Google Scholar 

  44. Hu, Y. & Smyth, G. K. ELDA: extreme limiting dilution analysis for comparing depleted and enriched populations in stem cells and other assays. J. Immunol. Methods 15, (70–78 (2009).

    Google Scholar 

  45. Medina-Rivera, A. et al. RSAT 2015: regulatory sequence analysis tools. Nucleic Acids Res. 43, W50–W56 (2015).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

Download references

Acknowledgements

We thank the ULB animal facility, the ULB genomic core facility (F. Libert and A. Lefort), G. Berx (Ghent University) and M. Mazzone (VIB-KUL) for MMTV-PyMT mice. I.P. is supported by TELEVIE. P.A.S. is supported by FNRS. A.S., D.B. and T.V. are supported by KULeuven SymBioSys, Stiching Tegen Kanker, FWO postdoctoral fellowship #12W7318N and Marie Sklodowska-Curie fellowship #12O5617N. The Department of Pathology acknowledges Fonds Yvonne Boel. The PDX platform is supported by Fonds Erasme. C.Bla. is supported by WELBIO, FNRS, Fondation Contre le Cancer, ULB Foundation, European Research Council, Worldwide Cancer Research and the Foundation Baillet Latour.

Reviewer information

Nature thanks B. E. Bernstein, E. Thompson and the other anonymous reviewer(s) for their contribution to the peer review of this work.

Author information

Authors and Affiliations

Authors

Contributions

I.P. and C.Bla. designed the experiments and performed data analysis. I.P. performed most of the biological experiments. P.A.S. performed part of the FACS analysis and histological characterization. A.B. performed the bioinformatic analysis. A.S., D.B. and T.V. performed scRNA-seq and analysis. M.F., A.V.K and J.-C.M. helped with breast cancer experiments. T.R. performed CTC experiments. S.B. helped with ATAC-seq. I.S., N.D’H., S.D.C., E.M., C.Bal., Y.S., A.L. and F.S. provided human samples. C.D. performed FACS sorting. V.M., S.L., F.D.C. and S.S. performed immunostaining, blocking antibody injections and follow-up of the mice. All authors read and approved the final manuscript.

Corresponding author

Correspondence to Cédric Blanpain.

Ethics declarations

Competing interests

The authors declare no competing interests.

Additional information

Publisher’s note: Springer Nature remains neutral with regard to jurisdictional claims in published maps and institutional affiliations.

Extended data figures and tables

Extended Data Fig. 1 Screening of cell surface markers homogeneously and heterogeneously expressed in Epcam+ and Epcam TCs in skin SCCs.

a, b, FACS profile of the isotype control and homogeneously or heterogeneously expressed markers in Epcam+ (a) and Epcam (b) TCs using BD lyoplate panel. c, Screening of all markers found to be heterogeneously expressed in at least one tumour in the initial BD lyoplate screen on a much larger cohort of tumours. Epcam TCs were more heterogeneous than Epcam+ TCs and CD51, CD61 and CD106 were heterogeneously expressed in at least 75% of the tumours analysed and therefore were selected and used in combination for further experiments.

Extended Data Fig. 2 Gating strategy for FACS analysis and cell sorting of the different tumour subpopulations.

a, FACS plots showing the gating strategy used to FACS-isolate Epcam+ and Epcam cells from YFP+ tumours (Lgr5CreER/KrasG12D/p53KO and K8CreER/Pik3ca/p53KO). b, FACS plots showing the six different subpopulations of Epcam TCs: EpcamCD106CD51CD61 (triple-negative), EpcamCD106+CD51CD61, EpcamCD106CD51+CD61, EpcamCD106+CD51+CD61, EpcamCD106CD51+CD61+ and EpcamCD106+CD51+CD61+ (triple-positive) populations. ch, Post-sort purity plots for triple-negative (c), CD106+ (d), CD51+ (e), CD106+CD51+ (f), CD51+CD61+ (g) and triple-positive (h) subpopulations (n = 1). i, FACS plots showing the gating strategy used to FACS-isolate Epcam+ and Epcam cells from MMTV-PyMT mammary tumours.

Extended Data Fig. 3 Epithelial, hybrid and mesenchymal marker expression by scRNA-seq.

ae, Dimensionality reduction of scRNA-seq data using PCA colouring for pure epithelial genes Esrp1 and Ovol1 (a), hybrid epithelial genes Krt5 and Trp63 (b), hybrid mesenchymal genes Prrx1, Snai1, Zeb1, Zeb2, Pdgfra and Col3a1 (c, d) and pure mesenchymal genes Aspn and Mmp19 (e). Every dot represents a single cell and its shape represents the FACS sorting criteria (Epcam+ or Epcam). The colour scale represents the normalized expression of the respective genes. Pure epithelial genes are expressed only in Epcam+ TCs, hybrid epithelial genes in both Epcam+ and hybrid Epcam TCs, hybrid mesenchymal genes in hybrid Epcam TCs and highly mesenchymal Epcam TCs, and pure mesenchymal genes only in highly mesenchymal Epcam TCs. ae, n = 66 Epcam+ and n = 277 Epcam cells from one tumour. f, Immunostaining showing the co-expression of Epcam and E-cadherin in the majority of epithelial TCs. Rare Epcam + cells are E-cadherin (arrowheads). Scale bars, 50 μm.

Extended Data Fig. 4 EMT transition states in Pik3caH1047R/p53KO metaplastic mammary tumours.

a, Percentage of Epcam+ cells in the two groups of tumours that differ by the frequency of the different CD106, CD51, CD61 subpopulations in metaplastic-like breast tumours resulting from activation of the oncogenic mutation Pik3caH1047R and p53 deletion in luminal cells of the mammary gland (n = 11 biological replicates, mean ± s.e.m.). b, FACS profile showing the six different CD106, CD51, CD61 subpopulations in the most frequent group 1 mixed tumours containing Epcam+ and Epcam TCs. c, d, Distribution of the different tumour subpopulations in group 1 (c; mixed tumours with Epcam+ and Epcam TCs) and in group 2 (d; mesenchymal Epcam TCs) (n = 10 biological replicates, mean ± s.e.m.). e, Immunostaining of K14 and vimentin in cystospin of FACS-isolated YFP+Epcam+, YFP+Epcam triple-negative and YFP+Epcam triple-positive populations. Scale bars, 20 μm; n = 3 biological replicates. f, Proportions of cells expressing K14 and vimentin based on cytospin counts; n = 3 biological replicates. g, Number of lung metastases arising from the injection of 2,000 YFP+ TCs from different EMT subpopulations. Dots represent the number of metastases per condition and per mouse (n = 2). h, qRT–PCR showing the downregulation of epithelial markers and the upregulation of EMT markers in the different Epcam tumour subpopulations. Histogram represents mean ( + s.e.m.) fold-change in gene expression compared to Epcam+ tumour cells for mesenchymal genes and compared to triple-positive tumour cells for epithelial genes (n = 7 Epcam+, n = 3 triple-negative, n = 5 CD106+, n = 3 CD51+, n = 6 CD106+CD51+, n = 3 CD51+CD61+, n = 6 triple-positive, all are biological replicates). il, Immunostaining showing the expression of K14 and vimentin, YFP and K14, YFP and vimentin, or YFP and CD61 in area 1 (K14+Vim CD61; i), area 2 (K14+Vim+CD61; j), area 3 (K14+Vim+CD61+; k) and area 4 (K14Vim+CD61+; l). Scale bars, 50 μm. mp, Immunostaining showing the expression of YFP and CD45, CD68, CD31 or Lyve1 in area 1 (m), area 2 (n), area 3 (o) and area 4 (p), showing that immune infiltration and the density of vascular and lymphatic vessels are associated with late stages of EMT. Scale bars, 50 μm, n = 3 biological replicates. In hair follicle EMT-derived tumours, the chromatin and transcriptional landscape of the cancer cell of origin prime the tumour to undergo EMT during tumorigenesis; in Pik3ca/p53KO mammary metaplastic tumours this is not always the case. Metaplastic EMT tumours are more frequent in luminal-derived tumours than in basal/myoepithelial cells, which under physiological conditions present much more EMT-like features do than luminal cells.

Source data

Extended Data Fig. 5 EMT transition states in MMTV-PyMT luminal-like mammary tumours.

a, Percentage of Epcam+ cells in two groups of tumours that differ by the frequency of the different CD106, CD51, CD61 subpopulations in MMTV-PyMT luminal-like mammary tumours (n = 5 mice, mean ± s.e.m.). b, Distribution of the different tumour subpopulations in group 2 (mixed tumours with Epcam+ and Epcam TCs) (n = 9 tumours, n = 5 mice, mean ± s.e.m.). c, FACS profile showing the six different CD106, CD51, CD61 subpopulations in the group containing mixed tumours with Epcam+ and Epcam TCs. d, Immunostaining of K8 and vimentin in cystospin of FACS-isolated YFP+Epcam+, YFP+Epcam triple-negative and YFP+Epcam triple-positive populations. Scale bars, 20 μm, n = 2 biological replicates. e, Proportion of cells expressing K8 and vimentin based on cytospin counts (n = 2 tumours). f, Number of lung metastases arising from the injection of 10,000 TCs from the different EMT subpopulations (Epcam+, n = 5; triple-negative, n = 5; CD106+, n = 5; triple-positive, n = 4). All are biological replicates, mean + s.e.m. Two-sided Mann–Whitney test: Epcam+ versus triple-negative, P = 0.0079; Epcam+ versus CD106+, P = 0.0079; triple-negative versus triple-positive, P = 0.0238; CD106+ versus triple-positive, P = 0.0476; mean + s.e.m. g, qRT–PCR showing the downregulation of epithelial markers and the upregulation of EMT markers in the different Epcam tumour subpopulations. Histogram represents mean ± s.e.m. fold-change in gene expression compared to Epcam+ tumour cells for mesenchymal genes and compared to triple-positive tumour cells for epithelial genes (Epcam+, n = 5; triple-negative, n = 3; CD106+, n = 5; triple-positive, n = 4; all are biological replicates). hj, Immunostaining of pancytokeratin (PanKRT) and vimentin, PyMT and pancytokeratin or PyMT and vimentin in area 1 (pancytokeratin+Vim; h), area 2 (pancytokeratin+Vim+; i) and area 3 (pancytokeratinVim+; j). Scale bars, 50 μm. km, Immunostaining of PyMT and CD68 or Lyve1 in area 1 (k), area 2 (l) and area 3 (m), showing that immune infiltration and the density of lymphatic vessels are associated with late-stage EMT. Scale bars, 50 μm, n = 2 biological replicates.

Source data

Extended Data Fig. 6 EMT transition states in human patient-derived xenotransplantation (PDX).

ad, Immunostaining of pancytokeratin and vimentin, human-specific marker Ku80 and pancytokeratin or Ku80 and vimentin in poorly differentiated human breast (a), lung (b) or oesophagus (c, d) PDX. In these examples, three different areas can be identified: area 1 (Ku80+pancytokeratin+vimentin), area 2 (Ku80+pancytokeratin+vimentin+) and area 3 (Ku80+pancytokeratinvimentin+), showing that TCs with different degrees of EMT can be identified in a subset of poorly differentiated human cancers. Scale bars, 50 μm. e, PDX derived from aggressive and highly metastatic pleomorphic lung carcinoma, showing the hybrid EMT state as defined by co-expression of pancytokeratin and vimentin by the majority of TCs.

Extended Data Fig. 7 Transcription factor binding motif enrichment in opened chromatin regions in the different EMT populations.

a, Heat map representing unsupervised clustering of RNA-seq data from the different tumour subpopulations, showing that Epcam+ epithelial TCs, Epcam hybrid TCs and Epcam mesenchymal TCs fall into different clusters (n = 3 biological replicates). b, Representation of chromatin remodelling and associated transcription factors (TFs) during EMT based on the transcription factor motifs that were statistically enriched in ATAC-seq peaks that differed between tumour subpopulations as determined by Homer analysis. cf, Transcription factor motifs enriched in the ATAC-seq peaks that were upregulated between the indicated subpopulations as determined by Homer analysis using known or de novo motif search (asterisk) or using JASPAR motif matrix (hash symbol). gj, Transcription factor motifs enriched in the ATAC-seq peaks that were upregulated between the indicated tumour subpopulations as determined by Homer analysis using known or de novo motif search (asterisk). Green, core transcription factors; pink, epithelial transcription factors; yellow, mesenchymal transcription factors. cj, Analysis based on ATAC-seq results from different subpopulations derived from one tumour.

Extended Data Fig. 8 Identification of EMT-related tumour heterogeneity by scRNA-seq.

a, Heat map of normalized scRNA-seq expression of markers. Markers are obtained from the scRNA-seq data using SC3 with clustering parameter k = 5 and selecting genes with an area under the curve (AUC) > 0.8 and P < 0.01. Columns represent cells and rows represent genes. Colouring represents the normalized expression in the scRNA-seq data for each cell. b, Dimensionality reduction of scRNA-seq data using PCA colouring for clusters identified by SC3 clustering. Dots represent single cells; shape represents FACS sorting criteria. c, Dimensionality reduction of scRNA-seq data using PCA colouring for clusters identified by Monocle using only the top 500 genes with the highest covariability with CD51, CD61 and CD106 expression. Dots represent single cells; colour represents Monocle cluster; shape represents FACS sorting criteria. d, Pseudotime-ordering trajectory of scRNA-seq data using Monocle. The top 500 differentially expressed genes across clusters identified in a semi-supervised approach using Monocle were used to order cells in pseudotime using the DDRTree method. Dots represent single cells; colours represent assigned Monocle clusters. e, Heat map representing normalized scRNA-seq data after branched expression analysis modelling (BEAM) using Monocle for the top 100 differentially expressed genes across the branch-point. The pre-branch here is set to be the state corresponding to the Epcam+ FACS-sorted cells. Cell fates 1 and 2 represent the two branches of the pseudotime trajectory corresponding to more hybrid and mesenchymal EMT phases, respectively. Heat map colours represent gene-wise normalized expression across pseudotime. Genes are hierarchically clustered according to their expression pattern. ae, n = 66 Epcam+ and n = 277 Epcam cells from one tumour.

Extended Data Fig. 9 Effect of constitutive overexpression of p63 and inhibition of TGF-β signalling in vivo on the different EMT subpopulations.

a, Immunostaining of K14 and vimentin, YFP and K14 or YFP and vimentin in hair follicle-derived skin SCCs with constitutive overexpression of p63, showing that EMT is blocked at an early stage and only area 1 epithelial TCs (K14+vimentin) and area 2 early hybrid TCs (K14+vimentin+) can be detected. Scale bars, 50 μm, n = 5 biological replicates. b, Experimental strategy to inhibit TGF-β signalling during hair follicle-derived SCC tumorigenesis. c, Immunostaining of K14 and vimentin or YFP K14, vimentin, CD61, CD45, CD68, CD31 or Lyve1 in area 1 (K14+vimentin), area 2 (K14+vimentin+), area 3 (K14+vimentin+CD61+) and area 4 (K14vimentin+CD61+) in mice treated with control antibodies. d, Immunostaining showing the expression of K14 and vimentin, or YFP and K14, vimentin, CD61, CD45, CD68, CD31 or Lyve1 in area 1 and area 2 of mice treated with anti-TGFβ antibodies, showing that inhibition of TGF-β or Smad2 signalling blocks EMT progression at the early stages of EMT and no areas corresponding to late EMT states (area 3 and 4) could be detected. c, d, Scale bars, 50 μm, n = 3 biological replicates.

Extended Data Fig. 10 Macrophage depletion inhibits EMT progression.

a, Experimental strategy to deplete macrophages by administrating anti-Csfr1 and anti-Ccl2 antibodies. b, Immunostaining of K14 and vimentin or YFP and K14, vimentin, YFP, CD45, CD68, CD31 or Lyve1 in area 1 (K14+vimentin), area 2 (K14+vimentin+), area 3 (K14+vimentin+CD61+) and area 4 (K14vimentin+CD61+) in mice treated with control antibodies. Scale bars, 50 μm. c, Immunostaining of K14 and vimentin or YFP and K14, vimentin, CD61, CD45, CD68, CD31 or Lyve1 in area 1 and area 2 of mice treated with anti-Csf1r and anti-Ccl2 antibodies, showing that EMT is blocked at its early stages and no areas corresponding to late EMT stages (areas 3 and 4) could be detected. Scale bars, 50 μm, n = 3 biological replicates.

Supplementary information

Source data

Rights and permissions

Reprints and permissions

About this article

Check for updates. Verify currency and authenticity via CrossMark

Cite this article

Pastushenko, I., Brisebarre, A., Sifrim, A. et al. Identification of the tumour transition states occurring during EMT. Nature 556, 463–468 (2018). https://doi.org/10.1038/s41586-018-0040-3

Download citation

  • Received:

  • Accepted:

  • Published:

  • Issue Date:

  • DOI: https://doi.org/10.1038/s41586-018-0040-3

This article is cited by

Comments

By submitting a comment you agree to abide by our Terms and Community Guidelines. If you find something abusive or that does not comply with our terms or guidelines please flag it as inappropriate.

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