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:

Snail-induced claudin-11 prompts collective migration for tumour progression

Abstract

Epithelial–mesenchymal transition (EMT) is a pivotal mechanism for cancer dissemination. However, EMT-regulated individual cancer cell invasion is difficult to detect in clinical samples. Emerging evidence implies that EMT is correlated to collective cell migration and invasion with unknown mechanisms. We show that the EMT transcription factor Snail elicits collective migration in squamous cell carcinoma by inducing the expression of a tight junctional protein, claudin-11. Mechanistically, tyrosine-phosphorylated claudin-11 activates Src, which suppresses RhoA activity at intercellular junctions through p190RhoGAP, maintaining stable cell–cell contacts. In head and neck cancer patients, the Snail–claudin-11 axis prompts the formation of circulating tumour cell clusters, which correlate with tumour progression. Overexpression of snail correlates with increased claudin-11, and both are associated with a worse outcome. This finding extends the current understanding of EMT-mediated cellular migration via a non-individual type of movement to prompt cancer progression.

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: Snail is involved in collective cell migration and invasion of SCC.
Fig. 2: Claudin-11 contributes to collective cell migration and invasion.
Fig. 3: Direct regulation of CLDN11 by acetylated Snail.
Fig. 4: Claudin-11 inactivates RhoA via Src-phosphorylated p190RhoGAP.
Fig. 5: Tyrosine-phosphorylated claudin-11 recruits and activates Src.
Fig. 6: Correlation between the number of CTC clusters and the clinical course of HNSCC patients.
Fig. 7: Expression of Snail or claudin-11 correlates with an advanced HNSCC and a worse prognosis.

Similar content being viewed by others

Data availability

cDNA microarray data that support the findings of this study have been deposited in the GEO under the accession code GSE87841 (https://www.ncbi.nlm.nih.gov/geo/query/acc.cgi?acc=GSE87841). The array-CGH data were deposited in the GEO database under the accession code GSE114122 (https://www.ncbi.nlm.nih.gov/geo/query/acc.cgi?acc=GSE114122). The whole-genome sequencing data were deposited in the Sequence Read Archive (SRA) database under the accession code SRP157020 (https://www.ncbi.nlm.nih.gov/sra/SRP157020). Snail-regulated cell-movement and cell-adhesion genes were analysed using the trailblazer gene signature under the accession code GSE58643 (https://www.ncbi.nlm.nih.gov/geo/query/acc.cgi?acc=GSE58643). The differentially expressed genes regulated by Snail were analysed using the lymph node metastasis gene signature of HNSCC under the accession code GSE36942 (https://www.ncbi.nlm.nih.gov/geo/query/acc.cgi?acc=GSE36942). Mass spectrometry data have been deposited in ProteomeXchange with the primary accession code PXD010908 (https://www.ebi.ac.uk/pride/archive/projects/PXD010908). The hyperlink to the PrognoScan dataset derived from this resource for human colorectal cancer is http://dna00.bio.kyutech.ac.jp/PrognoScan-cgi/PrognoScan.cgi?TITLE=Prognostic+value%20of%20CLDN11%20mRNA%20expression%20in%20Colorectal%20cancer&DATA_POSTPROCESSING=None&TEST_NUM=80&MODE=SHOW_GRAPH&PROBE_ID=4037590 and for ovarian cancer is http://dna00.bio.kyutech.ac.jp/PrognoScan-cgi/PrognoScan.cgi?TITLE=Prognostic+value%20of%20CLDN11%20mRNA%20expression%20in%20Ovarian%20cancer&DATA_POSTPROCESSING=None&TEST_NUM=53&MODE=SHOW_GRAPH&PROBE_ID=2006434. Statistical source data for Figs. 1b,c,f, 2a,d,g, 3c–f, 4a,f, 6c–f and 7b–f and Supplementary Figs. 1e,g,h,o, 2b,c,f–i, 3a, 4l,o, 5c, 6f–h and 7a–d have been provided as Supplementary Table 11. All other data supporting the findings of this study are available from the corresponding author upon reasonable request.

References

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

    Article  CAS  Google Scholar 

  2. Teddy, J. M. & Kulesa, P. M. In vivo evidence for short- and long-range cell communication in cranial neural crest cells. Development 131, 6141–6151 (2004).

    Article  CAS  Google Scholar 

  3. Chapnick, D. A. & Liu, X. Leader cell positioning drives wound-directed collective migration in TGFβ-stimulated epithelial sheets. Mol. Biol. Cell 25, 1586–1593 (2014).

    Article  Google Scholar 

  4. Yang, M. H. et al. Direct regulation of TWIST by HIF-1α promotes metastasis. Nat. Cell Biol. 10, 295–305 (2008).

    Article  CAS  Google Scholar 

  5. 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).

    Article  CAS  Google Scholar 

  6. Westcott, J. M. et al. An epigenetically distinct breast cancer cell subpopulation promotes collective invasion. J. Clin. Invest. 125, 1927–1943 (2015).

    Article  Google Scholar 

  7. Aceto, N. et al. Circulating tumor cell clusters are oligoclonal precursors of breast cancer metastasis. Cell 158, 1110–1122 (2014).

    Article  CAS  Google Scholar 

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

    Article  CAS  Google Scholar 

  9. Friedl, P. & Gilmour, D. Collective cell migration in morphogenesis, regeneration and cancer. Nat. Rev. Mol. Cell Biol. 10, 445–457 (2009).

    Article  CAS  Google Scholar 

  10. Omelchenko, T., Vasiliev, J. M., Gelfand, I. M., Feder, H. H. & Bonder, E. M. Rho-dependent formation of epithelial “leader” cells during wound healing. Proc. Natl Acad. Sci. USA 100, 10788–10793 (2003).

    Article  CAS  Google Scholar 

  11. Poujade, M. et al. Collective migration of an epithelial monolayer in response to a model wound. Proc. Natl Acad. Sci. USA 104, 1639–1651 (2007).

    Article  Google Scholar 

  12. Cheung, K. J., Gabrielson, E., Werb, Z. & Ewald, A. J. Collective invasion in breast cancer requires a conserved basal epithelial program. Cell 155, 1639–1651 (2013).

    Article  CAS  Google Scholar 

  13. Wolf, K. et al. Multi-step pericellular proteolysis controls the transition from individual to collective cancer cell invasion. Nat. Cell Biol. 9, 893–904 (2007).

    Article  CAS  Google Scholar 

  14. Nguyen-Ngoc, K. V. et al. ECM microenvironment regulates collective migration and local dissemination in normal and malignant mammary epithelium. Proc. Natl Acad. Sci. USA 109, E2595–E2604 (2012).

    Article  CAS  Google Scholar 

  15. Montell, D. J. Morphogenetic cell movements: diversity from modular mechanical properties. Science 322, 1502–1505 (2008).

    Article  CAS  Google Scholar 

  16. Friedl, P., Locker, J., Sahai, E. & Segall, J. E. Classifying collective cancer cell invasion. Nat. Cell Biol. 14, 777–783 (2012).

    Article  Google Scholar 

  17. Ewald, A. J., Brenot, A., Duong, M., Chan, B. S. & Werb, Z. Collective epithelial migration and cell rearrangements drive mammary branching morphogenesis. Dev. Cell 14, 570–581 (2008).

    Article  CAS  Google Scholar 

  18. Langbein, L. et al. Tight junction-related structures in the absence of a lumen: occludin, claudins and tight junction plaque proteins in densely packed cell formations of stratified epithelia and squamous cell carcinomas. Eur. J. Cell Biol. 82, 385–400 (2003).

    Article  CAS  Google Scholar 

  19. Smalley, K. S. et al. Up-regulated expression of zonula occludens protein-1 in human melanoma associates with N-cadherin and contributes to invasion and adhesion. Am. J. Pathol. 166, 1541–1554 (2005).

    Article  CAS  Google Scholar 

  20. Cheung, K. J. et al. Polyclonal breast cancer metastases arise from collective dissemination of keratin 14-expressing tumor cell clusters. Proc. Natl Acad. Sci. USA 113, E854–E863 (2016).

    Article  CAS  Google Scholar 

  21. Gaggioli, C. et al. Fibroblast-led collective invasion of carcinoma cells with differing roles for RhoGTPases in leading and following cells. Nat. Cell Biol. 9, 1392–1400 (2007).

    Article  CAS  Google Scholar 

  22. Macpherson, I. R. et al. p120-catenin is required for the collective invasion of squamous cell carcinoma cells via a phosphorylation-independent mechanism. Oncogene 26, 5214–5228 (2007).

    Article  CAS  Google Scholar 

  23. Nakashima, Y. et al. Podoplanin is expressed at the invasive front of esophageal squamous cell carcinomas and is involved in collective cell invasion. Cancer Sci. 104, 1718–1725 (2013).

    Article  CAS  Google Scholar 

  24. Giampieri, S. et al. Localized and reversible TGFbeta signalling switches breast cancer cells from cohesive to single cell motility. Nat. Cell Biol. 11, 1287–1296 (2009).

    Article  CAS  Google Scholar 

  25. Hsu, D. S. et al. Acetylation of snail modulates the cytokinome of cancer cells to enhance the recruitment of macrophages. Cancer Cell. 26, 534–548 (2014).

    Article  CAS  Google Scholar 

  26. McLean, G. W. et al. The role of focal-adhesion kinase in cancer – a new therapeutic opportunity. Nat. Rev. Cancer 5, 505–515 (2005).

    Article  CAS  Google Scholar 

  27. Chen, Y. J., Wang, Y. N. & Chang, W. C. ERK2-mediated C-terminal serine phosphorylation of p300 is vital to the regulation of epidermal growth factor-induced keratin 16 gene expression. J. Biol. Chem. 282, 27215–2728 (2007).

    Article  CAS  Google Scholar 

  28. Morita, K., Sasaki, H., Fujimoto, K., Furuse, M. & Tsukita, S. Claudin-11/OSP-based tight junctions of myelin sheaths in brain and Sertoli cells in testis. J. Cell Biol. 145, 579–588 (1999).

    Article  CAS  Google Scholar 

  29. 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. 116, 1959–1967 (2003).

    Article  CAS  Google Scholar 

  30. Wu, J. C. et al. Antibody conjugated supported lipid bilayer for capturing and purification of viable tumor cells in blood for subsequent cell culture. Biomaterials 34, 5191–5199 (2013).

    Article  CAS  Google Scholar 

  31. Chen, J. Y. et al. Sensitive and specific biomimetic lipid coated microfluidics to isolate viable circulating tumor cells and microemboli for cancer detection. PLoS ONE 11, e0149633 (2016).

    Article  Google Scholar 

  32. Huang, W. C., Ju, T. K., Hung, M. C. & Chen, C. C. Phosphorylation of CBP by IKKα promotes cell growth by switching the binding preference of CBP from p53 to NF-kappaB. Mol. Cell 26, 75–87 (2007).

    Article  Google Scholar 

  33. Etienne-Manneville, S. & Hall, A. Rho GTPases in cell biology. Nature 420, 629–635 (2002).

    Article  CAS  Google Scholar 

  34. Hidalgo-Carcedo, C. et al. Collective cell migration requires suppression of actomyosin at cell–cell contacts mediated by DDR1 and the cell polarity regulators Par3 and Par6. Nat. Cell Biol. 13, 49–58 (2011).

    Article  CAS  Google Scholar 

  35. Omelchenko, T. & Hall, A. Myosin-IXA regulates collective epithelial cell migration by targeting RhoGAP activity to cell–cell junctions. Curr. Biol. 22, 278–288 (2012).

    Article  CAS  Google Scholar 

  36. Ponik, S. M., Trier, S. M., Wozniak, M. A., Eliceiri, K. W. & Keely, P. J. RhoA is down-regulated at cell–cell contacts via p190RhoGAP-B in response to tensional homeostasis. Mol. Biol. Cell 24, 1688–1699 (2013).

    Article  CAS  Google Scholar 

  37. McCormack, J., Welsh, N. J. & Braga, V. M. Cycling around cell–cell adhesion with Rho GTPase regulators. J. Cell Sci. 126, 379–391 (2013).

    Article  CAS  Google Scholar 

  38. Hu, K. Q. & Settleman, J. Tandem SH2 binding sites mediate the RasGAP-RhoGAP interaction: a conformational mechanism for SH3 domain regulation. EMBO J. 16, 473–483 (1997).

    Article  CAS  Google Scholar 

  39. Hernández, S. E., Settleman, J. & Koleske, A. J. Adhesion-dependent regulation of p190RhoGAP in the developing brain by the Abl-related gene tyrosine kinase. Curr. Biol. 14, 691–696 (2004).

    Article  Google Scholar 

  40. Chang, J. H., Gill, S., Settleman, J. & Parsons, S. J. c-Src regulates the simultaneous rearrangement of actin cytoskeleton, p190RhoGAP, and p120RasGAP following epidermal growth factor stimulation. J. Cell Biol. 130, 355–368 (1995).

    Article  CAS  Google Scholar 

  41. Yeatman, T. J. A renaissance for SRC. Nat. Rev. Cancer 4, 470–480 (2004).

    Article  CAS  Google Scholar 

  42. Fincham, V. J., Chudleigh, A. & Frame, M. C. Regulation of p190 Rho-GAP by v-Src is linked to cytoskeletal disruption during transformation. J. Cell Sci. 112, 947–956 (1999).

    CAS  PubMed  Google Scholar 

  43. Mitra, S. K., Hanson, D. A. & Schlaepfer, D. D. Focal adhesion kinase: in command and control of cell motility. Nat. Rev. Mol. Cell Biol. 6, 56–68 (2005).

    Article  CAS  Google Scholar 

  44. Chen, Y. J. et al. Genome-wide profiling of oral squamous cell carcinoma. J. Pathol. 204, 326–332 (2004).

    Article  CAS  Google Scholar 

  45. Lawrence, M. S. et al. Comprehensive genomic characterization of head and neck squamous cell carcinomas. Nature 517, 576–582 (2015).

    Article  CAS  Google Scholar 

  46. Liu, C. J., Lin, S. C., Chen, Y. J., Chang, K. M. & Chang, K. W. Array-comparative genomic hybridization to detect genomewide changes in microdissected primary and metastatic oral squamous cell carcinomas. Mol. Carcinog. 45, 721–731 (2006).

    Article  CAS  Google Scholar 

  47. Edge, S. B. & Compton, C. C. The American Joint Committee on Cancer: the 7th edition of the AJCC Cancer Staging Manual and the future of TNM. Ann. Surg. Oncol. 17, 1471–1474 (2010).

    Article  Google Scholar 

  48. Dhawan, P. et al. Claudin-1 regulates cellular transformation and metastatic behavior in colon cancer. Nat. Cell Biol. 115, 1765–1776 (2005).

    CAS  Google Scholar 

  49. Tabariès, S. & Siegel, P. M. The role of claudins in cancer metastasis. Oncogene 36, 1176–1190 (2017).

    Article  Google Scholar 

  50. Shang, X., Lin, X., Alvarez, E., Manorek, G. & Howell, S. B. Tight junction proteins claudin-3 and claudin-4 control tumor growth and metastases. Neoplasia 14, 974–985 (2012).

    Article  CAS  Google Scholar 

  51. Molnar, B., Ladanyi, A., Tanko, L., Sréter, L. & Tulassay, Z. Circulating tumor cell clusters in the peripheral blood of colorectal cancer patients. Clin. Cancer Res. 7, 4080–4085 (2001).

    CAS  PubMed  Google Scholar 

  52. Crosbie, P. A. et al. Circulating tumor cells detected in the tumor-draining pulmonary vein are associated with disease recurrence after surgical resection of NSCLC. J. Thorac. Oncol. 11, 1793–1797 (2016).

    Article  Google Scholar 

  53. Ozkumur, E. et al. Inertial focusing for tumor antigen-dependent and -independent sorting of rare circulating tumor cells. Sci. Transl. Med. 5, 179ra47 (2013).

    Article  Google Scholar 

  54. Autebert, J. et al. High purity microfluidic sorting and analysis of circulating tumor cells: towards routine mutation detection. Lab Chip 15, 2090–2101 (2015).

    Article  CAS  Google Scholar 

  55. Harb, W. et al. Mutational analysis of circulating tumor cells using a novel microfluidic collection device and qPCR assay. Transl. Oncol. 6, 528–538 (2013).

    Article  Google Scholar 

  56. Yang, W. H. et al. RAC1 activation mediates Twist1-induced cancer cell migration. Nat. Cell Biol. 14, 366–374 (2012).

    Article  CAS  Google Scholar 

  57. Hsu, D. S. et al. Lymphotoxin-β interacts with methylated EGFR to mediate acquired resistance to cetuximab in head and neck cancer. Clin. Cancer Res. 23, 4388–4401 (2017).

    Article  CAS  Google Scholar 

  58. Lin, C. J., Wu, K. H., Yew, F. H. & Lee, T. C. Differential cytotoxicity of cadmium to rat embryonic fibroblasts and human skin fibroblasts. Toxicol. Appl. Pharmacol. 133, 20–26 (1995).

    Article  CAS  Google Scholar 

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

    Article  Google Scholar 

  60. Hwang, W. L. et al. MicroRNA-146a directs the symmetric division of Snail-dominant colorectal cancer stem cells. Nat. Cell Biol. 16, 268–280 (2014).

    Article  CAS  Google Scholar 

  61. Yoshioka, S. et al. Genomic profiling of oral squamous cell carcinoma by array-based comparative genomic hybridization. PLoS ONE 8, e56165 (2013).

    Article  CAS  Google Scholar 

  62. Mizuno, H., Kitada, K., Nakai, K. & Sarai, A. PrognoScan: a new database for meta-analysis of the prognostic value of genes. BMC Med. Genomics 2, 18–28 (2009).

    Article  Google Scholar 

  63. Yang, M. H. et al. Increased NBS1 expression is a marker of aggressive head and neck cancer and overexpression of NBS1 contributes to transformation. Clin. Cancer Res. 12, 507–515 (2006).

    Article  CAS  Google Scholar 

Download references

Acknowledgements

We thank C.-H. Lin and Y.-L. Huang (National Yang-Ming University, Taiwan) for help with mass spectrometry data analysis; Z.-F. Chang (National Taiwan University, Taiwan) for providing the pTriEx–RhoA FLARE.sc Biosensor WT plasmid; M.-C. Hung (The University of Texas M.D. Anderson Cancer Center, TX) for the pHA–CBP, pHA–EECBP and pHA–AACBP plasmids; Y.-J. Lee (National Yang-Ming University, Taiwan) for the LT-3R plasmid; T.-C. Lee (Academia Sinica, Taiwan) for providing the HFW cell line for organotypic culture; and M.-Y. Liao and H.-C. Wu (Academia Sinica, Taiwan) for providing the mouse anti-human EpCAM antibody. We thank H.-Y. Chen (Instrument Center of National Chung-Hsing University, Taiwan) for technical support of the mass spectrometry analysis; S.-R. Chiang (Taipei Veterans General Hospital, Taiwan) for technical support of the IHC assay; J.-I Lai, J.-R. Huang, T.-H. Jeng, Y.-A. Hsieh, Y.-J. Lin, Y. Ho, H.-Y. Yu, C.-Y. Chen, K.-C. Wu, S.-Y. Ho, K.-C. Kao and Y.-J. Chen (Taipei Veterans General Hospital, Taiwan) for collecting the blood from HNSCC patients for the CTC assay; and Y.-J. Zheng for technical support of the CTC assay (Academia Sinica, Taiwan). We thank Welgene Biotech Co., Ltd (Taipei, Taiwan) for technical support of array-CGH, and T.-T. Liu (National Yang-Ming University, Taiwan) for technical support and analysing the WGS data. This work was supported in part by the Division of Experimental Surgery of the Department of Surgery, Taipei Veterans General Hospital. This work was supported by grants from the Ministry of Science and Technology (103-2633-H-010-001, 104-2321-B-010-005, 104-0210-01-09-02, 105-0210-01-13-01, 106-0210-01-15-02 and 107-0210-01-19-01); National Health Research Institutes (NHRI-EX107-10622BI); Taipei Veterans General Hospital (V107C-071, V107D32-001-MY2-1, VTA105-V1-3-2, VTA106-V1-3-3 and 107-V1-3-2); Veterans General Hospital-University System of Taiwan Joint Research Program (VGHUST107-G4-1-3); the Cancer Progression Research Center of National Yang-Ming University granted by the Featured Areas Research Center Program within the framework of the Higher Education Sprout Project by the Ministry of Education; and the Ministry of Health and Welfare, Center of Excellence for Cancer Research (MOHW107-TDU-B-211-114019).

Author information

Authors and Affiliations

Authors

Contributions

M.-H.Y. and C.-F.L. conceived and designed the experiments. C.-F.L. performed most of the experiments with the help of H.-Y.L. and W.-H.H., and they all analysed the data. D.S.-S.H. generated the SAS-LN cells. J.-Y.C., L.-C.W. and Y.-H.H. isolated the CTCs, performed the antibody staining, counted the number of CTCs and analysed the data under the supervision of Y.-C.C. S.-K.T. and M.-H.Y. provided patient care, collected all demographic data, interpreted the IHC results, analysed the relevant clinical parameters and performed the statistical comparisons. C.-F.L. and M.-H.Y. wrote the paper with the help of Y.-C.C. and J.-Y.C.

Corresponding authors

Correspondence to Ying-Chih Chang or Muh-Hwa Yang.

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.

Integrated supplementary information

Supplementary Figure 1 Snail is involved in collective migration.

(a) Snapshots from phase-contrast videos of cells in 2.5D. Scale bars, 100 μm. The representative data were from two independent experiments with similar results. (b) Western blots of EMT-TFs. Red arrows indicate Slug/Snail. (c) Images of knockdown of EMT-TFs in SAS cells in 2.5D. Scale bars, 100 μm. (d) Upper: snapshots from phase-contrast videos of TE1-pLKO/TE1-shSNAI1 in 2.5D. Scale bars, 100 μm. Lower: directionality of migration (n = 15 cells). The data is representative of three independent experiments with similar results. (e) Upper: schema of the animal experiment. Middle: bioluminescence images of mice at the end of week 3. Lower: quantification of IVIS images (n = 10 mice). The mean values are shown (two-sided Mann-Whitney test). (f) GSEA for Snail-regulated gene signature and lymph node metastasis of HNSCC patients. P-values were calculated based on 1000 permutations by the GSEA algorithm and no adjustments were made for multiple comparisons. NES, normalized enrichment score; FDR, false discovery rate; FWER, family-wise error rate. (g) Left: representative western blots of E-cadherin and Snail in 2D/2.5D. Right: quantification of E-cadherin (two-sided Student’s t test). (h) Left: representative immunofluorescent images of E-cadherin in SAS-pLKO/SAS-shSNAI1 in 2D/2.5D. Scale bars, 10 μm. Right: quantification of E-cadherin mislocalization. (i) Immunoprecipitation-western blots and (j) western blots showing lysine-acetylated Snail in cells cultured in 2D/2.5D. (k) Western blots of indicated proteins in cells treated with DMSO or FAK inhibitor 14 (1 μM) for 24 h. (l), (m) and (n) Immunoprecipitation-western blots/western blots of indicated proteins in SAS cells treated with indicated chemicals for 24 h (FAK inhibitor 14, 1 μM; C646, 10 μM; PD98059, 10 μM). (o) RT-qPCR of the indicated genes in TE1 cells. The P value is estimated by ANOVA. b, c, f, and in was from one experiment. g, h and o were analysed from three independent experiments (n = 3). Data represent mean ± s.d. shown in g, h and o. NS, not significant. Uncropped images of all blots are shown in Supplementary Figure 8. Source data are provided in Supplementary Table 11.

Supplementary Figure 2 Claudin-11 contributes to collective cell migration and invasion.

(a) A heatmap show the expression of genes related to cell adhesion in SAS-pLKO vs. SAS-shSNAI1 cells. The data was from one experiment. (b) RT-qPCR indicated the expression of cell adhesion molecules and (c) claudins family genes in SAS-pLKO/SAS-shSNAI1 (n = 2 independent experiments). (d) Western blots of claudin-3, -4 and -7 in SAS-pLKO/SAS-shSNAI1 cells. The data was from one experiment. (e) Upper: snapshots from phase-contrast videos of TE1-pLKO/TE1-shCLDN11 cells moving on collagen gels. Scale bars, 100 μm. Lower: directionality of migration (n = 10 cells). (f) Left: representative images of 3D invasion assays. Yellow arrow indicates collectively invasive cells. The y-axis indicates invasion distance and y-axis heights, 60 μm. Right: the relative invasion index (two-sided Student’s t test). (g) MTT assay. The effect of CLDN11 knockdown on viability in SAS cells. The p-value is estimated by ANOVA. (h) Left: representative images of Transwell migration assay of SAS-pLKO/SAS-shCLDN11 cells. Scale bars, 100 μm. Right: quantification of Transwell migration assay (two-sided Student’s t test). (i) Left: schema of the mice experiment. Middle: three representative bioluminescence images of the mice at the end of week 3. Right: quantification of IVIS images (n = 10 mice; two-sided Mann-Whitney U test). (j) Representative images of H&E staining in lymph node samples from mice orthotopic model. Cropped regions are indicated by the yellow rectangle in the overview panels. Red arrows indicate the regions with tumor infiltration. Scale bars, 100 μm. (k) Western blots of claudin-11 in FaDu-WT vs. FaDu-LN cells. WT, wild-type; LN, lymph node. The representative data were from two independent experiments with similar results. (a), (b), (c), (d) and (e) were performed in 2.5D. (f), (g) and (h) were analyzed from three independent experiments (n = 3). Images of (e), (f), (h), and (j) are representative of three independent experiments with similar results. Data represent mean values shown in (b), (c), (g) and (i). Data represent mean ± S.D. shown in (f), (g) and (h). N.D., no detection; N.S., no significance. Uncropped images of all blots are shown in Supplementary Figure 8. Source data is provided in Supplementary Table 11.

Supplementary Figure 3 The relationship between the expression of claudin family members and Snail.

(a) RT-qPCR analysis of CLDN3, CLDN4, CLDN7, and SNAI1 in primary HNSCC cells expressing control (pCDH), wild-type Snail (Snail-WT) or unacetylatable Snail (Snail-2R). Caco2 cells was used as a positive control for the expressions of CLDN3, CLDN4 and CLDN7 (n = 2 independent experiments). Data represent mean values. (b) Western blots for showing claudin-3, -4, -7, and Snail in primary HNSCC cells expressing control (pCDH), wild-type Snail (Snail-WT) or unacetylatable Snail (Snail-2R). Caco2 cells was used as a positive control for the expressions of claudin-3, -4, and -7. β-actin was a loading control. The data was from one experiment. Uncropped images of all blots are shown in Supplementary Figure 8. Source data is provided in Supplementary Table 11.

Supplementary Figure 4 Claudin-11 inactivates RhoA via Src-phosphorylated p190RhoGAP.

(a) A pull down assay indicated the level of RhoA-GTP in pLKO/shCLDN11 cells and (b) in SAS-WT vs. SAS-LN cells. (c), (d) and (e) Immunoprecipitation-western blots of indicated proteins in SAS cells. (f) Left: representative snapshots from phase-contrast videos of TE1-pLKO/TE1-shGRLF1 cells moving on collagen gels. Scale bars, 100 μm. Lower: directionality of migration (n = 10 cells). The data is representative of three independent experiments with similar results. (g) Western blots analysis of the levels of indicated proteins in SAS-pLKO/SAS-shSNAI1. (h) A pull down assay for detecting the RhoA-GTP, and western blots for examining the levels of indicated proteins in SAS cells (imatinib, 10 μM; SU6656, 10 μM). (i) Western blots for the indicated proteins in pLKO/shSNAI1 cells. (j) Western blots for the indicated proteins in pLKO/shCLDN11 primary HNSCC cells in 3D for 3 h. (k) Immunoprecipitation-western blots for determining the phosphorylation status of Y418 residue of Src family in SAS-pLKO/SAS-shCLDN11 cells. (l) Left: FRET images of the pTriEx-RhoA biosensor-expressed SAS-pLKO/SAS-shSNAI1 cells. Right: quantification of normalized FRET efficiency ratio signal (n = 3 independent experiments). Data represent mean ± S.D. (two-sided Student’s t test). N.S., no significance. (m) Representative immunofluorescent images for showing the localization MLC2-pT18/S19 in SAS-pLKO/SAS-shSNAI1. Yellow arrows indicate the co-localization of MLC2-pT18/S19 and F-actin. Scale bars, 10 μm. (n) A heatmap showing the expression of genes encoding GEFs and GAPs in SAS-pLKO/SAS-shSNAI1. (o) RT-qPCR for validation of the expression of GEFs and GAPs in SAS-pLKO/SAS-shSNAI1 (n = 2 independent experiment). Data represent mean values. (p) Western blots of ARHGAP42 in SAS-pLKO/SAS-shSNAI1. Red arrows indicate the positions of ARHGAP42. All cells were cultured in 2.5D with the exception of (j) in 3D. (a), (b), (h), (k), (m) and (n) was from one experiment. The representative data (c), (d), (e), (g), (i), (j) and (p) were from two independent experiments with similar results. Uncropped images of all blots are shown in Supplementary Figure 8. Source data is provided in Supplementary Table 11.

Supplementary Figure 5 Tyrosine-phosphorylated claudin-11 recruits and activates Src.

(a) Amino acid sequence of claudin-11 (accession code: O75508). The cytosolic tail is highlighted in red. (b) Western blots of Y418-phosphorylated Src and Y530-phosphorylated Src in SAS-pLKO/SAS-shCLDN11. The representative data were from three independent experiments with similar results. (c) GAP activity assay for analyzing the p190RhoGAP activity in HEK293T cells co-transfected with the p190RhoGAP expression vector (pHA-GRLF1) and a vector expression wild-type (WT) or different tyrosine residue-mutated CLDN11, (n = 3 independent experiments). Data represent mean ± S.D. (two-sided Student’s t test). N.S., no significance. (d) Pull down assay/western blots for analyzing the GTP-bound/total RhoA in SAS cells transfected with plasmids expressing wild-type or different tyrosine residue-mutated FLAG-tagged claudin-11. The data was from one experiment. (e) Confirmation of the efficacy of different inhibitors with the western blots of T202/Y204-phosphorylated ERK1/2, and total ERK1/2 in SAS cells treated with different tyrosine kinase inhibitors. The representative data were from two independent experiments with similar results. (f) Immunoprecipitation-western blots for showing the level of tyrosine-phosphorylated claudin-11 in SAS treated with different tyrosine kinase inhibitors following by seeding on the thick collagen for 30 min (cetuximab, 10 μg/ml for 30 min; all other tyrosine kinases inhibitors, 10 μM for 30 min). The representative data were from two independent experiments with similar results. (g) Left: representative snapshots from phase-contrast videos SAS cells treated with DMSO, FAK inhibitor 14 or cetuximab moving on collagen gels. Solid line indicates the position of cell groups at t = 0 h; shading indicates the position of the same cell groups at t = 7 h. Scale bars, 100 μm. Right: directionality of migration presented in rose plot diagrams for SAS cells treated within DMSO, FAK inhibitor 14 or cetuximab (n = 20 cells). The data is representative of two independent experiments with similar results. (h) Western blots of integrin β1, FAK, and Y397-phosphorylated FAK in SAS-pLKO vs. SAS-shSNAI1. The data was from one experiment. Uncropped images of all blots are shown in Supplementary Figure 8. Source data is provided in Supplementary Table 11.

Supplementary Figure 6 Correlation between the number of CTC clusters and the clinical course of HNSCC patients.

(a) Images of the CTC clusters from one representative HNSCC patient. 208 CTC clusters were captured on the biomimetic supported lipid bilayer (SLB) coated microfluidic chip conjugated with a mouse anti-human EpCAM antibody in 2 ml whole blood. The cells were stained for pan-cytokeratin (Pan-CK, red), CD45 (green), and nuclei (DAPI, blue). Scale bars, 10 μm. The same CTC capturing experiment was performed in samples from 28 HNSCC patients. (b), (c), (d) and (e) Representative images of CTC clusters and WBCs from HNSCC patients. (b), the cells were stained with the antibody against pan-cytokeratin (Pan-CK, red), CD45 (green), and Snail (cyan). The images are representative of two independent CTC clusters with similar results. (c), the cells were stained with the antibodies against pan-cytokeratin (Pan-CK, red), CD45 (green), and Src-pY418 (cyan). The images are representative of five independent CTC clusters with similar results. (d), the cells were stained with the antibody against pan-cytokeratin (Pan-CK, red), CD45 (green), and p190RhoGAP-pY1087 (cyan). The images are representative of five independent CTC clusters with similar results. (e), the cells were stained with the antibody against pan-cytokeratin (Pan-CK, red), CD45 (green), and p190RhoGAP-pY1105 (cyan). The images are representative of six independent CTC clusters with similar results. All nuclei were stained with DAPI (blue). Scale bars, 10 μm. (f), (g) and (h) Quantification of single CTC in HNSCC patients with different (f) T stages, (g) lymph node metastasis status, and (h) recurrence. The center values indicate the mean values. The p-value is shown in each panel (two-sided Student’s t test). P, primary; R, recurrence; M, metastasis. (i), (j) and (k) Correlation between the amount of CTC clusters and clinical courses in three different HNSCC patients [patient 2 (i), 3 (j), 4 (k)] receiving treatment. Quantification of CTC clusters at different time points of treatment is illustrated. The treatment course of each patient is indicated at the bottom of the panel. Chemo, chemotherapy; CCRT, concurrent chemoradiotherapy; PR, partial response. Source data is provided in Supplementary Table 11.

Supplementary Figure 7 Expression of Snail or claudin-11 correlates with an advanced HNSCC and a worse prognosis.

(a) Immunohistochemical expression score of claudin-11 and Snail in adjacent normal tissues vs. HNSCC specimens. Y-axis displays the expression calculated by a modified H-score of claudin-11 (left) or Snail (right). The center values indicate the mean values. The p-value of each panel is shown (two-sided Student’s t test). N, normal; T, tumor. (b) Correlation between Snail expression and CTC counts in HNSCC patients. Left: correlation between Snail expression and clustering CTC counts. Right: correlation between Snail expression and single CTC counts. Each dot represents a HNSCC specimen (n = 21 patients). The Pearson correlation coefficient (r) and p-value are shown. (c) The correlation between the relative expression of SNAI1 and CLDN11 in 43 HNSCC specimens. Each red dot represents a specimen. The Pearson correlation coefficient (r) and p-value are shown. (d) Quantification of the relative CLDN11 (left) and SNAI1 (right) mRNA levels in tumor vs. normal parts of HNSCC patients. The p-value of each panel is shown (two-sided Student’s t test). N, normal; T, tumor. (e) The Kaplan-Meier survival plots to show the prognostic effect of CLDN11 expression in colorectal cancer (GSE17536) and ovarian cancer (DUKE-OC). The data were obtained from the public database PrognoScan. The corresponding p-value is indicated in each panel (two-sided Log-rank test). Source data is provided in Supplementary Table 11.

Supplementary Figure 8

Uncropped films showing the full blots displayed in the figures.

Supplementary information

Supplementary Information

Supplementary Figures 1–8, and Supplementary Video and Supplementary Table legends.

Reporting Summary

Supplementary Table 1

Upregulated and downregulated genes in cDNA microarray analysis of SAS-pLKO versus SAS-shSNAI1 cells.

Supplementary Table 2

List of DNA copy number aberrations in pan-CK+/CD45- CTCs and pan-CK-/CD45+ WBCs from a HNSCC patient.

Supplementary Table 3

Genetic alterations of 6 HNCC related genes detected in the pan-CK+/CD45- CTC but not in the matched WBC for patient A (Supplementary Table 3.1) and patient B (Supplementary Table 3.2).

Supplementary Table 4

Demographics, CTC counts and IHC results of 28 HNSCC patients.

Supplementary Table 5

Demographics and IHC results of 54 HNSCC patients.

Supplementary Table 6

Predictors of progression free survival; Cox multivariable analysis.

Supplementary Table 7

Demographics and RT-qPCR results of 43 HNSCC patients.

Supplementary Table 8

Relationships between CLDN11 expression and prognosis of patients with colorectal cancer and ovarian cancer were investigated using the PrognoScan database.

Supplementary Table 9

Information of nucleotide sequences used in the study.

Supplementary Table 10

Information of antibodies used in this study.

Supplementary Table 11

Statistics source data.

Supplementary Video 1

Time-lapse video microscopy of SAS, TE1, TE9 and HSC3 in 2.5D cultured system.

Supplementary Video 2

Time-lapse video microscopy of SAS-control and SAS-shSNAI1 in 2.5D cultured system.

Supplementary Video 3

Time-lapse video microscopy of TE1-control and TE1-shSNAI1 in 2.5D cultured system.

Supplementary Video 4

Movie illustrates a 3D invasion assay of 100% SAS-pLKO-GFP (green), 100% SAS-shSNAI1-RFP (red), or 2% SAS-pLKO-GFP/98% SAS-shSNAI1-RFP cells.

Supplementary Video 5

Time-lapse video microscopy of SAS-control and SAS-shCLDN11 in 2.5D cultured system.

Supplementary Video 6

Time-lapse video microscopy of TE1-control and TE1-shCLDN11 in 2.5D cultured system.

Supplementary Video 7

Time-lapse video microscopy of SAS-control and SAS-shGRLF1 in 2.5D cultured system.

Supplementary Video 8

Time-lapse video microscopy of TE1-control and TE1-shGRLF1 in 2.5D cultured system.

Supplementary Video 9

Time-lapse video microscopy of SAS treated with DMSO, FAK inhibitor 14 or cetuximab in 2.5D cultured system.

Rights and permissions

Reprints and permissions

About this article

Check for updates. Verify currency and authenticity via CrossMark

Cite this article

Li, CF., Chen, JY., Ho, YH. et al. Snail-induced claudin-11 prompts collective migration for tumour progression. Nat Cell Biol 21, 251–262 (2019). https://doi.org/10.1038/s41556-018-0268-z

Download citation

  • Received:

  • Accepted:

  • Published:

  • Issue Date:

  • DOI: https://doi.org/10.1038/s41556-018-0268-z

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