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.

  • Resource
  • Published:

A screen for morphological complexity identifies regulators of switch-like transitions between discrete cell shapes

Abstract

The way in which cells adopt different morphologies is not fully understood. Cell shape could be a continuous variable or restricted to a set of discrete forms. We developed quantitative methods to describe cell shape and show that Drosophila haemocytes in culture are a heterogeneous mixture of five discrete morphologies. In an RNAi screen of genes affecting the morphological complexity of heterogeneous cell populations, we found that most genes regulate the transition between discrete shapes rather than generating new morphologies. In particular, we identified a subset of genes, including the tumour suppressor PTEN, that decrease the heterogeneity of the population, leading to populations enriched in rounded or elongated forms. We show that these genes have a highly conserved function as regulators of cell shape in both mouse and human metastatic melanoma cells.

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

Access options

Buy this article

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

Figure 1: Automated morphological profiling.
Figure 2: N, L, C, T and R cells.
Figure 3: Hierarchical clustering of QMSs. Average linkage clustering of 284 5-feature QMSs comprising L, C, T and R SVM Z-scores as well as PZ scores.
Figure 4: Morphological complexity is a phenotype that can be altered by RNAi.
Figure 5: Kc cells exist as discrete subpopulations.
Figure 6: Melanoma cells make switch-like transitions between discrete morphologies on Col-I.
Figure 7: Loss of PTEN alters the exploration of shape space.
Figure 8: A conserved set of genes promotes rounding in Drosophila, mouse and human cells.

Similar content being viewed by others

References

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

    Article  CAS  Google Scholar 

  2. Keren, K. et al. Mechanism of shape determination in motile cells. Nature 453, 475–480 (2008).

    Article  CAS  Google Scholar 

  3. Mogilner, A. & Keren, K. The shape of motile cells. Curr. Biol. 19, R762–R771 (2009).

    Article  CAS  Google Scholar 

  4. Guarino, M., Rubino, B. & Ballabio, G. The role of epithelial-mesenchymal transition in cancer pathology. Pathology 39, 305–318 (2007).

    Article  CAS  Google Scholar 

  5. Sanz-Moreno, V. et al. Rac activation and inactivation control plasticity of tumour cell movement. Cell 135, 510–523 (2008).

    Article  CAS  Google Scholar 

  6. Wolf, K. et al. Compensation mechanism in tumour cell migration: mesenchymal-amoeboid transition after blocking of pericellular proteolysis. J. Cell Biol. 160, 267–277 (2003).

    Article  CAS  Google Scholar 

  7. Bakal, C., Aach, J., Church, G. & Perrimon, N. Quantitative morphological signatures define local signalling networks regulating cell morphology. Science 316, 1753–1756 (2007).

    Article  CAS  Google Scholar 

  8. Yin, Z. et al. Using iterative cluster merging with improved gap statistics to perform online phenotype discovery in the context of high-throughput RNAi screens. BMC Bioinformatics 9, 264 (2008).

    Article  Google Scholar 

  9. Waddington, C. H. The Strategy of Genes (Allen Unwin, 1957).

    Google Scholar 

  10. Gadea, G., Sanz-Moreno, V., Self, A., Godi, A. & Marshall, C. J. DOCK10-mediated Cdc42 activation is necessary for amoeboid invasion of melanoma cells. Curr. Biol. 18, 1456–1465 (2008).

    Article  CAS  Google Scholar 

  11. Sahai, E. & Marshall, C. J. Differing modes of tumour cell invasion have distinct requirements for Rho/ROCK signalling and extracellular proteolysis. Nat. Cell Biol. 5, 711–719 (2003).

    Article  CAS  Google Scholar 

  12. Sanz-Moreno, V. et al. ROCK and JAK1 signalling cooperate to controlactomyosin contractility in tumour cells and stroma. Cancer Cell 20, 229–245 (2011).

    Article  CAS  Google Scholar 

  13. Tan, C., Stronach, B. & Perrimon, N. Roles of myosin phosphatase during Drosophila development. Development 130, 671–681 (2003).

    Article  CAS  Google Scholar 

  14. Viros, A. et al. Improving melanoma classification by integrating genetic and morphologic features. PLoS Med. 5, e120 (2008).

    Article  Google Scholar 

  15. Sanz-Moreno, V. & Marshall, C. J. The plasticity of cytoskeletal dynamics underlying neoplastic cell migration. Curr. Opin. Cell Biol. 22, 690–696 (2010).

    Article  CAS  Google Scholar 

  16. Ridley, A. J. et al. Cell migration: integrating signals from front to back. Science 302, 1704–1709 (2003).

    Article  CAS  Google Scholar 

  17. Waddington, C. H. Canalization of development and genetic assimilation of acquired characters. Nature 183, 1654–1655 (1959).

    Article  CAS  Google Scholar 

  18. Kauffman, S. A. The Origins of Order. Self-Organization and Selection in Evolution (Oxford Univ. Press, 1993).

    Google Scholar 

  19. Dhomen, N. et al. Oncogenic Braf induces melanocyte senescence and melanoma in mice. Cancer Cell 15, 294–303 (2009).

    Article  CAS  Google Scholar 

  20. Dankort, D. et al. Braf(V600E) cooperates with Pten loss to induce metastatic melanoma. Nat. Genet. 41, 544–552 (2009).

    Article  CAS  Google Scholar 

  21. Huang da, W., Sherman, B. T. & Lempicki, R. A. Systematic and integrative analysis of large gene lists using DAVID bioinformatics resources. Nat. Protoc. 4, 44–57 (2009).

    Article  Google Scholar 

Download references

Acknowledgements

We are indebted to the Drosophila RNAi Screening Center staff at Harvard Medical School for their invaluable assistance. We especially thank I. Flockhart for assistance with data management. We thank J. Wang, X. Zhou and P. Bradley for their initial involvement in this study. We are grateful to N. Dhomen and R. Marais for melanoma cell lines. Work was financially supported in part by NCI grants (Grants R01CA121225 and U54CA149196) to S.T.C.W. and CRUK grants to C.B. (Grant 13478) and C.J.M. (Grant C107/A10433). N.P. is an Investigator of the Howard Hughes Medical Institute. A.S. is a Marie Curie Intra-European Fellow. C.J.M. is a Gibb Life Fellow of CRUK. C.B. is a Research Career Development Fellow of the Wellcome Trust.

Author information

Authors and Affiliations

Authors

Contributions

Z.Y. performed the bulk of statistical analysis of RNAi screening data and wrote the Supplementary Note. A.S. designed and performed all RNAi and cell line characterization experiments in mouse and human melanoma cells and contributed to writing of the manuscript. H.S. performed the analysis of live-cell melanoma cell imaging experiments and contributed to visualization of statistical results. A.M. performed all mouse work. X.X. and F.L. performed processing of images generated in Drosophila RNAi screen. M.A.G. and L.E. performed experiments describing penetrance of effects of different dsRNAs. A.R.B. contributed to writing and editing of the manuscript and the Supplementary Note. N.P. participated in the initial design of the study. S.T.C.W. coordinated image processing and statistical analysis. C.J.M. participated in design of melanoma experiments and contributed to writing the manuscript. C.B. participated in the design of experiments and statistical analysis, performed the Drosophila RNAi screen, performed the live-cell imaging assays, coordinated experimental and computational analysis, and wrote the manuscript.

Corresponding authors

Correspondence to Stephen T. C. Wong or Chris Bakal.

Ethics declarations

Competing interests

The authors declare no competing financial interests.

Supplementary information

Supplementary Information

Supplementary Information (PDF 1609 kb)

Supplementary Table 1

Supplementary Information (XLSX 10 kb)

Supplementary Table 2

Supplementary Information (XLSX 8 kb)

Supplementary Table 3

Supplementary Information (XLSX 107 kb)

Supplementary Table 4

Supplementary Information (XLSX 8 kb)

Supplementary Table 5

Supplementary Information (XLSX 146 kb)

Supplementary Table 6

Supplementary Information (XLSX 10 kb)

Supplementary Table 7

Supplementary Information (XLSX 31 kb)

Supplementary Table 8

Supplementary Information (XLSX 15 kb)

Rights and permissions

Reprints and permissions

About this article

Cite this article

Yin, Z., Sadok, A., Sailem, H. et al. A screen for morphological complexity identifies regulators of switch-like transitions between discrete cell shapes. Nat Cell Biol 15, 860–871 (2013). https://doi.org/10.1038/ncb2764

Download citation

  • Received:

  • Accepted:

  • Published:

  • Issue Date:

  • DOI: https://doi.org/10.1038/ncb2764

This article is cited by

Search

Quick links

Nature Briefing

Sign up for the Nature Briefing newsletter — what matters in science, free to your inbox daily.

Get the most important science stories of the day, free in your inbox. Sign up for Nature Briefing