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:

Wdr1-mediated cell shape dynamics and cortical tension are essential for epidermal planar cell polarity

Abstract

During mouse development, core planar cell polarity (PCP) proteins become polarized in the epidermal plane to guide angling/morphogenesis of hair follicles. How PCP is established is poorly understood. Here, we identify a key role for Wdr1 (also known as Aip1), an F-actin-binding protein that enhances cofilin/destrin-mediated F-actin disassembly. We show that cofilin and destrin function redundantly in developing epidermis, but their combined depletion perturbs cell adhesion, cytokinesis, apicobasal polarity and PCP. Although Wdr1 depletion accentuates single-loss-of-cofilin/destrin phenotypes, alone it resembles core PCP mutations. Seeking a mechanism, we find that Wdr1 and cofilin/destrin-mediated actomyosin remodelling are essential for generating or maintaining cortical tension within the developing epidermal sheet and driving the cell shape and planar orientation changes that accompany establishment of PCP in mammalian epidermis. Our findings suggest intriguing evolutionary parallels but mechanistic modifications to the distal wing hinge-mediated mechanical forces that drive cell shape change and orient PCP in the Drosophila wing disc.

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: Depletion of Wdr1 alters the cytoskeleton and yields a PCP phenotype.
Figure 2: Wdr1-depleted epidermis fails to establish PCP.
Figure 3: Molecular genetic analyses illuminate roles for the Wdr1-cofilin/destrin pathway in apicobasal and planar cell polarity.
Figure 4: Rescue of the planar polarity defects of Wdr1 mutants by expression of constitutively active cofilin.
Figure 5: Wdr1 is required for efficient F-actin severing in keratinocytes.
Figure 6: Wdr1 does not affect trafficking of PCP proteins, epidermal differentiation, or proliferation.
Figure 7: Cell shape dynamics are regulated by Wdr1 and coincide with PCP establishment.
Figure 8: Wdr1 activity is essential for the cell to generate/maintain cortical tension.

Similar content being viewed by others

References

  1. Eaton, S. & Jülicher, F. Cell flow and tissue polarity patterns. Curr. Opin. Genet. Dev. 21, 747–752 (2011).

    CAS  PubMed  Google Scholar 

  2. Goodrich, L. V. & Strutt, D. Principles of planar polarity in animal development. Development 138, 1877–1892 (2011).

    CAS  PubMed Central  PubMed  Google Scholar 

  3. McNeill, H. Planar cell polarity: keeping hairs straight is not so simple. Cold Spring Harb. Perspect. Biol. 2, a003376 (2010).

    PubMed Central  PubMed  Google Scholar 

  4. Devenport, D. & Fuchs, E. Planar polarization in embryonic epidermis orchestrates global asymmetric morphogenesis of hair follicles. Nat. Cell Biol. 10, 1257–1268 (2008).

    CAS  PubMed Central  PubMed  Google Scholar 

  5. Guo, N., Hawkins, C. & Nathans, J. Frizzled6 controls hair patterning in mice. Proc. Natl Acad. Sci. USA 101, 9277–9281 (2004).

    CAS  PubMed  Google Scholar 

  6. Aigouy, B. et al. Cell flow reorients the axis of planar polarity in the wing epithelium of Drosophila. Cell 142, 773–786 (2010).

    CAS  Google Scholar 

  7. Matis, M. & Axelrod, J. D. Regulation of PCP by the Fat signaling pathway. Genes Dev. 27, 2207–2220 (2013).

    CAS  PubMed Central  PubMed  Google Scholar 

  8. Luxenburg, C., Pasolli, H. A., Williams, S. E. & Fuchs, E. Developmental roles for Srf, cortical cytoskeleton and cell shape in epidermal spindle orientation. Nat. Cell Biol. 13, 203–214 (2011).

    CAS  PubMed Central  PubMed  Google Scholar 

  9. Fujibuchi, T. et al. AIP1/WDR1 supports mitotic cell rounding. Biochem. Biophys. Res. Commun. 327, 268–275 (2005).

    CAS  PubMed  Google Scholar 

  10. Chen, Q., Courtemanche, N. & Pollard, T. D. Aip1 promotes actin filament severing by cofilin and regulates the constriction of the cytokinetic contractile ring. J. Biol. Chem. 290, 2289–2300 (2014).

    PubMed Central  PubMed  Google Scholar 

  11. Aizawa, H. et al. Hyperosmotic stress-induced reorganization of actin bundles in Dictyostelium cells over-expressing cofilin. Genes Cells 4, 311–324 (1999).

    CAS  PubMed  Google Scholar 

  12. Augustine, R. C., Pattavina, K. A., Tuzel, E., Vidali, L. & Bezanilla, M. Actin interacting protein1 and actin depolymerizing factor drive rapid actin dynamics in Physcomitrella patens. Plant Cell 23, 3696–3710 (2011).

    CAS  PubMed Central  PubMed  Google Scholar 

  13. Chu, D. et al. AIP1 acts with cofilin to control actin dynamics during epithelial morphogenesis. Development 139, 3561–3571 (2012).

    CAS  PubMed  Google Scholar 

  14. Lin, M-C., Galletta, B. J., Sept, D. & Cooper, J. A. Overlapping and distinct functions for cofilin, coronin and Aip1 in actin dynamics in vivo. J. Cell Sci. 123, 1329–1342 (2010).

    CAS  PubMed Central  PubMed  Google Scholar 

  15. Mohri, K. & Ono, S. Actin filament disassembling activity of Caenorhabditis elegans actin-interacting protein 1 (UNC-78) is dependent on filament binding by a specific ADF/cofilin isoform. J. Cell Sci. 116, 4107–4118 (2003).

    CAS  PubMed  Google Scholar 

  16. Okada, K., Obinata, T. & Abe, H. XAIP1: a Xenopus homologue of yeast actin interacting protein 1 (AIP1), which induces disassembly of actin filaments cooperatively with ADF/cofilin family proteins. J. Cell Sci. 112 (Pt 10), 1553–1565 (1999).

    Google Scholar 

  17. Rodal, A. A., Tetreault, J. W., Lappalainen, P., Drubin, D. G. & Amberg, D. C. Aip1p interacts with cofilin to disassemble actin filaments. J. Cell Biol. 145, 1251–1264 (1999).

    CAS  PubMed Central  PubMed  Google Scholar 

  18. Kile, B. T. et al. Mutations in the cofilin partner Aip1/Wdr1 cause autoinflammatory disease and macrothrombocytopenia. Blood 110, 2371–2380 (2007).

    CAS  PubMed Central  PubMed  Google Scholar 

  19. Beronja, S., Livshits, G., Williams, S. & Fuchs, E. Rapid functional dissection of genetic networks via tissue-specific transduction and RNAi in mouse embryos. Nat. Med. 16, 821–827 (2010).

    CAS  PubMed Central  PubMed  Google Scholar 

  20. Curtin, J. A. et al. Mutation of celsr1 disrupts planar polarity of inner ear hair cells and causes severe neural tube defects in the mouse. Curr. Biol. 13, 1129–1133 (2003).

    CAS  PubMed  Google Scholar 

  21. Kibar, Z. et al. Ltap, a mammalian homolog of Drosophila Strabismus/Van Gogh, is altered in the mouse neural tube mutant Loop-tail. Nat. Genet. 28, 251–255 (2001).

    CAS  PubMed  Google Scholar 

  22. Chen, X. & Macara, I. G. Par-3 mediates the inhibition of LIM kinase 2 to regulate cofilin phosphorylation and tight junction assembly. J. Cell Biol. 172, 671–678 (2006).

    CAS  PubMed Central  PubMed  Google Scholar 

  23. Michelot, A. et al. Actin filament elongation in Arp2/3-derived networks is controlled by three distinct mechanisms. Dev. Cell 24, 182–195 (2013).

    CAS  PubMed Central  PubMed  Google Scholar 

  24. Nadkarni, A. V. & Brieher, W. M. Aip1 destabilizes cofilin-saturated actin filaments by severing and accelerating monomer dissociation from ends. Curr. Biol. 24, 2749–2757 (2014).

    CAS  PubMed Central  PubMed  Google Scholar 

  25. Ikeda, S. et al. Aberrant actin cytoskeleton leads to accelerated proliferation of corneal epithelial cells in mice deficient for destrin (actin depolymerizing factor). Hum. Mol. Genet. 12, 1029–1037 (2003).

    CAS  PubMed  Google Scholar 

  26. Smith, R. S. et al. Corn1: a mouse model for corneal surface disease and neovascularization. Invest. Ophthalmol. Vis. Sci. 37, 397–404 (1996).

    CAS  PubMed  Google Scholar 

  27. Agnew, B. J., Minamide, L. S. & Bamburg, J. R. Reactivation of phosphorylated actin depolymerizing factor and identification of the regulatory site. J. Biol. Chem. 270, 17582–17587 (1995).

    CAS  Google Scholar 

  28. Nagaoka, R., Abe, H. & Obinata, T. Site-directed mutagenesis of the phosphorylation site of cofilin: its role in cofilin-actin interaction and cytoplasmic localization. Cell. Motil. Cytoskeleton 35, 200–209 (1996).

    CAS  PubMed  Google Scholar 

  29. Kiuchi, T., Ohashi, K., Kurita, S. & Mizuno, K. Cofilin promotes stimulus-induced lamellipodium formation by generating an abundant supply of actin monomers. J. Cell Biol. 177, 465–476 (2007).

    CAS  PubMed Central  PubMed  Google Scholar 

  30. Mahaffey, J. P., Grego-Bessa, J., Liem, K. F. & Anderson, K. V. Cofilin and Vangl2 cooperate in the initiation of planar cell polarity in the mouse embryo. Development 140, 1262–1271 (2013).

    CAS  PubMed Central  PubMed  Google Scholar 

  31. Devenport, D., Oristian, D., Heller, E. & Fuchs, E. Mitotic internalization of planar cell polarity proteins preserves tissue polarity. Nat. Cell Biol. 13, 893–902 (2011).

    CAS  PubMed Central  PubMed  Google Scholar 

  32. Ren, N., Charlton, J. & Adler, P. N. The flare gene, which encodes the AIP1 protein of Drosophila, functions to regulate F-actin disassembly in pupal epidermal cells. Genetics 176, 2223–2234 (2007).

    CAS  PubMed Central  PubMed  Google Scholar 

  33. Shimada, Y., Yonemura, S., Ohkura, H., Strutt, D. & Uemura, T. Polarized transport of Frizzled along the planar microtubule arrays in Drosophila wing epithelium. Dev. Cell 10, 209–222 (2006).

    CAS  PubMed  Google Scholar 

  34. Ross, A. J. et al. Disruption of Bardet-Biedl syndrome ciliary proteins perturbs planar cell polarity in vertebrates. Nat. Genet. 37, 1135–1140 (2005).

    CAS  PubMed  Google Scholar 

  35. Reymann, A. C. et al. Actin network architecture can determine myosin motor activity. Science 336, 1310–1314 (2012).

    CAS  PubMed Central  PubMed  Google Scholar 

  36. Ono, K. & Ono, S. Two actin-interacting protein 1 isoforms function redundantly in the somatic gonad and are essential for reproduction in Caenorhabditis elegans. Cytoskeleton (Hoboken) 71, 36–45 (2014).

    CAS  Google Scholar 

  37. Yuan, B. et al. A cardiomyocyte-specific Wdr1 knockout demonstrates essential functional roles for actin disassembly during myocardial growth and maintenance in mice. Am. J. Pathol. 184, 1967–1980 (2014).

    CAS  PubMed  Google Scholar 

  38. Rhee, J. M. et al. In vivo imaging and differential localization of lipid-modified GFP-variant fusions in embryonic stem cells and mice. Genesis 44, 202–218 (2006).

    CAS  PubMed Central  PubMed  Google Scholar 

  39. Hutson, M. S. et al. Forces for morphogenesis investigated with laser microsurgery and quantitative modeling. Science 300, 145–149 (2003).

    CAS  PubMed  Google Scholar 

  40. Farhadifar, R., Röper, J-C., Aigouy, B., Eaton, S. & Jülicher, F. The influence of cell mechanics, cell–cell interactions, and proliferation on epithelial packing. Curr. Biol. 17, 2095–2104 (2007).

    CAS  PubMed  Google Scholar 

  41. Fernandez-Gonzalez, R., Simoes, S. D. E. M., Röper, J. C., Eaton, S. & Zallen, J. A. Myosin II dynamics are regulated by tension in intercalating cells. Dev. Cell 17, 736–743 (2009).

    CAS  PubMed Central  PubMed  Google Scholar 

  42. Straight, A. F. et al. Dissecting temporal and spatial control of cytokinesis with a myosin II inhibitor. Science 299, 1743–1747 (2003).

    CAS  Google Scholar 

  43. Ishizaki, T. et al. Pharmacological properties of Y-27632, a specific inhibitor of rho-associated kinases. Mol. Pharmacol. 57, 976–983 (2000).

    CAS  PubMed  Google Scholar 

  44. Bubb, M. R., Senderowicz, A. M., Sausville, E. A., Duncan, K. L. & Korn, E. D. Jasplakinolide, a cytotoxic natural product, induces actin polymerization and competitively inhibits the binding of phalloidin to F-actin. J. Biol. Chem. 269, 14869–14871 (1994).

    CAS  PubMed  Google Scholar 

  45. Heisenberg, C. P. & Bellaïche, Y. Forces in tissue morphogenesis and patterning. Cell 153, 948–962 (2013).

    CAS  PubMed  Google Scholar 

  46. Guillot, C. & Lecuit, T. Mechanics of epithelial tissue homeostasis and morphogenesis. Science 340, 1185–1189 (2013).

    CAS  PubMed  Google Scholar 

  47. Arber, S. et al. Regulation of actin dynamics through phosphorylation of cofilin by LIM-kinase. Nature 393, 805–809 (1998).

    CAS  PubMed  Google Scholar 

  48. Yang, N. et al. Cofilin phosphorylation by LIM-kinase 1 and its role in Rac-mediated actin reorganization. Nature 393, 809–812 (1998).

    CAS  Google Scholar 

  49. Moriyama, K. & Yahara, I. The actin-severing activity of cofilin is exerted by the interplay of three distinct sites on cofilin and essential for cell viability. Biochem. J. 365, 147–155 (2002).

    CAS  PubMed Central  PubMed  Google Scholar 

  50. Wiggan, O., Shaw, A. E., DeLuca, J. G. & Bamburg, J. R. ADF/cofilin regulates actomyosin assembly through competitive inhibition of myosin II binding to F-actin. Dev. Cell 22, 530–543 (2012).

    CAS  PubMed Central  PubMed  Google Scholar 

Download references

Acknowledgements

We thank D. Devenport, S. Williams, S. Beronja, A. R. Folgueras, D. Schramek, I. Matos and E. Ezratty for intellectual input; D. Oristian and A. Aldeguer as mouse specialists; Comparative Bioscience Center (AAALAC accredited) for care of mice in accordance with National Institutes of Health (NIH) guidelines; Bioimaging Center (A. North, director) for advice; Flow Cytometry facility (S. Mazel, director) for FACS sorting. Cfl–GFP was a generous gift from J. Condeelis (Albert Einstein college of Medicine, New York, USA); E.F. is an Investigator of the Howard Hughes Medical Institute. This research was supported by a grant from the NIH (R37-AR27883, E.F.), a Starr Stem Cell Postdoctoral Fellowship (C.L.) and a Genetics Training Grant by the NIH (E.H.).

Author information

Authors and Affiliations

Authors

Contributions

C.L. and E.F. conceived the study. C.L., E.H. and E.F. designed the experiments. C.L. and E.H. carried out the experiments and analysed the data. H.A.P. performed the ultrastructural analyses (Supplementary Fig. 1S). S.C. made the Wdr1-rescue construct, C.L. and N.S. performed the in utero injections. C.L. and M.N. prepared high-titre viruses. C.L., E.H. and E.F. wrote the paper. All authors provided intellectual input, vetted and approved the final manuscript.

Corresponding author

Correspondence to Elaine Fuchs.

Ethics declarations

Competing interests

The authors declare no competing financial interests.

Integrated supplementary information

Supplementary Figure 2 Wdr1 knockdown epidermis display normal intercellular adhesion and attachment to the basement membrane.

Correlative light and electron microscopy of E16.5 headskin in WT and Wdr1-368. Whole mount skin was immunolabeled for GFP (and subsequently embedded for electron microscopy. (a) and (b) Semithin sections (1 μm) were stained with toluidine blue and examined for the presence of GFP positive cells (brown nuclei). Dotted line indicated dermo-epidermal boundary. (b and b′) consecutive ultrathin sections were examined at the electron microscope. For Wdr1-368, the same exact cells that were GFP+ by immunohistochemistry were identified (pseudocolored in green) and photographed at high resolution. Red box enlarged in (c) and yellow box enlarged in (e). (c) Epidermal cells in Wdr1-368 display normal adhesion, with intact desmosomes (de) shown in boxed area enlarged in (d) No gaps were observed between the cells. Some inevitable cytoplasmic damage (asterisks) is due to detergent permeabilization necessary for immunolabelling. (e) Electron microscopic analysis showed intact basal lamina (bl) and hemidesmosomes (hd). Bars in (a) and (c), 2 μm, also valid for a′, b and b′. Bars in (d) and (e), 500 nm.

Supplementary Figure 3 Normal adhesion and apicobasal polarity in Wdr1 knockdown epidermis.

10 μm sagittal sections of E16.5 backskins were labelled for: (a) E-cadherin (E-cad) or ZO1 and co-labelled for integrin β4 (β4) and Laminin 5 (Lam5) (b) ABP markers Par3 and pericentrin. DAPI was used to label chromatin. Dotted line denotes the dermal-epidermal border. Inserts denote infected cells (H2B-GFP+ nuclei). Scale bar, 10 μm.

Supplementary Figure 4 Wdr1 knockdown by a second hairpin (Wdr1-1622) results in PCP defects analogous to Wdr1-368 and validation of Vangl2 shRNAs.

(a) 10 μm sagittal sections of E15.5 backskins were labelled for Par3 or Pericentrin (PC) or co-labelled for Keratin 5 (K5) and Keratin 10 (K10). (b) Whole-mount immunofluorescence of E15.5 backskins co-labelled for E-cadherin (E-cad) and Celsr1 and imaged in the mid-plane of the basal layer. (c) Whole-mount immunofluorescence of E18.5 HFs labelled for E-cadherin. (d) Quantifications of data shown in (c), n = 36 hair follicles from 3 embryos. Note similarities to data compiled on similarly aged Wdr1-368 knockdown embryonic epidermis (see Figs 1 and 2). Green arrows denote normal HF orientation, red arrows denote abnormal HF orientation. Red circles denote perpendicularly oriented HFs. Insets denote transduced cells (H2B-RFP+ nuclei). (e) In vitro Knockdown efficiencies of shRNAs against Vangl2 to achieve specific loss of PCP in the skin, n = 3 independent measurements/hairpin. Error bars indicate mean ± s.e.m. Scale bars, 10 μm.

Supplementary Figure 5 Generation and expression of a hairpin-resistant Wdr1-GFP lentiviral transgene.

(a) Wdr1 coding sequence recognized by Wdr1-368 hairpin and a mutant, shown in red, that renders it refactory to the hairpin. (b,c) Western blots of protein extracts from WT cells or cells transduced with lentiviruses that contain either Wdr1-368 shRNA, Scrambled control shRNA or the Wdr1-GFP hairpin-resistant mutant cDNA expression vector. Blots were probed with WDR1 antibodies (Ab) (b) or GFP Ab (c). Note the 93 kDa WDR1-GFP protein (endogenous WDR1 = 66 kDa, GFP = 27 kDa) detected with both Abs. Glyceraldehyde dehydrogenase (GAPDH) and hypoxanthine-guanine phosphoribosyltransferase (HPRT) Abs were used as loading controls.

Supplementary Figure 6 Normal localization of Cofilin and Destrin in Wdr1-368 KD epidermis.

(a) 10 μm sagittal sections of E15.5 control and WDR1-368 KD backskins were labelled for Cofilin (Cfl) or Destrin (Dstn). Inserts denote transduced cells (H2B-GFP+ nuclei). DAPI in blue. (c) Western blot analyses of protein extracts from control (Scr) and Wdr1-368 KD 10MK were probed with antibodies against: WDR1, Cofilin (Cfl) phospho-serine 3-cofilin (p-Cfl) and α-tubulin (αTub, loading control). (d) qPCR of mRNAs were from control and Cfl1KD/Dstn-null (KD/KO) 10MK. n = 3 samples/condition Cfl1 levels in Ctrl versus KD/KO p = 0.0026; Dstn levels in Ctrl. versus KD/KO p = 0.009. Dotted line denotes the dermal-epidermal border. Insets show transduced cells (H2B-GFP+ nuclei). Scale bars, 10 μm.

Supplementary Figure 7 Quantification of F-actin content, actin severing, and endogeneous levels of cofilin in Wdr1-KD and cofilin-overexpressing keratinocytes.

(a) Uncropped Western blots used to evaluate Wdr1 knockdown and endogeneous levels of actin in Fig. 1a. (b) Uncropped Western blots used to evalulate endogeneous cofilin and phosph-cofilin levels in transduced keratinocytes in Fig. 5a, b. Immunoblot for GFP indicates overexpression of histone 2B (H2B)-GFP (51 kDa), used as a marker of lentiviral transduction in scramble- and Wdr1-KD keratinocytes, and overexpression of GFP-CflS3A (52 kDa) in the others. Relative levels were quantified by fluorescence intensity on a LiCor Odyssey CLx imager and normalized to GAPDH levels. (c) Uncropped Western blots used to quantify F/G actin ratios. G-actin standards were loaded at the indicated quantities and used to construct a calibration curve. Actin levels were normalized to the cytoplasmic HPRT signal in each lysate.

Supplementary Figure 8 Defects in mitotic rounding in Wdr1-368 KD epidermis.

(a) Whole-mount immunofluorescence of E15.5 backskin labelled for Ecadherin (E-cad) and imaged at the basal layer plane. (b) Quantifications of data from (a). Control: 1.14 ± 0.088; Wdr1 KD: 1.39 ± 0.27, p = 5.27 × 10−5 (unpaired t-test), n = 23 cells (WT); n = 29 cells (Wdr1 KD) from 3 embryos per condition. Box and whiskers plot: line = median, box = 50% range, whiskers = 100% range. Arrow denotes early mitotic cells. Inserts denote infected cells (H2B-GFP+ nuclei). Scale bar, 10 μm.

Supplementary Table 1 Phenotypic analysis of the Wdr1/Destn/Cfl1 pathway.

Supplementary information

Supplementary Information

Supplementary Information (PDF 921 kb)

Rights and permissions

Reprints and permissions

About this article

Check for updates. Verify currency and authenticity via CrossMark

Cite this article

Luxenburg, C., Heller, E., Pasolli, H. et al. Wdr1-mediated cell shape dynamics and cortical tension are essential for epidermal planar cell polarity. Nat Cell Biol 17, 592–604 (2015). https://doi.org/10.1038/ncb3146

Download citation

  • Received:

  • Accepted:

  • Published:

  • Issue Date:

  • DOI: https://doi.org/10.1038/ncb3146

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