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:

Ciliary proteins link basal body polarization to planar cell polarity regulation

Abstract

Planar cell polarity (PCP) refers to coordinated polarization of cells within the plane of a cell sheet. A conserved signaling pathway is required for the establishment of PCP in epithelial tissues and for polarized cellular rearrangements known as convergent extension. During PCP signaling, core PCP proteins are sorted asymmetrically along the polarization axis; this sorting is thought to direct coordinated downstream morphogenetic changes across the entire tissue. Here, we show that a gene encoding a ciliary protein (a 'ciliary gene'), Ift88, also known as Polaris, is required for establishing epithelial PCP and for convergent extension of the cochlear duct of Mus musculus. We also show that the proper positioning of ciliary basal bodies and the formation of polarized cellular structures are disrupted in mice with mutant ciliary proteins ('ciliary mutants'), whereas core PCP proteins are partitioned normally along the polarization axis. Thus, our data uncover a distinct requirement for ciliary genes in basal body positioning and morphological polarization during PCP regulation.

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: Ift88 is expressed in primary cilia in the organ of Corti.
Figure 2: Misorientation of stereociliary bundles in Ift88 mutants.
Figure 3: Ift88 is required for cochlear extension.
Figure 4: PCP complexes are partitioned normally in Ift88 mutants.
Figure 5: Ciliary basal body positions are affected in Ift88 mutants.
Figure 6: Alteration of the apical cytoskeleton in Ift88 mutants.
Figure 7: Model of the role of ciliary genes and cilia in centriole and morphological polarization during the establishment of PCP in the organ of Corti.

Similar content being viewed by others

Accession codes

Accessions

GenBank/EMBL/DDBJ

References

  1. Wallingford, J.B. et al. Dishevelled controls cell polarity during Xenopus gastrulation. Nature 405, 81–85 (2000).

    Article  CAS  Google Scholar 

  2. Wang, J. et al. Dishevelled genes mediate a conserved mammalian PCP pathway to regulate convergent extension during neurulation. Development 133, 1767–1778 (2006).

    Article  CAS  Google Scholar 

  3. Wang, Y., Guo, N. & Nathans, J. The role of Frizzled3 and Frizzled6 in neural tube closure and in the planar polarity of inner-ear sensory hair cells. J. Neurosci. 26, 2147–2156 (2006).

    Article  CAS  Google Scholar 

  4. Ybot-Gonzalez, P. et al. Convergent extension, planar-cell-polarity signalling and initiation of mouse neural tube closure. Development 134, 789–799 (2007).

    Article  CAS  Google Scholar 

  5. Montcouquiol, M. et al. Identification of Vangl2 and Scrb1 as planar polarity genes in mammals. Nature 423, 173–177 (2003).

    Article  CAS  Google Scholar 

  6. 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).

    Article  CAS  Google Scholar 

  7. Lu, X. et al. PTK7/CCK-4 is a novel regulator of planar cell polarity in vertebrates. Nature 430, 93–98 (2004).

    Article  CAS  Google Scholar 

  8. Wang, J. et al. Regulation of polarized extension and planar cell polarity in the cochlea by the vertebrate PCP pathway. Nat. Genet. 37, 980–985 (2005).

    Article  CAS  Google Scholar 

  9. Qian, D. et al. Wnt5a functions in planar cell polarity regulation in mice. Dev. Biol. 306, 121–133 (2007).

    Article  CAS  Google Scholar 

  10. Cotanche, D.A. & Corwin, J.T. Stereociliary bundles reorient during hair cell development and regeneration in the chick cochlea. Hear. Res. 52, 379–402 (1991).

    Article  CAS  Google Scholar 

  11. Tilney, L.G., Tilney, M.S. & DeRosier, D.J. Actin filaments, stereocilia, and hair cells: how cells count and measure. Annu. Rev. Cell Biol. 8, 257–274 (1992).

    Article  CAS  Google Scholar 

  12. Denman-Johnson, K. & Forge, A. Establishment of hair bundle polarity and orientation in the developing vestibular system of the mouse. J. Neurocytol. 28, 821–835 (1999).

    Article  CAS  Google Scholar 

  13. Kikuchi, T., Tonosaki, A. & Takasaka, T. Development of apical-surface structures of mouse otic placode. Acta Otolaryngol. (Stockh.) 106, 200–207 (1988).

    Article  CAS  Google Scholar 

  14. Sobkowicz, H.M., Slapnick, S.M. & August, B.K. The kinocilium of auditory hair cells and evidence for its morphogenetic role during the regeneration of stereocilia and cuticular plates. J. Neurocytol. 24, 633–653 (1995).

    Article  CAS  Google Scholar 

  15. Kikuchi, K. & Hilding, D. The development of the organ of Corti in the mouse. Acta Otolaryngol. (Stockh.) 60, 207–222 (1965).

    Article  CAS  Google Scholar 

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

    Article  CAS  Google Scholar 

  17. Murcia, N.S. et al. The Oak Ridge Polycystic Kidney (orpk) disease gene is required for left-right axis determination. Development 127, 2347–2355 (2000).

    CAS  Google Scholar 

  18. Pazour, G.J. et al. Chlamydomonas IFT88 and its mouse homologue, polycystic kidney disease gene tg737, are required for assembly of cilia and flagella. J. Cell Biol. 151, 709–718 (2000).

    Article  CAS  Google Scholar 

  19. Pazour, G.J. et al. The intraflagellar transport protein, IFT88, is essential for vertebrate photoreceptor assembly and maintenance. J. Cell Biol. 157, 103–113 (2002).

    Article  CAS  Google Scholar 

  20. Yoder, B.K. et al. Polaris, a protein disrupted in orpk mutant mice, is required for assembly of renal cilium. Am. J. Physiol. Renal Physiol. 282, F541–F552 (2002).

    Article  CAS  Google Scholar 

  21. Cano, D.A., Murcia, N.S., Pazour, G.J. & Hebrok, M. orpk mouse model of polycystic kidney disease reveals essential role of primary cilia in pancreatic tissue organization. Development 131, 3457–3467 (2004).

    Article  CAS  Google Scholar 

  22. Taulman, P.D., Haycraft, C.J., Balkovetz, D.F. & Yoder, B.K. Polaris, a protein involved in left-right axis patterning, localizes to basal bodies and cilia. Mol. Biol. Cell 12, 589–599 (2001).

    Article  CAS  Google Scholar 

  23. Robert, A. et al. The intraflagellar transport component IFT88/polaris is a centrosomal protein regulating G1-S transition in non-ciliated cells. J. Cell Sci. 120, 628–637 (2007).

    Article  CAS  Google Scholar 

  24. Haycraft, C.J. et al. Intraflagellar transport is essential for endochondral bone formation. Development 134, 307–316 (2007).

    Article  CAS  Google Scholar 

  25. Hebert, J.M. & McConnell, S.K. Targeting of cre to the Foxg1 (BF-1) locus mediates loxP recombination in the telencephalon and other developing head structures. Dev. Biol. 222, 296–306 (2000).

    Article  CAS  Google Scholar 

  26. Pauley, S., Lai, E. & Fritzsch, B. Foxg1 is required for morphogenesis and histogenesis of the mammalian inner ear. Dev. Dyn. 235, 2470–2482 (2006).

    Article  CAS  Google Scholar 

  27. Badea, T.C., Wang, Y. & Nathans, J. A noninvasive genetic/pharmacologic strategy for visualizing cell morphology and clonal relationships in the mouse. J. Neurosci. 23, 2314–2322 (2003).

    Article  CAS  Google Scholar 

  28. Montcouquiol, M., Crenshaw, E.B.III & Kelley, M.W. Noncanonical Wnt signaling and neural polarity. Annu. Rev. Neurosci. 29, 363–386 (2006).

    Article  CAS  Google Scholar 

  29. Seifert, J.R. & Mlodzik, M. Frizzled/PCP signalling: a conserved mechanism regulating cell polarity and directed motility. Nat. Rev. Genet. 8, 126–138 (2007).

    Article  CAS  Google Scholar 

  30. Jones, C. & Chen, P. Planar cell polarity signaling in vertebrates. Bioessays 29, 120–132 (2007).

    Article  CAS  Google Scholar 

  31. Montcouquiol, M. et al. Asymmetric localization of Vangl2 and Fz3 indicate novel mechanisms for planar cell polarity in mammals. J. Neurosci. 26, 5265–5275 (2006).

    Article  CAS  Google Scholar 

  32. Deans, M.R. et al. Asymmetric distribution of prickle-like 2 reveals an early underlying polarization of vestibular sensory epithelia in the inner ear. J. Neurosci. 27, 3139–3147 (2007).

    Article  CAS  Google Scholar 

  33. Scholey, J.M. Intraflagellar transport. Annu. Rev. Cell Dev. Biol. 19, 423–443 (2003).

    Article  CAS  Google Scholar 

  34. Sandoz, D. et al. Organization and functions of cytoskeleton in metazoan ciliated cells. Biol. Cell 63, 183–193 (1988).

    Article  CAS  Google Scholar 

  35. Beisson, J. & Jerka-Dziadosz, M. Polarities of the centriolar structure: morphogenetic consequences. Biol. Cell 91, 367–378 (1999).

    Article  CAS  Google Scholar 

  36. Hagiwara, H., Kano, A., Aoki, T., Ohwada, N. & Takata, K. Localization of γ-tubulin to the basal foot associated with the basal body extending a cilium. Histochem. J. 32, 669–671 (2000).

    Article  CAS  Google Scholar 

  37. Park, T.J., Haigo, S.L. & Wallingford, J.B. Ciliogenesis defects in embryos lacking inturned or fuzzy function are associated with failure of planar cell polarity and Hedgehog signaling. Nat. Genet. 38, 303–311 (2006).

    Article  CAS  Google Scholar 

  38. Kondo, S. et al. KIF3A is a new microtubule-based anterograde motor in the nerve axon. J. Cell Biol. 125, 1095–1107 (1994).

    Article  CAS  Google Scholar 

  39. Lin, F. et al. Kidney-specific inactivation of the KIF3A subunit of kinesin-II inhibits renal ciliogenesis and produces polycystic kidney disease. Proc. Natl. Acad. Sci. USA 100, 5286–5291 (2003).

    Article  CAS  Google Scholar 

  40. Beech, P.L. et al. Localization of kinesin superfamily proteins to the connecting cilium of fish photoreceptors. J. Cell Sci. 109, 889–897 (1996).

    CAS  PubMed  Google Scholar 

  41. Liu, A., Wang, B. & Niswander, L.A. Mouse intraflagellar transport proteins regulate both the activator and repressor functions of Gli transcription factors. Development 132, 3103–3111 (2005).

    Article  CAS  Google Scholar 

  42. Huangfu, D. & Anderson, K.V. Cilia and Hedgehog responsiveness in the mouse. Proc. Natl. Acad. Sci. USA 102, 11325–11330 (2005).

    Article  CAS  Google Scholar 

  43. Schneider, L. et al. PDGFRα signaling is regulated through the primary cilium in fibroblasts. Curr. Biol. 15, 1861–1866 (2005).

    Article  CAS  Google Scholar 

  44. Simons, M. et al. Inversin, the gene product mutated in nephronophthisis type II, functions as a molecular switch between Wnt signaling pathways. Nat. Genet. 37, 537–543 (2005).

    Article  CAS  Google Scholar 

  45. Heisenberg, C.P. et al. Silberblick/Wnt11 mediates convergent extension movements during zebrafish gastrulation. Nature 405, 76–81 (2000).

    Article  CAS  Google Scholar 

  46. Boisvieux-Ulrich, E., Laine, M.C. & Sandoz, D. The orientation of ciliary basal bodies in quail oviduct is related to the ciliary beating cycle commencement. Biol. Cell 55, 147–150 (1985).

    Article  CAS  Google Scholar 

  47. Mitchell, B., Jacobs, R., Li, J., Chien, S. & Kintner, C. A positive feedback mechanism governs the polarity and motion of motile cilia. Nature 447, 97–101 (2007).

    Article  CAS  Google Scholar 

  48. Absalon, S. et al. Basal body positioning is controlled by flagellum formation in Trypanosoma brucei. PLoS ONE 2, e437 (2007).

    Article  Google Scholar 

  49. 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).

    Article  CAS  Google Scholar 

Download references

Acknowledgements

We thank W. Sale for discussions about the project, B. Shur, A. Fritz and T. Caspary for critical comments on the manuscript and S. Mark for technical support. We also thank S. Guadagnini and M.-C. Prévost from the Electron Microscopy Group (PFME) at the Pasteur Institute for their technical advice. Antibody against Fzd3 was a gift from J. Nathans (Johns Hopkins). This project is supported by NIH research grants to P.C. (RO1 DC005213 and DC007423) and B.K.Y. (RO1 DK065655), a postdoctoral fellowship to C.J. (F32DC008731), a Woodruff Foundation grant to P.C., a postdoctoral fellowship to I.F. (INSERM) and grants from Fondation R. & G. Strittmatter and from Ernst-Jung Stiftung für Medizin to C.P.

Author information

Authors and Affiliations

Authors

Contributions

C.J. carried out most of the analysis and played a pivotal role in progressively designing the experiments. V.C.R. and B.B. prepared Ift88flox/floxCreER and Kif3aflox/floxCreER samples. I.F. and C.P. contributed electron microscopy data. The study was initiated by B.K.Y and P.C., and carried out under the direction of P.C. C.J. and P.C. wrote the manuscript, which was critically commented upon and revised by B.K.Y. and C.P. D.Q. generated Vangl2GFP animals.

Corresponding author

Correspondence to Ping Chen.

Supplementary information

Supplementary Text and Figures

Supplementary Figures 1–5 (PDF 730 kb)

Supplementary Video 1

Three-dimensional images of cellular microtubules in a wild-type cochlea. Images of the cellular microtubules from three outer hair cells in a wild-type cochlea were acquired using the Zeiss LSM510 confocal microscope, as described in the text (Fig. 6). Serial 0.5μm optical sections were acquired from the apical-most cell surface moving basally through the organ of corti and projected into three dimensions using NIH ImageJ software. This video complements Figure 6 and provides a more complete view of the apical microtubules. (MOV 43 kb)

Supplementary Video 2

Three-dimensional images of cellular microtubules in an IFT88CKO/CKO mutant cochlea. Images of the cellular microtubules from three outer hair cells in an IFT88CKO/CKO mutant cochlea were acquired using the Zeiss LSM510 confocal microscope, as described in the text (Fig. 6). Serial 0.5μm optical sections were acquired from the apical-most cell surface moving basally through the organ of corti and projected into three dimensions using NIH ImageJ software. This video complements Figure 6 and provides a more complete view of the apical microtubules. (MOV 89 kb)

Rights and permissions

Reprints and permissions

About this article

Cite this article

Jones, C., Roper, V., Foucher, I. et al. Ciliary proteins link basal body polarization to planar cell polarity regulation. Nat Genet 40, 69–77 (2008). https://doi.org/10.1038/ng.2007.54

Download citation

  • Received:

  • Accepted:

  • Published:

  • Issue Date:

  • DOI: https://doi.org/10.1038/ng.2007.54

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