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:

The ciliopathy-associated CPLANE proteins direct basal body recruitment of intraflagellar transport machinery

A Corrigendum to this article was published on 27 July 2016

This article has been updated

Abstract

Cilia use microtubule-based intraflagellar transport (IFT) to organize intercellular signaling. Ciliopathies are a spectrum of human diseases resulting from defects in cilia structure or function. The mechanisms regulating the assembly of ciliary multiprotein complexes and the transport of these complexes to the base of cilia remain largely unknown. Combining proteomics, in vivo imaging and genetic analysis of proteins linked to planar cell polarity (Inturned, Fuzzy and Wdpcp), we identified and characterized a new genetic module, which we term CPLANE (ciliogenesis and planar polarity effector), and an extensive associated protein network. CPLANE proteins physically and functionally interact with the poorly understood ciliopathy-associated protein Jbts17 at basal bodies, where they act to recruit a specific subset of IFT-A proteins. In the absence of CPLANE, defective IFT-A particles enter the axoneme and IFT-B trafficking is severely perturbed. Accordingly, mutation of CPLANE genes elicits specific ciliopathy phenotypes in mouse models and is associated with ciliopathies in human patients.

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

Figure 1: The CPLANE interactome.
Figure 2: Jbts17 localizes to the base of cilia and is required for ciliogenesis and cilia-mediated patterning.
Figure 3: Jbts17 is necessary for recruitment of peripheral IFT-A proteins to basal bodies.
Figure 4: Jbts17 is required for bidirectional axonemal transport of IFT-B particles but not the IFT-A core.
Figure 5: CPLANE-mutant mice display diagnostic features of human OFD6.
Figure 6: CPLANE gene mutations in human ciliopathies.
Figure 7: Models for CPLANE function and structure.

Similar content being viewed by others

Change history

  • 30 May 2016

    In the version of this article initially published, the name of author Daniela A. Braun was misspelled. The error has been corrected in the HTML and PDF versions of the article.

References

  1. Hildebrandt, F., Benzing, T. & Katsanis, N. Ciliopathies. N. Engl. J. Med. 364, 1533–1543 (2011).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  2. Oh, E.C. & Katsanis, N. Cilia in vertebrate development and disease. Development 139, 443–448 (2012).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  3. Nachury, M.V. et al. A core complex of BBS proteins cooperates with the GTPase Rab8 to promote ciliary membrane biogenesis. Cell 129, 1201–1213 (2007).

    Article  CAS  PubMed  Google Scholar 

  4. Sang, L. et al. Mapping the NPHP–JBTS–MKS protein network reveals ciliopathy disease genes and pathways. Cell 145, 513–528 (2011).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  5. Garcia-Gonzalo, F.R. et al. A transition zone complex regulates mammalian ciliogenesis and ciliary membrane composition. Nat. Genet. 43, 776–784 (2011).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  6. Kobayashi, D. & Takeda, H. Ciliary motility: the components and cytoplasmic preassembly mechanisms of the axonemal dyneins. Differentiation 83, S23–S29 (2012).

    Article  CAS  PubMed  Google Scholar 

  7. Cole, D.G. et al. Chlamydomonas kinesin-II-dependent intraflagellar transport (IFT): IFT particles contain proteins required for ciliary assembly in Caenorhabditis elegans sensory neurons. J. Cell Biol. 141, 993–1008 (1998).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  8. Kozminski, K.G., Johnson, K.A., Forscher, P. & Rosenbaum, J.L. A motility in the eukaryotic flagellum unrelated to flagellar beating. Proc. Natl. Acad. Sci. USA 90, 5519–5523 (1993).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  9. Pazour, G.J., Wilkerson, C.G. & Witman, G.B. A dynein light chain is essential for the retrograde particle movement of intraflagellar transport (IFT). J. Cell Biol. 141, 979–992 (1998).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  10. Piperno, G. et al. Distinct mutants of retrograde intraflagellar transport (IFT) share similar morphological and molecular defects. J. Cell Biol. 143, 1591–1601 (1998).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  11. Iomini, C., Babaev-Khaimov, V., Sassaroli, M. & Piperno, G. Protein particles in Chlamydomonas flagella undergo a transport cycle consisting of four phases. J. Cell Biol. 153, 13–24 (2001).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  12. Pedersen, L.B., Geimer, S. & Rosenbaum, J.L. Dissecting the molecular mechanisms of intraflagellar transport in chlamydomonas. Curr. Biol. 16, 450–459 (2006).

    Article  CAS  PubMed  Google Scholar 

  13. Rompolas, P., Pedersen, L.B., Patel-King, R.S. & King, S.M. Chlamydomonas FAP133 is a dynein intermediate chain associated with the retrograde intraflagellar transport motor. J. Cell Sci. 120, 3653–3665 (2007).

    Article  CAS  PubMed  Google Scholar 

  14. Ou, G., Blacque, O.E., Snow, J.J., Leroux, M.R. & Scholey, J.M. Functional coordination of intraflagellar transport motors. Nature 436, 583–587 (2005).

    Article  CAS  PubMed  Google Scholar 

  15. Mukhopadhyay, S. et al. TULP3 bridges the IFT-A complex and membrane phosphoinositides to promote trafficking of G protein–coupled receptors into primary cilia. Genes Dev. 24, 2180–2193 (2010).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  16. Liem, K.F. Jr. et al. The IFT-A complex regulates Shh signaling through cilia structure and membrane protein trafficking. J. Cell Biol. 197, 789–800 (2012).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  17. Behal, R.H. et al. Subunit interactions and organization of the Chlamydomonas reinhardtii intraflagellar transport complex A proteins. J. Biol. Chem. 287, 11689–11703 (2012).

    Article  CAS  PubMed  Google Scholar 

  18. Lucker, B.F. et al. Characterization of the intraflagellar transport complex B core: direct interaction of the IFT81 and IFT74/72 subunits. J. Biol. Chem. 280, 27688–27696 (2005).

    Article  CAS  PubMed  Google Scholar 

  19. Taschner, M., Bhogaraju, S. & Lorentzen, E. Architecture and function of IFT complex proteins in ciliogenesis. Differentiation 83, S12–S22 (2012).

    Article  CAS  PubMed  Google Scholar 

  20. Craft, J.M., Harris, J.A., Hyman, S., Kner, P. & Lechtreck, K.F. Tubulin transport by IFT is upregulated during ciliary growth by a cilium-autonomous mechanism. J. Cell Biol. 208, 223–237 (2015).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  21. Bhogaraju, S. et al. Molecular basis of tubulin transport within the cilium by IFT74 and IFT81. Science 341, 1009–1012 (2013).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  22. Tarkar, A. et al. DYX1C1 is required for axonemal dynein assembly and ciliary motility. Nat. Genet. 45, 995–1003 (2013).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  23. Omran, H. et al. Ktu/PF13 is required for cytoplasmic pre-assembly of axonemal dyneins. Nature 456, 611–616 (2008).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  24. Mitchison, H.M. et al. Mutations in axonemal dynein assembly factor DNAAF3 cause primary ciliary dyskinesia. Nat. Genet. 44, 381–389, S1–S2 (2012).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  25. Horani, A. et al. Whole-exome capture and sequencing identifies HEATR2 mutation as a cause of primary ciliary dyskinesia. Am. J. Hum. Genet. 91, 685–693 (2012).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  26. Diggle, C.P. et al. HEATR2 plays a conserved role in assembly of the ciliary motile apparatus. PLoS Genet. 10, e1004577 (2014).

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  27. Seo, S. et al. BBS6, BBS10, and BBS12 form a complex with CCT/TRiC family chaperonins and mediate BBSome assembly. Proc. Natl. Acad. Sci. USA 107, 1488–1493 (2010).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  28. Zhang, Q., Yu, D., Seo, S., Stone, E.M. & Sheffield, V.C. Intrinsic protein–protein interaction-mediated and chaperonin-assisted sequential assembly of stable Bardet–Biedl syndrome protein complex, the BBSome. J. Biol. Chem. 287, 20625–20635 (2012).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  29. Goetz, S.C., Liem, K.F. Jr. & Anderson, K.V. The spinocerebellar ataxia-associated gene Tau tubulin kinase 2 controls the initiation of ciliogenesis. Cell 151, 847–858 (2012).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  30. Ye, X., Zeng, H., Ning, G., Reiter, J.F. & Liu, A. C2cd3 is critical for centriolar distal appendage assembly and ciliary vesicle docking in mammals. Proc. Natl. Acad. Sci. USA 111, 2164–2169 (2014).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  31. Singla, V., Romaguera-Ros, M., Garcia-Verdugo, J.M. & Reiter, J.F. Ofd1, a human disease gene, regulates the length and distal structure of centrioles. Dev. Cell 18, 410–424 (2010).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  32. Joo, K. et al. CCDC41 is required for ciliary vesicle docking to the mother centriole. Proc. Natl. Acad. Sci. USA 110, 5987–5992 (2013).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  33. Adler, P.N., Charlton, J. & Park, W.J. The Drosophila tissue polarity gene inturned functions prior to wing hair morphogenesis in the regulation of hair polarity and number. Genetics 137, 829–836 (1994).

    CAS  PubMed  PubMed Central  Google Scholar 

  34. Collier, S. & Gubb, D. Drosophila tissue polarity requires the cell-autonomous activity of the fuzzy gene, which encodes a novel transmembrane protein. Development 124, 4029–4037 (1997).

    CAS  PubMed  Google Scholar 

  35. Collier, S., Lee, H., Burgess, R. & Adler, P. The WD40 repeat protein fritz links cytoskeletal planar polarity to frizzled subcellular localization in the Drosophila epidermis. Genetics 169, 2035–2045 (2005).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  36. Kim, S.K. et al. Planar cell polarity acts through septins to control collective cell movement and ciliogenesis. Science 329, 1337–1340 (2010).

    Article  CAS  PubMed  PubMed Central  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  PubMed  Google Scholar 

  38. Huttlin, E.L. et al. The BioPlex network: a systematic exploration of the human interactome. Cell 162, 425–440 (2015).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  39. Rual, J.F. et al. Towards a proteome-scale map of the human protein–protein interaction network. Nature 437, 1173–1178 (2005).

    Article  CAS  PubMed  Google Scholar 

  40. Wang, Y., Yan, J., Lee, H., Lu, Q. & Adler, P.N. The proteins encoded by the Drosophila planar polarity effector genes inturned, fuzzy and fritz interact physically and can re-pattern the accumulation of “upstream” planar cell polarity proteins. Dev. Biol. 394, 156–169 (2014).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  41. Gray, R.S. et al. The planar cell polarity effector Fuz is essential for targeted membrane trafficking, ciliogenesis and mouse embryonic development. Nat. Cell Biol. 11, 1225–1232 (2009).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  42. Alazami, A.M. et al. Molecular characterization of Joubert syndrome in Saudi Arabia. Hum. Mutat. 33, 1423–1428 (2012).

    Article  CAS  PubMed  Google Scholar 

  43. Lopez, E. et al. C5orf42 is the major gene responsible for OFD syndrome type VI. Hum. Genet. 133, 367–377 (2014).

    Article  CAS  PubMed  Google Scholar 

  44. Shaheen, R. et al. Genomic analysis of Meckel–Gruber syndrome in Arabs reveals marked genetic heterogeneity and novel candidate genes. Eur. J. Hum. Genet. 21, 762–768 (2013).

    Article  CAS  PubMed  Google Scholar 

  45. Srour, M. et al. Mutations in C5orf42 cause Joubert syndrome in the French Canadian population. Am. J. Hum. Genet. 90, 693–700 (2012).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  46. Damerla, R.R. et al. Novel Jbts17 mutant mouse model of Joubert syndrome with cilia transition zone defects and cerebellar and other ciliopathy related anomalies. Hum. Mol. Genet. 24, 3994–4005 (2015).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  47. Brooks, E.R. & Wallingford, J.B. Control of vertebrate intraflagellar transport by the planar cell polarity effector Fuz. J. Cell Biol. 198, 37–45 (2012).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  48. Brooks, E.R. & Wallingford, J.B. The small GTPase Rsg1 is important for the cytoplasmic localization and axonemal dynamics of intraflagellar transport proteins. Cilia 2, 13 (2013).

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  49. Gurrieri, F., Franco, B., Toriello, H. & Neri, G. Oral-facial-digital syndromes: review and diagnostic guidelines. Am. J. Med. Genet. A. 143A, 3314–3323 (2007).

    Article  PubMed  Google Scholar 

  50. Tabler, J.M. et al. Fuz mutant mice reveal shared mechanisms between ciliopathies and FGF-related syndromes. Dev. Cell 25, 623–635 (2013).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  51. Merrill, A.E. et al. Ciliary abnormalities due to defects in the retrograde transport protein DYNC2H1 in short-rib polydactyly syndrome. Am. J. Hum. Genet. 84, 542–549 (2009).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  52. Mill, P. et al. Human and mouse mutations in WDR35 cause short-rib polydactyly syndromes due to abnormal ciliogenesis. Am. J. Hum. Genet. 88, 508–515 (2011).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  53. Hsieh, Y.C. & Hou, J.W. Oral-facial-digital syndrome with Y-shaped fourth metacarpals and endocardial cushion defect. Am. J. Med. Genet. 86, 278–281 (1999).

    Article  CAS  PubMed  Google Scholar 

  54. Panigrahi, I., Das, R.R., Kulkarni, K.P. & Marwaha, R.K. Overlapping phenotypes in OFD type II and OFD type VI: report of two cases. Clin. Dysmorphol. 22, 109–114 (2013).

    Article  PubMed  Google Scholar 

  55. Saari, J., Lovell, M.A., Yu, H.C. & Bellus, G.A. Compound heterozygosity for a frame shift mutation and a likely pathogenic sequence variant in the planar cell polarity–ciliogenesis gene WDPCP in a girl with polysyndactyly, coarctation of the aorta, and tongue hamartomas. Am. J. Med. Genet. A. 167A, 421–427 (2015).

    Article  PubMed  CAS  Google Scholar 

  56. Li, Y. et al. Global genetic analysis in mice unveils central role for cilia in congenital heart disease. Nature 521, 520–524 (2015).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  57. Cui, C. et al. Wdpcp, a PCP protein required for ciliogenesis, regulates directional cell migration and cell polarity by direct modulation of the actin cytoskeleton. PLoS Biol. 11, e1001720 (2013).

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  58. Butler, M.T. & Wallingford, J.B. Control of vertebrate core planar cell polarity protein localization and dynamics by Prickle 2. Development 142, 3429–3439 (2015).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  59. Zilber, Y. et al. The PCP effector Fuzzy controls cilial assembly and signaling by recruiting Rab8 and Dishevelled to the primary cilium. Mol. Biol. Cell 24, 555–565 (2013).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  60. van Dam, T.J. et al. Evolution of modular intraflagellar transport from a coatomer-like progenitor. Proc. Natl. Acad. Sci. USA 110, 6943–6948 (2013).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  61. Pan, X. et al. Mechanism of transport of IFT particles in C. elegans cilia by the concerted action of kinesin-II and OSM-3 motors. J. Cell Biol. 174, 1035–1045 (2006).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  62. Snow, J.J. et al. Two anterograde intraflagellar transport motors cooperate to build sensory cilia on C. elegans neurons. Nat. Cell Biol. 6, 1109–1113 (2004).

    Article  CAS  PubMed  Google Scholar 

  63. Harris, J.A., Liu, Y., Yang, P., Kner, P. & Lechtreck, K.F. Single particle imaging reveals IFT-independent transport and accumulation of EB1 in Chlamydomonas flagella. Mol. Biol. Cell 27, 295–307 (2016).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  64. Seo, J.H. et al. Mutations in the planar cell polarity gene, Fuzzy, are associated with neural tube defects in humans. Hum. Mol. Genet. 20, 4324–4333 (2011).

    Article  CAS  PubMed  Google Scholar 

  65. Ooi, Y.S., Stiles, K.M., Liu, C.Y., Taylor, G.M. & Kielian, M. Genome-wide RNAi screen identifies novel host proteins required for alphavirus entry. PLoS Pathog. 9, e1003835 (2013).

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  66. Giles, R.H., Ajzenberg, H. & Jackson, P.K. 3D spheroid model of mIMCD3 cells for studying ciliopathies and renal epithelial disorders. Nat. Protoc. 9, 2725–2731 (2014).

    Article  CAS  PubMed  Google Scholar 

  67. Moody, S.A. & Kline, M.J. Segregation of fate during cleavage of frog (Xenopus laevis) blastomeres. Anat. Embryol. (Berl.) 182, 347–362 (1990).

    Article  CAS  Google Scholar 

  68. Zeng, H., Hoover, A.N. & Liu, A. PCP effector gene Inturned is an important regulator of cilia formation and embryonic development in mammals. Dev. Biol. 339, 418–428 (2010).

    Article  CAS  PubMed  Google Scholar 

  69. Karpinka, J.B. et al. Xenbase, the Xenopus model organism database; new virtualized system, data types and genomes. Nucleic Acids Res. 43, D756–D763 (2015).

    Article  CAS  PubMed  Google Scholar 

  70. Park, T.J., Mitchell, B.J., Abitua, P.B., Kintner, C. & Wallingford, J.B. Dishevelled controls apical docking and planar polarization of basal bodies in ciliated epithelial cells. Nat. Genet. 40, 871–879 (2008).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  71. Nakayama, T. et al. Cas9-based genome editing in Xenopus tropicalis. Methods Enzymol. 546, 355–375 (2014).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  72. Lee, C., Kieserman, E., Gray, R.S., Park, T.J. & Wallingford, J. Whole-mount fluorescence immunocytochemistry on Xenopus embryos. CSH Protoc. 2008, pdb.prot4957 (2008).

  73. Sive, H.L., Grainger, R.M. & Harland, R.M. Early Development of Xenopus laevis: A Laboratory Manual (Cold Spring Harbor Press, 2000).

  74. Söding, J. Protein homology detection by HMM-HMM comparison. Bioinformatics 21, 951–960 (2005).

    Article  PubMed  Google Scholar 

  75. Lobley, A., Sadowski, M.I. & Jones, D.T. pGenTHREADER and pDomTHREADER: new methods for improved protein fold recognition and superfamily discrimination. Bioinformatics 25, 1761–1767 (2009).

    Article  CAS  PubMed  Google Scholar 

  76. Hasegawa, H. & Holm, L. Advances and pitfalls of protein structural alignment. Curr. Opin. Struct. Biol. 19, 341–348 (2009).

    Article  CAS  PubMed  Google Scholar 

Download references

Acknowledgements

We thank the patients and their families, the IntegraGen society for exome analysis and the NHLBI GO Exome Sequencing Project, which produced and provided exome variant calls for comparison: Lung GO Sequencing Project (HL-102923), WHI Sequencing Project (HL-102924), Broad GO Sequencing Project (HL-102925), Seattle GO Sequencing Project (HL-102926) and Heart GO Sequencing Project (HL-103010). We thank the Biological Resources Center–Ferdinand Cabanne (Dijon, France) for fibroblast centralization and storage. Sequencing was provided by the University of Washington Center for Mendelian Genomics (UW CMG) and was funded by the NHGRI and NHLBI (grant 1U54 HG006493) to D. Nickerson, J. Shendure and M. Bamshad. This work was supported by grants from the following: Uehara Memorial Foundation Fellowship to M.T.; an NIDCR NRSA to J.M.T.; the French Rare Diseases Foundation, the French Ministry of Health (PHRC national 2010-A01014-35 to C.T.-R.) and the Regional Council of Burgundy to C.T.-R.; NIDDK (DK1068306) to F.H., who is a Howard Hughes Medical Institute investigator, a Doris Duke Distinguished Clinical Scientist and the Warren E. Grupe Professor; NIAMS (AR061485) to J.C.; BBSRC (BB/K010492/1) and MRC (MR/L017237/1) to K.J.L.; NIH, NSF, CPRIT and the Welch Foundation (F-1515) to E.M.M.; R01 AR066124, March of Dimes and the Joseph Drown Foundation, NIH/NCATS UCLA CTSI grant UL1TR000124 to D.K.; R01 AR062651 to D.H.C.; NIGMS (GM114276), Baxter Laboratory, the Stanford Department of Research and NIGMS (GM114276) to P.K.J.; and NIGMS (GM104853) and NHLBI (HL117164) to J.B.W., who was a Howard Hughes Medical Institute Early Career Scientist.

Author information

Authors and Affiliations

Authors

Consortia

Contributions

M.T. contributed to the design, execution and interpretation of the overall research plan, with special emphasis on all Xenopus embryo experiments and in vitro binding assays. M.T. also contributed to writing the manuscript. C.L. designed, performed and interpreted live imaging of IFT particles in axonemes and contributed to other imaging experiments in Xenopus. K.D. and E.M.M. provided protein structural models. J.M.T., J.C. and K.J.L. contributed to the design, execution and interpretation of mouse genetic data. M.R.K. contributed to the execution and analysis of the proteomic data. S.K. contributed targeted coimmunoprecipitation data that confirmed CPLANE interactions. T.J.P. contributed to Xenopus studies. S.P.T., I.D., D.H.C., A.-L.B., D.A.B., G.P., A.B., K.W., A.M., I.P., B.F., H.A.A., Y.Y., Y.J.C., the University of Washington Center for Mendelian Genomics, Y.D., L.F. and J.-B.R. contributed to the collection of human patient and sequencing data. F.H., C.T.-R. and D.K. contributed to the design, execution and interpretation of human genetic data. P.K.J. contributed to coordinating the overall research effort with a focus on the design and interpretation of the proteomic screen and contributed to writing the manuscript. J.B.W. coordinated the overall research effort, oversaw experimental design and interpretation, and wrote the manuscript.

Corresponding author

Correspondence to John B Wallingford.

Ethics declarations

Competing interests

The authors declare no competing financial interests.

Additional information

A full list of members is available at http://uwcmg.org/#/.

Integrated supplementary information

Supplementary Figure 1 CPLANE pulldowns.

(a) Tandem affinity purification of Inturned interacting proteins. Lysates from mouse IMCD3 collecting duct cells stably expressing LAP-Intu were subjected to tandem affinity purification and silver stain to visualize major interacting species. Molecular mass markers are indicated on the left. (b) Tandem affinity purification of interacting proteins for Inturned, Fuz and Wdpcp. “sindicates the tagged proteins. Molecular mass markers are indicated on the left. The calculated molecular masses for these proteins are as follows: mIntu, 104.8 kDa; mFuz, 45.5 kDa; mWdpcp, 81.7 kDa. (ce) Tables showing an extracted subset of data from Supplementary Data 1 and highlighting key findings reported here. Numbers indicate peptide spectral matches for preys retrieved after pulldowns with the indicated baits. The “Other Ciliopathy” column reflects combined data from pulldowns of Ahi1, Cep290, Invs, Iqcb1 and Nphp4.

Supplementary Figure 2 CPLANE interactors.

(a) Venn diagram showing overlap between the interactomes of Intu, Fuz and Wdpcp. The intersection of the three (“combined CPLANE interactome”) contains ~250 proteins. (b) Additional components of the extended CPLANE protein network built using data from tandem affinity purification of Intu, Fuz, Wdpcp, IFT-A and the published NPHP network and thresholded for most likely network members. (c) The CPLANE proteins associate with IFT-A proteins and poorly characterized cilia-related proteins (arrows indicates bait → prey in Fuz, Wdpcp and Intu pulldowns). (d) The core CPLANE complex was identified by reciprocal pulldowns. (e) Coimmunoprecipitation with in vitro–translated proteins confirms CPLANE interactions with Jbts17 (a fragment of Jbts17 was used, as the very large size of this protein made in vitro translation intractable).

Supplementary Figure 3 Basal body localization of CPLANE proteins.

(a) Dorsal views of stage 19 Xenopus embryos show neural tube closure defects after Jbts17 knockdown that are rescued by expression of wild-type Jbts17 but not the Joubert syndrome–associated truncation (R1569*). The graph shows average distance between neural folds. (b) In situ hybridization to stage 30 Xenopus embryos shows loss of expression of vax1, a gene downstream of sonic hedgehog, after Jbts17 knockdown; numbers in each panel indicate the number of embryos showing the phenotype. (cf) The fluorescence micrographs show the localization of CPLANE proteins at basal bodies in multiciliated cells with Inturned (c), Fuzzy (d), Wdpcp (e) or Rsg1 (f) knockdown. Green and red signals represent GFP-tagged CPLANE proteins and centrin4-RFP, respectively. The graph to the right in each figure shows the fluorescence intensity of GFP signals normalized against Centrin4-RFP (ns, not significant; **P < 0.01, ***P < 0.001). (g) The fluorescence micrographs show the localization of GFP-tagged Cep164, Ofd1, Hook2 and Mks1 in control multiciliated cells and after Jbts17 knockdown. The graph to the right shows the normalized fluorescence intensity for the indicated proteins, as described for cf. (h) CRISPR-mediated disruption of Jbts17 phenocopies MO knockdown. The gels to the left demonstrate targeting of Jbts17 (see the Online Methods for details); the images to the right show disruption of ciliogenesis in MCCs.

Supplementary Figure 4 Effect of a Joubert syndrome–associated CPLANE allele.

(a) The Jbts17 truncated mutant and fragment constructs used in this study. (b) Fluorescence images of multiciliated cells expressing GFP-tagged wild-type Jbts17, R1569* and R2406* mutant Jbts17, and amino acids 1770–2318 of Jbts17. (c,d) Fluorescence images of GFP-tagged Inturned (c) and Ift43 (d) in control multiciliated cells and cells from Jbts17-knockdown embryos with no exogenous expression or expressing wild-type or truncated mutant (R1569*) Jbts17. The graphs show the normalized fluorescence intensity of GFP-Inturned (c) and GFP-IFT43 (d) at basal bodies, as described in Supplementary Figure 2. Scale bars, 10 μm.

Supplementary Figure 5 CPLANE and IFT.

(a,b) The fluorescence micrographs show the localization of the indicated GFP-tagged IFT-A (a) and IFT-B (b) proteins at basal bodies in multiciliated cells. The bottom panels in each figure show high-magnification images of basal bodies. (ce) Live images of multiciliated cells expressing GFP-tagged IFTs (green) and membrane-RFP (magenta) in control and Jbts17 morphants. (f,g) Kymographs representing the movements of IFT particles in control, Jbts17 and WDPCP morphants. (f) GFP-IFT20. (g) GFP-IFT122. (h) Plot showing the mean intensity of IFT protein fusions in axonemes. Scale bars, 10 μm.

Supplementary Figure 6 CPLANE mutations in human patients.

(a) The D54 residue of WDPCP that is mutated in OFD is invariant from human to fish. (b) Y-shaped metacarpals in a human patient with OFD harboring a mutation in INTU; this is patient 2 from Panigrahi et al. (2013). (c) An INTU mutation segregates with the OFD phenotype. (d) An INTU missense mutation segregates with NPHP in another family. (e) The A452 residue of INTU that is mutated in NPHP is not well conserved and may be hypomorphic, consistent with the more restricted phenotype in this patient. (f) The E500 residue of INTU that is mutated in SRP is invariant from human to fish. (g,h) Radiographs showing the phenotype of a patient with SRP transheterozygous for mutations in INTU and WDR35 (Ift121), as indicated by the pedigree in i. (j) The W311 residue of WDR35 that is mutated in SRP is invariant from human to fish.

Supplementary Figure 7 Models of CPLANE protein structures.

(a) Fuz. (b) Wdpcp. (c) Intu. (d) Rsg1. (e) Jbts17. As outlined in the Discussion, modeling predicts similarities between CPLANE proteins and vesicle trafficking machinery.

Supplementary information

Supplementary Text and Figures

Supplementary Figures 1–7 and Supplementary Tables 1–3. (PDF 3107 kb)

Supplementary Data 1

CPLANE pulldown interactions. (XLS 1176 kb)

Supplementary Data 2

Cytoscape network of CPLANE pulldown interactions. (ZIP 153 kb)

Rights and permissions

Reprints and permissions

About this article

Check for updates. Verify currency and authenticity via CrossMark

Cite this article

Toriyama, M., Lee, C., Taylor, S. et al. The ciliopathy-associated CPLANE proteins direct basal body recruitment of intraflagellar transport machinery. Nat Genet 48, 648–656 (2016). https://doi.org/10.1038/ng.3558

Download citation

  • Received:

  • Accepted:

  • Published:

  • Issue Date:

  • DOI: https://doi.org/10.1038/ng.3558

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