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:

Prostaglandin signalling regulates ciliogenesis by modulating intraflagellar transport

Abstract

Cilia are microtubule-based organelles that mediate signal transduction in a variety of tissues. Despite their importance, the signalling cascades that regulate cilium formation remain incompletely understood. Here we report that prostaglandin signalling affects ciliogenesis by regulating anterograde intraflagellar transport (IFT). Zebrafish leakytail (lkt) mutants show ciliogenesis defects, and the lkt locus encodes an ATP-binding cassette transporter (ABCC4). We show that Lkt/ABCC4 localizes to the cell membrane and exports prostaglandin E2 (PGE2), a function that is abrogated by the Lkt/ABCC4T804M mutant. PGE2 synthesis enzyme cyclooxygenase-1 and its receptor, EP4, which localizes to the cilium and activates the cyclic-AMP-mediated signalling cascade, are required for cilium formation and elongation. Importantly, PGE2 signalling increases anterograde but not retrograde velocity of IFT and promotes ciliogenesis in mammalian cells. These findings lead us to propose that Lkt/ABCC4-mediated PGE2 signalling acts through a ciliary G-protein-coupled receptor, EP4, to upregulate cAMP synthesis and increase anterograde IFT, thereby promoting ciliogenesis.

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: lkt mutants exhibit cilium loss and cilium-associated phenotypes.
Figure 2: Positional cloning identifies leakytail as the ABCC4 transporter.
Figure 3: lkt/abcc4 is expressed during embryonic development and regulates organ laterality in DFC/KV cells.
Figure 4: Lkt/ABCC4 exports PGE2 and regulates ciliogenesis.
Figure 5: The COX–EP4 signalling pathway is required for ciliogenesis.
Figure 6: Inhibition of the COX–EP4 signalling pathway using pharmacological agents leads to ciliogenesis defects.
Figure 7: Lkt/ABCC4-mediated PGE2 signalling promotes ciliogenesis in mammalian cells.
Figure 8: PGE2 signalling increases anterograde velocity of IFT.

Similar content being viewed by others

Accession codes

Accessions

GenBank/EMBL/DDBJ

References

  1. Smith, W. L., DeWitt, D. L. & Garavito, R. M. Cyclooxygenases: structural, cellular, and molecular biology. Annu. Rev. Biochem. 69, 145–182 (2000).

    Article  CAS  PubMed  Google Scholar 

  2. Langenbach, R., Loftin, C. D., Lee, C. & Tiano, H. Cyclooxygenase-deficient mice. A summary of their characteristics and susceptibilities to inflammation and carcinogenesis. Ann. N.Y. Acad. Sci. 889, 52–61 (1999).

    Article  CAS  PubMed  Google Scholar 

  3. Narumiya, S., Sugimoto, Y. & Ushikubi, F. Prostanoid receptors: structures, properties, and functions. Physiol. Rev. 79, 1193–1226 (1999).

    Article  CAS  PubMed  Google Scholar 

  4. Goetz, S. C. & Anderson, K. V. The primary cilium: a signalling centre during vertebrate development. Nat. Rev. Genet. 11, 331–344 (2010).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  5. Gerdes, J. M., Davis, E. E. & Katsanis, N. The vertebrate primary cilium in development, homeostasis, and disease. Cell 137, 32–45 (2009).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  6. 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 

  7. Omori, Y. et al. Elipsa is an early determinant of ciliogenesis that links the IFT particle to membrane-associated small GTPase Rab8. Nat. Cell Biol. 10, 437–444 (2008).

    Article  CAS  PubMed  Google Scholar 

  8. Fliegauf, M., Benzing, T. & Omran, H. When cilia go bad: cilia defects and ciliopathies. Nat. Rev. Mol. Cell Biol. 8, 880–893 (2007).

    Article  CAS  PubMed  Google Scholar 

  9. Neugebauer, J. M., Amack, J. D., Peterson, A. G., Bisgrove, B. W. & Yost, H. J. FGF signalling during embryo development regulates cilia length in diverse epithelia. Nature 458, 651–654 (2009).

    CAS  PubMed  PubMed Central  Google Scholar 

  10. Caron, A., Xu, X. & Lin, X. Wnt/beta-catenin signaling directly regulates Foxj1 expression and ciliogenesis in zebrafish Kupffer’s vesicle. Development 139, 514–524 (2012).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  11. Lopes, S. S. et al. Notch signalling regulates left-right asymmetry through ciliary length control. Development 137, 3625–3632 (2010).

    Article  CAS  PubMed  Google Scholar 

  12. Sarmah, B., Winfrey, V. P., Olson, G. E., Appel, B. & Wente, S. R. A role for the inositol kinase Ipk1 in ciliary beating and length maintenance. Proc. Natl Acad. Sci. USA 104, 19843–19848 (2007).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  13. Long, S., Ahmad, N. & Rebagliati, M. The zebrafish nodal-related gene southpaw is required for visceral and diencephalic left-right asymmetry. Development 130, 2303–2316 (2003).

    Article  CAS  PubMed  Google Scholar 

  14. Zhong, T. P., Rosenberg, M., Mohideen, M. A., Weinstein, B. & Fishman, M. C. Gridlock, an HLH gene required for assembly of the aorta in zebrafish. Science 287, 1820–1824 (2000).

    Article  CAS  PubMed  Google Scholar 

  15. Russel, F. G., Koenderink, J. B. & Masereeuw, R. Multidrug resistance protein 4 (MRP4/ABCC4): a versatile efflux transporter for drugs and signalling molecules. Trends Pharmacol. Sci. 29, 200–207 (2008).

    Article  CAS  PubMed  Google Scholar 

  16. Schuetz, J. D. et al. MRP4: a previously unidentified factor in resistance to nucleoside-based antiviral drugs. Nat. Med. 5, 1048–1051 (1999).

    Article  CAS  PubMed  Google Scholar 

  17. Slot, A. J., Molinski, S. V. & Cole, S. P. Mammalian multidrug-resistance proteins (MRPs). Essays Biochem. 50, 179–207 (2011).

    Article  CAS  PubMed  Google Scholar 

  18. Schuldiner, S. The ins and outs of drug transport. Nature 144, 156–157 (2006).

    Google Scholar 

  19. Essner, J. J., Amack, J. D., Nyholm, M. K., Harris, E. B. & Yost, H. J. Kupffer’s vesicle is a ciliated organ of asymmetry in the zebrafish embryo that initiates left-right development of the brain, heart and gut. Development 132, 1247–1260 (2005).

    Article  CAS  PubMed  Google Scholar 

  20. Kramer-Zucker, A. G. et al. Cilia-driven fluid flow in the zebrafish pronephros, brain and Kupffer’s vesicle is required for normal organogenesis. Development 132, 1907–1921 (2005).

    Article  CAS  PubMed  Google Scholar 

  21. Amack, J. D. & Yost, H. J. The T box transcription factor no tail in ciliated cells controls zebrafish left-right asymmetry. Curr. Biol. 14, 685–690 (2004).

    Article  CAS  PubMed  Google Scholar 

  22. Reid, G. et al. The human multidrug resistance protein MRP4 functions as a prostaglandin efflux transporter and is inhibited by nonsteroidal antiinflammatory drugs. Proc. Natl Acad. Sci. USA 100, 9244–9249 (2003).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  23. Fukuda, Y. et al. Human immunodeficiency virus protease inhibitors interact with ATP binding cassette transporter 4/multidrug resistance protein 4: a basis for unanticipated enhanced cytotoxicity. Mol. Pharmacol. 84, 361–371 (2013).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  24. Conseil, G., Deeley, R. G. & Cole, S. P. Functional importance of three basic residues clustered at the cytosolic interface of transmembrane helix 15 in the multidrug and organic anion transporter MRP1 (ABCC1). J. Biol. Chem. 281, 43–50 (2006).

    Article  CAS  PubMed  Google Scholar 

  25. Cha, Y. I. et al. Cyclooxygenase-1-derived PGE2 promotes cell motility via the G-protein-coupled EP4 receptor during vertebrate gastrulation. Genes Dev. 20, 77–86 (2006).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  26. Cha, Y. I., Kim, S. H., Solnica-Krezel, L. & Dubois, R. N. Cyclooxygenase-1 signaling is required for vascular tube formation during development. Dev. Biol. 282, 274–283 (2005).

    Article  CAS  PubMed  Google Scholar 

  27. North, T. E. et al. Prostaglandin E2 regulates vertebrate haematopoietic stem cell homeostasis. Nature 447, 1007–1011 (2007).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  28. Goessling, W. et al. Genetic interaction of PGE2 and Wnt signaling regulates developmental specification of stem cells and regeneration. Cell 136, 1136–1147 (2009).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  29. Besschetnova, T. Y. et al. Identification of signaling pathways regulating primary cilium length and flow-mediated adaptation. Curr. Biol. 20, 182–187 (2010).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  30. Yu, X., Ng, C. P., Habacher, H. & Roy, S. Foxj1 transcription factors are master regulators of the motile ciliogenic program. Nat. Genet. 40, 1445–1453 (2008).

    Article  CAS  PubMed  Google Scholar 

  31. Stubbs, J. L., Oishi, I., Belmonte, J. C. I. & Kintner, C. The forkhead protein Foxj1 specifies node-like cilia in Xenopus and zebrafish embryos. Nat. Genet. 40, 1454–1460 (2008).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  32. Cao, J. et al. miR-129-3p controls cilia assembly by regulating CP110 and actin dynamics. Nat. Cell Biol. 14, 697–706 (2012).

    Article  CAS  PubMed  Google Scholar 

  33. St-Jacques, B. & Ma, W. Prostaglandin E2/EP4 signalling facilitates EP4 receptor externalization in primary sensory neurons in vitro and in vivo. Pain 154, 313–323 (2013).

    Article  CAS  PubMed  Google Scholar 

  34. Tran, P. V. et al. THM1 negatively modulates mouse sonic hedgehog signal transduction and affects retrograde intraflagellar transport in cilia. Nat. Genet. 40, 403–410 (2008).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  35. Tsujikawa, M. & Malicki, J. Intraflagellar transport genes are essential for differentiation and survival of vertebrate sensory neurons. Neuron 42, 703–716 (2004).

    Article  CAS  PubMed  Google Scholar 

  36. Boyce, J. A. Mast cells and eicosanoid mediators: a system of reciprocal paracrine and autocrine regulation. Immunol. Rev. 217, 168–185 (2007).

    Article  CAS  PubMed  Google Scholar 

  37. Starner, R. J., McClelland, L., Abdel-Malek, Z., Fricke, A. & Scott, G. PGE(2) is a UVR-inducible autocrine factor for human melanocytes that stimulates tyrosinase activation. Exp. Dermatol. 19, 682–684 (2010).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  38. Wang, Z., Phan, T. & Storm, D. R. The type 3 adenylyl cyclase is required for novel object learning and extinction of contextual memory: role of cAMP signaling in primary cilia. J. Neurosci. 31, 5557–5561 (2011).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  39. Engel, B. D., Ludington, W. B. & Marshall, W. F. Intraflagellar transport particle size scales inversely with flagellar length: revisiting the balance-point length control model. J. Cell Biol. 187, 81–89 (2009).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  40. 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 

  41. Lord, A. M., North, T. E. & Zon, L. I. Prostaglandin E2: making more of your marrow. Cell Cycle 6, 3054–3057 (2007).

    Article  CAS  PubMed  Google Scholar 

  42. Regan, J. W. EP2 and EP4 prostanoid receptor signaling. Life Sci. 74, 143–153 (2003).

    Article  CAS  PubMed  Google Scholar 

  43. Youk, H. & Lim, W. A. Secreting and sensing the same molecule allows cells to achieve versatile social behaviors. Science 343, 1242782 (2014).

    Article  PubMed  PubMed Central  Google Scholar 

  44. Haxel, B. R., Schafer, D., Klimek, L. & Mann, W. J. Prostaglandin E2 activates the ciliary beat frequency of cultured human nasal mucosa via the second messenger cyclic adenosine monophosphate. Eur. Arch. Otorhinolaryngol. 258, 230–235 (2001).

    Article  CAS  PubMed  Google Scholar 

  45. Gross, G. A. et al. Opposing actions of prostaglandins and oxytocin determine the onset of murine labor. Proc. Natl Acad. Sci. USA 95, 11875–11879 (1998).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  46. Loftin, C. D. et al. Failure of ductus arteriosus closure and remodeling in neonatal mice deficient in cyclooxygenase-1 and cyclooxygenase-2. Proc. Natl Acad. Sci. USA 98, 1059–1064 (2001).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  47. Dinchuk, J. E. et al. Renal abnormalities and an altered inflammatory response in mice lacking cyclooxygenase II. Nature 378, 406–409 (1995).

    Article  CAS  PubMed  Google Scholar 

  48. Reese, J. et al. Coordinated regulation of fetal and maternal prostaglandins directs successful birth and postnatal adaptation in the mouse. Proc. Natl Acad. Sci. USA 97, 9759–9764 (2000).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  49. 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 

  50. Becker-Heck, A. et al. The coiled-coil domain containing protein CCDC40 is essential for motile cilia function and left-right axis formation. Nat. Genet. 43, 79–84 (2011).

    Article  CAS  PubMed  Google Scholar 

  51. Westerfield, M. The Zebrafish Book (Univ. Oregon Press, 2000).

    Google Scholar 

  52. Driever, W. et al. A genetic screen for mutations affecting embryogenesis in zebrafish. Development 123, 37–46 (1996).

    CAS  PubMed  Google Scholar 

  53. Zhong, T. P., Childs, S., Leu, J. P. & Fishman, M. C. Gridlock signalling pathway fashions the first embryonic artery. Nature 414, 216–220 (2001).

    Article  CAS  PubMed  Google Scholar 

  54. Williams, C. et al. Hedgehog signaling induces arterial endothelial cell formation by repressing venous cell fate. Dev. Biol. 341, 196–204 (2010).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  55. Higginbotham, H. et al. Arl13b-regulated cilia activities are essential for polarized radial glial scaffold formation. Nat. Neurosci. 16, 1000–1007 (2013).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  56. Bogard, A. S., Xu, C. & Ostrom, R. S. Human bronchial smooth muscle cells express adenylyl cyclase isoforms 2, 4, and 6 in distinct membrane microdomains. J. Pharmacol. Exp. Ther. 337, 209–217 (2011).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  57. Hoque, M. T. & Cole, S. P. Down-regulation of Na + /H + exchanger regulatory factor 1 increases expression and function of multidrug resistance protein 4. Cancer Res. 68, 4802–4809 (2008).

    Article  CAS  PubMed  Google Scholar 

  58. Leggas, M. et al. Mrp4 confers resistance to topotecan and protects the brain from chemotherapy. Mol. Cell Biol. 24, 7612–7621 (2004).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

Download references

Acknowledgements

We acknowledge P. Yuanyuan and J. Guan for assistance in fish care, X. Zhu for hRPE1 cells, J. Shah for IFT88–EYFP cells, C. Yi for cAMP assays, Q. Li for diagram drawing and L. Cai and G. Zhu for spin-disk confocal microscopy analysis. We are grateful to H. Ma, B. Appel, Z. Sun, J. Gamse and members of our laboratories for comments on the manuscript and helpful discussions. This research was supported in part by grants from the National Basic Research Program of China (CMST2013CB945301, CMST2012CB944501; T.P.Z.), National Natural Science Foundation of China (NSFC31172173; T.P.Z.) and Shanghai Pujiang Program (11PJ1401600; T.P.Z.) as well as the National Institute of Health of America (T.P.Z., J.D.S., J.M., I.A.D.) and Canadian Institutes of Health Research (MOP106513, S.P.C.C.).

Author information

Authors and Affiliations

Authors

Contributions

T.P.Z. conceived and directed the project. T.T.N. and H.W. initiated the project and discovered the lkt gene product as ABCC4. D.J. carried out most experiments and discovered the roles of COX, ABCC4 and EP4 in ciliogenesis. J.S. and G.Y. conducted cell culture experiments. J.D.A. carried out KV flow experiments. S.C. and J.D.S. conducted PGE2 efflux experiments. J.F., C.C. and B.Z. carried out some of the cell culture experiments. G.C. and S.P.C.C. carried out vesicular transport assays. W.L. conducted double in situ hybridization. A.P. and J.M. tested roles of PGE2 in ift mutants and were involved in early mutant analyses. I.A.D. carried out histology and provided reagents. T.P.Z., D.J. and J.M. prepared figures and wrote the paper.

Corresponding author

Correspondence to Tao P. Zhong.

Ethics declarations

Competing interests

The authors declare no competing financial interests.

Integrated supplementary information

Supplementary Figure 1 lkt/abcc4-deficient embryos display randomization of organ laterality.

(a) cmlc2 expression showing the laterality (left-jagging versus right-jagging) of heart tube in wild-type (wt) embryos and lkt mutants. (b) foxA3 expression depicting the sidedness of liver (black arrow) and pancreas (white arrow) in wt embryos and lkt mutants. (c) insulin expression displaying the pancreas laterality in wt embryos and lkt mutants. (d) lft1 expression showing brain laterality in wt embryos and lkt mutants. (e,f) Acetylated tubulin immunostaining reveals normal pronepheric cilia development in lkt mutants compared with wild-type embryos. Scale bar: 100 μm (af); 10 μm (e,f). (g,h) Analysis of sidedness of asymmetric gene expression in wt embryos, lkt mutants and lkt/abcc4 morphant embryos. The total numbers of embryos analysed pooled from n = 3 independent experiments are indicated on the top of each bar. The number of embryos with sided expression of each marker divided by total number of embryos. The percentages of embryos with LR defects were significantly higher in lkt mutants or abcc4 morphants as compared with wt embryos, but not statistically different among lkt mutants or abcc4 morphant embryos (Yates corrected Chi-square test, P < 0.0001, one degree of freedom). Statistics source data for Supplementary Fig. 1g, h can be found in Supplementary Table 2.

Supplementary Figure 2 Annotated alignment of deduced full-length zebrafish Lkt/ABCC4 protein with orthologs in other species.

Black: identical across four species. Grey: identical in 2 or more species or conservative amino acid substitution. IDs: human (NP005836), mouse (NP001028508), chicken (NP001025990) and zebrafish (EU586042). TM: transmembrane domain. NBD: nucleotide binding domain. Red star: leakytail zebrafish mutation site (T804M). Walker A/B (ATP-binding sites) and signature C are conserved motifs found within the NBD of ABC transporters.

Supplementary Figure 3 Injection of lkt/abcc4 splicing morpholinos phenocopies lkt mutants.

(a) Schematic diagram depicting zebrafish lkt/abcc4 gene structure, including exon 9, 10, 11, and intron 9, 10. Red bar: abcc4 morpholinos target site. Black arrows: primer sites for RT-PCR. (b) RT-PCR analyses showed that wild-type lkt/abcc4 transcripts contained fused exons 9, 10 and 11, whereas mis-spliced transcripts contained only fused exons 9 and 11 in embryos injected with different doses of lkt/abcc4-MO. Total RNAs were isolated from morphants and controls, and subjected to RT-PCR using primers located in exon 9 and 11. (ce) Acetylated tubulin immunostaining showing a reduction of KV cilia (c), a loss of OV short cilia (d) and lack of some ear kinocilia in lkt/abcc4 morphants (e), similar to those of lkt mutants. (fh) lkt/abcc4 morphants showing a curved body axis (f; 81%; n = 123), hydrocephalus (f; 78%, n = 123; red arrow), three otoliths (g; 80%; n = 123; red arrows) and reversed (left)-looped heart (h; 41%; n = 93). (i,j) In situ hybridization showing expression of abcc4 (e; arrowhead) and insulin, a pancreas marker (f; arrowhead). (k) Double in situ hybridization with abcc4 and insulin probes showing expression of both genes in the pancreas (arrowhead). Inset displays a high-magnification image of abcc4 and insulin expression patterns, which largely overlap. (l,m) In situ hybridization showing expression of abcc4 (l; arrowhead) and wt1b, a glomerulus marker (m; arrows). (n) Double in situ with abcc4 (arrowhead) and wt1b (arrows) probes showing expression in the pancreas and glomerulus, respectively. Inset shows an enlarged view of abcc4 and wt1b expression. Double in situ hybridization analyses were conducted using digoxingenin-labeled lkt antisense probe (i,k,l,n). insulin (j,k) and wt1b antisense probes (m,n) were fluorescein-labeled. Scale bar: 10 μm (ce); 100 μm (fh; in).

Supplementary Figure 4 Ectopic expression of abcc4 mRNA rescues lkt mutant phenotypes.

(ad) lkt mutants displaying a ventrally curved axis (a), three otoliths (b; red arrows), hydrocephalus (c; red arrow) and reversed (left)-looped heart (d). (eh) abcc4 mRNA injection rescued mutant phenotypes, including a straight body axis (e), two otoliths (f), normal brain (g) and normal (right)-looped heart (h). Scale bar: 100 μm (ah). (i) Percentages of rescued lkt mutants grouped in heart looping, otolith biogenesis, axis formation or brain development. Approximately 160 pg of abcc4 mRNA was injected into embryos derived from crosses of lkt heterozygouts. Injected embryos were genotyped using SSLP markers (Z17212 and Z6907). Embryos linking with the lkt mutant genotype but showing wild-type phenotypes were scored as rescued mutants. %: The number of rescued lkt mutants divided by number of all lkt mutants. The total numbers of embryos pooled from n = 3 independent experiments are indicated. Graph shows mean ± s.d.; Student’s t test: P < 0.001. (j) Bar graph depicting percentages of normal (rescued) brain phenotypes in embryos co-injected with control morpholinos, antisense morpholinos and antisense morpholinos with specific mRNAs. Wild-type embryos were co-injected with cox1-MO (8 ng) with cox1 mRNA (130 pg), cox2-MO (8 ng) with cox2 mRNA (125 pg), ep4-MO (1 ng) with ep4 mRNA (120 pg) or abcc4-MO (12 ng) with abcc4 mRNA (160 ng). Ctrl: Ctrl-MO (8 ng). The total numbers of embryos pooled from n = 3 independent experiments are indicated. Graph shows mean ± s.d.; Student’s t test: P < 0.001. Statistics source data for Supplementary Fig. 4i, j can be found in Supplementary Table 2.

Supplementary Figure 5 PGE2 signaling affects cilia-dependent processes.

(a) Bar chart depicting percentages of phenotypically wild-type embryos in several experimental groups as indicated. lkt mutants were treated using PGE2 (50μm) or PGF2〈 (50μm) to rescue reversed heart looping (3–24 hpf), otolith defects (10–48 hpf) and hydrocephalus (10–72 hpf). The total number of embryos pooled from n = 3 independent experiments are indicated. Error bars represent mean ± s.d.; Student’s t test: P < 0.001. NS: not significant. (b,c) cox2-MO injection (8 ng) caused hydrocephalus and curved axis (31%; n = 110), and three otoliths (21%; n = 110; arrows). (dg) Acetylated tubulin immunostaining showing a reduction of ependymal cell cilia in the spinal canal in embryos injected with cox1-MO (e; 8 ng, n = 24), cox2-MO (f; 8ng, n = 27) and ep4-MO (g; 1 ng, n = 25), compared with control embryos at 72 hpf (d). Scale bar: 100 μm (b,c); 10 μm (dg). (h,i) Stacked bar graphs depicting percentages of embryos with phenotypes grouped in otolith defect (h) and curved axis (i). cox1/2 morphants were incubated in PGE2 (50μM) to rescue otolith defect (10 to 48 hpf) and curved axis (10 to 72 hpf). Treatment of ep4 morphants with FSK (50 μM) but not PGE2 rescued otolith defect (10 to 48 hpf) and curved axis (10 to 72 hpf) in ep4 morphants. Ctrl: embryos injected with Ctrl-MO. The total numbers of embryos pooled from n = 3 independent experiments are indicated on the top of each bar. Yates corrected Chi-square test, P < 0.001, degree of freedom = 1. NS: not significant. Statistics source data for Supplementary Fig. 5a, d, e can be found in Supplementary Table 2.

Supplementary Figure 6 Expression of foxj1a and foxj1b are not altered in embryos deficient in lkt/abcc4, cox1/cox2 or ep4 activities.

(a,b) lkt+/− heterozygous and wild-type homozygous embryos injected with different doses of cox1-MO and ep4-MO display comparable percentages of LR randomization and hydrocephalus. Embryos derived from crosses between lkt+/− heterozygotes and wild-type zebrafish were injected with cox1-MO, ep4-MO or control-MO (Ctrl). The injected embryos were genotyped using SSLP markers (Z17212) to distinguish lkt+/− heterozygous embryos (50%) from wild-type homozygotes. Percentages reflect the frequency of phenotypic abnormalities. The total numbers of injected embryos pooled from n = 3 independent experiments are indicated above each bar. Yates corrected Chi-square test, P < 0.0001, one degree of freedom. NS: Not significant. Statistics source data for Supplementary Fig. 6a, b can be found in Supplementary Table 2. (cf) In situ hybridization analyses revealing the comparable expression of foxj1a in DFC cells in wild-type embryos, lkt mutants, cox1/cox2 morphants or ep4 morphants at 9 hpf. (gj) The expression of foxj1a in the spinal cord precursors (arrowhead) and rudiments of the pronephric ducts (arrows) was not altered in lkt mutants, cox1/cox2 morphants or ep4 morphants, compared with wild-type embryos at 13 hpf. (kn) The foxj1b expression in developing otic placodes was not altered in lkt mutants, cox1/cox2 morphants or ep4 morphants compared with wild-type embryos at 13 hpf. Scale bar: 100 μm (al).

Supplementary Figure 7 Anterograde IFT velocity is increased in PGE2-treated IMCD3 cells.

Histograms showing IFT88-EYFP particle velocity distribution in PGE2-treated IMCD3 cells and control cells (n > 50). Blue: Anterograde velocity; Red: Retrograde velocity. Arrows represent mean velocity in untreated cells. PGE2 treatment increased the anterograde velocity but not the retrograde velocity.

Supplementary Figure 8 ABCC4- or EP4-depletion causes ciliogenesis deficiency in IMCD3 cells.

(ad) Immunostaining analyses showing individual cilia in IMCD3 cells treated with control siRNA, ABCC4-siRNA, EP4-siRNA or PGE2. Insets reveal high-magnification images of cilia (red arrows). Green: ARL13B for cilium; Red: γ-tubulin for centrosome; Blue: DAPI; Arrow: cilium. PGE2 treatment: 10 μM; 12 h. Scale bar: 10 μm (ad). (e,f) Immunoblot analysis showing ABCC4 or EP4 protein levels in IMCD3 cells transfected with control siRNA, ABCC4-siRNA or EP4-siRNA. (g,h) Statistical analyses of percentages of ciliated cells and average length of cilia in IMCD3 cells transfected with control siRNA (470), ABCC4 siRNA (654), EP4 siRNA (594) and control siRNA plus PGE2 (371). Student’s t-test: P < 0.01,P < 0.001; error bar represents mean ± s.d.; n = 3 independent experiments with total number of cells provided in parentheses. (i,j) Bar graphs showing steady-state levels of intracellular cAMP (i) and intracellular Ca2+ (j) in IMCD3 cells treated with DMSO (314), PGE2 (303) and FSK (305). PGE2 treatment causes an increase of intracellular cAMP but not Ca2+ in IMCD3 cells. Forskolin (FSK) is used as a positive control. cAMP concentrations were measured using cAMP complete ELISA kit (ENZO, Life Science). Intracellular Ca2+ intensity levels (340/380nm) were measured using Fura-2 probe. DMSO: 0.5%; PGE2: 10 uM; Forskolin: 100 uM. Error bar represents mean ± s.d.; Student’s t-test: P < 0.05,P < 0.01,P < 0.001; n = 3 independent experiments with total number of cells analysed provided in parentheses. Statistics source data for Supplementary Fig. 8g–j can be found in Supplementary Table 2. Uncropped images of the immunoblots are shown in Supplementary Fig. 9.

Supplementary Figure 9 Scans of full-size immunoblots.

The red rectangles indicate the immunoblot fragments presented in the main body of the paper.

Supplementary information

Supplementary Information

Supplementary Information (PDF 2912 kb)

Supplementary Table 1

Supplementary Information (XLS 34 kb)

Supplementary Table 2

Supplementary Information (XLSX 110 kb)

KV fluid flow in wild-type embryos.

This is a time-lapse of fluorescent bead movement within the KV of an 8-somite wild-type embryo. Imaged were taken at 25 frames per second using a Zeiss Axio Imager M1 microscope with a 63 × Plan Apochromat objective. (MOV 9776 kb)

Absence of KV fluid flow in lkt mutants.

This is a time-lapse of fluorescent bead movement within the mutant KV at the 8-somite stage. KV fluid flow was imaged (25 frames per second) under a 63 × Plan Apochromat objective using a Zeiss Axio Imager M1 microscope. (MOV 13409 kb)

Rights and permissions

Reprints and permissions

About this article

Check for updates. Verify currency and authenticity via CrossMark

Cite this article

Jin, D., Ni, T., Sun, J. et al. Prostaglandin signalling regulates ciliogenesis by modulating intraflagellar transport. Nat Cell Biol 16, 841–851 (2014). https://doi.org/10.1038/ncb3029

Download citation

  • Received:

  • Accepted:

  • Published:

  • Issue Date:

  • DOI: https://doi.org/10.1038/ncb3029

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