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:

Shh-mediated centrosomal recruitment of PKA promotes symmetric proliferative neuroepithelial cell division

Abstract

Tight control of the balance between self-expanding symmetric and self-renewing asymmetric neural progenitor divisions is crucial to regulate the number of cells in the developing central nervous system. We recently demonstrated that Sonic hedgehog (Shh) signalling is required for the expansion of motor neuron progenitors by maintaining symmetric divisions. Here we show that activation of Shh/Gli signalling in dividing neuroepithelial cells controls the symmetric recruitment of PKA to the centrosomes that nucleate the mitotic spindle, maintaining symmetric proliferative divisions. Notably, Shh signalling upregulates the expression of pericentrin, which is required to dock PKA to the centrosomes, which in turn exerts a positive feedback onto Shh signalling. Thus, by controlling centrosomal protein assembly, we propose that Shh signalling overcomes the intrinsic asymmetry at the centrosome during neuroepithelial cell division, thereby promoting self-expanding symmetric divisions and the expansion of the progenitor pool.

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: Strong Shh/Gli activity in symmetrically dividing MN progenitors.
Figure 2: PKA localizes to the centrosome in dividing neural progenitors.
Figure 3: PKA docking to centrosomes changes during NT development.
Figure 4: Activation of the Shh/Gli pathway correlates with symmetric centrosomal docking of PKA.
Figure 5: Activation of the Shh/Gli pathway correlates with symmetric inheritance of apical membrane domains.
Figure 6: Asymmetric centrosomal docking of PKA is correlated with neurogenic divisions.
Figure 7: Shh signalling regulates pericentrin expression.
Figure 8: Pericentrin-mediated PKA docking to the centrosomes is necessary for Shh/Gli activation and inhibition of neurogenesis.

Similar content being viewed by others

References

  1. Cajal, R.y. Textura del Sistema Nervioso del Hombre y de los Vertebrados Ch. XXI (Nicolás Moya, 1899).

    Google Scholar 

  2. Gotz, M. & Huttner, W. B. The cell biology of neurogenesis. Nat. Rev. Mol. Cell Biol. 6, 777–788 (2005).

    Article  PubMed  CAS  Google Scholar 

  3. Lui, J. H., Hansen, D. V. & Kriegstein, A. R. Development and evolution of the human neocortex. Cell 146, 18–36 (2011).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  4. Franco, S. J. et al. Fate-restricted neural progenitors in the mammalian cerebral cortex. Science 337, 746–749 (2012).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  5. Franco, S. J. & Muller, U. Shaping our minds: stem and progenitor cell diversity in the mammalian neocortex. Neuron 77, 19–34 (2013).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  6. Delattre, M. & Gonczy, P. The arithmetic of centrosome biogenesis. J. Cell Sci. 117, 1619–1630 (2004).

    Article  CAS  PubMed  Google Scholar 

  7. Nigg, E. A. & Raff, J. W. Centrioles, centrosomes, and cilia in health and disease. Cell 139, 663–678 (2009).

    Article  CAS  PubMed  Google Scholar 

  8. Reina, J. & Gonzalez, C. When fate follows age: unequal centrosomes in asymmetric cell division. Phil. Trans. R. Soc. B 369, 20130466 (2014).

    Article  PubMed  CAS  PubMed Central  Google Scholar 

  9. Wang, X. et al. Asymmetric centrosome inheritance maintains neural progenitors in the neocortex. Nature 461, 947–955 (2009).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  10. Paridaen, J. T., Wilsch-Brauninger, M. & Huttner, W. B. Asymmetric inheritance of centrosome-associated primary cilium membrane directs ciliogenesis after cell division. Cell 155, 333–344 (2013).

    Article  CAS  PubMed  Google Scholar 

  11. Saade, M. et al. Sonic hedgehog signaling switches the mode of division in the developing nervous system. Cell Rep. 4, 492–503 (2013).

    Article  CAS  PubMed  Google Scholar 

  12. Lai, K., Kaspar, B. K., Gage, F. H. & Schaffer, D. V. Sonic hedgehog regulates adult neural progenitor proliferation in vitro and in vivo. Nat. Neurosci. 6, 21–27 (2003).

    Article  CAS  PubMed  Google Scholar 

  13. Machold, R. et al. Sonic hedgehog is required for progenitor cell maintenance in telencephalic stem cell niches. Neuron 39, 937–950 (2003).

    Article  CAS  PubMed  Google Scholar 

  14. Briscoe, J. Making a grade: sonic Hedgehog signalling and the control of neural cell fate. EMBO J. 28, 457–465 (2009).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  15. Cayuso, J., Ulloa, F., Cox, B., Briscoe, J. & Marti, E. The Sonic hedgehog pathway independently controls the patterning, proliferation and survival of neuroepithelial cells by regulating Gli activity. Development 133, 517–528 (2006).

    Article  CAS  PubMed  Google Scholar 

  16. Wong, W. & Scott, J. D. AKAP signalling complexes: focal points in space and time. Nat. Rev. Mol. Cell Biol. 5, 959–970 (2004).

    Article  CAS  PubMed  Google Scholar 

  17. Le Dreau, G., Saade, M., Gutierrez-Vallejo, I. & Marti, E. The strength of SMAD1/5 activity determines the mode of stem cell division in the developing spinal cord. J. Cell Biol. 204, 591–605 (2014).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  18. Briscoe, J. & Therond, P. P. The mechanisms of Hedgehog signalling and its roles in development and disease. Nat. Rev. Mol. Cell Biol. 14, 416–429 (2013).

    Article  PubMed  CAS  Google Scholar 

  19. Niewiadomski, P. et al. Gli protein activity is controlled by multisite phosphorylation in vertebrate Hedgehog signaling. Cell Rep. 6, 168–181 (2014).

    Article  CAS  PubMed  Google Scholar 

  20. Dzhindzhev, N. S. et al. Asterless is a scaffold for the onset of centriole assembly. Nature 467, 714–718 (2010).

    Article  CAS  PubMed  Google Scholar 

  21. Yan, X., Habedanck, R. & Nigg, E. A. A complex of two centrosomal proteins, CAP350 and FOP, cooperates with EB1 in microtubule anchoring. Mol. Biol. Cell 17, 634–644 (2006).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  22. Nigg, E. A., Schafer, G., Hilz, H. & Eppenberger, H. M. Cyclic-AMP-dependent protein kinase type II is associated with the Golgi complex and with centrosomes. Cell 41, 1039–1051 (1985).

    Article  CAS  PubMed  Google Scholar 

  23. Vandame, P. et al. The spatio-temporal dynamics of PKA activity profile during mitosis and its correlation to chromosome segregation. Cell Cycle 13, 3232–3240 (2014).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  24. Barzi, M., Berenguer, J., Menendez, A., Alvarez-Rodriguez, R. & Pons, S. Sonic-hedgehog-mediated proliferation requires the localization of PKA to the cilium base. J. Cell Sci. 123, 62–69 (2010).

    Article  CAS  PubMed  Google Scholar 

  25. Tuson, M., He, M. & Anderson, K. V. Protein kinase A acts at the basal body of the primary cilium to prevent Gli2 activation and ventralization of the mouse neural tube. Development 138, 4921–4930 (2011).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  26. Marthiens, V. & ffrench-Constant, C. Adherens junction domains are split by asymmetric division of embryonic neural stem cells. EMBO Rep. 10, 515–520 (2009).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  27. Sabherwal, N. et al. The apicobasal polarity kinase aPKC functions as a nuclear determinant and regulates cell proliferation and fate during Xenopus primary neurogenesis. Development 136, 2767–2777 (2009).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  28. Lesage, B., Gutierrez, I., Marti, E. & Gonzalez, C. Neural stem cells: the need for a proper orientation. Curr. Opin. Genet. Dev. 20, 438–442 (2010).

    Article  CAS  PubMed  Google Scholar 

  29. Brand, A. H. & Livesey, F. J. Neural stem cell biology in vertebrates and invertebrates: more alike than different? Neuron 70, 719–729 (2011).

    Article  CAS  PubMed  Google Scholar 

  30. Morin, X. & Bellaiche, Y. Mitotic spindle orientation in asymmetric and symmetric cell divisions during animal development. Dev. Cell 21, 102–119 (2011).

    Article  CAS  PubMed  Google Scholar 

  31. Cruz, C. et al. Foxj1 regulates floor plate cilia architecture and modifies the response of cells to sonic hedgehog signalling. Development 137, 4271–4282 (2010).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  32. Rabadan, M. A. et al. Jagged2 controls the generation of motor neuron and oligodendrocyte progenitors in the ventral spinal cord. Cell Death Differ. 19, 209–219 (2012).

    Article  CAS  PubMed  Google Scholar 

  33. Gillingham, A. K. & Munro, S. The PACT domain, a conserved centrosomal targeting motif in the coiled-coil proteins AKAP450 and pericentrin. EMBO Rep. 1, 524–529 (2000).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  34. Hausken, Z. E., Dell’Acqua, M. L., Coghlan, V. M. & Scott, J. D. Mutational analysis of the A-kinase anchoring protein (AKAP)-binding site on RII. Classification Of side chain determinants for anchoring and isoform selective association with AKAPs. J. Biol. Chem. 271, 29016–29022 (1996).

    Article  CAS  PubMed  Google Scholar 

  35. Peterson, K. A. et al. Neural-specific Sox2 input and differential Gli-binding affinity provide context and positional information in Shh-directed neural patterning. Genes Dev. 26, 2802–2816 (2012).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  36. Nishi, Y. et al. A direct fate exclusion mechanism by Sonic hedgehog-regulated transcriptional repressors. Development 142, 3286–3293 (2015).

    CAS  PubMed  PubMed Central  Google Scholar 

  37. Cohen, M., Kicheva, A. & Ribeiro, A. Ptch1 and Gli regulate Shh signalling dynamics via multiple mechanisms. Nat. Commun. 6, 6709 (2015).

    Article  CAS  PubMed  Google Scholar 

  38. Epstein, D. J., Marti, E., Scott, M. P. & McMahon, A. P. Antagonizing cAMP-dependent protein kinase A in the dorsal CNS activates a conserved Sonic hedgehog signaling pathway. Development 122, 2885–2894 (1996).

    Article  CAS  PubMed  Google Scholar 

  39. Marti, E., Bumcrot, D. A., Takada, R. & McMahon, A. P. Requirement of 19K form of Sonic hedgehog for induction of distinct ventral cell types in CNS explants. Nature 375, 322–325 (1995).

    Article  CAS  PubMed  Google Scholar 

  40. Marti, E., Takada, R., Bumcrot, D. A., Sasaki, H. & McMahon, A. P. Distribution of Sonic hedgehog peptides in the developing chick and mouse embryo. Development 121, 2537–2547 (1995).

    Article  CAS  PubMed  Google Scholar 

  41. Stamataki, D., Ulloa, F., Tsoni, S. V., Mynett, A. & Briscoe, J. A gradient of Gli activity mediates graded Sonic Hedgehog signaling in the neural tube. Genes Dev. 19, 626–641 (2005).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  42. Buchman, J. J. et al. Cdk5rap2 interacts with pericentrin to maintain the neural progenitor pool in the developing neocortex. Neuron 66, 386–402 (2010).

    Article  CAS  PubMed  Google Scholar 

  43. Thornton, G. K. & Woods, C. G. Primary microcephaly: do all roads lead to Rome? Trends Genet. 25, 501–510 (2009).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  44. Alkuraya, F. S. et al. Human mutations in NDE1 cause extreme microcephaly with lissencephaly [corrected]. Am. J. Hum. Genet. 88, 536–547 (2011).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  45. Kosodo, Y. et al. Asymmetric distribution of the apical plasma membrane during neurogenic divisions of mammalian neuroepithelial cells. EMBO J. 23, 2314–2324 (2004).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  46. Peyre, E. & Morin, X. An oblique view on the role of spindle orientation in vertebrate neurogenesis. Dev. Growth Differ. 54, 287–305 (2012).

    Article  CAS  PubMed  Google Scholar 

  47. Das, R. M. & Storey, K. G. Apical abscission alters cell polarity and dismantles the primary cilium during neurogenesis. Science 343, 200–204 (2014).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  48. Hamburger, V. & Hamilton, H. L. A series of normal stages in the development of the chick embryo. J. Morphol. 88, 49–92 (1951).

    Article  CAS  PubMed  Google Scholar 

  49. Caspary, T., Larkins, C. E. & Anderson, K. V. The graded response to Sonic Hedgehog depends on cilia architecture. Dev. Cell 12, 767–778 (2007).

    Article  CAS  PubMed  Google Scholar 

  50. Briscoe, J., Chen, Y., Jessell, T. M. & Struhl, G. A hedgehog-insensitive form of patched provides evidence for direct long-range morphogen activity of sonic hedgehog in the neural tube. Mol. Cell 7, 1279–1291 (2001).

    Article  CAS  PubMed  Google Scholar 

  51. Hynes, M. et al. The seven-transmembrane receptor smoothened cell-autonomously induces multiple ventral cell types. Nat. Neurosci. 3, 41–46 (2000).

    Article  CAS  PubMed  Google Scholar 

  52. Uchikawa, M., Ishida, Y., Takemoto, T., Kamachi, Y. & Kondoh, H. Functional analysis of chicken Sox2 enhancers highlights an array of diverse regulatory elements that are conserved in mammals. Dev. Cell 4, 509–519 (2003).

    Article  CAS  PubMed  Google Scholar 

  53. Sasaki, H., Hui, C., Nakafuku, M. & Kondoh, H. A binding site for Gli proteins is essential for HNF-3β floor plate enhancer activity in transgenics and can respond to Shh in vitro. Development 124, 1313–1322 (1997).

    Article  CAS  PubMed  Google Scholar 

  54. Acquaviva, C. et al. The centrosomal FOP protein is required for cell cycle progression and survival. Cell Cycle 8, 1217–1227 (2009).

    Article  CAS  PubMed  Google Scholar 

  55. Haydar, T. F., Ang, E. Jr & Rakic, P. Mitotic spindle rotation and mode of cell division in the developing telencephalon. Proc. Natl Acad. Sci. USA 100, 2890–2895 (2003).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  56. Roszko, I., Afonso, C., Henrique, D. & Mathis, L. Key role played by RhoA in the balance between planar and apico-basal cell divisions in the chick neuroepithelium. Dev. Biol. 298, 212–224 (2006).

    Article  CAS  PubMed  Google Scholar 

  57. Kosodo, Y. et al. Cytokinesis of neuroepithelial cells can divide their basal process before anaphase. EMBO J. 27, 3151–3163 (2008).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  58. Stamataki, D., Ulloa, F., Tsoni, S. V., Mynett, A. & Briscoe, J. A gradient of Gli activity mediates graded Sonic Hedgehog signaling in the neural tube. Genes Dev. 19, 626–641 (2005).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  59. Sievers, F. et al. Fast, scalable generation of high-quality protein multiple sequence alignments using Clustal Omega. Mol. Syst. Biol. 7, 539 (2011).

    Article  PubMed  PubMed Central  Google Scholar 

  60. Goujon, M. et al. A new bioinformatics analysis tools framework at EMBL-EBI. Nucleic Acids Res. 38, W695–W699 (2010).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  61. Marchler-Bauer, A. et al. CDD: NCBI’s conserved domain database. Nucleic Acids Res. 43, D222–D226 (2015).

    Article  CAS  PubMed  Google Scholar 

Download references

Acknowledgements

The authors are indebted to E. Rebollo for her invaluable technical assistance at the AFMU Facility (IBMB). For providing DNAs, we thank S. McKnight (University of Washington, USA), M. Uchikawa (Osaka University, Japan), M. Götz (Ludwig-Maximilians-University Munich, Germany), H. Lickert (GmbH, ISF, Neuherberg, Germany) and S. Pons (IBMB-CSIC). For providing antibodies, we also thank O. Rosnet (CRCM, Marseille, France), M. Bornens (Institut Curie, Paris, France) and S. Pons (IBMB-CSIC). The monoclonal antibodies were obtained from the Developmental Studies Hybridoma Bank, developed under the auspices of the NICHD and maintained by The University of Iowa, Department of Biological Sciences, Iowa City, Iowa 52242. The work in E.M.’s laboratory was supported by grants BFU2013-46477-P and BFU2014-55738-REDT.

Author information

Authors and Affiliations

Authors

Contributions

M.S. conceived and performed most experiments, analysed the data and discussed results. E.G.-G. contributed to image acquisition, image analysis and quantification, and statistics. R.E. performed the luciferase experiments. S.U. provided technical support to all experiments. E.M. conceived experiments, analysed the data, discussed results and wrote the manuscript.

Corresponding author

Correspondence to Elisa Martí.

Ethics declarations

Competing interests

The authors declare no competing financial interests.

Integrated supplementary information

Supplementary Figure 1 In dividing neuroepithelial cells, the primary cilia was not completely disassembled prior to mitosis.

(a) Transient expression of CEP152-GFP in the chick NT after electroporation (HH14, 16 h post electroporation, hpe) reliably labels the two centrosomes in dividing neural progenitors. CEP152-GFP (green) formed pairs of dots at the spindle poles during mitosis that co-localize with a-Tubulin-GFP (green) and that are immunostained with anti-α-Tubulin (red). DAPI (blue) stains the chromosomes (scale bars, 5 μm). (b) α-Tubulin-GFP (green) electroporation labelled the mitotic spindle. Immunostaining with anti-FOP (FGFR1 Oncogene Partner, red) revealed the centrosome pairs lining the NT lumen, as well as the nucleating mitotic spindles (Scale bars 5 μm). (c) Gli3-HA (red) localizes to the cilium tip (pink arrows) and the nucleus. Acetylated tubulin (green) stain the cilium shaft (yellow arrow) (d) Scheme showing the DNAs and timing of the co-electroporation (hpe = hours post electroporation) (scale bars, are 10 μm and 2 μm respectively). (e) Quantification of the subcellular Arl13b localization types as percentage of total anaphase/telophase H2B-GFP+ mitoses, at two developmental stages, showing that as neurogenesis progresses, the Arl13b-labelled ciliary remnant can lose its attachment to the old mother centriole during mitosis (HH10, n = 30 mitoses; HH14 n = 30 mitoses, from three independent experiments). (f,g) Selected images showing non-centrosomal (f) and centrosomal (g) Arl13b localization; H2B-GFP (green) labels chromosomes, anti-FOP (blue) revealed the centrosome pairs, Arl13b-RFP (red) labels the ciliary remnant. Yellow arrow shows centrosome localization (FOP+) and purple arrow shows ciliary remnant localization (Arl13b+) (Scale bars 5 μm). Images are representative of three independent experiments.

Supplementary Figure 2 PKA localizes to the centrosomes in dividing neural progenitors throughout mitosis.

(a,b) Transient expression of Arl13b-RFP in the chick NT (HH14, 16 hpe) reliably labels the primary cilia: centrosomes labelled with anti-FOP (purple) line the NT lumen; Arl13b-RFP (red) labels the cilia at the NT lumen; anti-FLAG staining (green) labels PKA; DAPI (blue) labels the nuclei (scale bar, 10 μm in a; scale bar, 0,5 μm in b). (c) RII-PKA and Cα-PKA are always co-electroporated in order to inhibit the enzyme’s kinase activity. Co-electroporation at low concentrations of RII-PKA + Cα-PKA was used to study the subcellular localization, and they do not activate Shh transcriptional responses, as assessed by the Gli-BS-Luc reporter activity. Both dnPKA and SmoM2 are assessed as activators of the pathway, and PtcΔ Loop2 is studied as an inhibitor of the pathway. Quantification of the Luc/Renilla activity of the Gli-BS-Luc reporter 24 hpe of the DNAs indicated (plots show the mean ± s.e.m., n = 8 embryos per condition; three independent experiments; one-way ANOVA; P < 0.05, P < 0.001) (scale bars, 5 μm). (di) PKA localizes to the centrosomes at different mitotic phases. RII-PKA + Cα-PKA-FLAG, revealed Cα-PKA by anti-FLAG staining (green) at centrosomes labelled with anti-FOP (red), from prophase to cytokinesis. DAPI (blue) labels the chromosomes. (j,k) Endogenous RII-PKA (green) and Cα-PKA (red) subunits symmetrically localize to centrosomes labelled with anti-FOP (purple), during mitosis. DAPI (blue) labels the chromosomes. (l,m) Endogenous RII-PKA (green) and Cα-PKA (red) subunits asymmetrically localize to centrosomes labelled with anti-FOP (purple), during mitosis. DAPI (blue) labels the chromosomes. (n,o) Endogenous RII-PKA (green) and Cα-PKA (red) subunits co-localize, either symmetrically (n) or asymmetrically (o) to centrosomes labelled with anti-FOP (purple), during mitosis. DAPI (blue) labels the chromosomes (scale bars, 5 μm). Images are representative of three independent experiments.

Supplementary Figure 3 PKA dissociates from centrosomes at the onset of neurogenesis,

(a) Scheme showing the DNAs electroporated and the timing of electroporation. (b) Selected images showing the symmetric centrosomal docking of PKA at the base of the cilium in Gli-BS-RFP+ (red) sister cells. Cα-PKA + RII-PKA-FLAG electroporation revealed Ca-PKA by anti-FLAG staining (green) and the centrosomes were labelled with anti-FOP (purple). (c,d) Selected images showing Gli-BS-RFP cells exiting the ventricular zone in which PKA is distributed in the cytosol but not predominantly associated to the FOP stained centrosome (yellow arrow). (e) Scheme showing the DNAs electroporated, the timing of electroporation, and the area (ROI) selected for fluorescence intensity measurement. (f) Quantification of centrosomal RII-PKA and Ca-PKA in Tis21-, plots show the mean ± s.e.m., Mann–Whitney U test, P < 0.05,P < 0.01, of cumulative fluoresce intensity in both centrosomes (RII-PKA-FLAG mean = 41 ± 9, n = 19 mitoses; Ca-PKA mean = 41 ± 7, n = 20 mitoses) and Tis21 + (RII-PKA-FLAG mean = 20 ± 3,n = 22 mitoses; Ca-PKA mean = 17 ± 2, n = 26 mitoses; from three independent experiments). (g) Scheme showing electroporated cDNAs, and the timing of electroporation. (h) Selected images showing asymmetric centrosomal docking of PKA in pTis21+ sister cells (red). Cα-PKA + RII-PKA-FLAG electroporation revealed Cα-PKA by anti-FLAG staining (green), showing the asymmetric docking of PKA at the centrosomes lining the NT lumen (yellow arrows), labelled with anti-FOP (purple) (scale bar, 0,5 μm). (i) Selected images showing pTis21+ cells exiting the ventricular zone where PKA is distributed in the cytosol and not associated to the FOP (purple) stained centrosomes (yellow arrow). (j) High magnification of the apical area in I, showing the centrosomal duplication (two yellow arrows) in the daughter cell that remains as a progenitor, in which PKA remains docked to the apical centrosomes (scale bars, 10 μm). (k) Scheme showing the area (ROI) selected for fluorescence intensity measurement. (l) Quantification of the endogenous centrosomal RII-PKA and endogenous ninein, plots show the mean ± s.e.m., Mann–Whitney U test, P < 0.01, P < 0.001 of cumulative fluoresce intensity in mother (high ninein) centrosomes (RII-PKA mean = 20, 6 ± 3,5 ninein mean = 9, 2 ± 1,6) and in dauther (low ninein) centrosomes (RII-PKA mean = 5, 8 ± 1, 3 ninein mean = 1, 8 ± 0, 8 n = 11 mitoses) Divisions were analysed from 6 independent embryos in one experiment. Images are representative of three independent experiments.

Supplementary Figure 4 Neurogenesis correlates with asymmetric inheritance of apical membrane domains.

(a) Scheme showing the split of AJs by the cleavage plane at anaphase, the cleavage plane being deduced by a line bisecting the two sets of condensed chromatin (blue plates). When the rectangles were oriented parallel to each other, the cleavage plane was positioned half way in between the rectangles (d1 = d2). When the rectangles were oriented at an angle to each other, the cleavage plane was positioned such that this angle was halved (a1 = a2), predicting the type of division made by distributing the N-cadherin hole (red outline) between the two daughter cells, and by partitioning the apical aPKC domain (green). (b) Quantification of the Rfi between inherited αPKC apical domains in mitotic cells at two developmental stages, where the lines and error bars correspond to the median ± s.e.m.; three independent experiments; Mann–Whitney U test; P < 0.001. The partitioning of αPKC was largely symmetric at 54 hpf; n = 32 mitosis), while it was asymmetric at the neurogenic phase 70 hpf (n = 42 mitosis). (c) Example of a symmetric mitosis in which the cleavage plane is deduced by drawing perpendicular lines (dashed line) bisecting the two sets of condensed chromatin (outlined). DAPI (blue) labels the chromosomes, N-cadherin is labelled in red, αPKC in green. (d) Example of an asymmetric mitosis (scale bars, 5 μm). (e) Scheme showing the DNAs electroporated and the timing of electroporation. (f) Example of two cells in anaphase and telophase in which the cleavage plane is deduced by drawing perpendicular lines (dashed line) bisecting the two sets of condensed chromatin (outlined). Symmetric partitioning of αPKC is correlated with Gli-BS activation (RFP+). GFP represents the control electroporation, FOP (purple) labels the centrosomes, DAPI (blue) labels the chromosomes (yellow arrow points to the N-cadherin hole). Black and white panel shows the isolation of αPKC domain for quantification. (g) Example of two cells in anaphase and telophase, in which asymmetric partitioning of αPKC is correlated with Gli-BS inactivation (RFP, yellow arrow points to the N-cadherin hole) (scale bars, 5 μm). (h) Scheme showing the DNAs electroporated and the timing of electroporation. (i) Example of two cells in telophase, in which asymmetric partitioning of αPKC is correlated with asymmetric centrosomal docking of RII-PKA. FOP (magenta) labels the centrosomes (magenta arrows point to centrosomes). RFP (red) labels the chromosomes (yellow arrow points to the N-cadherin hole)(Scale bars 5 μm). Images are representative of three independent experiments.

Supplementary Figure 5 Pericentrin mediates PKA docking to the centrosomes.

(a) Semi-quantitative PCR analysis of the AKAP9 transcripts expressed in control versus SmoM2-EP cells (plot shows the mean ± s.e.m., 25.000 cells from n = 8 independent embryos, three independent experiments, Mann–Whitney U test; NS). (b) Scheme showing the conservation of the PCNT (pericentrin) sequence, highlighting the RII-PKA-binding domain (blue) and the PCNT-AKAP9 Centrosomal Targeting (PACT) domain (purple). (c) Multiple sequence alignment of the RII-PKA binding domain of PCNT using Multaling version 5.4.1: red highlights the conserved leucines (L) critical for RII-PKA binding. (d) Multiple sequence alignment highlighting the conserved PACT binding domain in PCNT. (e) Selected images of RIIΔ2-6-PKA electroporation at mitotic phases: CEP152 (red) labels centrosomes (yellow arrows); DAPI (blue) labels chromosomes. Mutant RII-PKA (green) does not associate with the centrosomes at any phase of mitosis (scale bars, 5 μm Images are representative of three independent experiments.

Supplementary information

Supplementary Information

Supplementary Information (PDF 43620 kb)

Supplementary Information

Supplementary Information (XLSX 13 kb)

Supplementary Information

Supplementary Information (XLSX 12 kb)

Supplementary Information

Supplementary Information (XLSX 517 kb)

3D reconstruction of symmetric centrosomal docking of PKA in a Gli-BS-RFP+ mitoses.

Gli-BS-RFP+ mitosis (red) show symmetric RII-PKA/Cα-PKA-FLAG, revealed Cα-PKA by anti-FLAG staining (green) in centrosomes labelled with anti-FOP (purple). DAPI (blue) labels the chromosomes and yellow arrows point to PKA. (MP4 1798 kb)

3D reconstruction of asymmetric centrosomal docking of PKA in a pTis21-RFP+ mitoses.

pTis21-RFP+ mitoses (red) show asymmetric RII-PKA/Cα-PKA-FLAG, revealed Cα-PKA by anti-FLAG staining (green) in centrosomes labelled with anti-FOP (purple). DAPI (blue) labels the chromosomes and yellow arrows point to PKA. (MP4 4395 kb)

Rights and permissions

Reprints and permissions

About this article

Check for updates. Verify currency and authenticity via CrossMark

Cite this article

Saade, M., Gonzalez-Gobartt, E., Escalona, R. et al. Shh-mediated centrosomal recruitment of PKA promotes symmetric proliferative neuroepithelial cell division. Nat Cell Biol 19, 493–503 (2017). https://doi.org/10.1038/ncb3512

Download citation

  • Received:

  • Accepted:

  • Published:

  • Issue Date:

  • DOI: https://doi.org/10.1038/ncb3512

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