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.

  • Review Article
  • Published:

A gene regulatory network orchestrates neural crest formation

Key Points

  • The neural crest (NC) is a multipotent, migratory cell population that is unique to vertebrate embryos and that forms numerous derivatives ranging from melanocytes, peripheral neurons and glia, to bone and cartilage of the face. A multimodule gene regulatory network mediates the complex process of NC formation, involving signalling events and upregulation of transcription factors that regulate one another and effector genes that are involved in migration and differentiation.

  • Initial induction of the NC occurs at the neural plate border of the gastrula and is mediated by the combination of Wnt, bone morphogenetic protein (BMP) and fibroblast growth factor (FGF) signalling. These signals control the gene expression of transcription factors, such as Msx1, Msx2, Zic1, Pax3 and Pax7, which establish the competence of the neural plate border territory to respond to NC inducing signals.

  • Neural plate border genes in turn regulate NC specifier genes that maintain the NC progenitor pool by controlling the cell cycle, conferring survival properties to newly induced NC cells, controlling their emigration from the neural tube and migration along well-established pathways, and by regulating cell-fate decisions and events of terminal differentiation.

  • Bona fide NC cells delaminate from the neural tube once they have undergone an epithelial to mesenchymal transition and then embark on extensive migrations to sometimes distant sites in the periphery, where they settle and differentiate into various derivatives. NC specifier genes are thought to directly control these events by regulating various downstream-effector genes.

  • NC cells interact with and modify their environments during migration. They respond to diverse guidance cues, including ephrins and semaphorins, that help define their migratory pathways.

  • The NC gene regulatory network underlying the complex process of NC formation was present in the last common ancestor of jawless and jawed vertebrates and has been tightly conserved for over 500 million years.

Abstract

The neural crest is a multipotent, migratory cell population that is unique to vertebrate embryos and gives rise to many derivatives, ranging from the peripheral nervous system to the craniofacial skeleton and pigment cells. A multimodule gene regulatory network mediates the complex process of neural crest formation, which involves the early induction and maintenance of the precursor pool, emigration of the neural crest progenitors from the neural tube via an epithelial to mesenchymal transition, migration of progenitor cells along distinct pathways and overt differentiation into diverse cell types. Here, we review our current understanding of these processes and discuss the molecular players that are involved in the neural crest gene regulatory network.

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: Regulatory steps in neural crest formation.
Figure 2: Neural crest specification.

Similar content being viewed by others

References

  1. Scherson, T., Serbedzija, G., Fraser, S. & Bronner-Fraser, M. Regulative capacity of the cranial neural tube to form neural crest. Development 118, 1049–1062 (1993).

    Article  CAS  PubMed  Google Scholar 

  2. Hunt, P., Ferretti, P., Krumlauf, R. & Thorogood, P. Restoration of normal Hox code and branchial arch morphogenesis after extensive deletion of hindbrain neural crest. Dev. Biol. 168, 584–597 (1995).

    Article  CAS  PubMed  Google Scholar 

  3. Sechrist, J., Nieto, M. A., Zamanian, R. T. & Bronner-Fraser, M. Regulative response of the cranial neural tube after neural fold ablation: spatiotemporal nature of neural crest regeneration and up-regulation of Slug. Development 121, 4103–4115 (1995).

    Article  CAS  PubMed  Google Scholar 

  4. Couly, G., Grapin-Botton, A., Coltey, P. & Le Douarin, N. M. The regeneration of the cephalic neural crest, a problem revisited: the regenerating cells originate from the contralateral or from the anterior and posterior neural fold. Development 122, 3393–3407 (1996).

    Article  CAS  PubMed  Google Scholar 

  5. Crane, J. F. & Trainor, P. A. Neural crest stem and progenitor cells. Annu. Rev. Cell Dev. Biol. 22, 267–286 (2006).

    Article  CAS  PubMed  Google Scholar 

  6. Hall, B. K. The neural crest as a fourth germ layer and vertebrates as quadroblastic not triploblastic. Evol. Dev. 2, 3–5 (2000).

    Article  CAS  PubMed  Google Scholar 

  7. Gans, C. & Northcutt, R. G. Neural crest and the origin of vertebrates: a new head. Science 220, 268–273 (1983).

    Article  CAS  PubMed  Google Scholar 

  8. Hall, B. K. Hörstadius, S. The Neural Crest (Oxford University Press, Oxford, 1988).

    Google Scholar 

  9. Le Douarin, N., Kalcheim, C. The Neural Crest. 2nd edn (Cambridge University Press, New York, 1999).

    Book  Google Scholar 

  10. Meulemans, D. & Bronner-Fraser, M. Gene-regulatory interactions in neural crest evolution and development. Dev. Cell 7, 291–299 (2004).

    Article  CAS  PubMed  Google Scholar 

  11. Wawersik, S., Evola, C. & Whitman, M. Conditional BMP inhibition in Xenopus reveals stage-specific roles for BMPs in neural and neural crest induction. Dev. Biol. 277, 425–442 (2005).

    Article  CAS  PubMed  Google Scholar 

  12. Raven, C. P. & Kloos, J. Induction by medial and lateral pieces of the archenteron roof with special reference to the determination of the neural crest. Acta Neerl Morphol. 5, 348–362 (1945).

    Google Scholar 

  13. Marchant, L., Linker, C., Ruiz, P., Guerrero, N. & Mayor, R. The inductive properties of mesoderm suggest that the neural crest cells are specified by a BMP gradient. Dev. Biol. 198, 319–329 (1998).

    Article  CAS  PubMed  Google Scholar 

  14. LaBonne, C. & Bronner-Fraser, M. Neural crest induction in Xenopus: evidence for a two-signal model. Development 125, 2403–2414 (1998).

    Article  CAS  PubMed  Google Scholar 

  15. Garcia-Castro, M. I., Marcelle, C. & Bronner-Fraser, M. Ectodermal Wnt function as a neural crest inducer. Science 297, 848–851 (2002).

    Article  CAS  PubMed  Google Scholar 

  16. Mayor, R., Morgan, R. & Sargent, M. G. Induction of the prospective neural crest of Xenopus. Development 121, 767–777 (1995).

    Article  CAS  PubMed  Google Scholar 

  17. Mayor, R., Guerrero, N. & Martinez, C. Role of FGF and noggin in neural crest induction. Dev. Biol. 189, 1–12 (1997).

    Article  CAS  PubMed  Google Scholar 

  18. Monsoro-Burq, A. H., Fletcher, R. B. & Harland, R. M. Neural crest induction by paraxial mesoderm in Xenopus embryos requires FGF signals. Development 130, 3111–3124 (2003).

    Article  CAS  PubMed  Google Scholar 

  19. Selleck, M. A., Garcia-Castro, M. I., Artinger, K. B. & Bronner-Fraser, M. Effects of Shh and Noggin on neural crest formation demonstrate that BMP is required in the neural tube but not ectoderm. Development 125, 4919–4930 (1998).

    Article  CAS  PubMed  Google Scholar 

  20. Endo, Y., Osumi, N. & Wakamatsu, Y. Bimodal functions of Notch-mediated signaling are involved in neural crest formation during avian ectoderm development. Development 129, 863–873 (2002).

    Article  CAS  PubMed  Google Scholar 

  21. Glavic, A., Silva, F., Aybar, M. J., Bastidas, F. & Mayor, R. Interplay between Notch signaling and the homeoprotein Xiro1 is required for neural crest induction in Xenopus embryos. Development 131, 347–359 (2004).

    Article  CAS  PubMed  Google Scholar 

  22. Saint-Jeannet, J. P., He, X., Varmus, H. E. & Dawid, I. B. Regulation of dorsal fate in the neuraxis by Wnt-1 and Wnt-3a. Proc. Natl Acad. Sci. USA 94, 13713–13718 (1997).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  23. Bang, A. G., Papalopulu, N., Goulding, M. D. & Kintner, C. Expression of Pax-3 in the lateral neural plate is dependent on a Wnt-mediated signal from posterior nonaxial mesoderm. Dev. Biol. 212, 366–380 (1999).

    Article  CAS  PubMed  Google Scholar 

  24. Deardorff, M. A., Tan, C., Saint-Jeannet, J. P. & Klein, P. S. A role for frizzled 3 in neural crest development. Development 128, 3655–3663 (2001).

    Article  CAS  PubMed  Google Scholar 

  25. Lewis, J. L. et al. Reiterated Wnt signaling during zebrafish neural crest development. Development 131, 1299–1308 (2004).

    Article  CAS  PubMed  Google Scholar 

  26. Wu, J., Yang, J. & Klein, P. S. Neural crest induction by the canonical Wnt pathway can be dissociated from anterior–posterior neural patterning in Xenopus. Dev. Biol. 279, 220–232 (2005).

    Article  CAS  PubMed  Google Scholar 

  27. Jones, N. C. & Trainor, P. A. Role of morphogens in neural crest cell determination. J. Neurobiol. 64, 388–404 (2005).

    Article  CAS  PubMed  Google Scholar 

  28. Mansour, S. L., Goddard, J. M. & Capecchi, M. R. Mice homozygous for a targeted disruption of the proto-oncogene int-2 have developmental defects in the tail and inner ear. Development 117, 13–28 (1993).

    Article  CAS  PubMed  Google Scholar 

  29. Yamaguchi, T. P., Harpal, K., Henkemeyer, M. & Rossant, J. fgfr-1 is required for embryonic growth and mesodermal patterning during mouse gastrulation. Genes Dev. 8, 3032–3044 (1994).

    Article  CAS  PubMed  Google Scholar 

  30. Yamaguchi, T. P. & Rossant, J. Fibroblast growth factors in mammalian development. Curr. Opin. Genet. Dev. 5, 485–491 (1995).

    Article  CAS  PubMed  Google Scholar 

  31. Meyers, E. N., Lewandoski, M. & Martin, G. R. An Fgf8 mutant allelic series generated by Cre- and Flp-mediated recombination. Nature Genet. 18, 136–141 (1998).

    CAS  PubMed  Google Scholar 

  32. Reifers, F. et al. Fgf8 is mutated in zebrafish acerebellar (ace) mutants and is required for maintenance of midbrain–hindbrain boundary development and somitogenesis. Development 125, 2381–2395 (1998).

    Article  CAS  PubMed  Google Scholar 

  33. Sauka-Spengler, T., Meulemans, D., Jones, M. & Bronner-Fraser, M. Ancient evolutionary origin of the neural crest gene regulatory network. Dev. Cell 13, 405–420 (2007). This study tests the NC GRN in a collective fashion in a single vertebrate for the first time. The results demonstrate that the core of the network was present in the last common ancestor of jawless and jawed vertebrates and has been tightly conserved for over 500 million years.

    Article  CAS  PubMed  Google Scholar 

  34. Tribulo, C., Aybar, M. J., Nguyen, V. H., Mullins, M. C. & Mayor, R. Regulation of Msx genes by a Bmp gradient is essential for neural crest specification. Development 130, 6441–6452 (2003).

    Article  CAS  PubMed  Google Scholar 

  35. Monsoro-Burq, A. H., Wang, E. & Harland, R. Msx1 and Pax3 cooperate to mediate FGF8 and WNT signals during Xenopus neural crest induction. Dev. Cell 8, 167–178 (2005). This is the first study that shows parts of the regulatory cascade at the neural plate border. The authors show that Pax3 acts downstream of Msx1, and in synergistic action with Zic1, it induces Slug in a Wnt-dependent manner.

    Article  CAS  PubMed  Google Scholar 

  36. Sato, T., Sasai, N. & Sasai, Y. Neural crest determination by co-activation of Pax3 and Zic1 genes in Xenopus ectoderm. Development 132, 2355–2363 (2005). This work describes part of the NC regulatory cascade and demonstrates that Pax3 and Zic1 are essential for NC specification. Their concerted activity induces Slug and FoxD3 activity in a Wnt-dependent manner.

    Article  CAS  PubMed  Google Scholar 

  37. Litsiou, A., Hanson, S. & Streit, A. A balance of FGF, BMP and WNT signalling positions the future placode territory in the head. Development 132, 4051–4062 (2005).

    Article  CAS  PubMed  Google Scholar 

  38. Hong, C. S. & Saint-Jeannet, J. P. The activity of Pax3 and Zic1 regulates three distinct cell fates at the neural plate border. Mol. Biol. Cell 18, 2192–2202 (2007).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  39. Fuentealba, L. C. et al. Integrating patterning signals: Wnt/GSK3 regulates the duration of the BMP/Smad1 signal. Cell 131, 980–993 (2007).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  40. Vallin, J. et al. Cloning and characterization of three Xenopus Slug promoters reveal direct regulation by Lef/β-catenin signaling. J. Biol. Chem. 276, 30350–30358 (2001).

    Article  CAS  PubMed  Google Scholar 

  41. Sakai, D. et al. Regulation of Slug transcription in embryonic ectoderm by β-catenin-Lef/Tcf and BMP–Smad signaling. Dev. Growth Differ. 47, 471–482 (2005).

    Article  CAS  PubMed  Google Scholar 

  42. Werner, T., Hammer, A., Wahlbuhl, M., Bosl, M. R. & Wegner, M. Multiple conserved regulatory elements with overlapping functions determine Sox10 expression in mouse embryogenesis. Nucleic Acids Res. 35, 6526–6538 (2007).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  43. Bellmeyer, A., Krase, J., Lindgren, J. & LaBonne, C. The protooncogene c-Myc is an essential regulator of neural crest formation in Xenopus. Dev. Cell 4, 827–839 (2003).

    Article  CAS  PubMed  Google Scholar 

  44. Kee, Y. & Bronner-Fraser, M. To proliferate or to die: role of Id3 in cell cycle progression and survival of neural crest progenitors. Genes Dev. 19, 744–755 (2005).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  45. Light, W., Vernon, A. E., Lasorella, A., Iavarone, A. & LaBonne, C. Xenopus Id3 is required downstream of Myc for the formation of multipotent neural crest progenitor cells. Development 132, 1831–1841 (2005). References 43–45 show that the c-Myc–Id3 gene battery functions as a cell-cycle control switch, balancing cell proliferation versus cell death, and in cell-fate determination of early NC precursors.

    Article  CAS  PubMed  Google Scholar 

  46. Luo, T., Lee, Y. H., Saint-Jeannet, J. P. & Sargent, T. D. Induction of neural crest in Xenopus by transcription factor AP2α. Proc. Natl Acad. Sci. USA 100, 532–537 (2003).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  47. Panda, D. K., Miao, D., Lefebvre, V., Hendy, G. N. & Goltzman, D. The transcription factor SOX9 regulates cell cycle and differentiation genes in chondrocytic CFK2 cells. J. Biol. Chem. 276, 41229–41236 (2001).

    Article  CAS  PubMed  Google Scholar 

  48. Cheung, M. et al. The transcriptional control of trunk neural crest induction, survival, and delamination. Dev. Cell 8, 179–192 (2005).

    Article  CAS  PubMed  Google Scholar 

  49. Yan, Y. L. et al. A pair of Sox: distinct and overlapping functions of zebrafish Sox9 co-orthologs in craniofacial and pectoral fin development. Development 132, 1069–1083 (2005).

    Article  CAS  PubMed  Google Scholar 

  50. Thiery, J. P. & Sleeman, J. P. Complex networks orchestrate epithelial–mesenchymal transitions. Nature Rev. Mol. Cell Biol. 7, 131–142 (2006).

    Article  CAS  Google Scholar 

  51. Batlle, E. et al. The transcription factor Snail is a repressor of E-cadherin gene expression in epithelial tumour cells. Nature Cell Biol. 2, 84–89 (2000).

    Article  CAS  PubMed  Google Scholar 

  52. Cano, A. et al. The transcription factor Snail controls epithelial–mesenchymal transitions by repressing E-cadherin expression. Nature Cell Biol. 2, 76–83 (2000).

    Article  CAS  PubMed  Google Scholar 

  53. Nieto, M. A. The Snail superfamily of zinc-finger transcription factors. Nature Rev. Mol. Cell Biol. 3, 155–166 (2002).

    Article  CAS  Google Scholar 

  54. Ikenouchi, J., Matsuda, M., Furuse, M. & Tsukita, S. Regulation of tight junctions during the epithelium–mesenchyme transition: direct repression of the gene expression of claudins/occludin by Snail. J. Cell Sci. 116, 1959–1967 (2003).

    Article  CAS  PubMed  Google Scholar 

  55. Taneyhill, L. A., Coles, E. G. & Bronner-Fraser, M. Snail2 directly represses cadherin6B during epithelial-to-mesenchymal transitions of the neural crest. Development 134, 1481–1490 (2007).

    Article  CAS  PubMed  Google Scholar 

  56. Hatta, K., Takagi, S., Fujisawa, H. & Takeichi, M. Spatial and temporal expression pattern of N-cadherin cell adhesion molecules correlated with morphogenetic processes of chicken embryos. Dev. Biol. 120, 215–227 (1987).

    Article  CAS  PubMed  Google Scholar 

  57. Nakagawa, S. & Takeichi, M. Neural crest cell–cell adhesion controlled by sequential and subpopulation-specific expression of novel cadherins. Development 121, 1321–1332 (1995).

    Article  CAS  PubMed  Google Scholar 

  58. Chu, Y. S. et al. Prototypical type I E-cadherin and type II cadherin-7 mediate very distinct adhesiveness through their extracellular domains. J. Biol. Chem. 281, 2901–2910 (2006).

    Article  CAS  PubMed  Google Scholar 

  59. Hadeball, B., Borchers, A. & Wedlich, D. Xenopus cadherin-11 (Xcadherin-11) expression requires the Wg/Wnt signal. Mech. Dev. 72, 101–113 (1998).

    Article  CAS  PubMed  Google Scholar 

  60. Nakagawa, S. & Takeichi, M. Neural crest emigration from the neural tube depends on regulated cadherin expression. Development 125, 2963–2971 (1998).

    Article  CAS  PubMed  Google Scholar 

  61. Shin, K., Fogg, V. C. & Margolis, B. Tight junctions and cell polarity. Annu. Rev. Cell Dev. Biol. 22, 207–235 (2006).

    Article  CAS  PubMed  Google Scholar 

  62. Aaku-Saraste, E., Hellwig, A. & Huttner, W. B. Loss of occludin and functional tight junctions, but not ZO-1, during neural tube closure—remodeling of the neuroepithelium prior to neurogenesis. Dev. Biol. 180, 664–679 (1996).

    Article  CAS  PubMed  Google Scholar 

  63. Shook, D. & Keller, R. Mechanisms, mechanics and function of epithelial-mesenchymal transitions in early development. Mech. Dev. 120, 1351–1383 (2003).

    Article  CAS  PubMed  Google Scholar 

  64. Lo, C. W. et al. Cx43 gap junction gene expression and gap junctional communication in mouse neural crest cells. Dev. Genet. 20, 119–132 (1997).

    Article  CAS  PubMed  Google Scholar 

  65. Reaume, A. G. et al. Cardiac malformation in neonatal mice lacking connexin43. Science 267, 1831–1834 (1995).

    Article  CAS  PubMed  Google Scholar 

  66. Huang, G. Y. et al. Gap junction-mediated cell–cell communication modulates mouse neural crest migration. J. Cell Biol. 143, 1725–1734 (1998).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  67. Xu, X., Francis, R., Wei, C. J., Linask, K. L. & Lo, C. W. Connexin 43-mediated modulation of polarized cell movement and the directional migration of cardiac neural crest cells. Development 133, 3629–3639 (2006).

    Article  CAS  PubMed  Google Scholar 

  68. Liu, S. et al. Distinct cardiac malformations caused by absence of connexin 43 in the neural crest and in the non-crest neural tube. Development 133, 2063–2073 (2006).

    Article  CAS  PubMed  Google Scholar 

  69. Egeblad, M. & Werb, Z. New functions for the matrix metalloproteinases in cancer progression. Nature Rev. Cancer 2, 161–174 (2002).

    Article  CAS  Google Scholar 

  70. Chang, C. & Werb, Z. The many faces of metalloproteases: cell growth, invasion, angiogenesis and metastasis. Trends Cell Biol. 11, S37–43 (2001).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  71. Cai, D. H., Vollberg, T. M. Sr, Hahn-Dantona, E., Quigley, J. P. & Brauer, P. R. MMP-2 expression during early avian cardiac and neural crest morphogenesis. Anat. Rec. 259, 168–179 (2000).

    Article  CAS  PubMed  Google Scholar 

  72. Cai, D. H. & Brauer, P. R. Synthetic matrix metalloproteinase inhibitor decreases early cardiac neural crest migration in chicken embryos. Dev. Dyn. 224, 441–449 (2002).

    Article  CAS  PubMed  Google Scholar 

  73. Huh, M. I. et al. Roles of MMP/TIMP in regulating matrix swelling and cell migration during chick corneal development. J. Cell Biochem. 101, 1222–1237 (2007).

    Article  CAS  PubMed  Google Scholar 

  74. Alfandari, D. et al. Xenopus ADAM 13 is a metalloprotease required for cranial neural crest-cell migration. Curr. Biol. 11, 918–930 (2001).

    Article  CAS  PubMed  Google Scholar 

  75. Kee, Y., Hwang, B. J., Sternberg, P. W. & Bronner-Fraser, M. Evolutionary conservation of cell migration genes: from nematode neurons to vertebrate neural crest. Genes Dev. 21, 391–396 (2007).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  76. Lallier, T. & Bronner-Fraser, M. α1β1 integrin on neural crest cells recognizes some laminin substrata in a Ca2+-independent manner. J. Cell Biol. 119, 1335–1345 (1992).

    Article  CAS  PubMed  Google Scholar 

  77. Lallier, T. & Bronner-Fraser, M. Inhibition of neural crest cell attachment by integrin antisense oligonucleotides. Science 259, 692–695 (1993).

    Article  CAS  PubMed  Google Scholar 

  78. Delannet, M. et al. Specific roles of the α Vβ1, αVβ3 and αVβ5 integrins in avian neural crest cell adhesion and migration on vitronectin. Development 120, 2687–2702 (1994).

    Article  CAS  PubMed  Google Scholar 

  79. Kil, S. H., Lallier, T. & Bronner-Fraser, M. Inhibition of cranial neural crest adhesion in vitro and migration in vivo using integrin antisense oligonucleotides. Dev. Biol. 179, 91–101 (1996).

    Article  CAS  PubMed  Google Scholar 

  80. Alfandari, D., Cousin, H., Gaultier, A., Hoffstrom, B. G. & DeSimone, D. W. Integrin α5β1 supports the migration of Xenopus cranial neural crest on fibronectin. Dev. Biol. 260, 449–464 (2003).

    Article  CAS  PubMed  Google Scholar 

  81. Strachan, L. R. & Condic, M. L. Cranial neural crest recycle surface integrins in a substratum-dependent manner to promote rapid motility. J. Cell Biol. 167, 545–554 (2004).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  82. Burstyn-Cohen, T. & Kalcheim, C. Association between the cell cycle and neural crest delamination through specific regulation of G1/S transition. Dev. Cell 3, 383–395 (2002).

    Article  CAS  PubMed  Google Scholar 

  83. Vega, S. et al. Snail blocks the cell cycle and confers resistance to cell death. Genes Dev. 18, 1131–1143 (2004).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  84. Burstyn-Cohen, T., Stanleigh, J., Sela-Donenfeld, D. & Kalcheim, C. Canonical Wnt activity regulates trunk neural crest delamination linking BMP/noggin signaling with G1/S transition. Development 131, 5327–5339 (2004). This work, along with reference 82, shows that trunk NC cells undergo cell-cycle arrest upon delamination. This process is controlled by BMP-dependent Wnt signalling.

    Article  CAS  PubMed  Google Scholar 

  85. Theveneau, E., Duband, J. L. & Altabef, M. Ets-1 confers cranial features on neural crest delamination. PLoS ONE 2, e1142 (2007).

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  86. Kulesa, P. M. & Fraser, S. E. In ovo time-lapse analysis of chick hindbrain neural crest cell migration shows cell interactions during migration to the branchial arches. Development 127, 1161–1172 (2000).

    Article  CAS  PubMed  Google Scholar 

  87. Teddy, J. M. & Kulesa, P. M. In vivo evidence for short- and long-range cell communication in cranial neural crest cells. Development 131, 6141–6151 (2004).

    Article  CAS  PubMed  Google Scholar 

  88. Kasemeier-Kulesa, J. C., Kulesa, P. M. & Lefcort, F. Imaging neural crest cell dynamics during formation of dorsal root ganglia and sympathetic ganglia. Development 132, 235–245 (2005).

    Article  CAS  PubMed  Google Scholar 

  89. McLennan, R. & Kulesa, P. M. In vivo analysis reveals a critical role for neuropilin-1 in cranial neural crest cell migration in chick. Dev. Biol. 301, 227–239 (2007).

    Article  CAS  PubMed  Google Scholar 

  90. Krull, C. E. et al. Interactions of Eph-related receptors and ligands confer rostrocaudal pattern to trunk neural crest migration. Curr. Biol. 7, 571–580 (1997).

    Article  CAS  PubMed  Google Scholar 

  91. Smith, A., Robinson, V., Patel, K. & Wilkinson, D. G. The EphA4 and EphB1 receptor tyrosine kinases and ephrin-B2 ligand regulate targeted migration of branchial neural crest cells. Curr. Biol. 7, 561–570 (1997).

    Article  CAS  PubMed  Google Scholar 

  92. Wang, H. U. & Anderson, D. J. Eph family transmembrane ligands can mediate repulsive guidance of trunk neural crest migration and motor axon outgrowth. Neuron 18, 383–396 (1997).

    Article  CAS  PubMed  Google Scholar 

  93. Santiago, A. & Erickson, C. A. Ephrin-B ligands play a dual role in the control of neural crest cell migration. Development 129, 3621–3632 (2002).

    Article  CAS  PubMed  Google Scholar 

  94. De Bellard, M. E., Rao, Y. & Bronner-Fraser, M. Dual function of Slit2 in repulsion and enhanced migration of trunk, but not vagal, neural crest cells. J. Cell Biol. 162, 269–279 (2003).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  95. Jia, L., Cheng, L. & Raper, J. Slit/Robo signaling is necessary to confine early neural crest cells to the ventral migratory pathway in the trunk. Dev. Biol. 282, 411–421 (2005).

    Article  CAS  PubMed  Google Scholar 

  96. Gammill, L. S., Gonzalez, C., Gu, C. & Bronner-Fraser, M. Guidance of trunk neural crest migration requires neuropilin 2/semaphorin 3F signaling. Development 133, 99–106 (2006).

    Article  CAS  PubMed  Google Scholar 

  97. Davy, A., Aubin, J. & Soriano, P. Ephrin-B1 forward and reverse signaling are required during mouse development. Genes Dev. 18, 572–583 (2004).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  98. Gammill, L. S., Gonzalez, C. & Bronner-Fraser, M. Neuropilin 2/semaphorin 3F signaling is essential for cranial neural crest migration and trigeminal ganglion condensation. J. Neurobiol. 67, 47–56 (2006).

    Article  CAS  Google Scholar 

  99. Osborne, N. J., Begbie, J., Chilton, J. K., Schmidt, H. & Eickholt, B. J. Semaphorin/neuropilin signaling influences the positioning of migratory neural crest cells within the hindbrain region of the chick. Dev. Dyn. 232, 939–949 (2005).

    Article  CAS  PubMed  Google Scholar 

  100. Yu, H. H. & Moens, C. B. Semaphorin signaling guides cranial neural crest cell migration in zebrafish. Dev. Biol. 280, 373–385 (2005).

    Article  CAS  PubMed  Google Scholar 

  101. Young, H. M. et al. GDNF is a chemoattractant for enteric neural cells. Dev. Biol. 229, 503–516 (2001).

    Article  CAS  PubMed  Google Scholar 

  102. Asai, N. et al. Targeted mutation of serine 697 in the Ret tyrosine kinase causes migration defect of enteric neural crest cells. Development 133, 4507–4516 (2006).

    Article  CAS  PubMed  Google Scholar 

  103. Anderson, R. B. et al. Effects of different regions of the developing gut on the migration of enteric neural crest-derived cells: a role for Sema3A, but not Sema3F. Dev. Biol. 305, 287–299 (2007).

    Article  CAS  PubMed  Google Scholar 

  104. Lefebvre, V., Huang, W., Harley, V. R., Goodfellow, P. N. & de Crombrugghe, B. SOX9 is a potent activator of the chondrocyte-specific enhancer of the Proα1(II) collagen gene. Mol. Cell Biol. 17, 2336–2346 (1997).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  105. Kelsh, R. N. Sorting out Sox10 functions in neural crest development. Bioessays 28, 788–798 (2006).

    Article  PubMed  Google Scholar 

  106. Bondurand, N. et al. Interaction among SOX10, PAX3 and MITF, three genes altered in Waardenburg syndrome. Hum. Mol. Genet. 9, 1907–1917 (2000).

    Article  CAS  PubMed  Google Scholar 

  107. Potterf, S. B. et al. Analysis of SOX10 function in neural crest-derived melanocyte development: SOX10-dependent transcriptional control of dopachrome tautomerase. Dev. Biol. 237, 245–257 (2001).

    Article  CAS  PubMed  Google Scholar 

  108. Elworthy, S., Lister, J. A., Carney, T. J., Raible, D. W. & Kelsh, R. N. Transcriptional regulation of mitfa accounts for the sox10 requirement in zebrafish melanophore development. Development 130, 2809–2818 (2003).

    Article  CAS  PubMed  Google Scholar 

  109. Huber, W. E. et al. A tissue-restricted cAMP transcriptional response: SOX10 modulates alpha-melanocyte-stimulating hormone-triggered expression of microphthalmia-associated transcription factor in melanocytes. J. Biol. Chem. 278, 45224–45230 (2003).

    Article  CAS  PubMed  Google Scholar 

  110. Ludwig, A., Rehberg, S. & Wegner, M. Melanocyte-specific expression of dopachrome tautomerase is dependent on synergistic gene activation by the Sox10 and Mitf transcription factors. FEBS Lett. 556, 236–244 (2004).

    Article  CAS  PubMed  Google Scholar 

  111. Kim, J., Lo, L., Dormand, E. & Anderson, D. J. SOX10 maintains multipotency and inhibits neuronal differentiation of neural crest stem cells. Neuron 38, 17–31 (2003). This study shows that Sox10 controls sympathetic neuron fate during peripheral nervous system development.

    Article  CAS  PubMed  Google Scholar 

  112. Sonnenberg-Riethmacher, E. et al. Development and degeneration of dorsal root ganglia in the absence of the HMG-domain transcription factor Sox10. Mech. Dev. 109, 253–265 (2001).

    Article  CAS  PubMed  Google Scholar 

  113. Peirano, R. I., Goerich, D. E., Riethmacher, D. & Wegner, M. Protein zero gene expression is regulated by the glial transcription factor Sox10. Mol. Cell Biol. 20, 3198–3209 (2000).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  114. Stolt, C. C. et al. Terminal differentiation of myelin-forming oligodendrocytes depends on the transcription factor Sox10. Genes Dev. 16, 165–170 (2002).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  115. Bondurand, N. et al. Human Connexin 32, a gap junction protein altered in the X-linked form of Charcot–Marie–Tooth disease, is directly regulated by the transcription factor SOX10. Hum. Mol. Genet. 10, 2783–2795 (2001).

    Article  CAS  PubMed  Google Scholar 

  116. Schlierf, B., Werner, T., Glaser, G. & Wegner, M. Expression of connexin47 in oligodendrocytes is regulated by the Sox10 transcription factor. J. Mol. Biol. 361, 11–21 (2006).

    Article  CAS  PubMed  Google Scholar 

  117. Lang, D. et al. Pax3 is required for enteric ganglia formation and functions with Sox10 to modulate expression of c-Ret. J. Clin. Invest. 106, 963–971 (2000).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  118. Lang, D. & Epstein, J. A. Sox10 and Pax3 physically interact to mediate activation of a conserved c-RET enhancer. Hum. Mol. Genet. 12, 937–945 (2003).

    Article  CAS  PubMed  Google Scholar 

  119. Stanchina, L. et al. Interactions between Sox10, Edn3 and Ednrb during enteric nervous system and melanocyte development. Dev. Biol. 295, 232–249 (2006).

    Article  CAS  PubMed  Google Scholar 

  120. Zhu, L. et al. Spatiotemporal regulation of endothelin receptor-B by SOX10 in neural crest-derived enteric neuron precursors. Nature Genet. 36, 732–737 (2004).

    Article  CAS  PubMed  Google Scholar 

  121. Taylor, K. M. & Labonne, C. SoxE factors function equivalently during neural crest and inner ear development and their activity is regulated by SUMOylation. Dev. Cell 9, 593–603 (2005). This work highlights the importance of the post-translational regulation in the NC GRN. It shows that two members of the SoxE family, Sox9 and Sox10, undergo post-translational modification by sumoylation, providing a mechanism for differential function of these proteins in different cell types.

    CAS  Google Scholar 

  122. Kellerer, S. et al. Replacement of the Sox10 transcription factor by Sox8 reveals incomplete functional equivalence. Development 133, 2875–2886 (2006).

    Article  CAS  PubMed  Google Scholar 

  123. Schreiner, S. et al. Hypomorphic Sox10 alleles reveal novel protein functions and unravel developmental differences in glial lineages. Development 134, 3271–3281 (2007).

    Article  CAS  PubMed  Google Scholar 

  124. Martinez-Morales, J. R., Henrich, T., Ramialison, M. & Wittbrodt, J. New genes in the evolution of the neural crest differentiation program. Genome Biol. 8, R36 (2007).

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  125. Tachibana, M. MITF: a stream flowing for pigment cells. Pigment Cell Res. 13, 230–240 (2000).

    Article  CAS  PubMed  Google Scholar 

  126. Morrison, S. J. et al. Transient Notch activation initiates an irreversible switch from neurogenesis to gliogenesis by neural crest stem cells. Cell 101, 499–510 (2000).

    Article  CAS  PubMed  Google Scholar 

  127. Dupin, E., Glavieux, C., Vaigot, P. & Le Douarin, N. M. Endothelin 3 induces the reversion of melanocytes to glia through a neural crest-derived glial-melanocytic progenitor. Proc. Natl Acad. Sci. USA 97, 7882–7887 (2000).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  128. Leimeroth, R. et al. Membrane-bound neuregulin1 type III actively promotes Schwann cell differentiation of multipotent progenitor cells. Dev. Biol. 246, 245–258 (2002).

    Article  CAS  PubMed  Google Scholar 

  129. Kalcheim, C. The role of neurotrophins in development of neural-crest cells that become sensory ganglia. Philos. Trans. R. Soc. Lond. B Biol. Sci. 351, 375–381 (1996).

    Article  CAS  PubMed  Google Scholar 

  130. Sariola, H. & Saarma, M. Novel functions and signalling pathways for GDNF. J. Cell Sci. 116, 3855–3862 (2003).

    Article  CAS  PubMed  Google Scholar 

  131. Clouthier, D. E. et al. Signaling pathways crucial for craniofacial development revealed by endothelin-A receptor-deficient mice. Dev. Biol. 217, 10–24 (2000).

    Article  CAS  PubMed  Google Scholar 

  132. Schmidt, C., McGonnell, I. M., Allen, S., Otto, A. & Patel, K. Wnt6 controls amniote neural crest induction through the non-canonical signaling pathway. Dev. Dyn. 236, 2502–2511 (2007).

    Article  CAS  PubMed  Google Scholar 

  133. Basch, M. L., Bronner-Fraser, M. & Garcia-Castro, M. I. Specification of the neural crest occurs during gastrulation and requires Pax7. Nature 441, 218–222 (2006). This study demonstrates that avian NC cells are specified during gastrulation in a domain in the neural plate border that expresses Pax7. This transcription factor, in turn, is required for NC formation.

    Article  CAS  PubMed  Google Scholar 

  134. De Calisto, J., Araya, C., Marchant, L., Riaz, C. F. & Mayor, R. Essential role of non-canonical Wnt signalling in neural crest migration. Development 132, 2587–2597 (2005). This work demonstrates for the first time that migration of NC cells requires non-canonical Wnt signalling.

    Article  CAS  PubMed  Google Scholar 

  135. Garriock, R. J. & Krieg, P. A. Wnt11-R signaling regulates a calcium sensitive EMT event essential for dorsal fin development of Xenopus. Dev. Biol. 304, 127–140 (2007).

    Article  CAS  PubMed  Google Scholar 

  136. Huelsken, J. & Birchmeier, W. New aspects of Wnt signaling pathways in higher vertebrates. Curr. Opin. Genet. Dev. 11, 547–553 (2001).

    Article  CAS  PubMed  Google Scholar 

  137. Rosso, S. B., Sussman, D., Wynshaw-Boris, A. & Salinas, P. C. Wnt signaling through Dishevelled, Rac and JNK regulates dendritic development. Nature Neurosci. 8, 34–42 (2005).

    Article  CAS  PubMed  Google Scholar 

  138. Rupp, P. A. & Kulesa, P. M. A role for RhoA in the two-phase migratory pattern of post-otic neural crest cells. Dev. Biol. 311, 159–171 (2007).

    Article  CAS  PubMed  Google Scholar 

  139. Hall, A. Rho GTPases and the actin cytoskeleton. Science 279, 509–514 (1998).

    Article  CAS  PubMed  Google Scholar 

  140. Broders-Bondon, F. et al. Regulation of XSnail2 expression by Rho GTPases. Dev. Dyn. 236, 2555–2566 (2007).

    Article  CAS  PubMed  Google Scholar 

  141. Smolen, G. A. et al. A Rap GTPase interactor, RADIL, mediates migration of neural crest precursors. Genes Dev. 21, 2131–2136 (2007).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  142. Guémar, L. et al. The small GTPase RhoV is an essential regulator of neural crest induction in Xenopus. Dev. Biol. 310, 113–128 (2007).

    Article  PubMed  CAS  Google Scholar 

  143. de Boer, T. P. et al. Connexin43 repression following epithelium-to-mesenchyme transition in embryonal carcinoma cells requires Snail1 transcription factor. Differentiation 75, 208–218 (2007).

    Article  CAS  PubMed  Google Scholar 

  144. Miyoshi, A. et al. Snail accelerates cancer invasion by upregulating MMP expression and is associated with poor prognosis of hepatocellular carcinoma. Br. J. Cancer 92, 252–258 (2005).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

Download references

Acknowledgements

We thank S. Bhattacharyya and N. Nikitina for their suggestions and critical reading of the manuscript. This work was supported by California Institute for Regenerative Medicine (CIRM) fellowship to T.S.S. and National Institute of Health (NIH) grants, DE017919 and NS36585, to M.B.F.

Author information

Authors and Affiliations

Authors

Related links

Related links

FURTHER INFORMATION

Marianne Bronner-Fraser's homepage

Glossary

Synapomorphy

A derived or specialized characteristic that is shared by two or more groups of organisms and that was present in their last common ancestor and can be used to establish phylogenies.

Gastrulation

A series of morphogenetic movements that is observed during the early development of all triploblast animals. It leads to the formation of a multilayered embryo with an outer cell layer (ectoderm), an inner cell layer (endoderm) and an intermediate cell layer (mesoderm).

Neurulation

A morphogenetic process during which the progenitors of the nervous system segregate from the ectoderm. During neurulation, a flat sheet of thickened ectoderm (neural plate) is transformed into the rudiment of central nervous system, the neural tube.

Metalloprotease

A peptidase that depends on a coordinated metal ion (Zn2+) for its catalytic activity. These proteinases have an important role not only in extracellular matrix remodelling and degradation, but also in programmed cell death, cell proliferation and differentiation.

Paraxial mesoderm

Most dorsal region of the embryonic mesoderm in vertebrate embryos. After segmentation it forms paired somites that ultimately give rise to axial skeleton and skeletal muscles.

Animal cap assay

An assay to test the effects of selected regulatory factors in 'naive' cells. Factors are introduced into fertilized eggs and the animal region of the frog embryo at the blastula stage is dissected off and assayed for response. Conversely, dissected tissue can be directly treated with secreted factors or their inhibitors.

Preplacodal ectoderm

A unique region of the embryonic ectoderm in the future head territory in vertebrate embryos. It contains precursors of all cranial placodes that ultimately contribute to sense organs and cranial sensory ganglia.

Rohon-Beard primary neuron

A large transient sensory neuron that is located in the dorsal aspect of the neural tube in lower vertebrates. It is present in the embryo and in early larval stages, but subsequently disappears.

Epithelial to mesenchymal transition

The transformation of an epithelial cell into a mesenchymal cell with migratory and invasive properties.

Stress fibre

A component of the actin cytoskeleton that consists of contractile bundles of actin and myosin II, which terminate in adhesion plaques that link the actin cytoskeleton to the cell surface. Stress fibres are involved in cell adhesion and the generation of tensile force.

Delamination

Physical dissociation of NC cells from the neuroepithelium in the dorsal aspect of the neural tube. This process requires NC cells to lose their apicobasal polarity and also requires disassembly of the basement membranes, through which NC cells penetrate.

Branchial arches

(also known as pharyngeal arches). A series of bilateral outpouchings on each side of the developing pharynx and mouth cavity in the vertebrate embryo. Differentiated arches typically consist of pharyngeal clefts derived from ectoderm, pharyngeal pouches derived from endoderm and a bony or cartilaginous rod of neural crest origin.

Adherens junction

A specialized intercellular junction of the plasma membrane in which cadherin molecules of adjacent cells interact in a Ca2+-dependent manner. Actin filaments are linked to this structure through catenins that are located underneath the junction.

Tight junction

A circumferential ring at the apex of epithelial cells that seals adjacent cells to one another. Tight junctions function as selective (semi-permeable) diffusion barriers and regulate solute and ion flux between adjacent epithelial cells.

Gap junction

A communicating junction (permeable to molecules up to 1 kDa) that exists between adjacent cells. Gap junctions are composed of 12 connexin protein subunits, 6 of which form a connexon or hemichannel that is contributed to by each of the coupled cells.

Somite

A series of paired blocks of mesodermal cells that form during early vertebrate development and that give rise to the backbone and body muscle.

Dermomyotome

An epithelial cell layer that forms the dorsolateral region of the somite that faces the ectoderm and further differentiates into the most dorsal dermatome, which later differentiates into dermis and myotome — future skeletal muscles.

Rights and permissions

Reprints and permissions

About this article

Cite this article

Sauka-Spengler, T., Bronner-Fraser, M. A gene regulatory network orchestrates neural crest formation. Nat Rev Mol Cell Biol 9, 557–568 (2008). https://doi.org/10.1038/nrm2428

Download citation

  • Published:

  • Issue Date:

  • DOI: https://doi.org/10.1038/nrm2428

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