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.

Bone morphogenetic protein signalling and vertebrate nervous system development

Key Points

  • Signalling from bone morphogenetic proteins (BMPs), which are members of the transforming growth factor-β protein family, is crucial for the development of both the central and peripheral nervous systems in vertebrates.

  • BMPs act at different stages of neural development and in different regions of the CNS to regulate cell fate, proliferation and differentiation. The development of the CNS begins with the specialization of the ectoderm into either non-neural or neural ectoderm, a transformation that is actively directed by the presence or absence of BMP signalling, respectively.

  • BMPs help to define the region from which a vertebrate-specific population of neural cells, the neural crest, will be generated. The neural crest arises at the border between the neuroectoderm and the non-neural ectoderm and gives rise to many diverse cell populations, including the PNS, melanoctyes and head skeletal elements.

  • BMP signalling positively regulates CNS development at later stages of development. As the neural tube closes, BMP proteins secreted from the ectoderm and the signalling centre at the dorsal-most region of the CNS, the roof plate, are important for patterning of the CNS. BMP signalling patterns the dorsal and intermediate regions of the spinal cord. Furthermore, active repression of BMP signalling is also crucial for the normal patterning of the ventral spinal cord.

  • BMP proteins are implicated in the regulation of axonal projections; in particular, in the repulsion of commissural axons from the dorsal aspect of the neural tube.

  • BMPs are negative regulators of proliferation and cell survival in the brain, which is the opposite of their effects in the spinal cord. BMPs also downregulate the expression of the anterior forebrain markers brain factor 1 (Bf1), fibroblast growth factor 8 (Fgf8) and sonic hedgehog (Shh). In addition, BMPs are important regulators of cerebellar granule neuron fate determination.

  • In a Smad-independent process, BMP signalling activates the LIM-domain-containing protein kinase 1 (LIMK1), which regulates the actin cytoskeleton and leads to changes in dendrite morphogenesis and the stabilization of synapses.

Abstract

Transforming growth factor-β (TGFβ) signalling, particularly signalling from the bone morphogenetic protein (BMP) members of this protein family, is crucial for the development of both the central and peripheral nervous systems in vertebrates. Experimental embryology and genetics performed in a range of organisms are providing insights into how BMPs establish the neural tissue and control the types and numbers of neurons formed. These studies also highlight the interactions between different developmental signals that are necessary to form a functional nervous system. The challenges ahead will be to uncover functions of TGFβ signalling in later stages of CNS development, as well as to determine possible associations with neurological diseases.

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: Bone morphogenetic protein (BMP) receptors mediate BMP signalling by activating Smad transcription factors.
Figure 2: Formation of the neural tube by neurulation.
Figure 3: The spinal cord is patterned along its dorsoventral axis.
Figure 4: Expression of bone morphogenetic proteins in the brain.

Similar content being viewed by others

References

  1. Nohe, A., Keating, E., Knaus, P. & Petersen, N. O. Signal transduction of bone morphogenetic protein receptors. Cell Signal. 16, 291–299 (2004).

    Article  CAS  PubMed  Google Scholar 

  2. Shi, Y. & Massague, J. Mechanisms of TGF-β signaling from cell membrane to the nucleus. Cell 113, 685–700 (2003).

    Article  CAS  PubMed  Google Scholar 

  3. Foletta, V. C. et al. Direct signaling by the BMP type II receptor via the cytoskeletal regulator LIMK1. J. Cell Biol. 162, 1089–1098 (2003).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  4. Lee-Hoeflich, S. T. et al. Activation of LIMK1 by binding to the BMP receptor, BMPRII, regulates BMP-dependent dendritogenesis. EMBO J. 23, 4792–4801 (2004).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  5. Eaton, B. A. & Davis, G. W. LIM kinase1 controls synaptic stability downstream of the type II BMP receptor. Neuron 47, 695–708 (2005).

    Article  CAS  PubMed  Google Scholar 

  6. Kretzschmar, M., Doody, J. & Massague, J. Opposing BMP and EGF signalling pathways converge on the TGF-β family mediator Smad1. Nature 389, 618–622 (1997).

    Article  CAS  PubMed  Google Scholar 

  7. Spemann, H. & Mangold, H. Uber Induktion von Embryonanlagen durch Implantation artfremder Organisatoren. Arch. Mikrosk. Anat. 100, 599–638 (1924).

    Google Scholar 

  8. Smith, W. C. & Harland, R. M. Expression cloning of noggin, a new dorsalizing factor localized to the Spemann organizer in Xenopus embryos. Cell 70, 829–840 (1992). References 8 and 10 describe the identification of noggin as a dorsalizing signal from the Spemann organizer and demonstrate that noggin functions as a BMP antagonist.

    Article  CAS  PubMed  Google Scholar 

  9. Smith, W. C., Knecht, A. K., Wu, M. & Harland, R. M. Secreted noggin protein mimics the Spemann organizer in dorsalizing Xenopus mesoderm. Nature 361, 547–549 (1993).

    Article  CAS  PubMed  Google Scholar 

  10. Zimmerman, L. B., De Jesus-Escobar, J. M. & Harland, R. M. The Spemann organizer signal noggin binds and inactivates bone morphogenetic protein 4. Cell 86, 599–606 (1996).

    Article  CAS  PubMed  Google Scholar 

  11. Sasai, Y. et al. Xenopus chordin: a novel dorsalizing factor activated by organizer-specific homeobox genes. Cell 79, 779–790 (1994).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  12. Piccolo, S., Sasai, Y., Lu, B. & De Robertis, E. M. Dorsoventral patterning in Xenopus: inhibition of ventral signals by direct binding of chordin to BMP-4. Cell 86, 589–598 (1996).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  13. Lamb, T. M. et al. Neural induction by the secreted polypeptide noggin. Science 262, 713–718 (1993).

    Article  CAS  PubMed  Google Scholar 

  14. Wilson, P. A. & Hemmati-Brivanlou, A. Induction of epidermis and inhibition of neural fate by Bmp-4. Nature 376, 331–333 (1995).

    Article  CAS  PubMed  Google Scholar 

  15. Sasai, Y., Lu, B., Steinbeisser, H. & De Robertis, E. M. Regulation of neural induction by the Chd and Bmp-4 antagonistic patterning signals in Xenopus. Nature 376, 333–336 (1995).

    Article  CAS  PubMed  Google Scholar 

  16. Hawley, S. H. et al. Disruption of BMP signals in embryonic Xenopus ectoderm leads to direct neural induction. Genes Dev. 9, 2923–2935 (1995).

    Article  CAS  PubMed  Google Scholar 

  17. Kuroda, H., Wessely, O. & De Robertis, E. M. Neural induction in Xenopus: requirement for ectodermal and endomesodermal signals via Chordin, Noggin, β-Catenin, and Cerberus. PLoS Biol. 2, e92 (2004).

    Article  PubMed  PubMed Central  Google Scholar 

  18. Oelgeschlager, M., Kuroda, H., Reversade, B. & De Robertis, E. M. Chordin is required for the Spemann organizer transplantation phenomenon in Xenopus embryos. Dev. Cell 4, 219–230 (2003).

    Article  CAS  PubMed  Google Scholar 

  19. Khokha, M. K., Yeh, J., Grammer, T. C. & Harland, R. M. Depletion of three BMP antagonists from Spemann's organizer leads to a catastrophic loss of dorsal structures. Dev. Cell 8, 401–411 (2005).

    Article  CAS  PubMed  Google Scholar 

  20. Hild, M. et al. The smad5 mutation somitabun blocks Bmp2b signaling during early dorsoventral patterning of the zebrafish embryo. Development 126, 2149–2159 (1999).

    CAS  PubMed  Google Scholar 

  21. Kishimoto, Y., Lee, K. H., Zon, L., Hammerschmidt, M. & Schulte-Merker, S. The molecular nature of zebrafish swirl: BMP2 function is essential during early dorsoventral patterning. Development 124, 4457–4466 (1997).

    CAS  PubMed  Google Scholar 

  22. Dick, A. et al. Essential role of Bmp7 (snailhouse) and its prodomain in dorsoventral patterning of the zebrafish embryo. Development 127, 343–354 (2000).

    CAS  PubMed  Google Scholar 

  23. Bachiller, D. et al. The organizer factors Chordin and Noggin are required for mouse forebrain development. Nature 403, 658–661 (2000).

    Article  CAS  PubMed  Google Scholar 

  24. Delaune, E., Lemaire, P. & Kodjabachian, L. Neural induction in Xenopus requires early FGF signalling in addition to BMP inhibition. Development 132, 299–310 (2005).

    Article  CAS  PubMed  Google Scholar 

  25. Londin, E. R., Niemiec, J. & Sirotkin, H. I. Chordin, FGF signaling, and mesodermal factors cooperate in zebrafish neural induction. Dev. Biol. 279, 1–19 (2005).

    Article  CAS  PubMed  Google Scholar 

  26. Wilson, S. I., Graziano, E., Harland, R., Jessell, T. M. & Edlund, T. An early requirement for FGF signalling in the acquisition of neural cell fate in the chick embryo. Curr. Biol. 10, 421–429 (2000).

    Article  CAS  PubMed  Google Scholar 

  27. Streit, A., Berliner, A. J., Papanayotou, C., Sirulnik, A. & Stern, C. D. Initiation of neural induction by FGF signalling before gastrulation. Nature 406, 74–78 (2000).

    Article  CAS  PubMed  Google Scholar 

  28. Lamb, T. M. & Harland, R. M. Fibroblast growth factor is a direct neural inducer, which combined with noggin generates anterior–posterior neural pattern. Development 121, 3627–3636 (1995).

    CAS  PubMed  Google Scholar 

  29. Launay, C., Fromentoux, V., Shi, D. L. & Boucaut, J. C. A truncated FGF receptor blocks neural induction by endogenous Xenopus inducers. Development 122, 869–880 (1996).

    CAS  PubMed  Google Scholar 

  30. Pera, E. M., Wessely, O., Li, S. Y. & De Robertis, E. M. Neural and head induction by insulin-like growth factor signals. Dev. Cell 1, 655–665 (2001).

    Article  CAS  PubMed  Google Scholar 

  31. Pera, E. M., Ikeda, A., Eivers, E. & De Robertis, E. M. Integration of IGF, FGF, and anti-BMP signals via Smad1 phosphorylation in neural induction. Genes Dev. 17, 3023–3028 (2003). Shows that Smad1 phosphorylation by MAPK, which is activated by the IGF and FGF pathways, is crucial for the neural inhibiting activity of BMP, providing the molecular basis for the crosstalk between these signalling pathways.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  32. Farlie, P. G., McKeown, S. J. & Newgreen, D. F. The neural crest: basic biology and clinical relationships in the craniofacial and enteric nervous systems. Birth Defects Res. C Embryo Today 72, 173–189 (2004).

    Article  CAS  PubMed  Google Scholar 

  33. Selleck, M. A. & Bronner-Fraser, M. Origins of the avian neural crest: the role of neural plate–epidermal interactions. Development 121, 525–538 (1995).

    CAS  PubMed  Google Scholar 

  34. Moury, J. D. & Jacobson, A. G. The origins of neural crest cells in the axolotl. Dev. Biol. 141, 243–253 (1990).

    Article  CAS  PubMed  Google Scholar 

  35. Moury, J. D. & Jacobson, A. G. Neural fold formation at newly created boundaries between neural plate and epidermis in the axolotl. Dev. Biol. 133, 44–57 (1989).

    Article  CAS  PubMed  Google Scholar 

  36. Dickinson, M. E., Selleck, M. A., McMahon, A. P. & Bronner-Fraser, M. Dorsalization of the neural tube by the non-neural ectoderm. Development 121, 2099–2106 (1995).

    CAS  PubMed  Google Scholar 

  37. Liem, K. F. Jr, Tremml, G., Roelink, H. & Jessell, T. M. Dorsal differentiation of neural plate cells induced by BMP-mediated signals from epidermal ectoderm. Cell 82, 969–979 (1995).

    Article  CAS  PubMed  Google Scholar 

  38. Schmid, B. et al. Equivalent genetic roles for bmp7/snailhouse and bmp2b/swirl in dorsoventral pattern formation. Development 127, 957–967 (2000).

    CAS  PubMed  Google Scholar 

  39. Nguyen, V. H. et al. Dorsal and intermediate neuronal cell types of the spinal cord are established by a BMP signaling pathway. Development 127, 1209–1220 (2000). Describes the analysis of three zebrafish mutants ( swr, snh and sbn ) that have defective BMP signalling and shows that BMP signalling is essential for spinal cord patterning and neural crest induction. The authors also show that cell fates in the spinal cord are determined by a BMP gradient.

    CAS  PubMed  Google Scholar 

  40. Nguyen, V. H. et al. Ventral and lateral regions of the zebrafish gastrula, including the neural crest progenitors, are established by a bmp2b/swirl pathway of genes. Dev. Biol. 199, 93–110 (1998).

    Article  CAS  PubMed  Google Scholar 

  41. 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 

  42. Chang, C. & Hemmati-Brivanlou, A. Neural crest induction by Xwnt7B in Xenopus. Dev. Biol. 194, 129–134 (1998).

    Article  CAS  PubMed  Google Scholar 

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

    CAS  PubMed  Google Scholar 

  44. 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).

    CAS  PubMed  Google Scholar 

  45. 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 

  46. Ikeya, M., Lee, S. M., Johnson, J. E., McMahon, A. P. & Takada, S. Wnt signalling required for expansion of neural crest and CNS progenitors. Nature 389, 966–970 (1997).

    Article  CAS  PubMed  Google Scholar 

  47. Garcia-Castro, M. & Bronner-Fraser, M. Induction and differentiation of the neural crest. Curr. Opin. Cell Biol. 11, 695–698 (1999).

    Article  CAS  PubMed  Google Scholar 

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

    Article  CAS  PubMed  Google Scholar 

  49. 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).

    Article  CAS  PubMed  Google Scholar 

  50. 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 

  51. Sela-Donenfeld, D. & Kalcheim, C. Regulation of the onset of neural crest migration by coordinated activity of BMP4 and Noggin in the dorsal neural tube. Development 126, 4749–4762 (1999).

    CAS  PubMed  Google Scholar 

  52. Sela-Donenfeld, D. & Kalcheim, C. Inhibition of noggin expression in the dorsal neural tube by somitogenesis: a mechanism for coordinating the timing of neural crest emigration. Development 127, 4845–4854 (2000).

    CAS  PubMed  Google Scholar 

  53. Graham, A., Francis-West, P., Brickell, P. & Lumsden, A. The signalling molecule BMP4 mediates apoptosis in the rhombencephalic neural crest. Nature 372, 684–686 (1994).

    Article  CAS  PubMed  Google Scholar 

  54. Marazzi, G., Wang, Y. & Sassoon, D. Msx2 is a transcriptional regulator in the BMP4-mediated programmed cell death pathway. Dev. Biol. 186, 127–138 (1997).

    Article  CAS  PubMed  Google Scholar 

  55. Smith, A. & Graham, A. Restricting Bmp-4 mediated apoptosis in hindbrain neural crest. Dev. Dyn. 220, 276–283 (2001).

    Article  CAS  PubMed  Google Scholar 

  56. Shah, N. M., Groves, A. K. & Anderson, D. J. Alternative neural crest cell fates are instructively promoted by TGFβ superfamily members. Cell 85, 331–343 (1996).

    Article  CAS  PubMed  Google Scholar 

  57. Varley, J. E., McPherson, C. E., Zou, H., Niswander, L. & Maxwell, G. D. Expression of a constitutively active type I BMP receptor using a retroviral vector promotes the development of adrenergic cells in neural crest cultures. Dev. Biol. 196, 107–118 (1998).

    Article  CAS  PubMed  Google Scholar 

  58. Varley, J. E. & Maxwell, G. D. BMP-2 and BMP-4, but not BMP-6, increase the number of adrenergic cells which develop in quail trunk neural crest cultures. Exp. Neurol. 140, 84–94 (1996).

    Article  CAS  PubMed  Google Scholar 

  59. 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).

    Article  CAS  PubMed  Google Scholar 

  60. de Melker, A. A., Desban, N. & Duband, J. L. Cellular localization and signaling activity of β-catenin in migrating neural crest cells. Dev. Dyn. 230, 708–726 (2004).

    Article  CAS  PubMed  Google Scholar 

  61. Hari, L. et al. Lineage-specific requirements of β-catenin in neural crest development. J. Cell Biol. 159, 867–880 (2002).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  62. Kleber, M. et al. Neural crest stem cell maintenance by combinatorial Wnt and BMP signaling. J. Cell Biol. 169, 309–320 (2005).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  63. Goulding, M. D., Chalepakis, G., Deutsch, U., Erselius, J. R. & Gruss, P. Pax-3, a novel murine DNA binding protein expressed during early neurogenesis. EMBO J. 10, 1135–1147 (1991).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  64. Jostes, B., Walther, C. & Gruss, P. The murine paired box gene, Pax7, is expressed specifically during the development of the nervous and muscular system. Mech. Dev. 33, 27–37 (1990).

    Article  CAS  PubMed  Google Scholar 

  65. Walther, C. & Gruss, P. Pax-6, a murine paired box gene, is expressed in the developing CNS. Development 113, 1435–1449 (1991).

    CAS  PubMed  Google Scholar 

  66. Briscoe, J. et al. Homeobox gene Nkx2.2 and specification of neuronal identity by graded Sonic hedgehog signalling. Nature 398, 622–627 (1999).

    Article  CAS  PubMed  Google Scholar 

  67. Lee, K. J., Mendelsohn, M. & Jessell, T. M. Neuronal patterning by BMPs: a requirement for GDF7 in the generation of a discrete class of commissural interneurons in the mouse spinal cord. Genes Dev. 12, 3394–3407 (1998).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  68. Basler, K., Edlund, T., Jessell, T. M. & Yamada, T. Control of cell pattern in the neural tube: regulation of cell differentiation by dorsalin-1, a novel TGF β family member. Cell 73, 687–702 (1993).

    Article  CAS  PubMed  Google Scholar 

  69. Lee, K. J., Dietrich, P. & Jessell, T. M. Genetic ablation reveals that the roof plate is essential for dorsal interneuron specification. Nature 403, 734–740 (2000).

    Article  CAS  PubMed  Google Scholar 

  70. Millonig, J. H., Millen, K. J. & Hatten, M. E. The mouse Dreher gene Lmx1a controls formation of the roof plate in the vertebrate CNS. Nature 403, 764–769 (2000).

    Article  CAS  PubMed  Google Scholar 

  71. Liem, K. F. Jr, Tremml, G. & Jessell, T. M. A role for the roof plate and its resident TGFβ-related proteins in neuronal patterning in the dorsal spinal cord. Cell 91, 127–138 (1997). Using an intermediate spinal cord explant assay, the authors showed that the roof plate provides signals that regulate dorsal spinal cord patterning. Furthermore, TGFβ proteins, including BMPs, which are expressed by roof plate cells, can mediate this activity.

    Article  CAS  PubMed  Google Scholar 

  72. Timmer, J. R., Wang, C. & Niswander, L. BMP signaling patterns the dorsal and intermediate neural tube via regulation of homeobox and helix–loop–helix transcription factors. Development 129, 2459–2472 (2002). Provides in vivo evidence that BMP signalling is essential for patterning the dorsal and intermediate spinal cord in the chick embryo.

    CAS  PubMed  Google Scholar 

  73. Panchision, D. M. et al. Sequential actions of BMP receptors control neural precursor cell production and fate. Genes Dev. 15, 2094–2110 (2001).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  74. Chesnutt, C., Burrus, L. W., Brown, A. M. & Niswander, L. Coordinate regulation of neural tube patterning and proliferation by TGFβ and WNT activity. Dev. Biol. 274, 334–347 (2004).

    Article  CAS  PubMed  Google Scholar 

  75. Timmer, J., Chesnutt, C. & Niswander, L. The Activin signaling pathway promotes differentiation of dI3 interneurons in the spinal neural tube. Dev. Biol. 285, 1–10 (2005).

    Article  CAS  PubMed  Google Scholar 

  76. Wine-Lee, L. et al. Signaling through BMP type 1 receptors is required for development of interneuron cell types in the dorsal spinal cord. Development 131, 5393–5403 (2004).

    Article  CAS  PubMed  Google Scholar 

  77. Chiang, C. et al. Cyclopia and defective axial patterning in mice lacking Sonic hedgehog gene function. Nature 383, 407–413 (1996).

    Article  CAS  PubMed  Google Scholar 

  78. McMahon, J. A. et al. Noggin-mediated antagonism of BMP signaling is required for growth and patterning of the neural tube and somite. Genes Dev. 12, 1438–1452 (1998).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  79. Liem, K. F. Jr, Jessell, T. M. & Briscoe, J. Regulation of the neural patterning activity of sonic hedgehog by secreted BMP inhibitors expressed by notochord and somites. Development 127, 4855–4866 (2000).

    CAS  PubMed  Google Scholar 

  80. Mekki-Dauriac, S., Agius, E., Kan, P. & Cochard, P. Bone morphogenetic proteins negatively control oligodendrocyte precursor specification in the chick spinal cord. Development 129, 5117–5130 (2002).

    CAS  PubMed  Google Scholar 

  81. Hammerschmidt, M., Serbedzija, G. N. & McMahon, A. P. Genetic analysis of dorsoventral pattern formation in the zebrafish: requirement of a BMP-like ventralizing activity and its dorsal repressor. Genes Dev. 10, 2452–2461 (1996).

    Article  CAS  PubMed  Google Scholar 

  82. Hatta, K., Kimmel, C. B., Ho, R. K. & Walker, C. The cyclops mutation blocks specification of the floor plate of the zebrafish central nervous system. Nature 350, 339–341 (1991).

    Article  CAS  PubMed  Google Scholar 

  83. Strahle, U. et al. one-eyed pinhead is required for development of the ventral midline of the zebrafish (Danio rerio) neural tube. Genes Funct. 1, 131–148 (1997).

    Article  CAS  PubMed  Google Scholar 

  84. Albert, S. et al. Cyclops-independent floor plate differentiation in zebrafish embryos. Dev. Dyn. 226, 59–66 (2003).

    Article  PubMed  Google Scholar 

  85. Gritsman, K. et al. The EGF-CFC protein one-eyed pinhead is essential for nodal signaling. Cell 97, 121–132 (1999).

    Article  CAS  PubMed  Google Scholar 

  86. Rebagliati, M. R., Toyama, R., Haffter, P. & Dawid, I. B. cyclops encodes a nodal-related factor involved in midline signaling. Proc. Natl Acad. Sci. USA 95, 9932–9937 (1998).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  87. Sampath, K. et al. Induction of the zebrafish ventral brain and floorplate requires cyclops/nodal signalling. Nature 395, 185–189 (1998).

    Article  CAS  PubMed  Google Scholar 

  88. Feldman, B. et al. Zebrafish organizer development and germ-layer formation require nodal-related signals. Nature 395, 181–185 (1998).

    Article  CAS  PubMed  Google Scholar 

  89. Megason, S. G. & McMahon, A. P. A mitogen gradient of dorsal midline Wnts organizes growth in the CNS. Development 129, 2087–2098 (2002).

    CAS  PubMed  Google Scholar 

  90. Zechner, D. et al. β-Catenin signals regulate cell growth and the balance between progenitor cell expansion and differentiation in the nervous system. Dev. Biol. 258, 406–418 (2003).

    Article  CAS  PubMed  Google Scholar 

  91. Butler, S. J. & Dodd, J. A role for BMP heterodimers in roof plate-mediated repulsion of commissural axons. Neuron 38, 389–401 (2003).

    Article  CAS  PubMed  Google Scholar 

  92. Augsburger, A., Schuchardt, A., Hoskins, S., Dodd, J. & Butler, S. BMPs as mediators of roof plate repulsion of commissural neurons. Neuron 24, 127–141 (1999). Together, references 91 and 92 showed that BMP proteins secreted by the roof plate have pivotal roles in guiding the commissural axons.

    Article  CAS  PubMed  Google Scholar 

  93. Furuta, Y., Piston, D. W. & Hogan, B. L. Bone morphogenetic proteins (BMPs) as regulators of dorsal forebrain development. Development 124, 2203–2212 (1997).

    CAS  PubMed  Google Scholar 

  94. Anderson, R. M., Lawrence, A. R., Stottmann, R. W., Bachiller, D. & Klingensmith, J. Chordin and noggin promote organizing centers of forebrain development in the mouse. Development 129, 4975–4987 (2002).

    CAS  PubMed  Google Scholar 

  95. Hebert, J. M., Mishina, Y. & McConnell, S. K. BMP signaling is required locally to pattern the dorsal telencephalic midline. Neuron 35, 1029–1041 (2002).

    Article  CAS  PubMed  Google Scholar 

  96. D'Alessandro, J. S. & Wang, E. A. Bone morphogenetic proteins inhibit proliferation, induce reversible differentiation and prevent cell death in astrocyte lineage cells. Growth Factors 11, 45–52 (1994).

    Article  CAS  PubMed  Google Scholar 

  97. D'Alessandro, J. S., Yetz-Aldape, J. & Wang, E. A. Bone morphogenetic proteins induce differentiation in astrocyte lineage cells. Growth Factors 11, 53–69 (1994).

    Article  CAS  PubMed  Google Scholar 

  98. Gomes, W. A., Mehler, M. F. & Kessler, J. A. Transgenic overexpression of BMP4 increases astroglial and decreases oligodendroglial lineage commitment. Dev. Biol. 255, 164–177 (2003).

    Article  CAS  PubMed  Google Scholar 

  99. Gross, R. E. et al. Bone morphogenetic proteins promote astroglial lineage commitment by mammalian subventricular zone progenitor cells. Neuron 17, 595–606 (1996).

    Article  CAS  PubMed  Google Scholar 

  100. Mehler, M. F., Mabie, P. C., Zhu, G., Gokhan, S. & Kessler, J. A. Developmental changes in progenitor cell responsiveness to bone morphogenetic proteins differentially modulate progressive CNS lineage fate. Dev. Neurosci. 22, 74–85 (2000).

    Article  CAS  PubMed  Google Scholar 

  101. Grinspan, J. B. et al. Stage-specific effects of bone morphogenetic proteins on the oligodendrocyte lineage. J. Neurobiol. 43, 1–17 (2000).

    Article  CAS  PubMed  Google Scholar 

  102. Mabie, P. C. et al. Bone morphogenetic proteins induce astroglial differentiation of oligodendroglial–astroglial progenitor cells. J. Neurosci. 17, 4112–4120 (1997).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  103. Alder, J., Lee, K. J., Jessell, T. M. & Hatten, M. E. Generation of cerebellar granule neurons in vivo by transplantation of BMP-treated neural progenitor cells. Nature Neurosci. 2, 535–540 (1999). Using both in vitro explant assays and in vivo cell transplantion, the authors showed that BMP signalling is crucial for the determination of cerebellar granule neurons.

    Article  CAS  PubMed  Google Scholar 

  104. Rios, I., Alvarez-Rodriguez, R., Marti, E. & Pons, S. Bmp2 antagonizes sonic hedgehog-mediated proliferation of cerebellar granule neurones through Smad5 signalling. Development 131, 3159–3168 (2004).

    Article  CAS  PubMed  Google Scholar 

  105. Angley, C., Kumar, M., Dinsio, K. J., Hall, A. K. & Siegel, R. E. Signaling by bone morphogenetic proteins and Smad1 modulates the postnatal differentiation of cerebellar cells. J. Neurosci. 23, 260–268 (2003).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  106. Meng, Y. et al. Abnormal spine morphology and enhanced LTP in LIMK-1 knockout mice. Neuron 35, 121–133 (2002).

    Article  CAS  PubMed  Google Scholar 

  107. Endo, M. et al. Control of growth cone motility and morphology by LIM kinase and Slingshot via phosphorylation and dephosphorylation of cofilin. J. Neurosci. 23, 2527–2537 (2003).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  108. Suzuki, A., Ueno, N. & Hemmati-Brivanlou, A. Xenopus msx1 mediates epidermal induction and neural inhibition by BMP4. Development 124, 3037–3044 (1997).

    CAS  PubMed  Google Scholar 

  109. Binato, R., Martinez, C. E., Pizzatti, L., Robert, B. & Abdelhay, E. SMAD 8 binding to mice Msx-1 basal promoter is required for transcriptional activation. Biochem. J. 16 Aug 2005 (10.1042/BJ20050327).

  110. Alvarez Martinez, C. E. et al. Characterization of a Smad motif similar to Drosophila mad in the mouse Msx1 promoter. Biochem. Biophys. Res. Commun. 291, 655–662 (2002).

    Article  CAS  PubMed  Google Scholar 

  111. Liu, Y., Helms, A. W. & Johnson, J. E. Distinct activities of Msx1 and Msx3 in dorsal neural tube development. Development 131, 1017–1028 (2004).

    Article  CAS  PubMed  Google Scholar 

  112. 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 

  113. Bach, A. et al. Msx1 is required for dorsal diencephalon patterning. Development 130, 4025–4036 (2003).

    Article  CAS  PubMed  Google Scholar 

  114. Fernandez-Llebrez, P. et al. Msx1-deficient mice fail to form prosomere 1 derivatives, subcommissural organ, and posterior commissure and develop hydrocephalus. J. Neuropathol. Exp. Neurol. 63, 574–586 (2004).

    Article  CAS  PubMed  Google Scholar 

  115. Ramos, C., Fernandez-Llebrez, P., Bach, A., Robert, B. & Soriano, E. Msx1 disruption leads to diencephalon defects and hydrocephalus. Dev. Dyn. 230, 446–460 (2004).

    Article  CAS  PubMed  Google Scholar 

  116. Linker, C. & Stern, C. D. Neural induction requires BMP inhibition only as a late step, and involves signals other than FGF and Wnt antagonists. Development 131, 5671–5681 (2004).

    Article  CAS  PubMed  Google Scholar 

  117. Hussein, S. M., Duff, E. K. & Sirard, C. Smad4 and β-catenin co-activators functionally interact with lymphoid-enhancing factor to regulate graded expression of Msx2. J. Biol. Chem. 278, 48805–48814 (2003).

    Article  CAS  PubMed  Google Scholar 

Download references

Acknowledgements

We thank K. Artinger, L. Sussel and I. Zohn for critical reading of the manuscript. We gratefully acknowledge all individuals who have contributed to this field and apologize to those who were not included in this review due to space limitations. L.N. is an Investigator of the Howard Hughes Medical Institute.

Author information

Authors and Affiliations

Authors

Corresponding author

Correspondence to Lee A. Niswander.

Ethics declarations

Competing interests

The authors declare no competing financial interests.

Supplementary information

Related links

Related links

DATABASES

Entrez Gene

BMP4

BMP7

BMPRIA

BMPRIB

BMPRII

chordin

gremlin

LIMK1

Math1

Msx1

NGN1

Nkx2.2

noggin

Pax3

Pax6

Pax7

SHH

Wnt3a

Zic1

ZFIN

one-eyed pinhead

snh

sbn

swr

Glossary

EXPRESSION CLONING

A function-based gene discovery method. In Xenopus spp., this is done by injecting mRNA of candidate genes into the egg or early embryo and screening for a phenotype of interest: for example, the formation of a secondary body axis or the loss of head structures.

MORPHOLINO

A non-ionic DNA analogue of 25 nucleotides. When targeted to the 5′ untranslated region or start codon, a morpholino may prevent the translation of its target mRNA, generating a partial loss-of-function phenotype.

EMBRYONIC ORGANIZER

(Also known as Spemann's organizer.) Originally defined as the tissue in the gastrulating embryo that has the potential to induce a secondary body axis when grafted ectopically. Now it is recognized as an important signalling centre that regulates the basic body plan through the action of various signalling molecules, including antagonists of BMP.

EPIBLAST

The layer of a blastula that gives rise to the ectoderm after gastrulation.

SMALL INTERFERING RNA

(siRNA). A sequence-specific gene-silencing tool used in RNA interference. siRNAs are short fragments of synthetic double-stranded RNA with 21–23 pairs of nucleotides that have sequence specificity to the gene of interest (the target). These small double-stranded RNAs trigger degradation of the target RNA, thereby creating a partial loss-of-function by decreasing the amount of translatable RNA. As such, siRNA can reveal the requirement for the gene of interest in targeted cells.

HEDGEHOG SIGNALLING

Hedghog (HH) is a family of secreted proteins that regulate key developmental processes in both vertebrates and invertebrates through a receptor-mediated signalling cascade. In the vertebrate CNS in particular, HH proteins, especially sonic hedgehog, have many roles, including ventral patterning, axon guidance, cell survival and differentiation.

SERUM-FREE MOUSE EMBRYO CELLS

(SFME cells). A cell line derived in a defined serum-free medium. These cells have been cultured for more than 200 generations and show properties of neural progenitor cells.

Rights and permissions

Reprints and permissions

About this article

Cite this article

Liu, A., Niswander, L. Bone morphogenetic protein signalling and vertebrate nervous system development. Nat Rev Neurosci 6, 945–954 (2005). https://doi.org/10.1038/nrn1805

Download citation

  • Issue Date:

  • DOI: https://doi.org/10.1038/nrn1805

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