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:

Retinoid signalling in the development of the central nervous system

Key Points

  • Vitamin A is an essential component of the diet; in its absence, animals show characteristic changes, including keratinization of epithelia, decreased immune function, anaemia and blindness. Dietary deficiency during pregnancy also causes congenital malformations in the embryonic central nervous system (CNS).

  • Retinoic acid (RA) is the most biologically active member of the retinoids — a family of molecules that are derived from vitamin A. RA is highly teratogenic when administered in excess to pregnant mammals, and it has been shown to cause patterning defects in the CNS.

  • The primary neurons in fish and amphibians coordinate escape movements, and their numbers are regulated by RA.

  • In addition to regulating the genes that control neuronal differentiation, RA switches on genes that pattern the neural plate along the anteroposterior (AP) axis. Initial experiments implied that it acts throughout the AP extent of the neural tube, but subsequent experiments have focused on a more localized role in the hindbrain and anterior spinal cord.

  • There are two theories to explain how RA organizes AP patterning. There could be a head-to-tail gradient, with a high point at the posterior end, or there could be a localized source of RA at the posterior end of the hindbrain. The existence of a gradient has proved difficult to demonstrate, and recent findings seem to point towards the latter model.

  • RA is also required for the patterning of neuronal populations along the dorsoventral axis of the neural tube. It seems to suppress ventral neuronal genes and to induce dorsal genes, allowing the generation of interneurons in the centre of the spinal cord. In addition, RA is required for the specification of lateral motor column neurons.

  • As RA is an essential component of the adult diet, it is likely that RA signalling also occurs in the adult. The failure of any component of the RA signalling pathway would be expected to cause a malfunction in the neurons concerned. Indeed, defects in RA signalling have been implicated in several neurological diseases, including movement disorders, schizophrenia and motor neuron disease.

Abstract

Retinoids — a family of molecules that are derived from vitamin A — have been implicated in many developmental processes. In the embryonic vertebrate central nervous system (CNS), retinoic acid (RA) has a role in patterning both the anteroposterior and dorsoventral axes. Initially, RA was thought to be involved in generating the entire anteroposterior extent of the CNS, but more recent experiments have identified its main sites of action as the hindbrain and anterior spinal cord. RA also regulates interneuron and motor neuron development along the dorsoventral axis. This review describes the studies that led to these conclusions, and discusses how understanding the mechanisms of RA action in the developing CNS might provide insights into neurological disease.

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: Pathways for the synthesis and mechanism of action of RA.
Figure 2: Primary neurons in Xenopus and the effect of altered RA signalling.
Figure 3: Chart to show where RA fits into the scheme of neural development.
Figure 4: The effects of altered RA signalling on Xenopus embryos.
Figure 5: The effects of increasing and decreasing RA signalling on the hindbrain of chick and mouse embryos.
Figure 6: The hindbrain structure after gradually decreasing RA signalling.
Figure 7: Endogenous RA in the chick and mouse embryo.
Figure 8: Role of RA in the dorsoventral axis of the spinal cord.

Similar content being viewed by others

References

  1. Wolbach, S. B. & Howe, P. R. Tissue changes following deprivation of fat soluble A vitamin. J. Exp. Med. 42, 753–777 (1925).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  2. Hart, E. B., Miller, W. S. & McCollum, E. V. Further studies on the nutritive deficiencies of wheat and grain mixtures and the pathological conditions produced in swine by their use. J. Biol. Chem. 25, 239–260 (1916).

    Article  CAS  Google Scholar 

  3. Hughes, J. S., Lienhardt, H. F. & Aubel, C. E. Nerve degeneration resulting from avitaminosis A. J. Nutr. 2, 183–186 (1929).

    Article  CAS  Google Scholar 

  4. Hale, F. Pigs born without eye balls. J. Hered. 24, 105–106 (1933).

    Article  Google Scholar 

  5. Cohlan, S. Q. Excessive intake of vitamin A as a cause of congenital abnormalities in the rat. Science 117, 535–536 (1953).

    Article  CAS  PubMed  Google Scholar 

  6. Langman, J. & Welch, G. W. Effect of vitamin A on development of the central nervous system. J. Comp. Neurol. 128, 1–16 (1967).

    Article  Google Scholar 

  7. Shenfelt, R. E. Morphogenesis of malformations in hamsters caused by retinoic acid: relation to dose and stage at treatment. Teratology 5, 103–118 (1972).

    Article  Google Scholar 

  8. Shum, A. S. W. et al. Retinoic acid induces down-regulation of Wnt-3a, apoptosis and diversion of tail bud cells to a neural fate in the mouse embryo. Mech. Dev. 84, 17–30 (1999).

    Article  CAS  PubMed  Google Scholar 

  9. Tibbles, L. & Wiley, M. J. A comparative study of the effects of retinoic acid given during the critical period for inducing spina bifida in mice and hamsters. Teratology 37, 113–125 (1988).

    Article  CAS  PubMed  Google Scholar 

  10. Jones-Villeneuve, E. M. V., McBurney, M. W., Rogers, K. A. & Kalnins, V. I. Retinoic acid induces embryonal carcinoma cells to differentiate into neurons and glial cells. J. Cell Biol. 94, 253–262 (1982).

    Article  CAS  PubMed  Google Scholar 

  11. McBurney, M. W., Jones-Villeneuve, E. M. V., Edwards, M. K. S. & Anderson, P. J. Control of muscle and neuronal differentiation in a cultured embryonal carcinoma cell line. Nature 299, 165–167 (1982).

    Article  CAS  PubMed  Google Scholar 

  12. Maden, M. Role and distribution of retinoic acid during CNS development. Int. Rev. Cytol. 209, 1–77 (2001).

    Article  CAS  PubMed  Google Scholar 

  13. Duester, G. Families of retinoid dehydrogenases regulating vitamin A function. Production of visual pigment and retinoic acid. Eur. J. Biochem. 267, 4315–4324 (2000).

    Article  CAS  PubMed  Google Scholar 

  14. Fujii, H. et al. Metabolic inactivation of retinoic acid by a novel P450 differentially expressed in developing mouse embryos. EMBO J. 16, 4163–4173 (1997).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  15. White, J. A. et al. Identification of the retinoic acid-inducible all-trans-retinoic acid 4-hydroxylase. J. Biol. Chem. 271, 29922–29927 (1996).

    Article  CAS  PubMed  Google Scholar 

  16. White, J. A. et al. Identification of the human cytochrome P450, P450RAI-2, which is predominantly expressed in the adult cerebellum and is responsible for all-trans-retinoic acid metabolism. Proc. Natl Acad. Sci. USA 97, 6403–6408 (2000).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  17. Sonneveld, E., van den Brink, C. E., Tertoolen, L. G. J., van der Burg, B. & van der Saag, P. T. Retinoic acid hydroxylase (CYP26) is a key enzyme in neuronal differentiation of embryonal carcinoma cells. Dev. Biol. 213, 390–404 (1999).

    Article  CAS  PubMed  Google Scholar 

  18. Niederreither, K. et al. Genetic evidence that oxidative derivatives of retinoic acid are not involved in retinoid signaling during mouse development. Nature Genet. 31, 84–88 (2002).

    Article  CAS  PubMed  Google Scholar 

  19. Kastner, P., Chambon, P. & Leid, M. in Vitamin A in Health and Disease (ed. Blomhoff, R.) 189–238 (Marcel Dekker, New York, 1994).

    Google Scholar 

  20. Kliewer, S. A., Umesono, K., Evans, R. M. & Mangelsdorf, D. J. in Vitamin A in Health and Disease (ed. Blomhoff, R.) 239–255 (Marcel Dekker, New York, 1994).

    Google Scholar 

  21. Sharpe, C. & Goldstone, K. The control of Xenopus embryonic primary neurogenesis is mediated by retinoid signalling in the neurectoderm. Mech. Dev. 91, 69–80 (2000).

    Article  CAS  PubMed  Google Scholar 

  22. Franco, P. G., Paganelli, A. R., Lopez, S. L. & Carrasco, A. E. Functional association of retinoic acid and hedgehog signaling in Xenopus primary neurogenesis. Development 126, 4257–4265 (1999).This paper considers the involvement of RA in primary neurogenesis, and how prepattern genes, proneural genes and neural differentiation genes are linked together by a cascade that starts with RA.

    Article  CAS  PubMed  Google Scholar 

  23. Papalopulu, N. & Kintner, C. A posteriorising factor, retinoic acid, reveals that anteroposterior patterning controls the timing of neuronal differentiation in Xenopus neuroectoderm. Development 122, 3409–3418 (1996).

    Article  CAS  PubMed  Google Scholar 

  24. Sharpe, C. R. & Goldstone, K. Retinoid receptors promote primary neurogenesis in Xenopus. Development 124, 515–523 (1997).

    Article  CAS  PubMed  Google Scholar 

  25. Sharpe, C. & Goldstone, K. Retinoid signalling acts during the gastrula stages to promote primary neurogenesis. Int. J. Dev. Biol. 44, 463–470 (2000).

    CAS  PubMed  Google Scholar 

  26. Blumberg, B. et al. An essential role for retinoid signalling in anteroposterior neural patterning. Development 124, 373–379 (1997).An analysis of the effects of dominant-negative and constitutively active RARs on the AP patterning of gene expression in the Xenopus CNS.

    Article  CAS  PubMed  Google Scholar 

  27. Bertrand, N., Castro, D. S. & Guillemot, F. Proneural genes and the specification of neural cell types. Nature Rev. Neurosci. 3, 517–530 (2002).

    Article  CAS  Google Scholar 

  28. Gomez-Skarmeta, J., Glavic, A., de la Calle-Mustienes, E., Modolell, J. & Mayor, R. Xiro, a Xenopus homolog of the Drosophila Iroquois complex genes, control development at the neural plate. EMBO J. 17, 181–190 (1998).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  29. Cho, K. W. Y. & De Robertis, E. M. Differential activation of Xenopus homeobox genes by mesoderm-inducing growth factors and retinoic acid. Genes Dev. 4, 1910–1916 (1990).

    Article  CAS  PubMed  Google Scholar 

  30. Dekker, E.-J. et al. Xenopus Hox-2 genes are expressed sequentially after the onset of gastrulation and are differentially inducible by retinoic acid. Dev. Suppl., 195–202 (1992).

  31. Durston, A. J. et al. Retinoic acid causes an anteroposterior transformation in the developing nervous system. Nature 340, 140–144 (1989).A study of the effect of excess RA on Xenopus embryos, showing a CNS action that reignited interest in RA and CNS development.

    Article  CAS  PubMed  Google Scholar 

  32. Leroy, P. & De Robertis, E. M. Effects of lithium chloride and retinoic acid on the expression of genes from the Xenopus Hox 2 complex. Dev. Dyn. 194, 21–32 (1992).

    Article  CAS  PubMed  Google Scholar 

  33. Lopez, S. L. & Carrasco, A. E. Retinoic acid induces changes in the localization of homeobox proteins in the antero-posterior axis of Xenopus laevis embryos. Mech. Dev. 36, 153–164 (1992).

    Article  CAS  PubMed  Google Scholar 

  34. Pannese, M. et al. The Xenopus homologue of Otx2 is a maternal homeobox gene that demarcates and specifies anterior body regions. Development 121, 707–720 (1995).

    Article  CAS  PubMed  Google Scholar 

  35. Sive, H. L., Draper, B. W., Harland, R. M. & Weintrub, H. Identification of a retinoic acid-sensitive period during primary axis formation in Xenopus laevis. Genes Dev. 4, 932–942 (1990).

    Article  CAS  PubMed  Google Scholar 

  36. Creech-Kraft, J., Schuh, T., Juchau, M. & Kimelman, D. The retinoid X receptor ligand, 9-cis-retinoic acid, is a potential regulator of early Xenopus development. Proc. Natl Acad. Sci. USA 91, 3067–3071 (1994).

    Article  Google Scholar 

  37. Drysdale, T. A. & Crawford, M. J. Effects of localized application of retinoic acid on Xenopus laevis development. Dev. Biol. 162, 394–401 (1994).

    Article  CAS  PubMed  Google Scholar 

  38. Kolm, P. J. & Sive, H. L. Regulation of the Xenopus labial homeodomain genes, HoxA1 and HoxD1: activation by retinoids and peptide growth factors. Dev. Biol. 167, 34–49 (1995).

    Article  CAS  PubMed  Google Scholar 

  39. Pijnappel, W. W. M. et al. The retinoid ligand 4-oxo-retinoic acid is a highly active modulator of positional specification. Nature 366, 340–344 (1993).

    Article  CAS  PubMed  Google Scholar 

  40. Saha, M. S., Michel, R. B., Gulding, K. M. & Grainger, R. M. A Xenopus homeobox gene defines dorsal–ventral domains in the developing brain. Development 118, 193–202 (1993).

    Article  CAS  PubMed  Google Scholar 

  41. Taira, M., Otani, H., Jamrich, M. & Dawid, I. B. Expression of the LIM class homeobox gene Xlim-1 in pronephros and CNS cell lineages of Xenopus embryos is affected by retinoic acid and exogastrulation. Development 120, 1525–1536 (1994).

    Article  CAS  PubMed  Google Scholar 

  42. von Bubnoff, A., Schmidt, J. E. & Kimelman, D. The Xenopus laevis homeobox gene Xgbx-2 is an early marker of anteroposterior patterning in the ectoderm. Mech. Dev. 54, 149–160 (1995).

    Article  Google Scholar 

  43. Maden, M., Gale, E., Horton, C. & Smith, J. C. in Retinoids in Normal Development and Teratogenesis (ed. Morriss-Kay, G. M.) 119–134 (Oxford Univ. Press, Oxford, UK, 1992).

    Google Scholar 

  44. Zhang, Z., Balmer, J. E., Lovlie, A., Fromm, S. H. & Blomhoff, R. Specific teratogenic effects of different retinoic acid isomers and analogs in the developing anterior central nervous system of zebrafish. Dev. Dyn. 206, 73–86 (1996).

    Article  CAS  PubMed  Google Scholar 

  45. Avantaggiato, V., Acampora, D., Tuorto, F. & Simeone, A. Retinoic acid induces stage-specific repatterning of the rostral central nervous system. Dev. Biol. 175, 347–357 (1996).

    Article  CAS  PubMed  Google Scholar 

  46. Cunningham, M. L., MacAuley, A. & Mirkes, P. E. From gastrulation to neurulation: transition in retinoic acid sensitivity identifies distinct stages of neural patterning in the rat. Dev. Dyn. 200, 227–241 (1994).

    Article  CAS  PubMed  Google Scholar 

  47. Simeone, A. et al. Retinoic acid induces stage-specific antero-posterior transformation of rostral central nervous system. Mech. Dev. 51, 83–98 (1995).

    Article  CAS  PubMed  Google Scholar 

  48. Agarwal, V. R. & Sato, S. M. Retinoic acid affects central nervous system development of Xenopus by changing cell fate. Mech. Dev. 44, 167–173 (1993).

    Article  CAS  PubMed  Google Scholar 

  49. Ruiz i Altaba, A. & Jessell, T. M. Retinoic acid modifies the pattern of cell differentiation in the central nervous system of neurula stage Xenopus embryos. Development 112, 945–958 (1991).

    Article  CAS  PubMed  Google Scholar 

  50. Ruiz i Altaba, A. & Jessell, T. Retinoic acid modifies mesodermal patterning in early Xenopus embryos. Genes Dev. 5, 175–187 (1991).

    Article  CAS  PubMed  Google Scholar 

  51. Sive, H. L. & Cheng, P. F. Retinoic acid perturbs the expression of Xhox.lab genes and alters mesodermal determination in Xenopus laevis. Genes Dev. 5, 1321–1332 (1991).

    Article  CAS  PubMed  Google Scholar 

  52. Taira, M., Saint-Jeannet, J.-P. & Dawid, I. B. Role of the Xlim-1 and Xbra genes in anteroposterior patterning of neural tissue by the head and trunk organiser. Proc. Natl Acad. Sci. USA 94, 895–900 (1997).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  53. Dekker, E.-J. et al. Overexpression of a cellular retinoic acid binding protein (xCRABP) causes anteroposterior defects in developing Xenopus embryos. Development 120, 973–985 (1994).

    Article  CAS  PubMed  Google Scholar 

  54. van der Wees, J. et al. Inhibition of retinoic acid receptor-mediated signalling alters positional identity in the developing hindbrain. Development 125, 545–556 (1998).

    Article  CAS  PubMed  Google Scholar 

  55. Hollemann, T., Chen, Y., Grunz, H. & Pieler, T. Regionalized metabolic activity establishes boundaries of retinoic acid signalling. EMBO J. 17, 7361–7372 (1998).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  56. Godsave, S. F. et al. Graded retinoid responses in the developing hindbrain. Dev. Dyn. 213, 39–49 (1998).

    Article  CAS  PubMed  Google Scholar 

  57. Holder, N. & Hill, J. Retinoic acid modifies development of the midbrain–hindbrain border and affects cranial ganglion formation in zebrafish embryos. Development 113, 1159–1170 (1991).

    Article  CAS  PubMed  Google Scholar 

  58. Lopez, S. L., Dono, R., Zeller, R. & Carrasco, A. E. Differential effects of retinoic acid and a retinoid antagonist on the spatial distribution of the homeoprotein Hoxb-7 in vertebrate embryos. Dev. Dyn. 204, 457–471 (1995).

    Article  CAS  PubMed  Google Scholar 

  59. Papalopulu, N. et al. Retinoic acid causes abnormal development and segmental patterning of the anterior hindbrain in Xenopus embryos. Development 113, 1145–1158 (1991).

    Article  CAS  PubMed  Google Scholar 

  60. Sundin, O. & Eichele, G. An early marker of axial pattern in the chick embryo and its respecification by retinoic acid. Development 114, 841–852 (1992).

    Article  CAS  PubMed  Google Scholar 

  61. Lee, Y. M. et al. Retinoic acid stage-dependently alters the migration pattern and identity of hindbrain neural crest cells. Development 121, 825–837 (1995).

    Article  CAS  PubMed  Google Scholar 

  62. Leonard, L., Horton, C., Maden, M. & Pizzey, J. A. Anteriorization of CRABP-I expression by retinoic acid in the developing mouse central nervous system and its relationship to teratogenesis. Dev. Biol. 168, 514–528 (1995).

    Article  CAS  PubMed  Google Scholar 

  63. Morriss-Kay, G. M., Murphy, P., Hill, R. E. & Davidson, D. R. Effects of retinoic acid excess on expression of Hox-2.9 and Krox-20 and on morphological segmentation in the hindbrain of mouse embryos. EMBO J. 10, 2985–2995 (1991).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  64. Morriss, G. M. Morphogenesis of the malformations induced in rat embryos by maternal hypervitaminosis A. J. Anat. 113, 241–250 (1972).

    CAS  PubMed  PubMed Central  Google Scholar 

  65. Wood, H., Pall, G. & Morriss-Kay, G. Exposure to retinoic acid before or after the onset of somitogenesis reveals separate effects on rhombomeric segmentation and 3′ HoxB gene expression domains. Development 120, 2279–2285 (1994).

    Article  CAS  PubMed  Google Scholar 

  66. Conlon, R. A. & Rossant, J. Exogenous retinoic acid rapidly induces anterior ectopic expression of murine Hox-2 genes in vivo. Development 116, 357–368 (1992).A thorough analysis of the effect of RA on the induction of ectopic Hox gene expression in mouse embryos, relating it to the concentration, time and position of the gene in the Hox cluster.

    Article  CAS  PubMed  Google Scholar 

  67. Mallo, M. & Brandlin, I. Segmental identity can change independently in the hindbrain and rhombencephalic neural crest. Dev. Dyn. 210, 146–156 (1997).

    Article  CAS  PubMed  Google Scholar 

  68. Hill, J., Clarke, J. D. W., Vargesson, N., Jowett, T. & Holder, N. Exogenous retinoic acid causes specific alterations in the development of the midbrain and hindbrain of the zebrafish embryo including positional respecification of the Mauthner neuron. Mech. Dev. 50, 3–16 (1995).

    Article  CAS  PubMed  Google Scholar 

  69. Marshall, H. et al. Retinoic acid alters hindbrain Hox code and induces transformation of rhombomeres 2/3 into a 4/5 identity. Nature 360, 737–741 (1992).This study shows the remarkable effect of RA on mouse embryos — transforming rhombomeres into a different fate (r2 and r3 into r4 and r5).

    Article  CAS  PubMed  Google Scholar 

  70. Manns, M. & Fritzsch, B. Retinoic acid affects the organization of reticulospinal neurons in developing Xenopus. Neurosci. Lett. 139, 253–256 (1992).

    Article  CAS  PubMed  Google Scholar 

  71. Kessel, M. Reversal of axonal pathways from rhombomere 3 correlates with extra Hox expression domains. Neuron 10, 379–393 (1993).

    Article  CAS  PubMed  Google Scholar 

  72. Gale, E., Zile, M. & Maden, M. Hindbrain respecification in the retinoid-deficient quail. Mech. Dev. 89, 43–54 (1999).

    Article  CAS  PubMed  Google Scholar 

  73. Maden, M., Gale, E., Kostetskii, I. & Zile, M. Vitamin A-deficient quail embryos have half a hindbrain and other neural defects. Curr. Biol. 6, 417–426 (1996).This paper shows that a complete lack of RA in the embryo results in loss of the posterior hindbrain rhombomeres, as well as other neural defects.

    Article  CAS  PubMed  Google Scholar 

  74. White, J. C. et al. Defects in embryonic hindbrain development and fetal resorption resulting from vitamin A deficiency in the rat are prevented by feeding pharmacological levels of all-trans-retinoic acid. Proc. Natl Acad. Sci. USA 95, 13459–13464 (1998).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  75. White, J. C., Highland, M., Kaiser, M. & Clagett-Dame, M. Vitamin A deficiency results in the dose-dependent acquisition of anterior character and shortening of the caudal hindbrain of the rat embryo. Dev. Biol. 220, 263–284 (2000).

    Article  CAS  PubMed  Google Scholar 

  76. Niederreither, K., Subbarayan, V., Dolle, P. & Chambon, P. Embryonic retinoic acid synthesis is essential for early mouse post-implantation development. Nature Genet. 21, 444–448 (1999).This study locates the source of RA for hindbrain development in the paraxial mesoderm, showing that the Raldh2 mutant mouse has the same hindbrain defects as animals that are subject to complete vitamin A deficiency.

    Article  CAS  PubMed  Google Scholar 

  77. Niederreither, K., Vermot, J., Schubaur, B., Chambon, P. & Dolle, P. Retinoic acid synthesis and hindbrain patterning in the mouse embryo. Development 127, 75–85 (2000).

    Article  CAS  PubMed  Google Scholar 

  78. Begemann, G., Schilling, T. F., Rauch, G.-J., Geisler, R. & Ingham, P. W. The zebrafish neckless mutation reveals a requirement for raldh2 in mesodermal signals that pattern the hindbrain. Development 128, 3081–3094 (2001).

    Article  CAS  PubMed  Google Scholar 

  79. Grandel, H. et al. Retinoic acid signalling is the zebrafish embryo is necessary during pre-segmentation stages to pattern the anterior–posterior axis of the CNS and to induce a pectoral fin bud. Development 129, 2851–2865 (2002).

    Article  CAS  PubMed  Google Scholar 

  80. Dupe, V. & Lumsden, A. Hindbrain patterning involves graded responses to retinoic acid signalling. Development 128, 2199–2208 (2001).This paper shows that the hindbrain responds in a graded way to loss of RA by gradually losing rhombomeres, rather than in an all-or-nothing manner.

    Article  CAS  PubMed  Google Scholar 

  81. Dupe, V., Ghyselinck, N., Wendling, O., Chambon, P. & Mark, M. Key roles of retinoic acid receptors α and β in the patterning of the caudal hindbrain, pharyngeal arches and otocyst in the mouse. Development 126, 5051–5059 (1999).

    Article  CAS  PubMed  Google Scholar 

  82. Wendling, O., Ghyselinck, N., Chambon, P. & Mark, M. Roles of retinoic acid receptors in early embryonic morphogenesis and hindbrain patterning. Development 128, 2031–2038 (2001).Different combinations of RAR knockouts result in different types of hindbrain defect in mouse embryos.

    Article  CAS  PubMed  Google Scholar 

  83. Gavalas, A. ArRAnging the hindbrain. Trends Neurosci. 25, 61–64 (2002).

    Article  CAS  PubMed  Google Scholar 

  84. Chen, Y.-P., Huang, L. & Solursh, M. A concentration gradient of retinoids in the early Xenopus laevis embryo. Dev. Biol. 161, 70–76 (1994).

    Article  PubMed  Google Scholar 

  85. Horton, C. & Maden, M. Endogenous distribution of retinoids during normal development and teratogenesis in the mouse embryo. Dev. Dyn. 202, 312–323 (1995).

    Article  CAS  PubMed  Google Scholar 

  86. Maden, M., Sonneveld, E., van der Saag, P. T. & Gale, E. The distribution of endogenous retinoic acid in the chick embryo: implications for developmental mechanisms. Development 125, 4133–4144 (1998).A detailed analysis of endogenous retinoids in the chick embryo, showing the anteroposterior boundary of RA at very early stages, as well as data on later stages.

    Article  CAS  PubMed  Google Scholar 

  87. Ang, H. L., Deltour, L., Hayamizu, T. F., Zgombic-Knight, M. & Duester, G. Retinoic acid synthesis in mouse embryos during gastrulation and craniofacial development linked to class IV alcohol dehydrogenase gene expression. J. Biol. Chem. 271, 9526–9534 (1996).

    Article  CAS  PubMed  Google Scholar 

  88. LaMantia, A. S., Colbert, M. C. & Linney, E. Retinoic acid induction and regional differentiation prefigure olfactory pathway formation in the mammalian forebrain. Neuron 10, 1035–1048 (1993).

    Article  CAS  PubMed  Google Scholar 

  89. Wagner, M., Han, B. & Jessell, T. M. Regional differences in retinoid release from embryonic neural tissue detected by an in vitro reporter assay. Development 116, 55–66 (1992).

    Article  CAS  PubMed  Google Scholar 

  90. Balkan, W., Colbert, M., Bock, C. & Linney, E. Transgenic indicator mice for studying activated retinoic acid receptors during development. Proc. Natl Acad. Sci. USA 89, 3347–3351 (1992).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  91. Mendelsohn, C., Ruberte, E., LeMeur, M., Morriss-Kay, G. & Chambon, P. Developmental analysis of the retinoic acid-inducible RAR-β2 promoter in transgenic animals. Development 113, 723–734 (1991).

    Article  CAS  PubMed  Google Scholar 

  92. Reynolds, K., Mezey, E. & Zimmer, A. Activity of the β-retinoic acid receptor promoter in transgenic mice. Mech. Dev. 36, 15–29 (1991).

    Article  CAS  PubMed  Google Scholar 

  93. Rossant, J., Zirngibl, R., Cado, D., Shago, M. & Giguere, V. Expression of a retinoic acid response element–hsplacZ transgene defines specific domains of transcriptional activity during mouse embryogenesis. Genes Dev. 5, 1333–1344 (1991).

    Article  CAS  PubMed  Google Scholar 

  94. Shen, S., van den Brink, C. E., Kruijer, W. & van der Saag, P. T. Embryonic stem cells stably transfected with mRARb2lacZ exhibit specific expression in chimeric embryos. Int. J. Dev. Biol. 36, 465–476 (1992).

    CAS  PubMed  Google Scholar 

  95. Niederreither, K., McCaffery, P., Drager, U. C., Chambon, P. & Dolle, P. Restricted expression and retinoic acid-induced downregulation of the retinaldehyde dehydrogenase type 2 (RALDH-2) gene during mouse development. Mech. Dev. 62, 67–78 (1997).

    Article  CAS  PubMed  Google Scholar 

  96. Swindell, E. C. et al. Complementary domains of retinoic acid production and degradation in the early chick embryo. Dev. Biol. 216, 282–296 (1999).This study shows that CYP26A1 and RALDH2 form complementary domains to potentially generate a source/sink diffusion gradient of RA across a field of cells that constitute the presumptive hindbrain.

    Article  CAS  PubMed  Google Scholar 

  97. Maden, M. Heads or tails? Retinoic acid will decide. Bioessays 21, 809–812 (1999).

    Article  CAS  PubMed  Google Scholar 

  98. Grapin-Botton, A., Bonnin, M.-A., Sieweke, M. & Le Douarin, N. M. Defined concentrations of a posteriorizing signal are critical for MafB/Kreisler segmental expression in the hindbrain. Development 125, 1173–1181 (1998).

    Article  CAS  PubMed  Google Scholar 

  99. MacLean, G. et al. Cloning of a novel retinoic-acid metabolizing cytochrome P450, Cyp26B1, and comparative expression analysis with Cyp26A1 during early murine development. Mech. Dev. 107, 195–201 (2001).

    Article  CAS  PubMed  Google Scholar 

  100. Berggren, K., McCaffery, P., Drager, U. & Forehand, C. J. Differential distribution of retinoic acid synthesis in the chicken embryo as determined by immunolocalization of the retinoic acid synthetic enzyme, RALDH-2. Dev. Biol. 210, 288–304 (1999).

    Article  CAS  PubMed  Google Scholar 

  101. Mic, F. A., Haselbeck, R. J., Cuenca, A. E. & Duester, G. Novel retinoic acid generating activities in the neural tube and heart identified by conditional rescue of Raldh2 null mutant mice. Development 129, 2271–2282 (2002).

    Article  CAS  PubMed  Google Scholar 

  102. Solomin, L. et al. Retinoid-X receptor signalling in the developing spinal cord. Nature 395, 398–402 (1998).

    Article  CAS  PubMed  Google Scholar 

  103. Grapin-Botton, A., Bonnin, M.-A. & Le Douarin, N. M. Hox gene induction in the neural tube depends on three parameters: competence, signal supply and paralogue group. Development 124, 849–859 (1997).

    Article  CAS  PubMed  Google Scholar 

  104. Itasaki, N., Sharpe, J., Morrison, A. & Krumlauf, R. Reprogramming Hox expression in the vertebrate hindbrain: influence of paraxial mesoderm and rhombomere transposition. Neuron 16, 487–500 (1996).

    Article  CAS  PubMed  Google Scholar 

  105. Gould, A., Itasaki, N. & Krumlauf, R. Initiation of rhombomeric Hoxb4 expression requires induction by somites and a retinoid pathway. Neuron 21, 39–51 (1998).

    Article  CAS  PubMed  Google Scholar 

  106. La Mantia, A.-S. Forebrain induction, retinoic acid, and vulnerability to schizophrenia: insights from molecular and genetic analysis in developing mice. Biol. Psychiatry 46, 19–30 (1999).

    Article  CAS  Google Scholar 

  107. Abu-Abed, S. S. et al. The retinoic acid-metabolising enzyme, CYP26A1, is essential for normal hindbrain patterning, vertebral identity, and development of posterior structures. Genes Dev. 15, 226–240 (2001).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  108. Sakai, Y. et al. The retinoic acid-inactivating enzyme CYP26 is essential for establishing an uneven distribution of retinoic acid along the anterio-posterior axis within the mouse embryo. Genes Dev. 15, 213–225 (2001).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  109. Pierani, A., Brenner-Morton, S., Chiang, C. & Jessell, T. M. A sonic hedgehog-independent, retinoid-activated pathway of neurogenesis in the ventral spinal cord. Cell 97, 903–915 (1999).

    Article  CAS  PubMed  Google Scholar 

  110. Liu, J.-P., Laufer, E. & Jessell, T. M. Assigning the positional identity of spinal motor neurons: rostrocaudal patterning of Hox-c expression by FGFs, Gdf11, and retinoids. Neuron 32, 997–1012 (2001).

    Article  CAS  PubMed  Google Scholar 

  111. Zhao, D. et al. Molecular identification of a major retinoic acid-synthesising enzyme, a retinaldehyde-specific dehydrogenase. Eur. J. Biochem. 240, 15–22 (1996).

    Article  CAS  PubMed  Google Scholar 

  112. Sockanathan, S. & Jessell, T. M. Motor neuron-derived retinoid signalling specifies the subtype identity of spinal motor neurons. Cell 94, 503–514 (1998).This study reveals the role of RA in generating a subset of motor neurons by the paracrine action of RALDH2 in the ventral horn.

    Article  CAS  PubMed  Google Scholar 

  113. McCaffery, P. & Drager, U. C. Hot spots of retinoic acid synthesis in the developing spinal cord. Proc. Natl Acad. Sci. USA 91, 7194–7197 (1994).This paper reveals the presence of a RA-synthesizing enzyme (RALDH1) in a specific set of neurons in the adult brain, and shows that these neurons are involved in Parkinson's disease. Could loss of RALDH1 be an aetiological factor in Parkinson's disease?

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  114. Ensini, M., Tsuchida, T., Betling, H.-G. & Jessell, T. The control of rostrocaudal pattern in the developing spinal cord: specification of motor neuron subtype identity is initiated by signals from paraxial mesoderm. Development 125, 969–982 (1998).

    Article  CAS  PubMed  Google Scholar 

  115. Cullingford, T. E. et al. Distribution of mRNAs encoding the peroxisome proliferator-activated receptor α, β, and γ and the retinoid X receptor α, β, and γ in rat central nervous system. J. Neurochem. 70, 1366–1375 (1998).

    Article  CAS  PubMed  Google Scholar 

  116. Krezel, W., Kastner, P. & Chambon, P. Differential expression of retinoid receptors in the adult mouse central nervous system. Neuroscience 89, 1291–1300 (1999).

    Article  CAS  PubMed  Google Scholar 

  117. Zetterstrom, R. H., Simon, A., Giacobini, M. M. J., Eriksson, U. & Olson, L. Localization of cellular retinoid-binding proteins suggests specific roles for retinoids in the adult central nervous system. Neuroscience 62, 899–918 (1994).

    Article  CAS  PubMed  Google Scholar 

  118. Zetterstrom, R. H. et al. Role of retinoids in the CNS: differential expression of retinoid binding protein and receptors and evidence for presence of retinoic acid. Eur. J. Neurosci. 11, 407–416 (1999).

    Article  CAS  PubMed  Google Scholar 

  119. McCaffery, P. & Drager, U. C. High levels of a retinoic acid-generating dehydrogenase in the meso-telencephalic dopamine system. Proc. Natl Acad. Sci. USA 91, 7772–7776 (1994).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  120. Farooqui, S. M. Induction of adenylyl cyclase sensitive dopamine D2-receptors in retinoic acid induced differentiated human neuroblastoma SHSY-5Y cells. Life Sci. 55, 1887–1893 (1994).

    Article  CAS  PubMed  Google Scholar 

  121. Samad, T. A., Krezel, W., Chambon, P. & Borrelli, E. Regulation of dopaminergic pathways by retinoids: activation of the D2 receptor promoter by members of the retinoic acid receptor-retinoid X receptor family. Proc. Natl Acad. Sci. USA 94, 14349–14354 (1997).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  122. Valdenaire, O., Maus-Moatti, M., Vincent, J. D., Mallet, J. & Vernier, P. Retinoic acid regulates the developmental expression of dopamine D2 receptors in rat striatal primary cultures. J. Neurochem. 71, 929–936 (1998).

    Article  CAS  PubMed  Google Scholar 

  123. Krezel, W. et al. Impaired locomotion and dopamine signalling in retinoid receptor mutant mice. Science 279, 863–867 (1998).

    Article  CAS  PubMed  Google Scholar 

  124. Malaspina, A., Kaushik, N. & De Belleroche, J. Differential expression of 14 genes in amyotrophic lateral sclerosis spinal cord detected using gridded cDNA arrays. J. Neurochem. 77, 132–145 (2001).

    Article  CAS  PubMed  Google Scholar 

  125. Corcoran, J., So, P.-L. & Maden, M. Absence of retinoids can induce motoneuron disease in the adult rat and a retinoid defect is present in motoneuron disease patients. J. Cell Sci. (in the press).

  126. Chiang, M.-Y. et al. An essential role for retinoid receptors RARβ and RXRγ in long-term potentiation and depression. Neuron 21, 1353–1361 (1998).

    Article  CAS  PubMed  Google Scholar 

  127. Goodman, A. B. Three independent lines of evidence suggest retinoids as causal to schizophrenia. Proc. Natl Acad. Sci. USA 95, 7240–7244 (1998).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  128. Goodman, A. B. Chromosomal locations and modes of action of genes of the retinoid (vitamin A) system support their involvement in the etiology of schizophrenia. Am. J. Med. Genet. 60, 335–348 (1995).

    Article  CAS  PubMed  Google Scholar 

  129. Schneider, R. A., Hu, D., Rubenstein, J. L. R., Maden, M. & Helms, J. A. Local retinoid signalling coordinates forebrain and facial morphogenesis by maintaining FGF8 and SHH. Development 128, 2755–2767 (2001).

    Article  CAS  PubMed  Google Scholar 

Download references

Author information

Authors and Affiliations

Authors

Related links

Related links

DATABASES

LocusLink

ADHs

CRABP1

CRABP2

CRBP1

CRBP2

CYP26A1

CYP26B1

Eph

MafB

Meis2

RALDH1

RALDH2

RALDH3

RARα

RARβ

RARγ

RoDHs

RXRα

RXRβ

RXRγ

OMIM

Parkinson's disease

schizophrenia

Swiss-Prot

CEK8

chordin

En2

follistatin

Gli3

Hoxb3

Hoxb4

Hoxb9

Krox20

noggin

Otx2

Pax2

Pax6

XASH-3

X-delta-1

X-MyT1

X-ngnr-1

X-shh

Zic2

FURTHER INFORMATION

Encyclopedia of Life Sciences

vertebrate central nervous system: pattern formation

Glossary

XEROPHTHALMIA

An excessive dryness of the conjunctiva and cornea.

HYDROCEPHALUS

A condition, marked by an expansion of the cerebral ventricles and a compression of neural structures, that is caused by a block in the flow of cerebral spinal fluid or by its overproduction.

SPINA BIFIDA

Failure of neural tube closure at the posterior neuropore, which causes the spinal cord tissue to protrude through the vertebral column.

ANOPHTHALMIA

Absence of the eyeballs.

MICROPHTHALMIA

A reduction in the size of the eyeballs.

EXENCEPHALY

Failure of the cranial component of the neural tube to close.

ENCEPHALOCOEL

A neural tube defect that causes the herniation of brain tissue.

MICROCEPHALY

An abnormally small head, caused by reduced growth of the brain or skull.

TAIL BUD

A blastema-like structure at the posterior end of the embryo that gives rise to all the structures of the sacrocaudal body region: neural tube, notochord, paraxial mesoderm and gut endoderm.

TERATOCARCINOMA CELLS

A cell line derived from a malignant germ-cell tumour, which arises from the ovary or testis and is composed of embryonal carcinoma cells.

NEUROBLASTOMA CELLS

An immortalized cell line derived from tumours that arise from the neural crest.

ANIMAL CAPS

An explant cut from an amphibian embryo at the blastula stage, comprising a 'cap' of about 60° that is centred on the animal pole. These explants consist of uncommitted ectodermal tissue, and they are often used to test the activities of putative neuralizing factors.

GASTRULATION

The process by which the embryo becomes regionalized into three layers: ectoderm, mesoderm and endoderm.

PRONEURAL GENES

Genes that encode transcription factors of the basic helix–loop–helix class that specify neural progenitor cells and promote their differentiation.

PREPATTERN GENES

Also known as pre-proneural genes, these genes seem to provide a link between the patterning of the nervous system and the specification of neurons.

BLASTULA

An embryo before the gastrulation stage, consisting of a hollow ball of epithelial cells that surround a fluid-filled cavity.

NEURULA

The stage of development that follows gastrulation, when the neural plate starts to develop from the ectoderm.

PRIMITIVE STREAK

An elongated depression of reptile, bird and mammalian embryos, through which mesodermal and endodermal cells migrate into the interior of the embryo. The most anterior tip of the primitive streak forms Hensen's node. The streak is functionally homologous to the amphibian blastopore.

OTOCYST

An ectodermal invagination that constitutes the primordium of the internal ear.

NODE

A major organizing centre in primitive-streak-stage embryos that regulates pattern formation. It is known as Hensen's node in chick and the Spemann organizer in frog.

PARAXIAL MESENCHYME

A region of the mesoderm adjacent to the notochord, which becomes segmented rostrocaudally to give rise to the somites early in development.

BRACHIAL

At the level of the forelimbs.

LUMBAR

At the level of the lower back.

SUBSTANTIA NIGRA

A part of the midbrain that contains dopamine-producing neurons, the axons of which innervate the striatum and thereby control body movements.

MORRIS WATER MAZE

A learning task in which an animal is placed in a pool filled with opaque water and has to learn to escape to a hidden platform that is placed at a constant position. The animal must learn to use distal cues, and the spatial relationship between them and the platform. Learning in this task involves the hippocampus.

Rights and permissions

Reprints and permissions

About this article

Cite this article

Maden, M. Retinoid signalling in the development of the central nervous system. Nat Rev Neurosci 3, 843–853 (2002). https://doi.org/10.1038/nrn963

Download citation

  • Issue Date:

  • DOI: https://doi.org/10.1038/nrn963

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