Skip to main content
Log in

Regulation of Cortical Neuron Migration by the Reelin Signaling Pathway

  • Original Paper
  • Published:
Neurochemical Research Aims and scope Submit manuscript

Abstract

Reeler is a mutant mouse with defects in layered structures of the central nervous system, such as the cerebral cortex, hippocampus, and cerebellum, and has been extensively examined for more than half a century. The full-length cDNA for the responsible gene for reeler, reelin, was serendipitously identified, revealing that Reelin encodes a large secreted protein. So far, two Reelin receptors, apolipoprotein E receptor 2 and very low-density lipoprotein receptor, and the cytoplasmic adaptor protein Disabled homolog 1 (Dab1) have been shown to be essential for Reelin signaling. Although a number of downstream cascades of Dab1 have also been reported using various experimental systems, the physiological functions of Reelin in vivo remain controversial. Here, we review recent advances in the understanding of the Reelin-Dab1 signaling pathway in the developing cerebral cortex.

This is a preview of subscription content, log in via an institution to check access.

Access this article

Price excludes VAT (USA)
Tax calculation will be finalised during checkout.

Instant access to the full article PDF.

Fig. 1
Fig. 2

Similar content being viewed by others

References

  1. Tabata H, Kanatani S, Nakajima K (2009) Differences of migratory behavior between direct progeny of apical progenitors and basal progenitors in the developing cerebral cortex. Cereb Cortex 19:2092–2105

    PubMed  Google Scholar 

  2. Marin O, Valiente M, Ge X et al (2010) Guiding neuronal cell migrations. Cold Spring Harb Perspect Biol 2:a001834

    PubMed  Google Scholar 

  3. Nakajima K (2007) Control of tangential/non-radial migration of neurons in the developing cerebral cortex. Neurochem Int 51:121–131

    PubMed  CAS  Google Scholar 

  4. Heng JI, Chariot A, Nguyen L (2010) Molecular layers underlying cytoskeletal remodelling during cortical development. Trends Neurosci 33:38–47

    PubMed  CAS  Google Scholar 

  5. Valiente M, Marin O (2010) Neuronal migration mechanisms in development and disease. Curr Opin Neurobiol 20:68–78

    PubMed  CAS  Google Scholar 

  6. D’Arcangelo G (2005) The reeler mouse: anatomy of a mutant. Int Rev Neurobiol 71:383–417

    PubMed  Google Scholar 

  7. Falconer DS (1951) Two new mutants trembler and reeler, with neurological actionss in the house mouse. J Genet 50:192–201

    Google Scholar 

  8. Hamburgh M (1963) Analysis of the postnatal developmental effects of “Reeler”, a neurological mutation in mice. A study in developmental genetics. Dev Biol 19:165–185

    PubMed  CAS  Google Scholar 

  9. Mikoshiba K, Nagaike K, Kohsaka S et al (1980) Developmental studies on the cerebellum from reeler mutant mouse in vivo and in vitro. Dev Biol 79:64–80

    PubMed  CAS  Google Scholar 

  10. Martin MR (1981) Morphology of the cochlear nucleus of the normal and reeler mutant mouse. J Comp Neurol 197:141–152

    PubMed  CAS  Google Scholar 

  11. Goffinet AM (1983) The embryonic development of the inferior olivary complex in normal and reeler (rlORL) mutant mice. J Comp Neurol 219:10–24

    PubMed  CAS  Google Scholar 

  12. Yip JW, Yip YP, Nakajima K et al (2000) Reelin controls position of autonomic neurons in the spinal cord. Proc Natl Acad Sci U S A 97:8612–8616

    PubMed  CAS  Google Scholar 

  13. Caviness VS Jr, Sidman RL (1973) Time of origin or corresponding cell classes in the cerebral cortex of normal and reeler mutant mice: an autoradiographic analysis. J Comp Neurol 148:141–151

    PubMed  Google Scholar 

  14. Sheppard AM, Pearlman AL (1997) Abnormal reorganization of preplate neurons and their associated extracellular matrix: an early manifestation of altered neocortical development in the reeler mutant mouse. J Comp Neurol 378:173–179

    PubMed  CAS  Google Scholar 

  15. Valverde F, De Carlos JA, Lopez-Mascaraque L (1995) Time of origin and early fate of preplate cells in the cerebral cortex of the rat. Cereb Cortex 5:483–493

    PubMed  CAS  Google Scholar 

  16. Goffinet AM (1979) An early development defect in the cerebral cortex of the reeler mouse. A morphological study leading to a hypothesis concerning the action of the mutant gene. Anat Embryol (Berl) 157:205–216

    CAS  Google Scholar 

  17. Bayer SA, Altman J (1991) Development of the cortical plate. In: Neocortical development. Raven Press, New York, pp 73–81

  18. Caviness VS Jr (1982) Neocortical histogenesis in normal and reeler mice: a developmental study based upon [3H]thymidine autoradiography. Brain Res 256:293–302

    PubMed  Google Scholar 

  19. Pinto Lord MC, Caviness VS Jr (1979) Determinants of cell shape and orientation: a comparative Golgi analysis of cell-axon interrelationships in the developing neocortex of normal and reeler mice. J Comp Neurol 187:49–69

    PubMed  CAS  Google Scholar 

  20. Terashima T, Inoue K, Inoue Y et al (1983) Distribution and morphology of corticospinal tract neurons in reeler mouse cortex by the retrograde HRP method. J Comp Neurol 218:314–326

    PubMed  CAS  Google Scholar 

  21. Terashima T, Inoue K, Inoue Y et al (1985) Distribution and morphology of callosal commissural neurons within the motor cortex of normal and reeler mice. J Comp Neurol 232:83–98

    PubMed  CAS  Google Scholar 

  22. Tabata H, Nakajima K (2002) Neurons tend to stop migration and differentiate along the cortical internal plexiform zones in the Reelin signal-deficient mice. J Neurosci Res 69:723–730

    PubMed  CAS  Google Scholar 

  23. Ogawa M, Miyata T, Nakajima K et al (1995) The reeler gene-associated antigen on Cajal-Retzius neurons is a crucial molecule for laminar organization of cortical neurons. Neuron 14:899–912

    PubMed  CAS  Google Scholar 

  24. Barinaga M (1996) Guiding neurons to the cortex. Science 274:1100–1101

    PubMed  CAS  Google Scholar 

  25. D’Arcangelo G, Miao GG, Chen SC et al (1995) A protein related to extracellular matrix proteins deleted in the mouse mutant reeler. Nature 374:719–723

    PubMed  Google Scholar 

  26. Montgomery JC, Guarnieri MH, Tartaglia KE et al (1994) High-resolution genetic map and YAC contig around the mouse neurological locus reeler. Mamm Genome 5:756–761

    PubMed  CAS  Google Scholar 

  27. Bar I, Lambert De Rouvroit C, Royaux I et al (1995) A YAC contig containing the reeler locus with preliminary characterization of candidate gene fragments. Genomics 26:543–549

    PubMed  CAS  Google Scholar 

  28. Hirotsune S, Takahara T, Sasaki N et al (1995) The reeler gene encodes a protein with an EGF-like motif expressed by pioneer neurons. Nat Genet 10:77–83

    PubMed  CAS  Google Scholar 

  29. D’Arcangelo G, Nakajima K, Miyata T et al (1997) Reelin is a secreted glycoprotein recognized by the CR-50 monoclonal antibody. J Neurosci 17:23–31

    PubMed  Google Scholar 

  30. Hong SE, Shugart YY, Huang DT et al (2000) Autosomal recessive lissencephaly with cerebellar hypoplasia is associated with human RELN mutations. Nat Genet 26:93–96

    PubMed  CAS  Google Scholar 

  31. Kikkawa S, Yamamoto T, Misaki K et al (2003) Missplicing resulting from a short deletion in the reelin gene causes reeler-like neuronal disorders in the mutant shaking rat Kawasaki. J Comp Neurol 463:303–315

    PubMed  CAS  Google Scholar 

  32. Miyata T, Nakajima K, Mikoshiba K et al (1997) Regulation of Purkinje cell alignment by reelin as revealed with CR-50 antibody. J Neurosci 17:3599–3609

    PubMed  CAS  Google Scholar 

  33. Del Rio JA, Heimrich B, Borrell V et al (1997) A role for Cajal-Retzius cells and reelin in the development of hippocampal connections. Nature 385:70–74

    PubMed  CAS  Google Scholar 

  34. Sinagra M, Verrier D, Frankova D et al (2005) Reelin, very-low-density lipoprotein receptor, and apolipoprotein E receptor 2 control somatic NMDA receptor composition during hippocampal maturation in vitro. J Neurosci 25:6127–6136

    PubMed  CAS  Google Scholar 

  35. Nakajima K, Mikoshiba K, Miyata T et al (1997) Disruption of hippocampal development in vivo by CR-50 mAb against reelin. Proc Natl Acad Sci U S A 94:8196–8201

    PubMed  CAS  Google Scholar 

  36. Heinrich C, Nitta N, Flubacher A et al (2006) Reelin deficiency and displacement of mature neurons, but not neurogenesis, underlie the formation of granule cell dispersion in the epileptic hippocampus. J Neurosci 26:4701–4713

    PubMed  CAS  Google Scholar 

  37. Utsunomiya-Tate N, Kubo K, Tate S et al (2000) Reelin molecules assemble together to form a large protein complex, which is inhibited by the function-blocking CR-50 antibody. Proc Natl Acad Sci U S A 97:9729–9734

    PubMed  CAS  Google Scholar 

  38. Kubo K, Mikoshiba K, Nakajima K (2002) Secreted Reelin molecules form homodimers. Neurosci Res 43:381–388

    PubMed  CAS  Google Scholar 

  39. Yoneshima H, Nagata E, Matsumoto M et al (1997) A novel neurological mutant mouse, yotari, which exhibits reeler-like phenotype but expresses CR-50 antigen/reelin. Neurosci Res 29:217–223

    PubMed  CAS  Google Scholar 

  40. Sweet HO, Bronson RT, Johnson KR et al (1996) Scrambler, a new neurological mutation of the mouse with abnormalities of neuronal migration. Mamm Genome 7:798–802

    PubMed  CAS  Google Scholar 

  41. Howell BW, Gertler FB, Cooper JA (1997) Mouse disabled (mDab1): a Src binding protein implicated in neuronal development. EMBO J 16:121–132

    PubMed  CAS  Google Scholar 

  42. Howell BW, Hawkes R, Soriano P et al (1997) Neuronal position in the developing brain is regulated by mouse disabled-1. Nature 389:733–737

    PubMed  CAS  Google Scholar 

  43. Sheldon M, Rice DS, D’Arcangelo G et al (1997) Scrambler and yotari disrupt the disabled gene and produce a reeler- like phenotype in mice. Nature 389:730–733

    PubMed  CAS  Google Scholar 

  44. Ware ML, Fox JW, Gonzalez JL et al (1997) Aberrant splicing of a mouse disabled homolog, mdab1, in the scrambler mouse. Neuron 19:239–249

    PubMed  CAS  Google Scholar 

  45. Kojima T, Nakajima K, Mikoshiba K (2000) The disabled 1 gene is disrupted by a replacement with L1 fragment in yotari mice. Brain Res Mol Brain Res 75:121–127

    PubMed  CAS  Google Scholar 

  46. Rice DS, Sheldon M, D’Arcangelo G et al (1998) Disabled-1 acts downstream of Reelin in a signaling pathway that controls laminar organization in the mammalian brain. Development 125:3719–3729

    PubMed  CAS  Google Scholar 

  47. Howell BW, Herrick TM, Cooper JA (1999) Reelin-induced tryosine phosphorylation of disabled 1 during neuronal positioning. Genes Dev 13:643–648

    PubMed  CAS  Google Scholar 

  48. Howell BW, Herrick TM, Hildebrand JD et al (2000) Dab1 tyrosine phosphorylation sites relay positional signals during mouse brain development. Curr Biol 10:877–885

    PubMed  CAS  Google Scholar 

  49. Honda T, Nakajima K (2006) Mouse Disabled1 (DAB1) is a nucleocytoplasmic shuttling protein. J Biol Chem 281:38951–38965

    PubMed  CAS  Google Scholar 

  50. Trommsdorff M, Gotthardt M, Hiesberger T et al (1999) Reeler/Disabled-like disruption of neuronal migration in knockout mice lacking the VLDL receptor and ApoE receptor 2. Cell 97:689–701

    PubMed  CAS  Google Scholar 

  51. D’Arcangelo G, Homayouni R, Keshvara L et al (1999) Reelin is a ligand for lipoprotein receptors. Neuron 24:471–479

    PubMed  Google Scholar 

  52. Hiesberger T, Trommsdorff M, Howell BW et al (1999) Direct binding of Reelin to VLDL receptor and ApoE receptor 2 induces tyrosine phosphorylation of disabled-1 and modulates tau phosphorylation. Neuron 24:481–489

    PubMed  CAS  Google Scholar 

  53. Yasui N, Nogi T, Takagi J (2010) Structural basis for specific recognition of reelin by its receptors. Structure 18:320–331

    PubMed  CAS  Google Scholar 

  54. Weeber EJ, Beffert U, Jones C et al (2002) Reelin and ApoE receptors cooperate to enhance hippocampal synaptic plasticity and learning. J Biol Chem 277:39944–39952

    PubMed  CAS  Google Scholar 

  55. Forster E, Tielsch A, Saum B et al (2002) Reelin, Disabled 1, and beta 1 integrins are required for the formation of the radial glial scaffold in the hippocampus. Proc Natl Acad Sci U S A 99:13178–13183

    PubMed  CAS  Google Scholar 

  56. Luque JM, Morante-Oria J, Fairen A (2003) Localization of ApoER2, VLDLR and Dab1 in radial glia: groundwork for a new model of reelin action during cortical development. Brain Res Dev Brain Res 140:195–203

    PubMed  CAS  Google Scholar 

  57. Hartfuss E, Forster E, Bock HH et al (2003) Reelin signaling directly affects radial glia morphology and biochemical maturation. Development 130:4597–4609

    PubMed  CAS  Google Scholar 

  58. Niu S, Renfro A, Quattrocchi CC et al (2004) Reelin promotes hippocampal dendrite development through the VLDLR/ApoER2-Dab1 pathway. Neuron 41:71–84

    PubMed  CAS  Google Scholar 

  59. Olson EC, Kim S, Walsh CA (2006) Impaired neuronal positioning and dendritogenesis in the neocortex after cell-autonomous Dab1 suppression. J Neurosci 26:1767–1775

    PubMed  CAS  Google Scholar 

  60. Cooper JA (2008) A mechanism for inside-out lamination in the neocortex. Trends Neurosci 31:113–119

    PubMed  CAS  Google Scholar 

  61. Chai X, Forster E, Zhao S et al (2009) Reelin stabilizes the actin cytoskeleton of neuronal processes by inducing n-Cofilin phosphorylation at Serine3. J Neurosci 29:288–299

    PubMed  CAS  Google Scholar 

  62. Pujadas L, Gruart A, Bosch C et al (2010) Reelin regulates postnatal neurogenesis and enhances spine hypertrophy and long-term potentiation. J Neurosci 30:4636–4649

    PubMed  CAS  Google Scholar 

  63. Pearlman AL, Sheppard AM (1996) Extracellular matrix in early cortical development. Prog Brain Res 108:117–134

    PubMed  CAS  Google Scholar 

  64. Frotscher M (1997) Dual role of Cajal-Retzius cells and reelin in cortical development. Cell Tissue Res 290:315–322

    PubMed  CAS  Google Scholar 

  65. Dulabon L, Olson EC, Taglienti MG et al (2000) Reelin binds alpha3beta1 integrin and inhibits neuronal migration. Neuron 27:33–44

    PubMed  CAS  Google Scholar 

  66. Sanada K, Gupta A, Tsai LH (2004) Disabled-1-regulated adhesion of migrating neurons to radial glial fiber contributes to neuronal positioning during early corticogenesis. Neuron 42:197–211

    PubMed  CAS  Google Scholar 

  67. Belvindrah R, Graus-Porta D, Goebbels S et al (2007) Beta1 integrins in radial glia but not in migrating neurons are essential for the formation of cell layers in the cerebral cortex. J Neurosci 27:13854–13865

    PubMed  CAS  Google Scholar 

  68. Graus-Porta D, Blaess S, Senften M et al (2001) Beta1-class integrins regulate the development of laminae and folia in the cerebral and cerebellar cortex. Neuron 31:367–379

    PubMed  CAS  Google Scholar 

  69. Magdaleno S, Keshvara L, Curran T (2002) Rescue of ataxia and preplate splitting by ectopic expression of reelin in reeler mice. Neuron 33:573–586

    PubMed  CAS  Google Scholar 

  70. Tissir F, Goffinet AM (2003) Reelin and brain development. Nat Rev Neurosci 4:496–505

    PubMed  CAS  Google Scholar 

  71. Meyer G, Cabrera Socorro A, Perez Garcia CG et al (2004) Developmental roles of p73 in Cajal-Retzius cells and cortical patterning. J Neurosci 24:9878–9887

    PubMed  CAS  Google Scholar 

  72. Jossin Y, Ignatova N, Hiesberger T et al (2004) The central fragment of Reelin, generated by proteolytic processing in vivo, is critical to its function during cortical plate development. J Neurosci 24:514–521

    PubMed  CAS  Google Scholar 

  73. Yoshida M, Assimacopoulos S, Jones KR et al (2006) Massive loss of Cajal-Retzius cells does not disrupt neocortical layer order. Development 133:537–545

    PubMed  CAS  Google Scholar 

  74. Miyata T, Nakajima K, Aruga J et al (1996) Distribution of a reeler gene-related antigen in the developing cerebellum: an immunohistochemical study with an allogeneic antibody CR-50 on normal and reeler mice. J Comp Neurol 372:215–228

    PubMed  CAS  Google Scholar 

  75. Kubo K, Honda T, Tomita K et al (2010) Ectopic Reelin induces neuronal aggregation with a normal birthdate-dependent “inside-out” alignment in the developing neocortex. J Neurosci 30:10953–10966

    PubMed  CAS  Google Scholar 

  76. Uchida T, Baba A, Perez-Martinez FJ et al (2009) Downregulation of functional Reelin receptors in projection neurons implies that primary Reelin action occurs at early/premigratory stages. J Neurosci 29:10653–10662

    PubMed  CAS  Google Scholar 

  77. Hammond V, Howell B, Godinho L et al (2001) disabled-1 functions cell autonomously during radial migration and cortical layering of pyramidal neurons. J Neurosci 21:8798–8808

    PubMed  CAS  Google Scholar 

  78. Morimura T, Ogawa M (2009) Relative importance of the tyrosine phosphorylation sites of Disabled-1 to the transmission of Reelin signaling. Brain Res 1304:26–37

    PubMed  CAS  Google Scholar 

  79. Simo S, Jossin Y, Cooper JA (2010) Cullin 5 regulates cortical layering by modulating the speed and duration of Dab1-dependent neuronal migration. J Neurosci 30:5668–5676

    PubMed  CAS  Google Scholar 

  80. Keshvara L, Benhayon D, Magdaleno S et al (2001) Identification of reelin-induced sites of tyrosyl phosphorylation on disabled 1. J Biol Chem 276:16008–16014

    PubMed  CAS  Google Scholar 

  81. Feng L, Cooper JA (2009) Dual functions of Dab1 during brain development. Mol Cell Biol 29:324–332

    PubMed  CAS  Google Scholar 

  82. Bock HH, Jossin Y, Liu P et al (2003) Phosphatidylinositol 3-kinase interacts with the adaptor protein Dab1 in response to Reelin signaling and is required for normal cortical lamination. J Biol Chem 278:38772–38779

    PubMed  CAS  Google Scholar 

  83. Feng L, Allen NS, Simo S et al (2007) Cullin 5 regulates Dab1 protein levels and neuron positioning during cortical development. Genes Dev 21:2717–2730

    PubMed  CAS  Google Scholar 

  84. Pramatarova A, Ochalski PG, Chen K et al (2003) Nck beta interacts with tyrosine-phosphorylated disabled 1 and redistributes in Reelin-stimulated neurons. Mol Cell Biol 23:7210–7221

    PubMed  CAS  Google Scholar 

  85. Assadi AH, Zhang G, Beffert U et al (2003) Interaction of reelin signaling and Lis1 in brain development. Nat Genet 35:270–276

    PubMed  CAS  Google Scholar 

  86. Ballif BA, Arnaud L, Arthur WT et al (2004) Activation of a Dab1/CrkL/C3G/Rap1 pathway in Reelin-stimulated neurons. Curr Biol 14:606–610

    PubMed  CAS  Google Scholar 

  87. Chen K, Ochalski PG, Tran TS et al (2004) Interaction between Dab1 and CrkII is promoted by Reelin signaling. J Cell Sci 117:4527–4536

    PubMed  CAS  Google Scholar 

  88. Huang Y, Magdaleno S, Hopkins R et al (2004) Tyrosine phosphorylated Disabled 1 recruits Crk family adapter proteins. Biochem Biophys Res Commun 318:204–212

    PubMed  CAS  Google Scholar 

  89. Kuo G, Arnaud L, Kronstad-O’Brien P et al (2005) Absence of Fyn and Src causes a reeler-like phenotype. J Neurosci 25:8578–8586

    PubMed  CAS  Google Scholar 

  90. Park TJ, Curran T (2008) Crk and Crk-like play essential overlapping roles downstream of disabled-1 in the Reelin pathway. J Neurosci 28:13551–13562

    PubMed  CAS  Google Scholar 

  91. Thomas SM, Brugge JS (1997) Cellular functions regulated by Src family kinases. Annu Rev Cell Dev Biol 13:513–609

    PubMed  CAS  Google Scholar 

  92. Jossin Y, Ogawa M, Metin C et al (2003) Inhibition of SRC family kinases and non-classical protein kinases C induce a reeler-like malformation of cortical plate development. J Neurosci 23:9953–9959

    PubMed  CAS  Google Scholar 

  93. Arnaud L, Ballif BA, Forster E et al (2003) Fyn tyrosine kinase is a critical regulator of Disabled-1 during brain development. Curr Biol 13:9–17

    PubMed  CAS  Google Scholar 

  94. Bock HH, Herz J (2003) Reelin activates SRC family tyrosine kinases in neurons. Curr Biol 13:18–26

    PubMed  CAS  Google Scholar 

  95. Feller SM (2001) Crk family adaptors-signalling complex formation and biological roles. Oncogene 20:6348–6371

    PubMed  CAS  Google Scholar 

  96. Tabata H, Nakajima K (2003) Multipolar migration: the third mode of radial neuronal migration in the developing cerebral cortex. J Neurosci 23:9996–10001

    PubMed  CAS  Google Scholar 

  97. Voss AK, Britto JM, Dixon MP et al (2008) C3G regulates cortical neuron migration, preplate splitting and radial glial cell attachment. Development 135:2139–2149

    PubMed  CAS  Google Scholar 

  98. Hashimoto-Torii K, Torii M, Sarkisian MR et al (2008) Interaction between Reelin and Notch signaling regulates neuronal migration in the cerebral cortex. Neuron 60:273–284

    PubMed  CAS  Google Scholar 

  99. Gaiano N, Fishell G (2002) The role of notch in promoting glial and neural stem cell fates. Annu Rev Neurosci 25:471–490

    PubMed  CAS  Google Scholar 

  100. Yoon K, Gaiano N (2005) Notch signaling in the mammalian central nervous system: insights from mouse mutants. Nat Neurosci 8:709–715

    PubMed  CAS  Google Scholar 

  101. Fairen A, Cobas A, Fonseca M (1986) Times of generation of glutamic acid decarboxylase immunoreactive neurons in mouse somatosensory cortex. J Comp Neurol 251:67–83

    PubMed  CAS  Google Scholar 

  102. Peduzzi JD (1988) Genesis of GABA-immunoreactive neurons in the ferret visual cortex. J Neurosci 8:920–931

    PubMed  CAS  Google Scholar 

  103. Yozu M, Tabata H, Nakajima K (2004) Birth-date dependent alignment of GABAergic neurons occurs in a different pattern from that of non-GABAergic neurons in the developing mouse visual cortex. Neurosci Res 49:395–403

    PubMed  CAS  Google Scholar 

  104. Hevner RF, Daza RA, Englund C et al (2004) Postnatal shifts of interneuron position in the neocortex of normal and reeler mice: evidence for inward radial migration. Neuroscience 124:605–618

    PubMed  CAS  Google Scholar 

  105. Hammond V, So E, Gunnersen J et al (2006) Layer positioning of late-born cortical interneurons is dependent on Reelin but not p35 signaling. J Neurosci 26:1646–1655

    PubMed  CAS  Google Scholar 

  106. Pla R, Borrell V, Flames N et al (2006) Layer acquisition by cortical GABAergic interneurons is independent of Reelin signaling. J Neurosci 26:6924–6934

    PubMed  CAS  Google Scholar 

Download references

Acknowledgments

This work was supported by the Strategic Research Program for Brain Sciences (“Understanding of molecular and environmental bases for brain health”), the Ministry of Education, Culture, Sports, Science, and Technology of Japan, the Japan Society for the Promotion of Science, the Japan Brain Foundation, the Takeda Science Foundation, the Keio Gijuku Academic Development Funds, and the Promotion and Mutual Aid Corporation for Private Schools of Japan.

Author information

Authors and Affiliations

Authors

Corresponding author

Correspondence to Kazunori Nakajima.

Additional information

Special Issue: In Honor of Dr.Mikoshiba.

Rights and permissions

Reprints and permissions

About this article

Cite this article

Honda, T., Kobayashi, K., Mikoshiba, K. et al. Regulation of Cortical Neuron Migration by the Reelin Signaling Pathway. Neurochem Res 36, 1270–1279 (2011). https://doi.org/10.1007/s11064-011-0407-4

Download citation

  • Accepted:

  • Published:

  • Issue Date:

  • DOI: https://doi.org/10.1007/s11064-011-0407-4

Keywords

Navigation