Doublecortin is necessary for the migration of adult subventricular zone cells from neurospheres

https://doi.org/10.1016/j.mcn.2006.06.014Get rights and content

Abstract

Mutations in human doublecortin (DCX) and knockdown of Dcx in rodents cause radial migration defects in the embryonic cerebral cortex. However, the brain phenotype of Dcx knockout mice is largely normal suggesting that Dcx is not necessary for most migration events. Adult subventricular zone (SVZ) cells migrate tangentially in the rostral migratory stream to the olfactory bulbs. Dcx is expressed in the SVZ but it is unknown if it is necessary for migration. We show that Dcx RNAi reduced SVZ cell migration in vitro, both cell autonomously and non-cell autonomously. Overexpression of Dcx rescued migration after knockdown, but did not increase migration by itself. Thus, Dcx is necessary not only for embryonic radial migration but also migration of adult SVZ cells.

Introduction

Doublecortin is a 40-kDa protein specifically expressed in the leading processes of migrating neurons (Friocourt et al., 2003). Although DCX functions as a microtubule-associated protein (MAP) (Francis et al., 1999, Gleeson et al., 1999), its specific mechanism of action in neuronal migration is still poorly understood. Mutations in the human DCX gene (which is X-linked) cause lissencephaly in males and a less severe phenotype, subcortical laminar heterotopia (SCLH), in heterozygous females (des Portes et al., 1998b, Gleeson et al., 1998). Because of random X-inactivation in females, migrating neurons are separated according to which copy of DCX is active. Therefore, in SCLH, the cerebral cortex appears normal, but a second layer of cortical neurons remain in the subcortical white matter. The hippocampus of both heterozygous females and hemizygous male mice with a targeted mutation in the Dcx gene shows mildly disrupted lamination in the CA3 region (Corbo et al., 2002). Nevertheless, the cerebral cortex and other brain areas, including the olfactory bulb, appear to be normal, suggesting that Dcx is not necessary for all types of neuronal migration and that similar genes have redundant functions (Corbo et al., 2002, Deuel et al., 2006, Koizumi et al., 2006).

Interestingly, when Dcx levels are knocked down at E14 with in utero electroporation of short hairpin RNAs (shRNAs) targeted to Dcx mRNA, radial migration of cortical neurons decreases (Bai et al., 2003). Four days after Dcx RNAi, the majority of electroporated cortical neurons have prematurely terminated migration within the intermediate zone. They remain there, and postnatally produce the characteristic band of ectopic neurons seen in SCLH. Dcx also seems to modulate cell-to-cell communication; neurons that did not take up the shRNA plasmids were unable to migrate through the field of cells affected by RNAi, but were able to migrate past regions with normal cells (Bai et al., 2003). This suggests that Dcx works both cell autonomously and in a non-cell autonomous fashion.

Dcx is expressed in the adult SVZ by neuroblasts that migrate to the olfactory bulbs (Gleeson et al., 1999, Nacher et al., 2001, Yang et al., 2004). The olfactory bulb is normal in the Dcx knockout mouse, suggesting that Dcx is not necessary for SVZ motility. Dcx is also expressed in the neurogenic dentate gyrus and in the piriform cortex (Nacher et al., 2001), regions in which there is little to no migration, suggesting that it may have functions not related to motility. Adult SVZ cell migration is different from embryonic radial migration in the cerebral cortex in a number of ways. (1) SVZ cells migrate much longer distances on average (Lois and Alvarez-Buylla, 1994). (2) SVZ neuroblasts migrate as longitudinal arrays of cells (chains), whereas cortical neurons migrate individually (Lois et al., 1996). (3) SVZ cells migrate through a meshwork of specialized astrocyte processes and extracellular matrix (tangential migration); cortical migration occurs along radial glia (radial migration) (Thomas et al., 1996). (4) SVZ neuroblasts are capable of proliferation, but migrating cortical neurons are postmitotic (Thomas et al., 1996). SVZ cells can be cultured as floating neurospheres which, when plated migrate away from the neurospheres. Therefore, we decided to use this system to ask if Dcx knockdown also diminishes SVZ cell migration. We cultured SVZ cells as neurospheres from adult mice and showed that knockdown of Dcx by RNAi significantly decreases migration of cells, in both a cell autonomous and non cell-autonomous fashion.

Section snippets

Doublecortin mRNA in the adult subventricular zone

Anti-Dcx immunoreactivity is found in migrating neuroblasts of the adult brain SVZ, and is downregulated during granule and periglomerular neuron differentiation in the olfactory bulb (Brown et al., 2003, Gleeson et al., 1999, Nacher et al., 2001, Yang et al., 2004). To further characterize the developmental expression of Dcx mRNA, we used reverse transcriptase-polymerase chain reaction (RT-PCR) to confirm its expression in the adult SVZ (Fig. 1A). A major band of approximately 1000 bp was

Discussion

This study revealed the following novel observations. Dcx mRNA is identical in embryonic and adult migrating neuroblasts. Dcx is required for migration of SVZ cells from neurospheres and Dcx’s effects on adult SVZ migration are both cell autonomous and non-cell autonomous. Overexpression of Dcx does not increase SVZ migration but rescues the migration lost after knockdown. Since adult SVZ migration is different in multiple respects from embryonic cortical migration, these results expand the

Plasmid constructs

The molecular and cellular methods used in this study followed standard protocols.

Three RNAi constructs were the kind gift of Dr. Joseph LoTurco (University of Connecticut) and were described previously (Bai et al., 2003). The shRNA sequences targeted to different sequences on the Dcx transcript, driven by the mouse U6 promoter, were inserted into the mU6pro vector: Doublecortin (DCX) 3′ untranslated region (DCX 3′UTR) hairpin RNAi sequence (DCX 3′UTRhp; 5′-GCUCAAGUGACCAACAAGGCUAUAGACACA

Acknowledgments

The authors would like to thank Joe LoTurco for the Dcx RNAi plasmids. We thank Honglin Li and Christine DiDonato for advice throughout the project. We would also like to thank, Hans-Georg Simon, Jill Morris, and members of the Szele laboratory for critical readings of the manuscript. FGS supported by NIH RO1 NS/AG42253.

References (44)

  • P. Rakic

    Guidance of neurons migrating to the fetal monkey neocortex

    Brain Res.

    (1971)
  • B.T. Schaar et al.

    Doublecortin microtubule affinity is regulated by a balance of kinase and phosphatase activity at the leading edge of migrating neurons

    Neuron

    (2004)
  • T. Tanaka et al.

    Cdk5 phosphorylation of doublecortin ser297 regulates its effect on neuronal migration

    Neuron

    (2004)
  • H. Wichterle et al.

    Direct evidence for homotypic, glia-independent neuronal migration

    Neuron

    (1997)
  • S.A. Anderson et al.

    Interneuron migration from basal forebrain to neocortex: dependence on Dlx genes

    Science

    (1997)
  • J. Bai et al.

    RNAi reveals doublecortin is required for radial migration in rat neocortex

    Nat. Neurosci.

    (2003)
  • A.J. Bolteus et al.

    GABA release and uptake regulate neuronal precursor migration in the postnatal subventricular zone

    J. Neurosci.

    (2004)
  • J.P. Brown et al.

    Transient expression of doublecortin during adult neurogenesis

    J. Comp. Neurol.

    (2003)
  • G. Chazal et al.

    Consequences of neural cell adhesion molecule deficiency on cell migration in the rostral migratory stream of the mouse

    J. Neurosci.

    (2000)
  • J.C. Corbo et al.

    Doublecortin is required in mice for lamination of the hippocampus but not the neocortex

    J. Neurosci.

    (2002)
  • V. des Portes et al.

    Doublecortin is the major gene causing X-linked subcortical laminar heterotopia (SCLH)

    Hum. Mol. Genet.

    (1998)
  • V. des Portes et al.

    A novel CNS gene required for neuronal migration and involved in X-linked subcortical laminar heterotopia and lissencephaly syndrome

    Cell

    (1998)
  • Cited by (39)

    • BKM120 alters the migration of doublecortin-positive cells in the dentate gyrus of mice

      2022, Pharmacological Research
      Citation Excerpt :

      For instance, DCX is not just a marker of migrating neuroblasts/immature neurons but also regulates migration by interacting with microtubules [23]. In this context, the genetic inhibition of DCX has been reported to alter the migration of neuroblasts during adult neurogenesis [24,25]. DCX activity is regulated by the phosphorylation of several serine residues within its C-terminus, which can be mediated by c-Jun N-terminal kinase (JNK) or Cdk5 [26–28].

    • Crosstalk between anticancer drugs and mitochondrial functions

      2021, Current Research in Pharmacology and Drug Discovery
      Citation Excerpt :

      Mutation in double cortin causes defective neuronal migration. The expression level of double cortin is mainly seen in the neurogenic zone such as the subgranular and subventricular zone (Gleeson et al., 1999; Rivest, 2006; Tint et al., 2009; Ocbina et al., 2006). The nerve growth factor BDNF belongs to the family of neurotrophins, which has a dominant role in the brain, which promotes the survival, growth, maturation, and axonal plasticity of neurons.

    • Mutations in USP9X are associated with x-linked intellectual disability and disrupt neuronal cell migration and growth

      2014, American Journal of Human Genetics
      Citation Excerpt :

      We isolated neural progenitor cells (NPCs) from both wild-type and knockout embryonic brains and grew them in vitro as nonadherent neurosphere cultures. Next we employed an in vitro neuronal migration assay, wherein neurospheres are adhered to a poly-L-lysine substrate and the migration of neurons outward from the sphere boundary is recorded.15–17 We found a highly significant decrease in the migration of neurons from the neurospheres in the absence of Usp9x (Figure 2).

    • Systemic hypoxia differentially affects neurogenesis during early mouse brain maturation

      2012, Brain and Development
      Citation Excerpt :

      Disrupted microtubule development and anomalies of cortical structure such as lissencephaly in males and subcortical heterotopy in females caused by DCX gene mutations reflect the essential role of DCX for physiological brain development [29,32]. In addition, DCX plays a role in adult neurogenesis of the hippocampus and subventricular zone [30,31]. Under conditions of perinatal cerebral hypoxia/ischemia, activation of neurogenesis estimated from accumulation of DCX-positive cells has been shown in postnatal (P6) rat brain [33].

    • Regulation of adult neural precursor cell migration

      2011, Neurochemistry International
      Citation Excerpt :

      This phenotype ultimately amounts to a delay of cell migration and accumulation of neuroblasts within the RMS (Kappeler et al., 2006; Koizumi et al., 2006). In vitro RNA interference data also supported the role of DCX in SVZ cell migration, as specific knockdown of DCX obstructed cell migration, which could be reverted by reintroduction of DCX (Ocbina et al., 2006). Slit proteins, a family of molecules known to chemo-repel axons (Li et al., 1999; Nguyen Ba-Charvet et al., 1999), are expressed in neonatal and adult brains.

    View all citing articles on Scopus
    View full text