Vous êtes sur la page 1sur 12

Current Molecular Medicine 2001, 1, 677-688 677

Molecular Mechanisms of Neuronal Migration Disorders, Quo Vadis?

S. Couillard-Despres1,2, J. Winkler2,3, G. Uyanik2 and L. Aigner*1,2

1Volkswagen Foundation Junior Group, University of Regensburg, Regensburg, Germany


2Department of Neurology, University of Regensburg, Regensburg, Germany
3Department of Neurosciences, University of California at San Diego, La Jolla, USA

Abstract: Following terminal mitosis, neuronal precursor cells leave their site of origin and migrate
towards their definitive site of residency. In order to establish the intricate cytoarchitecture described
in the adult human brain, neuronal migration must be finely regulated. In humans, brain malformations
can result from neuronal migration defects. The spectrum of migration disorder severity extends from
few heterotopic neurons, as observed in periventricular heterotopia, to a complete cortical
disorganization, as observed in cases of lissencephaly. Recently, specific migration disorders have
been linked to mutations/deletions in the doublecortin , filamin-1, LIS1 and reelin genes. These proteins
act at different levels of the signaling cascades transducing extracellular guiding cues into
cytoskeletal reorganization. Here, we summarize the data concerning these four molecules and
speculate on their functions and interaction partners during neuronal development.

INTRODUCTION disorders with genetic defects. Several genes have


been linked over recent years to neuronal migration
The adult human brain is an organ bearing a disorders, but their function in normal and
fascinating degree of complexity. The establish- pathological conditions is not fully elucidated. In
ment of the three-dimensional topography this review, we will summarize the recent findings
described in the adult brain requires a tight control on genes involved in human neuronal migration
of neural progenitor cell proliferation and disorders in the context of cortical development.
differentiation as well as a precise guidance of the
neuroblasts from their site of origin to their final
destination. The cerebral cortex, with its six layered DEVELOPMENT OF THE CEREBRAL CORTEX
structure, is an area of the brain where the
developmental program must be particularly fine Cell Proliferation
tuned. Following their last mitosis in the ventricular
zone, the future cortical neurons migrate towards Many neurons present in the neocortex originate
the pial surface, through distances corresponding to from the pseudostratified ventricular epithelium that
hundreds of times their own size. Extracellular cues lines the embryonic cerebral ventricle wall [1-3].
from the surrounding extracellular matrix and Prior to neurogenesis, the cells from this epithelium
neighboring cells, as well as the internal proliferate and undergo mitosis in a symmetric
cytoskeletal machinery, are required to achieve a mode, resulting in two identical daughter cells that
proper migration. Genetic defects in any of these will continue to proliferate. At the onset of
components can provoke neuronal migration neurogenesis, on embryonic day 11 in mouse and
disorders. These disorders cover a wide spectrum of approximately the 5th week of gestation in human
severity. In the milder forms, the affected [4, 5], some of the mitoses occur in an asymmetric
individuals have normal cognitive functions and mode. In the present situation, the process results in
rare episodes of epilepsy, whereas the most severely one cell maintaining the "mother cell"
affected cases will suffer from intractable epilepsy, characteristics, the so-called stem cell, and a sister
severe mental retardation and shortened life span. cell which will not undergo further mitosis, but will
The recent progress in routinely performed rather migrate towards the pial aspect of the
neuroimaging techniques allowed for a more developing cortex. The proportion of asymmetric
distinct classification of developmental brain division increases during the entire time of
aberrations and facilitated the linkage of specific neurogenesis [2], which ends at approximately
embryonic day 17 in mouse and the 5th month of
gestation in human [4, 6]. Further proliferation of
*Address correspondence to this author at the Department of Neurology, the stem cells essentially results in an increase in
VW-Foundation Junior Group, University of Regensburg, Franz-Josef- the glial cell population. The transition from
Strauss-Allee 11, D-93053 Regensburg, Germany; Tel: (+49) (941) 944 symmetric to asymmetric division implicates that
8950; Fax: (+49) (941) 944 8951; the proliferation before the onset of neurogenesis
email: ludwig.aigner@klinik.uni-regensburg.de

1566-5240/01 $20.00+.00 2001 Bentham Science Publishers Ltd.


678 Current Molecular Medicine, 2001, Vol. 1, No. 6 Couillard-Despres et al.

enlarges the surface of the future cortex, whereas their final site of residency. During corticogenesis,
the proliferation during the neurogenic period one mode of neuroblasts migration is the so-called
influences the thickness of the future cortex [7, 8]. A "radial migration" [4, 12, 13]. This process is
recent study using retroviral infection for clonal characterized by the use of radial glia as guiding
lineage analysis demonstrated that radial glia tracks for migrating neurons. In contrast, the
constitute the neural stem cell population during tangential migration does not rely on radial glia
the period of corticogenesis [9]. These bipolar cells guidance. This type of migration is responsible for
have their somata in the ventricular zone and the lateral dispersion of clonally related neurons
extend two fibers reaching the ventricular wall and within the developing cortex and also for the
the pial surface. During mitosis, the somata of the colonization of the latter by neurons originating
radial glia migrate to the luminal surface where from the ganglionic eminence [14-18]. In this
cells divide. The somata of the two daughter cells review, we will mainly address issues related to
then return to more basal regions of the ventricular radial migration. In humans, neuroblast migration
zone, according to a process called interkinetic mainly occurs between the 8th and the 24th week
nuclear migration. Surprisingly, the radial glia do of gestation [4]. During this process, the neuroblasts
not need to retract their processes in order to migrate along their "mother" radial glia towards the
undergo mitosis [9]. In cases of asymmetric division, pia [9, 12, 19]. Extracellular molecules, such as
uneven distribution of certain molecules along the laminin, fibronectin, astrotactin and cerebroglycan,
basal-apical axis of radial glia, such as Notch1 [10] are thought to be particularly important for the
or Numb [11], is thought to be responsible for the interaction between neuroblasts and radial glia [20-
differential fate of the daughter cells. 23].

The first step in cortical development is the


Neuronal Cell Migration establishment of a pre-plate (Fig. (1)). This early
structure, located above the ventricular zone, can
Following terminal mitosis, neuroblasts must be observed during the 5th to 8th week of gestation.
migrate considerable distances in order to reach It consists of the first wave of post-mitotic

Figure (1). Development of the cerebral cortex.


Neuroepithelial cells constitute the pseudostratified ventricular epithelium lining the wall of the neural tube. The radial
oriented progenitor cells (blue) divide according to a mode typical for pseudostratified epithelia termed interkinetic nuclear
migration. The cell cycle is closely associated with the repetitive migration of the nuclei between the basal (facing the
future pial surface) and the apical (facing the lumen of the ventricle) part of the ventricular zone. Symmetric division
produces identical progeny and increases the number of neural progenitor cells resulting in an expansion of the surface
area of the ventricular zone. At the fifth week of gestation, cells start to divide asymmetrically and produce postmitotic
neuronal precursor cells (orange) that will migrate along the radial process of their mother cells (yellow) towards the pial
surface. The first waves of neuronal precursors migrate to the surface to form the preplate (PP). Subsequently, migrating
neuronal precursors form the cortical plate by separating the pre-plate into the marginal zone (MZ) (future layer I) and the
subplate (SP). Additional neural precursors migrate into the cortical plate and generate the layers II to VI. In the adult, the
ventricle is lined by the ependymal cell layer (E). Radial glial cells have differentiated into astrocytes (not shown) and a
band of white matter containing axons and oligodendrocytes (W) separates the cortex from the ventricle.
Neuronal Migration Disorders, Quo Vadis Current Molecular Medicine, 2001, Vol. 1, No. 6 679

neuroblasts [5]. The following wave of neuroblasts thickened and typically lacks or bears a significant
migrating into the neocortex will intercalate in this reduction of gyri (Fig. (2d)). In type 1 lissencephaly,
pre-plate to form a structure called the cortical also named classical lissencephaly, the cerebral
plate. The pre-plate is thereby bisected in two wall architecture is similar to the one observed in a
entities, the marginal zone and the sub-plate (Fig. 12 week-old foetus [25]. Instead of the characteristic
(1)). Pial to the cortical plate, the marginal zone six cortical layers, only four layers can be
contains the Cajal-Retzius neurons that will form the described. The outermost layer, the molecular
future layer 1 of the mature neocortex. As layer, consists of the former marginal zone, which is
neuroblasts initiate their migration in the ventricular settled on a layer of neurons, resembling those
zone, the radial glia fibers on which they migrate normally found in the deepest layers of the
are arranged into fascicles. However, the radial glia neocortex. Underneath is a cell-sparse layer, and
processes defasciculate at the level of the sub-plate the deepest layer consists in columns of
and penetrate into the cortical plate as single fibers heterotropic neurons [27]. A fine ribbon of white
[24]. Interestingly, this level is an important site of matter separates the gray matter from the
neuronal heterotopia in neuronal migration ventricular region. The neurons forming the
disorders. heterotopia are likely to have arrested during their
migration and never migrated through the previous
As more neuroblasts migrate into the cortical layers of neurons. The clinical symptoms associated
plate, the latter undergoes an important increase in with lissencephaly comprise epileptic seizures,
thickness. The addition of new neurons into the cognitive impairment and focal neurological
cortical plate proceeds according to an inside- deficits, resulting in most cases in premature death
outside sequence. Each new wave of neuroblasts [28, 29].
migrates through the previous cell generation to
occupy a more superficial position (Fig. (1)). The Lissencephaly constitutes one of the most severe
need for a greater surface area in the outer cortical forms of impaired migration described in patients.
layers, as compared to the inner layers, is thought to On the other hand, several milder defects can now
provoke a compressive force involved in the be detected using standard magnetic resonance
formation of gyri [25]. These gyri develop imaging (MRI) techniques. It is estimated that
significantly between the 26th and 28th week of neuronal heterotopia could be one of the major
gestation, but continue to be further elaborated up cause of idiopathic epileptic seizures. Two of the
to the early postnatal period [26]. milder neuronal migration defects that have
recently attracted attention are periventricular
nodular heterotopia (PNH) and subcortical laminar
NEURONAL MIGRATION DISORDERS heterotopia (SCLH), also termed subcortical band
heterotopia or double cortex syndrome. In these
Neuronal migration can be affected at different disorders, only a fraction of neurons experiences
levels, each defect producing a characteristic migration defects, while on the contrary unaffected
pathological condition. In lissencephaly, for neurons establish a characteristic cortical structure.
example, the majority of neurons fail to reach their The underlying causes of these disorders are
proper cortical layers and interrupt prematurely their described below in more detail.
migration. The resulting cortex is therefore

Figure (2). Coronal Magnetic Resonance Images (MRI) of different neuronal migration disorders.

In contrast to a normal brain a) the brain of individuals suffering of PNH contains nodules or bands of gray matter lining
the ventricle (arrows) b). In cases of subcortical band heterotopia (double cortex) a band of heterotopic neurons can be
detected between the normal cortex and the ventricles (arrows) c) and in lissencephaly d) a significant reduction of the gyri
and a thickening of the cortical gray matter can be observed. Mutations in some genes, mentioned underneath each panel,
have been linked to these disorders.
680 Current Molecular Medicine, 2001, Vol. 1, No. 6 Couillard-Despres et al.

MOLECULAR ASPECTS OF MIGRATION impairing the establishment of a proper postnatal


DISORDERS blood circulation [34].

Filamin-1 and Periventricular Nodular Heterotopia Filamin-1 is a large protein of 280 kDa, encoded
by a gene composed of 47 exons spanning 26 kbp
Following terminal mitosis, newly formed of genomic sequences [40]. An actin-binding
neurons of the ventricular zone leave their site of domain similar to -actinin and dystrophin is
origin and migrate towards the marginal zone at the localized at the amino-terminus of filamin-1,
surface of the developing cerebrum. In cases where followed by 24 repeats of a 96 amino acids
neurons fail to initiate the migratory process, they sequence forming Ig-like -sheet motifs. The last
accumulate in the ventricular wall as nodules, repeat is truncated and contains sequences
causing a pathological condition referred to as allowing for the dimerization of filamin-1
periventricular nodular heterotopia (PNH) (Fig. (2b)). monomers. The 24 repeats are also interrupted
Histological examination of individuals affected by between the repeats 15-16 and 23-24 by sequences
PNH revealed the presence of differentiated creating hinges in the rod domain [41].
neurons inside the nodules and normal cortical
layering [30, 31]. Interestingly, heterotopic neurons Previous reports demonstrated that filamin-1 can
seem to be incorporated in the motor circuitry. A bind the intracellular domain of transmembrane
recent study using positron emission tomography receptors, such as the 1 and 2 integrins [42-44].
(PET) and oxygen-15-labeled water revealed an Therefore, filamin-1 can constitute a link
increased cerebral blood flow during a finger transducing extracellular signals into actin network
tapping task in the nodular heterotopic gray matter reorganization. The function of filamin-1 is
located in the white matter below the frontal regulated by phosphorylation, which alters its
neocortex, suggesting that neuronal activation subcellular localization and binding affinity to
occurs in subcortical gray matter heterotopia. [32]. membrane receptors and actin filaments [41].
Using an identical paradigm during MRI scanning, Interestingly, phosphorylation consensus sequence
we observed an increased blood flow in the gray analysis revealed that filamin-1 is very likely to be a
matter lining the ventricular surface in patients substrate for Cdk5 phosphorylation [34]. Null
suffering of PNH [33]. These preliminary functional mutation of the Cdk5 is known to provoke profound
neuroimaging studies suggest that heterotopic neuronal migration disorder, in a manner related,
pyramidal neurons, located in the subcortical white but not identical, to the reeler phenotype [45]. The
matter and along the ventricular wall, project to the reeler phenotype has been linked to a null mutation
spinal cord and could therefore be functionally of the reelin gene (see below) [46]. The reelin
integrated in motor circuits required during motor protein is secreted into the extracellular matrix by a
task performance. subpopulation of neurons and binds to the 31
integrins. Thus, reelin and filamin-1 could be part
In studying various pedigrees, some cases of of the same signaling pathway via their common
PNH were linked to mutations in the filamin-1 (FLN1) interaction partner, the 1 integrin.
gene [34]. The mutations identified are likely to
provoke a loss of function of filamin-1 proteins. A role of filamin-1 in the formation of filopodia
Filamin-1 is an actin-binding protein producing was recently reported [47]. Filamin-1 was found to
orthogonal branching on the actin filaments and bind the activated RalA protein, a small GTPase,
can induce filament bundles similar to those with its carboxy-terminal region. The microinjection
observed in filopodia [35, 36]. The function of of constitutively active RalA in cells causes the
filamin-1 is, however, not restricted to the nervous extension of filopodia. Taking advantage of a
system. A role in the development of the immune human melanoma cell line lacking filamin-1, Ohta
and vascular system is supported by recent studies and colleagues [47] were able to demonstrate that
[37-39]. In males, mutations in filamin-1 are the ability of RalA, and other proteins upstream of
normally embryonically lethal, but in cases where the RalA pathway, to promote filipodia extension
the prenatal development reached advanced was dependent on the presence of filamin-1. In
stages, deficient myeloid and erythroid vitro, the presence of RalA does not affect the
differentiations were reported, together with binding or cross-linking activity of filamin-1 on actin
systemic bleeding [34]. In females, the phenotype is filaments. In vivo, however, activated RalA is able
significantly milder. The reason for this discrepancy to recruit filamin-1 beneath the cellular membrane.
is the localization of the filamin-1 gene on the X- This concentration of filamin-1 could result in the
chromosome (Xq28). Therefore, as a result of transition of its actin filament branching activity
random X-chromosome inactivation, females observed at low concentrations to the filament
bearing a mutation in filamin-1 express the mutant bundling activity reported for high concentrations of
gene in a mosaic pattern. Yet, these females also filamin-1, thereby promoting the formation of
show some vascular anomalies as revealed by a filopodia [35, 36]. Similarly, in individuals lacking
higher incidence of patent ductus arteriosus, viz. filamin-1 protein, the neurons, having completed
the shunt between the aorta and the pulmonary their last mitosis, would be impaired in their
artery does not close properly after birth, thus capacity to extend filopodia. The filopodia are an
Neuronal Migration Disorders, Quo Vadis Current Molecular Medicine, 2001, Vol. 1, No. 6 681

essential element of the cell migration machinery, and thereby contribute to an increase of the
probing the environment and transducing guiding maximal polymer length measured in the
cues to the soma. In the absence of appropriate microtubules pool.
guiding cues, the newly born neurons could remain
anchored at their site of origin, forming such The LIS1 protein is extremely conserved
nodules as observed in PNH. Moreover, between mouse and human, with only one amino
modification of filamin-1 subcellular distribution via acid difference [52]. LIS1 was also reported to be
protein-protein interaction, with RalA and 1 for serine-phosphorylated [53]. Phosphorylation is likely
example, or via phosphorylation through kinases to modulate the binding properties of LIS1 to
such as Cdk5, could also be at the origin of microtubules. The presence of multiple LIS1
migration disorders. phosphorylation isoforms was described when the
fraction of protein tightly bound to microtubules was
enriched. Therefore, the phosphorylation of LIS1 by
Doublecortin and LIS1, Molecules Involved in protein kinases could constitute a rapid mechanism
Lissencephaly and Subcortical Laminar to control locally the polymerization dynamic of the
Heterotopia microtubules.

LIS1 haplo-insufficiency is the first specific gene As mentioned above, the LIS1 protein has been
defect demonstrated to be causative of identified as a -subunit for the PAFAH1B. Although
lissencephaly [48]. Microdeletions of chromosome the purification of microtubules allows for the
17p13.3 were already reported to be present in enrichment of LIS1 protein, the resulting fraction is
more than 90% of individuals suffering of Miller- devoid of PAFAH activity [51]. Hence, LIS1 proteins
Dieker syndrome, a type I lissencephaly associated seem to be involved in two distinct pathways: first as
with dysmorphic facial appearance [49]. Although a subunit of the PAFAH1B, leading to the
the LIS1 gene is ubiquitously expressed, the levels inactivation of the platelet-activating factor (PAF),
of expression are particularly high in the brain, and second as a microtubule-associated protein
heart, muscle and kidney [48]. In humans, the ~80 (MAP), promoting the stabilization and elongation
kb gene is organized into 11 exons and two major of microtubules.
transcripts of 5.5 kb and 7.5 kb can be detected
[48]. The use of two different polyadenylation The presence of functional PAFAH1B is
signals is likely to be at the origin of the two essential during development as revealed by the
transcripts. The 45 kDa protein resulting from LIS1 post-implantation embryonic lethality of mice
expression has been identified as a non-catalytic bearing a null mutation for the PAFAH1B -subunit,
subunit of the platelet-activating factor i.e. the LIS1 protein [54]. Mice heterozygous for the
acetylhydrolase isoform 1b (PAFAH1B), a brain LIS1 gene present a blurred cortical layering, as
specific isoform of PAFAH [50]. The main structural compared to the normal mice, and a trabeculated
feature of the protein is the presence of seven neuronal distribution [54]. Nevertheless, the cortex,
WD40 repeats, often found in G-protein -subunits marginal and white matter zones were not
[51]. However, the degree of similarity between significantly affected with regard to size and
LIS1 and the other conventional G-protein - cellularity. Since the mouse is a priori an animal
subunits is relatively low (55%) [48]. The WD40 with lissencephaly, the study of the cortex is not
repeats in LIS1 suggest that the latter is involved in very informative in this respect. However, another
multiple protein-protein interactions. region of the human brain severely affected by
mutation of the LIS1 gene is the hippocampus. The
Detection of LIS1 proteins by immunohisto- inactivation of one allele of LIS1 in mice leads to a
chemistry revealed a subcellular distribution that disorganization of the hippocampus, in which the
closely resembled the microtubule network [51]. pyramidal cells do not pack properly and appear as
Further investigation demonstrated that LIS1 could discontinuous split layers, especially in the CA1
bind microtubules with an affinity similar to typical region [54]. These histological abnormalities are
microtubule-associated proteins (MAPs). In dosage dependent, since mating using different
addition, the use of amino-truncated LIS1 protein LIS1 mutant alleles showed that the cerebral
indicates that this segment participates directly in defects are proportional to the decrease of LIS1
microtubule binding [51]. The influence of LIS1 expression.
protein on the microtubule dynamic was therefore
investigated. In vivo, microtubules constantly The hemidepletion of LIS1 does not lead to an
oscillate between a polymerization phase and a inversion of the cortical layering as is observed in
depolymerization phase. The duration of each the Reeler mouse (see below). Labeling the neurons
phase determines the average polymer length of born at specific time points using bromo-
the microtubule pool. Using differential interference deoxyuridine (BrdU) revealed a typical inside-out
contrast microscopy (DIC), Sapir and colleagues construction of the cortical layers. However, the
[51] reported that LIS1 proteins could reduce the waves of migrating neurons appear broader when
frequency of polymerization to depolymerization examined in newborn animals lacking one allele of
transition, referred to as the "catastrophe event", LIS1. In one month-old animals, however, the
682 Current Molecular Medicine, 2001, Vol. 1, No. 6 Couillard-Despres et al.

neurons reached their appropriate residing to be elucidated, the protein is clearly involved in
positions, suggesting that migration was only slowed the fundamental aspects of neuronal cell biology.
down [54]. To investigate this issue, an in vitro assay
was performed using unilateral migration of Recently, a new gene, doublecortin, linked with
reaggregated cerebellar cells on a laminin another type of lissencephaly has been recently
substrate [55]. In this setting, the migration of reported. This type of lissencephaly is characterized
neurons hemizigous for the LIS1 gene was severely by the presence within the same pedigree of males
impaired when monitored 24h after plating. affected by classical lissencephaly and females
Moreover, chimeric reaggregates demonstrated that affected by a milder condition called subcortical
the LIS1 deficiency affects neurons in a cell- laminar heterotopia (SCLH). The females affected
autonomous fashion [54]. by SCLH present a heterotopic band of neurons
located between the ventricles and the cortex,
The mechanism by which LIS1 deficiency can producing a second band of gray matter, the so-
lead to migration disorders is not yet understood. It called double cortex (Fig. (2c)). Although the real
was reported that extracellular application of PAF cortex appears normal, the neurons within the
provokes the collapse of the growth cones [56]. supplementary band of gray matter appear
Therefore, an insufficient activity of the PAFAH1B disorganized, with their apical dendrites pointing
complex, caused by a reduction in LIS1 protein, either towards the cortex or towards the ventricles
would result in an accumulation of PAF and [61]. Moreover, in the lower aspect of the
thereby to an impaired migration. The fact, heterotopic band, the neurons are disposed in a
however, that LIS1 reduction affected neuronal columnar organization, suggesting an arrest during
migration in a cell-autonomous fashion in the in radial migration towards the cortex. Finally, at the
vitro migration assay does not support this lower edge of the heterotopic neuron band, some
hypothesis. However, accumulation of PAF cannot heterotopic neurons forming small nodules are
be ruled out as an aggravating factor leading to frequently found [61].
neuronal migration disorder in vivo.
The pattern of inheritance of this type of
On the other hand, some lines of evidence point lissencephaly is characteristic for a X-linked genetic
to perturbation in mitosis and nucleokinesis of defect and is referred to as SCLH/X-LIS. In 1998,
proliferating neuronal precursors as a result of two groups reported simultaneously that mutations
reduction in LIS1 protein content. As mentioned in the doublecortin gene associated with inherited
above, the timing of terminal mitosis and the and sporadic SCLH/X-LIS cases [62, 63]. Together,
migration of neuronal nuclei are crucial aspects of mutations/deletions in the LIS1 and the
the cerebral development. In Drosophila, deletion doublecortin genes account for approximately 76%
of one allele of LIS1 leads to inappropriate nucleus of all the sporadic cases of classical lissencephally
positioning in the developing embryo [57]. [64]. This new gene is localized on chromosome X
Moreover, immunoprecipitation experiments at the position X922.3-23, where it covers
revealed that LIS1 proteins can interact with approximately 85 kbp of genomic sequences. The
dynein, a microtubule-anchored motor complex milder pathology observed in females bearing
[58, 59]. The interaction of LIS1 with the dynein doublecortin mutations is believed to be the
heavy-chain stimulated the activity of the dynein consequence of random X-chromosomal
motor complex, thereby modifying the distribution inactivation. The neuroblasts in which the X-
of different organelles and also influencing the chromosome bearing the wild type allele of the
microtubule-organizing center (MTOC) [59]. Since gene is active will migrate properly to the cortex,
the migration/positioning of the nucleus is proposed whereas the neuroblasts expressing the mutated
to be a MTOC-dependent process [60], hindered copy will display aberrant migration and form the
nuclear positioning could be at the origin of the additional band of gray matter observed between
impeded migration reported. Furthermore, LIS1 was the cortex and the ventricles. This explanation is
shown to influence different aspects of mitosis [58]. based on a cell autonomous effect of the
For instance, cells with increased or decreased mutations.
LIS1 protein content display a significantly longer
duration of mitosis and problems with the proper The doublecortin transcription product of 9.5 kb
alignment of chromosomes on the metaphase originates from the alternative splicing of 9 exons
plate. Moreover, the overexpression of LIS1 clearly [62, 63]. Three exons (1A, 1B or 1C) can form the 5'
perturb the mitotic spindle, leading to a end of the mRNA. The second exon possesses the
randomization of its orientation [58]. Together, initial ATG of a 1080 bp long open reading frame,
these observations indicate that aberrant levels of i.e. encoding a polypeptide of 360 amino acids.
LIS1 can perturbed the timing of the last neuronal Another potential ATG is located in the exon 1C.
mitosis, its plane of symmetry and the rate of Although not yet reported, the use of this codon as
migration towards the superior aspects of the translation start point would produce a polypeptide
developing cortex. Although, the mechanism by bearing an additional 42 amino acids [62]. Two
which LIS1 mutation leads to lissencephaly remains other polymorphisms have also been observed in
doublecortin cDNAs. The first is an insertion that
Neuronal Migration Disorders, Quo Vadis Current Molecular Medicine, 2001, Vol. 1, No. 6 683

would result in 6 additional residues, Gly-Asn-Asp- eliminated by the pre-treatment of the protein
Glu-Asp-Ala, at the amino acid position 311, extract with alkaline phosphatase [65]. The sites
whereas an alternative use of the splicing junctions and kinases responsible for this in vivo
between exons 8 and 9 could result in the presence phosphorylation have not been identified to date.
of an additional valine residue [63]. One Interestingly, the level of doublecortin
particularity of the doublecortin transcript is its very phosphorylation appears to be related to its
long 3' untranslated region. This sequence of interaction with the microtubules. For example, in
approximately 7.9 kb is encoded by a single exon vitro the depolymerization of the microtubule
and contains two AU-rich elements (AREs). The network using nocodazole led to the disappearance
ARE sequences are often found within the 3' region of the phosphorylated doublecortin protein doublet
of labile mRNAs and are likely to influence their [65].
stability [62].
Reports on the intra-cellular distribution of
In situ hybridizations and northern blot analysis doublecortin have been divergent. Using primary
revealed that the doublecortin transcript is specific neurons in culture, Francis and colleagues [65]
for young neurons, and it is found to be particularly observed an enrichment of doublecortin in the
abundant in neurons forming the cortical plate and distal part of the neurites. On the other hand,
the subventricular region [62]. In mouse embryos, Gleeson and colleagues [66] reported the detection
the expression of the doublecortin transcript is first of doublecortin in migrating Purkinje cells and
detectable in the neuroepithelium at day E10.5 cultured neurons, predominantly at the periphery of
and can later be detected in various regions of the the cell soma with little immunoreactivity in the
CNS and PNS [65]. The fact that doublecortin neurites. Using cultures of human embryonic
mutations predominantly affect the cortex, as cortical neural stem cells, we could detect high
compared to the rest of the CNS and PNS, is levels of doublecortin along the entire length of
intriguing considering the widespread expression of processes in newly differentiated neurons. There is
doublecortin in migrating neurons. nevertheless agreement on the co-localization of
doublecortin and the microtubule network, without
A polypeptide of 40 kDa is predicted by the apparent association to the actin or neurofilament
analysis of the doublecortin open reading frame. networks [65, 66]. In vitro experiments revealed that
The protein sequence does not bear significant doublecortin could interact directly with purified
homology with known functional domains. It is, tubulin subunits and thereby reduce the critical
however, highly homologous to another protein concentration required for microtubule assembly
designated as KIAA0369 [62, 63]. The amino- [65, 67]. Doublecortin facilitation of microtubule in
terminal of this protein shares 75% similarity at the vitro polymerization results in an increase of total
amino acids level with the doublecortin number of microtubules, without an apparent effect
polypeptide. Interestingly the carboxy-terminal of on the average length of the polymers [66]. This
KIAA0369 encodes a calcium calmodulin- activity is believed to originate from the
dependent kinase (CaM kinase) domain. microtubule-bundling capacity of doublecortin.
Doublecortin sequence analysis also revealed This capacity can readily be observed in COS-7
phosphorylation target sequences for MAP kinases cells overexpressing doublecortin (Fig. (3)). Upon
and Abl [62, 63]. In brains of embryonic day 12.5 doublecortin expression, the microtubule network
and older mouse embryos, a fraction of the reorganizes into large bundles. These microtubule
doublecortin proteins has a slower electrophoretic bundles become more resistant to depolymerizing
mobility on SDS-PAGE. The presence of this agents, such as colchicine [66], as well as to cold-
fraction, forming a band-doublet, can be induced depolymerization [66, 68]. In parallel,

Figure (3). Doublecortin affects the microtubular network.

COS-7 cells transiently expressing a doublecortin / red fluorescent (RFP) fusion protein. The same microscopic field is
shown for a) phase contrast, b) doublecortin-RFP, c) -tubulin and d) and overlay of doublecortin-RFP and -tubulin.
Doublecortin colocalizes with and bundles microtubules.
684 Current Molecular Medicine, 2001, Vol. 1, No. 6 Couillard-Despres et al.

over-expression of doublecortin also increases the leading to an outside-in addition of new neurons
percentage of tubulin protein recovered in the [73].
insoluble fraction. Whereas approximately half of
the tubulin dimers are normally recovered in the In humans, the reelin gene is localized on
soluble fraction when cells are homogenized in chromosome 7q22 [46]. The 12kb reelin transcript is
mild conditions, the overexpression of doublecortin encoded by 65 exons translated into a pro-protein
increases this percentage to more than 80% [67]. containing a cleavable signal peptide. At the
The amino-terminus half of the protein appears to amino-terminus of the 388 kDa mature protein, a
encode the microtubule binding-domain. Although domain sharing homology with F-sponding is
no canonic microtubule-binding domain has been described, followed by 8 repeats containing EGF-
detected by sequence analysis, a polypeptide like motifs [46]. Reelin possesses a secretion signal
containing the first 198 amino acid can efficiently located at its carboxy-terminus, allowing for the
bind to microtubules, whereas a polypeptide accumulation of the protein into the extracellular
representing the last 110 amino acids does matrix [74]. There are two potential reelin mRNA
apparently not interact [67, 69, 70]. Expression of polyadenylation signals reported, as well as two
several doublecortin mutants in non-neuronal cells alternative splice isoforms, only distinguished by the
yielded variable phenotypes. While some mutants presence or absence of 6 nucleotides from exon 64
induced similar microtubule bundling as observed [74, 75]. The presence of exon 64 adds two amino
following overexpression of the wild type allele, acids, valine and serine, and therefore offers an
other mutants did not alter the microtubule extra phosphorylation site. More substantial
distribution, whereas a third group of doublecortin changes are predicted for the use of the most 5'
mutants provoked protein aggregates [69, 70]. polyadenylation signal, since the resulting mRNA
Therefore, the mechanism by which doublecortin would encode a protein devoid of a secretion
mutations lead to lissencephaly remains to be signal. The significance of these various reelin
elucidated. mRNA isoforms is still unknown. In vivo, a N-terminal
directed antibody detects three forms of reelin,
Interestingly, a direct interaction between the approx. 400 kDa, 300 kDa and 180 kDa. The two
LIS1 and doublecortin proteins has been reported smaller bands detected are likely to be cleavage
[71]. It has been proposed that LIS1 can form products of the full length reelin by a protease
heterodimers with doublecortin, thereby competing located either in the post-endoplasmic reticulum
with doublecortin homodimerization. The impact of secretory pathway or in the extracellular space [76].
doublecortin mutations was also scrutinized in
respect to the doublecortin-LIS1 interaction, but no The Cajal-Retzius cells residing in the marginal
difference was observed when compared to the zone, i.e. layer 1 of the cortex, are among the first
behavior of doublecortin wild type allele [71]. post-mitotic neurons. They also represent one of the
main sources of reelin in the developing embryo.
Yet, the next generations of migrating neurons
Reelin and Lissencephaly building the cortical plate do not, or at very low
levels, express this protein [46]. Therefore, in
Some cases of autosomal recessive contrast to molecules presented in the two previous
lissencephaly with cerebellar hypoplasia from two sections, mutations in reelin cause migration
different pedigrees have recently been linked to disorders in a non-cell autonomous fashion.
mutations in the reelin gene [72]. Deletion of this
protein was already reported to cause migration Recent studies shed more light on the possible
disorders in an animal model, the reeler mouse [46]. mechanisms of action of reelin on neuronal
The pathological similarities between some migration. Dulabon and colleagues [77] reported
individuals affected by recessive lissencephaly with that reelin interacts with the 31 integrins present
cerebellar hypoplasia and reeler mice prompted the on the migrating neurons of the cortical plate. In
investigators to look for reelin mutations in the response to reelin, the rate of neuronal migration
selected families. The "reeler phenotype" consists decreases and neurons detach from the radial glia.
of a hypoplasia of the cerebellum and migration Hence the vicinity of Cajal-Retzius cells producing
disorders affecting the neocortex, the cerebellum, a reelin-filled extracellular milieu is interpreted by
the hippocampal formation and the brain stem. neurons as a stop signal during their outward
One hallmark of this phenotype is the "inversion" of migration on the radial glia scaffold.
the cortical layers, where the earlier born neurons Immunoprecipitation experiments established that
occupy a more superficial position in respect to the 31 integrins can specifically bind reelin [77]. In
later born neurons. At the origin of this aberrant absence of this integrin type, the radial glia-guided
lamination is the failure of the first cortical plate migration of embryonic neurons becomes
neurons to bisect the pre-plate into its two unresponsive to the presence of reelin. In neurons,
components: the marginal zone and the subplate. the 31integrin colocalizes with a cytoplasmic
Each subsequent cohort of migrating neurons molecule named mammalian Disabled (mDab1)
simply accumulate beneath the preceding ones, [77]. The spontaneous null mutation of this
molecule described in the scrambler mouse
Neuronal Migration Disorders, Quo Vadis Current Molecular Medicine, 2001, Vol. 1, No. 6 685

provokes a phenotype equivalent to the reeler lacking 31 integrin nevertheless led to extensive
phenotype, suggesting that the molecules are phosphorylation of mDab1. This indicates that in
components of the same signaling pathway [78, addition to 31 integrin, other molecules are
79]. Two other observations substantiate this involved in the signal transduction pathway of
hypothesis. First, in response to the presence of reelin across the cytoplasmic membrane.
reelin, mDab1 becomes tyrosine-phosphorylated
[80]. In contrast, in the absence of reelin, such as in The very low-density lipoprotein receptor
the reeler mouse, mDab1 accumulated at levels (VLDLR) and the apolipoprotein E receptor 2
several times higher than those observed in normal (ApoER2), two members of the low-density
mice [81]. Although mDab1 is a cytoplasmic lipoprotein receptor family, have also been
protein, it possesses a protein interaction / presented as receptors for reelin [83]. These two
phosphotyrosine-binding domain, allowing for receptors are closely related, suggesting possible
interaction with the cytoplasmic moiety of functional overlap. Pull-down experiments
transmembrane receptors [82]. In response to reelin, demonstrated the mDab1 could directly bind to the
the phosphorylated mDab1 can interact with non- cytoplasmic tail of the VLDLR and the ApoER2
receptor tyrosine kinases such as Abl, Fyn and Src [84]. Although the disruption of either receptor
[82]. Surprisingly, in the absence of 31 integrin leads to a mild alteration of the brain
as a consequence of 3 integrin targeted cytoarchitecture, the simultaneous gene
disruption, a reduction of approximately 50% in inactivation of the VLDLR and ApoER2 lead to an
mDab1 protein content is described [77]. This aberrant neuronal development analogous to the
contrasts with the reeler mice, in which an reeler/scrambler phenotype, comprising of a
accumulation of mDab1 is reported [81]. Therefore, hypoplasia of the cerebellum and inversion of the
the integrins and mDab1 are likely to be part of a cortical layers [84]. Moreover, similarly to the reeler
common reelin-signaling pathway, where 31 and mice, the double-knockout animals for VLDLR and
reelin directly or indirectly stabilize mDab1. Yet, ApoE2R show a significant increase in the
application of reelin on cortical embryonic neurons accumulated levels of mDab1 proteins.

Figure (4). Schematic representation of molecules involved in the regulation of neuronal precursor cell migration.
Doublecortin, LIS1 and FLN1 are intracellular molecules and mutations in their genes will cause cell-autonomous migration
impairment. In contrast, Reelin is a protein secreted in the extracellular matrix and arrests the migration of the incoming
neuroblasts.
686 Current Molecular Medicine, 2001, Vol. 1, No. 6 Couillard-Despres et al.

Unfortunately, the level of phosphorylation of MRI = Magnetic resonance imaging


mDab1 was not investigated in these various
knockout animals [84]. However, in a parallel study, MTOC = Microtubule-organizing center
indirect evidence was presented to demonstrate
that competition for the binding of reelin on VLDLR mDab1 = Mammalian Disabled
and ApoER2 reduces the levels of mDab1 tyrosine PET = Positron emission tomography
phosphorylation [85]. Hiesberger and colleagues
also report in the same study that in absence of SCLH = Subcortical laminar heterotopia
reelin, as well as in the double VLDLR/ApoER2
knockout, the level of phosphorylation of Tau PNH = Periventricular nodular heterotopia
protein in the brain was greatly increased when
compared to the normal mouse. It is hypothesized VLDLR = Very low-density lipoprotein receptor
that an increase of Tau phosphorylation would lead
to a decrease in microtubule stability and therefore
affect the axonal cytoskeleton. However, the REFERENCES
histological analysis of mice in which the Tau gene
is selectively targeted did not reveal neuronal [1] Caviness, V.S., Jr.. and Sidman, R.L. (1973) J. Comp. Neurol.,
migration impairment [86]. 148(2), 141-51.

[2] Caviness, V.S., Jr., Takahashi, T. and Nowakowski, R.S. (1995)


Figure (4) constitutes an attempt to link the Trends Neurosci., 18(9), 379-83.
various molecules described in this review into a
schematic representation. Animals bearing [3] Takahashi, T., Nowakowski, R.S. and Caviness, V.S., Jr. (1996)
disruption of the p35/cdk5 complex present a J. Neurosci., 16(18), 5762-76.
phenotype similar to the reeler mouse [45, 87]. To
[4] Sidman, R.L. and Rakic, P. (1973) Brain Res., 62(1), 1-35.
date, however, no mutations in these genes were
reported to be linked with migration disorders in [5] Meyer, G., Schaaps, J.P., Moreau, L. and Goffinet, A.M. (2000)
human. Still, animal models bearing genetic J. Neurosci., 20(5), 1858-68.
defects that cause neuronal migration disorders are
extremely valuable tools. For instance, some [6] Dobbing, J. and Sands, J. (1973) Arch. Dis. Child., 48757-761.
spontaneous mutations causing migration disorders [7] Rakic, P. (1995) Trends Neurosci., 18(9), 383-8.
may originate from mutations in pathways also
affected in human migrations disorders, albeit at a [8] Rakic, P. (2000) Novartis Found. Symp., 22830-42.
different level. Caution must be taken, however,
because defective pathways could affect human [9] Noctor, S.C., Flint, A.C., Weissman, T.A., Dammerman, R.S.
and Kriegstein, A.R. (2001) Nature, 409(6821), 714-20.
corticogenesis in a different way due to intrinsic
molecular and cytoarchitectural differences. For [10] Chenn, A. and McConnell, S.K. (1995) Cell, 82(4), 631-41.
example, cortex development in mice bearing a
hemi-depletion of LIS1 is only delayed, whereas in [11] Chenn, A., Zhang, Y.A., Chang, B.T. and McConnell, S.K. (1998)
human, cortical development is completely Mol. Cell Neurosci., 11(4), 183-93.
disturbed. [12] Rakic, P. (1972) J. Comp. Neurol., 145(1), 61-83.

[13] Tan, S.S. and Breen, S. (1993) Nature, 362(6421), 638-40.


ACKNOWLEDGEMENT
[14] O'Rourke, N.A., Chenn, A. and McConnell, S.K. (1997)
Development, 124(5), 997-1005.
We would like to thank Christiana Cooper-Kuhn
helping in preparing the manuscript. We thank Dr. [15] O'Rourke, N.A., Sullivan, D.P., Kaznowski, C.E., Jacobs, A.A.
Ulrich Bogdahn as the head of the Dept. for and McConnell, S.K. (1995) Development, 121(7), 2165-76.
Neurology, University of Regensburg, for support.
The work was further supported by the Volkswagen- [16] Anderson, S.A., Marin, O., Horn, C., Jennings, K. and
Foundation, Hannover, Germany and by the Fritz- Rubenstein, J.L. (2001) Development, 128(3), 353-63.
Thyssen Foundation, Kln, Germany. [17] Lavdas, A.A., Grigoriou, M., Pachnis, V. and Parnavelas, J.G.
(1999) J. Neurosci., 19(18), 7881-8.

LIST OF ABBREVIATIONS [18] Anderson, S.A., Eisenstat, D.D., Shi, L. and Rubenstein, J.L.
(1997) Science, 278(5337), 474-6.
AREs = AU-rich elements [19] Rakic, P. (1971) Brain Res., 33(2), 471-6.
ApoER2 = Apolipoprotein E receptor 2 [20] Liesi, P. (1990) Experientia, 46(9), 900-7.

BrdU = Bromo-deoxyuridine [21] Fishell, G. and Hatten, M.E. (1991) Development, 113(3), 755-
65.
MAP = Microtubule-associated protein
[22] Stipp, C.S., Litwack, E.D. and Lander, A.D. (1994) J. Cell Biol.,
124(1-2), 149-60.
Neuronal Migration Disorders, Quo Vadis Current Molecular Medicine, 2001, Vol. 1, No. 6 687

[23] Hatten, M.E. (1999) Annu. Rev. Neurosci., 22511-39. [45] Gilmore, E.C., Ohshima, T., Goffinet, A.M., Kulkarni, A.B. and
Herrup, K. (1998) J. Neurosci., 18(16), 6370-7.
[24] Gadisseux, J.F., Kadhim, H.J., van den Bosch de Aguilar, P.,
Caviness, V.S. and Evrard, P. (1990) Brain Res., Dev Brain [46] D'Arcangelo, G., Miao, G.G., Chen, S.C., Soares, H.D., Morgan,
Res., 52(1-2), 39-56. J.I. and Curran, T. (1995) Nature, 374(6524), 719-23.

[25] Volpe, (1995) Neurology of the newborn. W.B. Saunders [47] Ohta, Y., Suzuki, N., Nakamura, S., Hartwig, J.H. and Stossel,
Company. Philadelphia. T.P. (1999) Proc. Natl. Acad. Sci. USA, 96(5), 2122-8.

[26] Chi, J.G., Dooling, E.C. and Gilles, F.H. (1977) Ann. Neurol., [48] Reiner, O., Carrozzo, R., Shen, Y., Wehnert, M., Faustinella, F.,
1(1), 86-93. Dobyns, W.B., Caskey, C.T. and Ledbetter, D.H. (1993) Nature,
364(6439), 717-21.
[27] Sarnat, H.B. (1991) Developmental disorders of the nervous
system. Neurology in Clinical Practice. Bradley WG, Daroff RB, [49] Ledbetter, S.A., Kuwano, A., Dobyns, W.B. and Ledbetter, D.H.
Fenichel GM, Marsden CD. Butterworth-Heinemann. (1992) Am. J. Hum. Genet., 50(1), 182-9.
Stoneham.
[50] Gambello, M.J., Hirotsune, S. and Wynshaw-Boris, A. (1999)
[28] Livingston, J.H. and Aicardi, J. (1990) J. Neurol. Neurosurg. Neurogenetics, 2(2), 77-86.
Psychiatry, 53(7), 617-20.
[51] Sapir, T., Elbaum, M. and Reiner, O. (1997) EMBO J., 16(23),
[29] Dobyns, W.B. and Truwit, C.L. (1995) Neuropediatrics, 26(3), 6977-84.
132-47.
[52] Peterfy, M., Gyuris, T., Basu, R. and Takacs, L. (1994) Gene,
[30] Eksioglu, Y.Z., Scheffer, I.E., Cardenas, P., et al. (1996) Neuron, 150(2), 415-6.
16(1), 77-87.
[53] Sapir, T., Cahana, A., Seger, R., Nekhai, S. and Reiner, O.
[31] Fox, J.W. and Walsh, C.A. (1999) Am. J. Hum. Genet., 65(1), (1999) Eur. J. Biochem., 265(1), 181-8.
19-24.
[54] Hirotsune, S., Fleck, M.W., Gambello, M.J., Bix, G.J., Chen, A.,
[32] Hatazawa, J., Sasajima, T., Shimosegawa, E., Fujita, H., Clark, G.D., Ledbetter, D.H., McBain, C.J. and Wynshaw-Boris,
Okudera, T., Kanno, I., Mineura, K. and Uemura, K. (1996) A. (1998) Nat. Genet., 19(4), 333-9.
AJNR Am. J. Neuroradiol., 17(3), 479-82.
[55] Asou, H., Miura, M., Kobayashi, M., Uyemura, K. and Itoh, K.
[33] Winner, B. and Winkler, J. MRI. Deutsche Gesellschaft fuer (1992) Neurosci. Lett., 144(1-2), 221-4.
Neurologie. Aachen, 2001.
[56] Clark, G.D., McNeil, R.S., Bix, G.J. and Swann, J.W. (1995)
[34] Fox, J.W., Lamperti, E.D., Eksioglu, Y.Z., et al. (1998) Neuron, Neuroreport, 6(18), 2569-75.
21(6), 1315-25.
[57] Lei, Y. and Warrior, R. (2000) Dev. Biol., 226(1), 57-72.
[35] Hartwig, J.H., Tyler, J. and Stossel, T.P. (1980) J. Cell Biol.,
87(3 Pt 1), 841-8. [58] Faulkner, N.E., Dujardin, D.L., Tai, C.Y., Vaughan, K.T.,
O'Connell, C.B., Wang, Y. andVallee, R.B. (2000) Nat. Cell
[36] Hartwig, J.H. and Stossel, T.P. (1981) J. Mol. Biol., 145(3), 563- Biol., 2(11), 784-791.
81.
[59] Smith, D.S, Niethammer, M., Ayala, R., Zhou, Y., Gambello,
[37] Ott, I., Fischer, E.G., Miyagi, Y., Mueller, B.M. and Ruf, W. M.J., Wynshaw-Boris, A. and Tsai, L.H. (2000) Nat. Cell Biol.,
(1998) J. Cell Biol., 140(5), 1241-53. 2(11), 767-775.

[38] Yoshida, M., Westlin, W.F., Wang, N., Ingber, D.E., [60] Morris, N.R. (2000) J. Cell Biol., 148(6), 1097-101.
Rosenzweig, A., Resnick, N. and Gimbrone, M.A., Jr. (1996) J.
Cell Biol., 133(2), 445-55. [61] Harding, B. Gray matter heterotopia. In Guerrini R, Canapicchi
R, Zifkin BG, et al., eds. Dysplasia of cerebral cortex and
[39] Muller, M., Albrecht, S., Golfert, F., Hofer, A., Funk, R.H., epilepsy. new York: Lippincott-Raven, 1996. pp. 81-88.
Magdolen, V., Flossel, C. and Luther, T. (1999) Exp. Cell Res.,
248(1), 136-47. [62] des Portes, V., Pinard, J.M., Billuart, P., et al. (1998) Cell, 92(1),
51-61.
[40] Patrosso, M.C., Repetto, M., Villa, A., et al. (1994) Genomics,
21(1), 71-6. [63] Gleeson, J.G., Allen, K.M., Fox, J.W., et al. (1998) Cell, 92(1),
63-72.
[41] Gorlin, J.B., Yamin, R., Egan, S., Stewart, M., Stossel, T.P.,
Kwiatkowski, D.J. and Hartwig, J.H. (1990) J. Cell Biol., 111(3), [64] Pilz, D.T., Matsumoto, N., Minnerath, S., et al. (1998) Hum.
1089-105. Mol. Genet., 7(13), 2029-37.

[42] Liu, S., Calderwood, D.A. and Ginsberg, M.H. (2000) J. Cell [65] Francis, F., Koulakoff, A., Boucher, D., et al. (1999) Neuron,
Sci., 113(Pt 20), 3563-71. 23(2), 247-56.

[43] Loo, D.T., Kanner, S.B. and Aruffo, A. (1998) J. Biol. Chem., [66] Gleeson, J.G., Lin, P.T., Flanagan, L.A. and Walsh, C.A. (1999)
273(36), 23304-12. Neuron, 23(2), 257-71.

[44] Sharma, C.P., Ezzell, R.M. and Arnaout, M.A. (1995) J. [67] Horesh, D., Sapir, T., Francis, F., Wolf, S.G., Caspi, M.,
Immunol., 154(7), 3461-70. Elbaum, M., Chelly, J. andReiner, O. (1999) Hum. Mol. Genet.,
8(9), 1599-610.
688 Current Molecular Medicine, 2001, Vol. 1, No. 6 Couillard-Despres et al.

[68] Taylor, K.R., Holzer, A.K., Bazan, J.F., Walsh, C.A. and [78] Ware, M.L., Fox, J.W., Gonzalez, J.L., Davis, N.M., Lambert de
Gleeson, J.G. (2000) J. Biol. Chem., 275(44), 34442-50. Rouvroit, C., Russo, C.J., Chua, S.C., Jr., Goffinet, A.M. and
Walsh, C.A. (1997) Neuron, 19(2), 239-49.
[69] Yoshiura, K., Noda, Y., Kinoshita, A. and Niikawa, N. (2000) J.
Neurobiol., 43(2), 132-9. [79] Sheldon, M., Rice, D.S. and D'Arcangelo G, et al. (1997) Nature,
389(6652), 730-3.
[70] Sapir, T., Horesh, D., Caspi, M., et al. (2000) Hum. Mol. Genet.,
9(5), 703-12. [80] Howell, B.W., Herrick, T.M. and Cooper, J.A. (1999) Genes
Dev., 13(6), 643-8.
[71] Caspi, M., Atlas, R., Kantor, A., Sapir, T. and Reiner, O. (2000)
Hum. Mol. Genet., 9(15), 2205-13. [81] Rice DS, Sheldon M, D'Arcangelo G, Nakajima K, Goldowitz D.
and Curran, T. (1998) Development, 125(18), 3719-29.
[72] Hong, S.E., Shugart, Y.Y., Huang, D.T., Shahwan, S.A., Grant,
P.E., Hourihane, J.O., Martin, N.D. and Walsh, C.A. (2000) Nat. [82] Howell BW, Gertler FB, Cooper JA. (1997) EMBO J., 16(1),
Genet., 26(1), 93-6. 121-32.

[73] Caviness, V.S., Jr. (1982) Brain Res., 256(3), 293-302. [83] D'Arcangelo, G., Homayouni, R., Keshvara, L., Rice, D.S.,
Sheldon, M. and Curran T. (1999) Neuron, 24(2), 471-9.
[74] DeSilva, U., D'Arcangelo, G., Braden, VV., Chen, J., Miao, G.G.,
Curran, T. and Green ED. (1997) Genome. Res., 7(2), 157-64. [84] Trommsdorff, M., Gotthardt, M., Hiesberger, T., Shelton, J.,
Stockinger, W., Nimpf, J., Hammer, R.E., Richardson, J.A. and
[75] Royaux, I., Lambert de Rouvroit, C., D'Arcangelo, G., Demirov, Herz, J. (1999) Cell, 97(6), 689-701.
D. and Goffinet, A.M. (1997) Genomics, 46(2), 240-50.
[85] Hiesberger, T., Trommsdorff, M., Howell, B.W., Goffinet, A.,
[76] Lambert de Rouvroit, C., de Bergeyck, V., Cortvrindt, C., Bar, I., Mumby, M.C., Cooper, J.A. and Herz, J. (1999) Neuron, 24(2),
Eeckhout, Y. and Goffinet, A.M. (1999) Exp. Neurol., 156(1), 481-9.
214-7.
[86] Takei, Y., Teng, J., Harada, A. and Hirokawa, N. (2000) J. Cell
[77] Dulabon, L., Olson, E.C., Taglienti, M.G., Eisenhuth, S., Biol., 150(5), 989-1000.
McGrath, B., Walsh, C.A., Kreidberg, J.A. and Anton, E.S.
(2000) Neuron, 27(1), 33-44. [87] Kwon, Y.T. and Tsai, L.H. (1998) J. Comp. Neurol., 395(4), 510-
22.

Vous aimerez peut-être aussi