Vous êtes sur la page 1sur 9

Clin Genet 2007: 72: 296304 # 2007 The Author

Printed in Singapore. All rights reserved Journal compilation # 2007 Blackwell Munksgaard
CLINICAL GENETICS
doi: 10.1111/j.1399-0004.2007.00888.x

Developmental Biology: Frontiers for Clinical Genetics


Section Editors:
Roderick R. McInnes, email: mcinnes@sickkids.ca
Jacques L. Michaud, email: jacques.michaud@recherche-ste-justine.qc.ca

Lissencephaly and LIS1: insights into the


molecular mechanisms of neuronal migration
and development
Wynshaw-Boris A. Lissencephaly and LIS1: insights into the molecular A Wynshaw-Boris
mechanisms of neuronal migration and development. Departments of Pediatrics and Medicine,
Clin Genet 2007: 72: 296304. # Blackwell Munksgaard, 2007 UCSD School of Medicine,
La Jolla, CA, USA
Lissencephaly is a severe human neuronal migration defect characterized
by a smooth cerebral surface, mental retardation and seizures. LIS1 was
first gene cloned in an organism important for neuronal migration, as it
was deleted or mutated in patients with lissencephaly in a heterozygous
fashion. Studies in model organisms, particularly Aspergillus nidulans, as
well as those in the mouse, have uncovered an evolutionarily conserved
pathway that involves LIS1 and cytoplasmic dynein. This pathway codes
for proteins in a complex with cytoplasmic dynein and positively Key words: 14-3-3e dynein
regulates its conserved function in nuclear migration. This complex interkinetic nuclear migration isolated
appears to be important for proliferation and neuronal survival as well as lissencephaly sequence LIS1
neuronal migration. One of the components of this complex, NDEL1, is lissencephaly MillerDieker
a phosphoprotein that is a substrate for CDK5 (or CDK2 in fibroblasts) syndrome NDEL1 neuronal
and Aurora-A, two mitotic kinases. CDK5-phosphorylated NDEL1 migration nuclear migration
binds to 14-3-3e, which protects it from phosphatase attack. Corresponding author: Professor Anthony
Interestingly, 14-3-3e is located 1 Mb from LIS1 and is heterozygously Wynshaw-Boris, MD, PhD,
deleted with LIS1 in patients with a severe form of lissencephaly, Miller Departments of Pediatrics and Medicine,
Dieker syndrome. Mouse models confirm that 14-3-3e plays an important UCSD School of Medicine,
role in neuronal migration, and mice that are double heterozygotes for La Jolla, CA 92-93-0627,
mutations in Lis1 and 14-3-3e, display more severe neuronal migration USA.
defects. The identification of LIS1 as the first lissencephaly gene, and the Tel.: 11 858 822 3400;
first gene required for neuronal migration has revealed the importance of fax: 11 858 822 3409;
the regulation of cytoplasmic dynein in the control of neuronal migration e-mail: awynshawboris@ucsd.edu
by modulating nuclear migration in a pathway conserved in virtually all Received 6 July 2007, revised and
eukaryotes. accepted for publication 10 July 2007

The human brain is the most elaborate structure display a precise connectivity to provide specific
known and comprised of an integrated network of inputs and outputs to and from the cerebral
more than 100 billion neuronal cells. The forma- cortex.
tion of this intricate neuronal network requires the Much has been learned about neuronal devel-
precise choreography of neurogenesis, neuronal opment by studying the variations from the basic
migration, pruning and synaptogenesis during pattern of forebrain organization in different
development. The mammalian cerebral cortex is mammalian organisms, including the human.
highly patterned and divided into anatomically One of the most striking abnormalities of human
distinct, functionally specialized areas for cogni- neuronal migration is lissencephaly (or smooth
tive, executive, motor, somatosensory auditory brain), a term originally used to describe the
and visual functions. These different cortical areas smooth cerebral surface of lower mammals.

296
Lissencephaly and LIS1
Lissencephaly of the human brain is a severe (a)
malformation in which the brain has a smooth BASAL
cerebral surface rather than the characteristic gyri
and sulci that make the human and primate brain
instantly recognizable. Lissencephaly can be
further specified as agyria (absence of gyri) or
pachygyria (reduced numbers of broadened gyri). APICAL
NESCs G1 S G2 M NESCs
In this review, I will focus on LIS1, the first
neuronal migration gene cloned in any organism. (b)
The discovery of LIS1 as the causative gene in
CP
a subset of human lissencephaly led to the
elucidation of a complex conserved from yeast
to human that regulates the activity of cytoplas- IZ
mic dynein. This complex is crucial for neuronal
migration, but more recent data indicates that it is
also broadly important during neurogenesis and VZ
for neuronal survival during development. The
reader is referred to recent reviews for more com- RGs G1 S G2 M RGs
prehensive insights into the control of neuronal
migration (13) and LIS1 function (4). Fig. 1. Interkinetic nuclear migration in neuroepithelial
stem cells and radial glial progenitor cells. (a) Neuro-
epithelial stem cells (NESC) exist as a pseudostratified
Neuronal migration during cortical epithelium with each cell attached at both apical and basal
development surfaces. Interkinetic nuclear migration occurs throughout
the length of the cell, with nuclei from cells in G1 moving
Neuronal migration has been studied extensively from the apical to the basal surface (arrow above G1),
for over 30 years in diverse mammalian species where cells synthesize DNA (S). Nuclei from cells in G2
from the mouse to human. The basic sequence of move from the basal to apical surface (arrow above G2),
where the cells divide (M), to produce two daughter NESCs.
events that occurs during cortical development is (b) Interkinetic nuclear migration occurs in the more
shared by all of these species, although the timing restricted radial glial progenitor cells (RG), but this
of events is species dependent. For example, these migration is confined to the ventricular zone (VZ), and
events occur between 5th and 22nd gestational excluded from the intermediate zone (IZ) and cortical plate
weeks of human development, while in the mouse (CP). Note that RG cells divide at the apical surface.
they occur between embryonic day 11 (E11) and
19 (E19). (Fig. 2). The first migrating cells give rise to the
Prior to E11 in the mouse, the neural tube marginal zone or layer 1, which is made up largely
consists of a single layer of pseudostratified of the earliest differentiated neurons of the cortex,
neuroepithelial stem cells that proliferate rapidly. the CajalRetzius cells. Between E13 and 19 in the
Each of these stem cells stretches from the apical mouse, neurons generated in the VZ migrate
to the basal surface of the neuroepithelium, and as across the intermediate zone toward the cortical
the cell progresses through the cell cycle, the nuclei plate at the pial surface and become the projection
display interkinetic nuclear migration (Fig. 1). neurons of the cortex. As a result, several
Nuclei divide at the apical surface and synthesize embryonic laminas are established over the next
DNA at the basal surface of the neural epithelium. few days. Interestingly, the proliferating radial
Consequently, nuclei from cells in G1 migrate glial progenitors display interkinetic nuclear
from the apical to basal surface, while nuclei from migration, similar to the neuroepithelial stem
cells in G2 move from the basal to apical surface. cells, but only in the VZ, a proliferative zone at the
At about E11, shortly after closure of the rostral apical surface (Fig. 1). The embryonic cortical
neural tube, the neuroepithelial stem cells further plate forms layers 2 through 6 in the post-natal
differentiate into a more restricted progenitor, period. Classical studies have established that the
radial glial progenitor cells, and in a subpopula- cerebral cortex is formed by an insideout
tion of these cells, the daughter cells of the radial migration of cells from the VZ. The early arising
glial progenitors become post-mitotic, immature neuroblasts that migrate first to the cortical plate
neuroblasts while in and shortly after leaving the occupy the deepest cortical layers in the adult
ventricular zone (VZ). Young neurons begin to (layers 4, 5 and 6). The later born migrating young
move out from the VZ at about E12 radially along neurons migrate past the earlier established
the extensions of the radial glial progenitors that cortical plate cells to occupy more superficial
act as both progenitor and scaffold for migration layers (layers 2 and 3) of the adult cortex.

297
Wynshaw-Boris
and generalized neuronal heterotopias (5). There
are two major types of classical lissencephaly:
3V isolated lissencephaly sequence (ILS) and Miller
Cortex
Dieker syndrome (MDS). ILS is a heterogeneous
disorder consisting of variably severe lissence-
HIP HIP LV NCX phaly with no other major malformations such as
craniofacial dysmorphism. Children with ILS are
also severely retarded and typically have epilepsy
LGE (6). MDS consists of more severe lissencephaly
than ILS patients, characteristic facial anomalies,
Caudate, MGE and occasionally other malformations. Children
putamen with MDS are severely retarded and suffer from
epilepsy. Craniofacial dysmorphism includes mi-
Globus crocephaly with bitemporal narrowing, a high
pallidus forehead, a small nose with anteverted nares, thin
vermilion border, and micrognathia (7). The
Fig. 2. Direction of migration of neurons during mamma- disorder is invariably fatal in early childhood.
lian cortical development. The schematic represents a coro- The major pathology in both disorders is usually
nal section through an E12-13 mouse cortex. Arrows
represent radial migration from the hippocampus (HIP,
considered to be in the cerebral cortex, but
green), cortex (aqua), lateral ganglionic eminence (LGE, cerebellar and olivary heterotopias have been
red) to the caudate, putamen and medial ganglionic described in MDS patients as well.
eminence (MGE, black) to form the projection neurons. MDS (100%) and some cases of ILS (40%) are
Additionally, cells migrate tangentially from the LGE (blue) the result of haploinsufficiency at human chromo-
to the caudate, putamen and MGE (pink) to form the some 17p13.3, with visible or submicroscopic
inhibitory interneurons in the cortex. These tangentially
migrating cells interact with radially migrating cells born at deletions detectable by fluorescence in situ hybrid-
the same time in the intermediate zone of the cortex, and ization (8, 9). The LIS1 gene was cloned from this
then migrate together to the proper layer in the cortex. LV, region (10). LIS1 was disrupted in an ILS patient
lateral ventricle; NCX, neocortex; 3V, third ventricle. with a translocation, and several other key MDS
patients (9). Point mutations and an intragenic
deletion in LIS1 were identified in ILS patients who
In addition to radial migration of the projection showed no gross structural chromosomal rear-
neurons of the cortex, the inhibitory interneurons rangements, confirming that mutations or dele-
migrate tangentially from the medial ganglionic tions of the LIS1 gene are responsible for classical
eminences (MGE) and lateral (LGE) ganglionic lissencephaly in ILS and MDS patients (11, 12).
eminences (Fig. 2). As one example of the tight The more severe neuronal migration phenotype
choreography of neuronal migration, cells born and facial dysmorphisms displayed by MDS
in the cortical VZ meet cells born at the same time patients suggest that genes other than LIS1, such
in the MGE and LGE in the IZ, where they in- as 14-3-3e are responsible for these phenotypes.
teract and migrate together to the appropriate
cortical layer.
Mouse models for Lis1 functional studies

Human lissencephaly
Mouse models for these disorders have aided in
the understanding of the function of LIS1 and the
Lissencephaly is one of the best characterized pathways associated with it during brain devel-
and studied human genetic malformations of the opment. Although human studies demonstrated
cerebral cortex. The primary defect underlying the that LIS1 mutations are responsible for lissence-
smoothening of the brain of lissencephalic pa- phaly, it is not possible to study the neuronal
tients is defective neuronal migration. The inabil- migration defects that occur embryologically
ity of post-mitotic neurons to reach their final during human development. To examine the
destination and correctly populate the cortical consequences of dosage reduction and complete
plate of the cerebral cortex consequently leads to deficiency of Lis1 in vivo, we produced two
abnormal cortical thickness and reduced or absent knockout and one conditional knockout Lis1
gyri and sulci of its surface. mutant alleles by gene targeting in the mouse (13).
Classical lissencephaly defines a subgroup of The two knockout alleles (one from germline
human neuronal migration disorders character- Cre-mediated deletion of the conditional knock-
ized by generalized agyria/pachygyria, four out allele) are nulls, while the conditional allele
abnormal cortical layers, enlarged ventricles, is hypomorphic, because of the disruption of

298
Lissencephaly and LIS1
transcription or splicing by the insertion of PGKneo Thus, the Dynein/Lis1 complex plays an impor-
in intron 3. By mating mice with various Lis1 tant role in the regulation of spindle orientation.
alleles, mice were produced with graded reduction More recently, it has been possible to directly
in LIS1 dosage. Mice with decreased levels of Lis1 examine neuronal migration in slice cultures from
exhibited dose-dependent disorganized cortical embryonic cortex. Lipophilic dyes or in utero tran-
layers, hippocampus, cerebellum and olfactory sfection with green fluorescent protein are used to
bulb because of cell autonomous neuronal migra- label the migrating cells, and migration is recorded
tion defects (13) and are a good model for the by confocal time-lapse video microscopy. The use
human disorder. Complete loss of LIS1 results in of RNAi knockdown of Lis1 definitively demon-
peri-implantation lethality, a result confirmed in strated that LIS1 is required for neuronal migration
another Lis1 knockout (14), demonstrating that (20, 21). Additionally, LIS1 knockdown was
Lis1 is an essential gene. associated with neural stem cell division (21),
Further studies demonstrated impairments of consistent with an important role for LIS1 in
motor coordination and cognition in Lis1-null proliferation.
heterozygotes (15), the types of deficits expected
based on the human phenotype. The disruption of LIS1, MTs and dynein
hippocampal cellular and synaptic physiology
(16) seen in the Lis1 mutant mice were the result Neuronal migration is driven by complex mech-
of neuronal migration defects. Golgi impregna- anisms that are orchestrated by many proteins
tion studies demonstrated that the dendritic arbor that interact mainly, but not exclusively, to
of neurons from both wild type and Lis11/KO promote the recruitment, organization, stability
cells located within stratum pyramidale were not and movement/function of microtubules and
significantly different, but the dendritic arbor actin cytoskeleton. LIS1 is an atypical microtu-
of heterotopic pyramidal cells was significantly bule associated protein. LIS1 consists of seven
smaller compared with both cell types, with spaced WD-40 repeats, and probably assumes the
stunted dendrites that make fewer dendritic structure of a propeller wheel similar to other
branches. The enhanced excitability displayed by WD-40 repeat proteins. Bovine and murine LIS1
slices from Lis11/KO mice provides a potential are almost identical in amino acid sequence. In
explanation for the intractable seizures seen in ILS mammals, LIS1 has at least two identified func-
patients. tions. LIS1 is a non-catalytic subunit of platelet-
Several lines of evidence support the conclusion activating factor acetylhydrolase (PAFAH)
that there are in vivo migrational defects in mice isoform Ib, an inactivating enzyme for platelet-
with reduction of LIS1 dosage (13). These include activating factor (PAF) (22), so the formal gene
histological analysis during development, BrdU name for LIS1 is PAFAH1B1. The catalytic alpha
birthdating experiments, and in vitro migration of subunits (a1 and a2) inactivate PAF, although the
granule cells from cerebellar cell re-aggregates. role of LIS1 in regulating enzymatic function is
Other studies from our laboratory have extended unknown. In addition, LIS1 has a non-enzymatic
and confirmed these findings (17, 18). The function; it is part of a highly conserved pathway
development of the pre-plate, CajalRetzius cells that regulates nuclear migration in fungi (23). The
and the radial glial scaffold appeared unaffected LIS1 homologue in the slime mold Aspergillus
by LIS1 levels. LIS1 also was required for the nidulans, NudF is part of a signaling pathway that
normal generation and survival of cortical VZ regulates nuclear migration along microtubules
neuroblasts. This study suggests that LIS1 is via the regulation of dynein motor function. In
important not only for neuronal migration, but this organism, nuclei migrate toward the growing
also for cell proliferation during neurogenesis as tip of hyphae to establish regular spacing, a pro-
well as for neuronal survival. Consistent with cess termed nucleokinesis (24, 25). Several nuclear
these studies, Lis1 is co-localized with cytoplasmic distribution mutants (nud mutants) have been
dynein at the mitotic kinetochores, indicating generated in this organism (25, 26). One of these,
a role of Lis1 in chromosomal behavior (19). For nudF is the fungal homologue of LIS1 (27), while
example, microinjection of anti-Lis1 antibody other nud mutants directly implicated cytoplasmic
resulted in a delay of mitotic progression accom- dynein and dynactin in this process (28, 29).
panied with chromosomal defects at the meta- The nud pathway is remarkably conserved in
phase plate. Lis1 is also expressed at the cortical eukaryotes, and the regulation of dynein motor
(marginal) area of proliferating epithelial cells, function by LIS1 and microtubule organization is
and its overexpression results in disruption of conserved in mammalian cells (Fig. 3). LIS1
the mitotic spindle organization as a result of associates with the microtubule fraction of cell
reductions in cortical distribution of dynein. supernatants (30) and is present in regions of high

299
Wynshaw-Boris
microtubule density in neurons and fibroblasts, provided a direct link between LIS1 and dynein
especially at the centrosome and microtubule- motors. There are at least two mammalian NudE
organizing center (MTOC) (19, 31). A large body homologues: mNudE (now termed NDE1) and
of data provide strong evidence that LIS1 dosage NUDEL (now termed NDEL1). Both are coiled
regulates cytoplasmic dynein motor function, but coil proteins of 344-345 amino acids and contain
how does LIS1 regulate neuronal migration? 10% serines. NDEL1 and NDE1 proteins co-
It appears that LIS1 is required for nuclear localize with LIS1 at the centrosome or MTOC in
movement during neuronal migration by coupling mitotic embryonic neuroblasts or fibroblasts. Both
the nucleus to the centrosome (18). Cerebellar NDEL1 and LIS1 directly interact with the CDHC,
granule cells from Lis11/2 mice displayed slowed providing a direct link between LIS1 and cytoplas-
migration and a lengthening of the distance mic dynein motors. mNudE (NDE1) binds to the
between the centrosome and the nucleus. LIS1 is dynein light chain, and its mechanism of dynein
localized to the centrosome with some extension regulation has not been as extensively studied at this
toward the nucleus. Consistent with an important time as NDEL1. Overall, these results are consistent
role in dynein function, inhibition of dynein by with the hypothesis that LIS1 and NDEL1 are
overexpression of dynamitin resulted in a similar responsible for the localization of CDHC (with
phenotype. Arrest of nuclear movement have been associated proteins) and regulate dynein motor
observed using RNAi knockdown of LIS1 in function and organelle transport in the cell.
embryonic cortical slice cultures (20, 21). These NDEL1 is a phosphoprotein. It is a bona fide
studies strongly suggest that nuclear movement substrate for Cdk5/p35 in vitro and in vivo at three
during neuronal migration is mediated by a LIS1 sites: S198, T219 and S231 (34, 35). Cdk5 phos-
complex regulating cytoplasmic dynein function, phorylation of NDEL1 controls its cellular locali-
a function that is conserved through evolution zation and probably influences dynein motor
from Aspergillus to mammals. function. These same serine residues are conserved
in mNudE (NDE1), suggesting that NDE1 may be
Central role of NudE homologs in LIS1 function
a target of Cdk5 as well. However, direct in vitro
and in vivo experiments can address whether or not
The nud pathway is conserved in all eukaryotes, NDE1 is a substrate for the Cdk5 complex.
including mammals. Several groups, including our In addition to CDK5 (or CDK1 in fibroblasts),
own, reported the cloning of mammalian NudE NDEL1 is phosphorylated by the polo-like
homologues from yeast two-hybrid screens using mitotic kinase Aurora-A (36). Polo-like kinases
LIS1 as bait (3235), and some of these studies are conserved in all eukaryotes (37). At the G2/M

Fig. 3. The LIS1/NDE/dynein com-


PP2A plex. LIS1 interacts directly with
NDE1, which interacts with the cyto-
plasmic dynein light chain (CDLC) to
PR65
LIS1
regulate centrosomal protein localiza-
tion and function. LIS1 also directly
NDE1 LIS1 NDEL1 P interacts with NDEL1, and LIS1 and
14-3-3
NDEL1 directly interact with the cyto-
P Ser198
LIS1 plasmic dynein heavy chain (CDHC) to
Thr219
Ser231
regulate centrosomal protein localiza-
Centrosomal CDLC CDHC
tion and function as well as microtubule
Proteins
dynamics. These interactions are critical
for nuclear movements and neuronal
CDK5 p35
migration. NDEL1 appears to be the
focus of regulation, as it is phosphory-
Centrosome lated by two different mitotic kinases:
function CDK5 with its required co-activator p35
Microtubule Ser251 phosphorylates NDEL1 on three sites
dynamics (S198, T219 and S231); and Aurora-A
kinase phosphorylates NDEL1 on S251.
CDK5-phosphorylated NDEL1 binds
Aurora to 14-3-3e, which protects NDEL1 from
A de-phosphorylation, perhaps by phos-
Nuclear movement phatase 2A (PP2A), as NDEL1 binds to
neuronal migration the PP2A regulatory subunit PR65a.

300
Lissencephaly and LIS1
transition and during early stages of mitosis, Plks made a knockout and conditional knockout of
have been implicated in the maturation of Ndel1 (40). Complete loss of Ndel1 results in early
centrosomes, the activation of Cdk1/cyclin B, embryonic lethality, demonstrating that Ndel1 is
disassembly of the Golgi complex and removal of an essential gene. RNAi knockdown studies of
cohesin subunits from chromatin. Aurora-A kinase Ndel1, Lis1 or dynein were performed by in utero
efficiently phosphorylates NDEL1 at serine 251 at electroporation into the developing neocortex
the beginning of mitotic entry coincidently with (20). RNAi knockdown of any of these three
Aurora-A activation at the G2-prophase transi- genes impaired neuronal positioning and caused
tion, while S251 phosphorylation suddenly disap- the uncoupling of the centrosome and nucleus,
peared after prophase (36). Comparing Aurora-A similar to our studies with Lis1 mutants (18).
mediated S251 phosphorylation with CDK1 Overexpression of LIS1 partially rescued the
mediated T219 NDEL1 phosphorylation, it ap- positioning defect caused by Ndel1 RNAi but
pears that phosphorylation by CDK1 inhibited not dynein RNAi, whereas overexpression of
further phosphorylation by Aurora-A, suggesting NDEL1 did not rescue the phenotype induced
that Aurora-A mediated phosphorylation of by Lis1 RNAi. These findings with NDEL1 are in
NDEL1 is transient and may trigger the entrance contrast to genetic ablation of the LIS1-interacting
into prophase, followed by degradation and protein NDE1 in mouse (41). Complete loss of
CDK1 mediated phosphorylation. Persistent and Nde1 results in viable mice with microcephaly,
simultaneous phosphorylation of NDEL1 by both especially in the cerebral cortex. Cortical lamina-
Aurora-A and CDK1 appeared to be inhibitory to tion was mostly preserved, but the mutant cortex
the generation of normal spindle and chromo- had fewer neurons and very thin superficial
some segregation. Phosphorylation of NDEL1 by cortical layers (layers 24), with dramatic defects
CDK1 facilitates katanin p60 recruitment to the in mitotic progression, mitotic orientation, and
centrosome to trigger microtubule remodeling mitotic chromosome localization in cortical pro-
and shortening (38). NDEL1 phosphorylation by genitors. The small cerebral cortex seems to reflect
Aurora-A is essential for mitotic entry and both reduced progenitor cell division and altered
centrosome separation by recruitment of TACC3 neuronal cell fates. Taken together, these results
and XMAP215 at the centrosome (36), critical provide strong evidence that NDEL1 and NDE1
events for centrosome-mediated MT assembly in interaction with LIS1 are particularly important
mitosis. This coordinate regulation of NDEL1 for microtubule organization, nuclear transloca-
phosphorylation by two mitotic kinases suggests tion, neuronal positioning and development.
that NDEL1 is a central component of the LIS1
complex to regulate proliferation, and the activity NDEL1 and 14-3-3e: a molecular explanation for
of NDEL1 and the LIS1/dynein complex appears MillerDieker syndrome
to be tightly regulated by a typical signal trans-
duction pathway (Fig. 3). The role of these To further investigate the potential differences
phosphorylation events during neuronal migra- between ILS and MDS, we examined the function
tion require further investigation. of genes deleted in MDS, but not ILS. We found
Recent structural studies have suggested that that 14-3-3e binds to P-NDEL1 (42), and that
subunits of PAF-AH compete with NDEL1 for 14-3-3e is always deleted in MDS patients (43).
LIS1 binding (39). They solved the structure of The 14-3-3 family of genes is found in all
LIS1 in complex with the a2/ a2 PAF-AH eukaryotic cells, and there are seven separate
homodimer. One LIS1 homodimer binds sym- genes or isoforms present in mammalian cells
metrically to one a2/ a2 homodimer via the highly (denoted by Greek letters). These highly con-
conserved top faces of the LIS1 b propellers. The served regulatory molecules bind to sequence-
same surface of LIS1 contains sites of mutations specific phosphoserine and phosphothreonine
causing lissencephaly and overlaps with a putative motifs. More than 100 binding partners for 14-3-3
dynein binding surface. NDEL1 competes with proteins have been discovered, resulting in mod-
the a2/ a2 homodimer for LIS1 via a complex ulation of signal transduction, cell cycle control,
interaction that requires both the N- and C- apoptosis, stress responses, vesicular transport
terminal domains of LIS1. and malignant transformation (44). We produced
mice deficient for 14-3-3e (42). These mice display
developmental brain and neuronal migration
Functional studies of NDEL1 and NDE1
defects similar to Lis1 heterozygous mutant mice.
The studies in the previous section suggest that 14-3-3e1/2 mice showed mild disorganization
NDEL1 has a critical and central role in mediating in the hippocampus and thinning of the cortex
the effects of LIS1. Consistent with this notion, we that was more severe in the 14-3-3e2/2 mice.

301
Wynshaw-Boris
Fig. 4. Similarity between interkinetic
CP
nuclear migration and neuronal migra-
tion. Labeling is the same as Fig. 1,
IZ except the centrosome is represented as
a red circle. A cell undergoing interki-
netic nuclear migration is enlarged on
VZ the left, while a migrating neuron is
enlarged on the right. Microtubules that
Interkinetic Neuronal stretch from the centrosome to the
nuclear migration nucleus are red.
migration

Neuronal migration was also quantitated by (Fig. 4). During interkinetic nuclear migration,
BrdU analysis and revealed that, similar to Lis1 the centrosome is at the apical surface and the
mutant mice, as the dose of 14-3-3e is reduced, the nucleus is more basal. The nuclei move in a basal
rate of neuronal migration decreases. Double direction during G1, and during G2 toward the
heterozygotes of 14-3-3e and Lis1 display more apical surface, where they divide. The nucleus is
severe migration defects in both the cortex and surrounded by microtubules anchored at the
hippocampus than single heterozygotes, provid- centrosome. During neuronal migration, the
ing genetic evidence that 14-3-3e may be respon- migration of the nucleus takes place in a similar
sible for most severe form of LIS, complete agyria, fashion, but reversed by 180. The centrosome is
seen in MDS. 14-3-3e binds directly to CDK5/ ahead of the nucleus and is attached to the nucleus
p35-phosphorylated NDEL1, and this binding by microtubules. The centrosome moves forward
maintains NDEL1 phosphorylation (Fig. 3). As into the leading process, followed by the nucleus
NDEL1 also binds to a regulatory subunit of the that is tethered to the centrosome by micro-
PP2A phosphatase (Fig. 3), it is likely that tubules. Interkinetic nuclear migration and neu-
binding of 14-3-3e to P-NDEL1 protects it from ronal (nuclear) migration both involved an
phosphatase attack. Similar to LIS1, reduction of oscillation in which the distance between the
14-3-3e results in mislocalization of NDEL1 and nucleus and centrosome lengthens and then
LIS1, consistent with reduction of cytoplasmic contracts. As LIS1 and the dynein complex are
dynein function. These results establish a crucial essential for interkinetic nuclear migration and
role for 14-3-3e in neuronal development by neuronal migration by coupling the centrosome to
sustaining the effects of CDK5 phosphorylation. the nucleus, it is reasonable to hypothesize that
It also provides a molecular explanation for the both processes act in similar cell biological
differences in severity of human neuronal migra- fashions. As the major difference between these
tion defects with 17p13.3 deletions, namely ILS processes is the position of the centrosome and
and MDS. In addition, it places 14-3-3e in the nucleus relative to the apical surface, it is likely
LIS1 pathway. that apical-basal polarity is an important factor in
controlling the position of the centrosome and the
action of the LIS1 complex on nuclear-centrosomal
Conclusions and future directions
coupling.
The identification of LIS1 as the first lissence-
phaly gene and the first gene required for neuronal
migration has illustrated the importance of Acknowledgements
cytoplasmic dynein in the regulation of neuronal The author is grateful for all those who have worked in his
migration by modulating nuclear migration in laboratory on neuronal migration, and for his colleagues in the
a pathway conserved in virtually all eukaryotes. neuronal migration field. Because of restraints on the number
By examining the effect of reduction of LIS1 either of references, it was impossible to cite all relevant papers. This
genetically or by RNAi knockdown, it is clear that work was supported by grants from the National Institutes of
LIS1 has a broader effect on proliferation and Health (NS41030 and HD047380).
survival. Does LIS1 and its complex regulate
distinct aspects of proliferation, migration and
survival? Or is there a similarity between these References
cellular processes that LIS1 and its complex
1. Gupta A, Tsai LH, Wynshaw-Boris A. Life is a journey:
regulates? Although direct experiments have not a genetic look at neocortical development. Nat Rev Genet
been performed to address these questions, it is 2002: 3: 342355.
apparent that there is similarity between inter- 2. Tsai LH, Gleeson JG. Nucleokinesis in neuronal migration.
kinetic nuclear migration and neuronal migration Neuron 2005: 46: 383388.

302
Lissencephaly and LIS1
3. Ayala R, Shu T, Tsai LH. Trekking across the brain: the plasmic dynein to regulate cortical neuronal positioning.
journey of neuronal migration. Cell 2007: 128: 2943. Neuron 2004: 44: 263277.
4. Vallee RB, Tsai JW. The cellular roles of the lissencephaly 21. Tsai JW, Chen Y, Kriegstein AR, Vallee RB. LIS1 RNA
gene LIS1, and what they tell us about brain development. interference blocks neural stem cell division, morphogenesis,
Genes Dev 2006: 20: 13841393. and motility at multiple stages. J Cell Biol 2005: 170:
5. Barkovich AJ, Kuzniecky RI, Jackson GD, Guerrini R, 935945.
Dobyns WB. A developmental and genetic classification for 22. Hattori M, Adachi H, Tsujimoto M, Arai H, Inoue K.
malformations of cortical development. Neurology 2005: Miller-Dieker lissencephaly gene encodes a subunit of brain
65: 18731887. platelet-activating factor acetylhydrolase. Nature 1994:
6. Dobyns WB, Elias ER, Newlin AC, Pagon RA, Ledbetter 370: 216218.
DH. Causal heterogeneity in isolated lissencephaly. 23. Morris NR. Nuclear migration: from fungi to the
Neurology 1992: 42: 13751388. mammalian brain. J Cell Biol 2000: 148: 10971101.
7. Dobyns WB, Stratton RF, Greenberg F. Syndromes with 24. Oakley BR, Morris NR. Nuclear movement is beta
lissencephaly. I: Miller-Dieker and Norman-Roberts syn- tubulin-dependent in Aspergillus nidulans. Cell 1980: 19:
dromes and isolated lissencephaly. Am J Med Genet 1984: 255262.
18: 509526. 25. Xiang X, Morris NR. Hyphal tip growth and nuclear
8. Dobyns WB, Carrozzo R, Ledbetter DH. Frequent migration. Curr Opin Microbiol 1999: 2: 636640.
deletions of the LIS1 gene in classic lissencephaly. Ann 26. Morris NR. Mitotic mutants of Aspergillus nidulans.
Neurol 1994: 36: 489490. Genet Res 1975: 26: 237254.
9. Chong SS, Pack SD, Roschke AV et al. A revision of 27. Xiang X, Osmani AH, Osmani SA, Xin M, Morris NR.
the lissencephaly and Miller-Dieker syndrome critical NudF, a nuclear migration gene in Aspergillus nidulans, is
regions in chromosome 17p13.3. Hum Mol Genet 1997: 6: similar to the human LIS-1 gene required for neuronal
147155. migration. 1995: 6: 297310.
10. Reiner O, Carrozzo R, Shen Y et al. Isolation of a Miller- 28. Xiang X, Beckwith SM, Morris NR. Cytoplasmic dynein is
Dieker lissencephaly gene containing G protein beta- involved in nuclear migration in Aspergillus nidulans. Proc
subunit-like repeats. Nature 1993: 364: 717721. Natl Acad Sci U S A 1994: 91: 21002104.
11. LoNigro C, Chong CS, Smith AC, Dobyns WB, Carrozzo 29. Beckwith SM, Rohgi CH, Liu B, Morris NR. The 8-kD
R, Ledbetter DH. Point mutations and an intragenic cytoplasmic dynein light chain is required for nuclear
deletion in LIS1, the lissencephaly causative gene in migration and for dynein heavy chain localization in
isolated lissencephaly sequence and Miller-Dieker syn- Aspergillus nidulans. J Cell Biol 1998: 143: 12391247.
drome. Hum Mol Genet 1997: 6: 157164. 30. Sapir T, Elbaum M, Reiner O. Reduction of microtubule
12. Pilz DT, Matsumoto N, Minnerath S et al. LIS1 and XLIS catastrophe events by LIS1, platelet- activating factor
(DCX) mutations cause most classical lissencephaly, but acetylhydrolase subunit. EMBO J 1997: 16: 69776984.
different patterns of malformation. Hum Mol Genet 1998: 31. Smith DS, Niethammer M, Zhou Y, Gambello MJ,
7: 20292037. Wynshaw-Boris A, Tsai LH. Regulation of cytoplasmic
13. Hirotsune S, Fleck MW, Gambello MJ et al. Graded dynein behavior and mirotubule organization by mamma-
reduction of Pafah1b1 (Lis1) activity results in neuronal lian Lis1. Nat Cell Biol 2000: 2: 767775.
migration defects and early embryonic lethality. Nat Genet 32. Feng Y, Olson EC, Stukenberg PT, Flanagan LA,
1998: 19: 333339. Kirschner MW, Walsh CA. Interactions between LIS1
14. Cahana A, Escamez T, Nowakowski RS et al. Targeted and mNudE, a central component of the centrosome, are
mutagenesis of Lis1 disrupts cortical development and LIS1 required for CNS lamination. Neuron 2000: 28: 665679.
homodimerization. Proc Natl Acad Sci U S A 2001: 98: 33. Kitagawa M, Umezu M, Aoki J, Koizumi H, Arai H,
64296434. Inoue K. Direct association of LIS1, the lissencephaly gene
15. Paylor R, Hirotsune S, Gambello MJ, Yuva-Paylor L, product, with a mammalian homologue of a fungal nu-
Crawley J, Wynshaw-Boris A. Impaired learning and clear distribution protein, rNUDE. FEBS Lett 2000: 479:
motor behavior in heterozygous Pafah1b1 (Lis1) mutant 5762.
mice. Learn Mem 1999: 6: 521537. 34. Niethammer M, Smith DS, Ayala R et al. The Lis1
16. Fleck MW, Hirotsune S, Gambello MJ et al. Hippocampal interacting protein Nudel is a Cdk5 substrate that interacts
abnormalities in Lis1 mutant mice provide a neuronal basis with cytoplasmic dynein. Neuron 2000: 28: 697711.
for epileptogenesis in neuronal migration defects. J Neurosci 35. Sasaki S, Shionoya A, Ishida M et al. A LIS1/NUDEL/
2000: 20: 24392450. cytoplasmic dynein heavy chain complex in the develop-
17. Gambello MJ, Darling DL, Yingling J, Tanaka T, Gleeson ing and adult central nervous system. Neuron 2000: 28:
JG, Wynshaw-Boris A. Multiple dose-dependent effects of 681696.
Lis1 on cerebral cortical development. J Neurosci 2003: 23: 36. Mori D, Yano Y, Toyo-oka K et al. Phosphorylation of
17191729. NDEL1 by aurora-A kinase regulates centrosome matura-
18. Tanaka T, Serneo FF, Paczkowski C, Gambello MJ, tion and separation, which are essential for G2-M pro-
Wynshaw-Boris A, Gleeson JG. Lis1 and doublecortin gression. Mol Cell Biol 2006: 27: 352367.
function with dynein to mediate coupling of the nucleus to 37. Nigg EA. Mitotic kinases as regulators of cell division and
the centrosome in neuronal migration. J Cell Biol 2004: its checkpoints. Nat Rev Mol Cell Biol 2001: 2: 2132.
165: 709721. 38. Toyo-oka K, Sasaki S, Yano Y et al. Recruitment of
19. Faulkner NE, Dujardin DL, Tai C et al. A role for the katanin p60 by phosphorylated NDEL1, an LIS1 interact-
lissencephaly gene LIS1 in mitosis and cytoplasmic dynein ing protein, is essential for mitotic cell division and neu-
function. Nat Cell Biol 2000: 2: 784791. ronal migration. Hum Mol Genet 2005: 14: 31133128.
20. Shu T, Ayala R, Nguyen MD, Xie Z, Gleeson JG, Tsai LH. 39. Tarricone C, Perrina F, Monzani S et al. Coupling
Ndel1 operates in a common pathway with LIS1 and cyto- PAF signaling to dynein regulation: structure of LIS1

303
Wynshaw-Boris
in complex with PAF-acetylhydrolase. Neuron 2004: 44: via binding to NUDEL: a molecular explanation for
809821. Miller-Dieker syndrome. Nat Genet 2003: 34: 274285.
40. Sasaki S, Mori D, Toyo-oka K et al. Disruption of Ndel1 43. Cardoso C, Leventer RJ, Ward HL et al. Refinement of
causes neuronal migration defects and early embryonic a 400-kb critical region allows genotypic differentiation
lethality. Mol Cell Biol 2005: 25: 78127827. between isolated lissencephaly, Miller-Dieker syndrome
41. Feng Y, Walsh CA. Mitotic spindle regulation by and other phenotypes secondary to deletions of 17p13.3.
Nde1 controls cerebral cortical size. Neuron 2004: 44: Am J Hum Genet 2003: 72: 918930.
279293. 44. Darling DL, Yingling J, Wynshaw-Boris A. Role of 14-3-3
42. Toyo-oka K, Shionoya A, Gambello MJ et al. Wynshaw- proteins in eukaryotic signaling and development. Curr
Boris A. 14-3-3e is important for neuronal migration Top Dev Biol 2005: 68: 281315.

304

Vous aimerez peut-être aussi