ArticlePDF Available

Abstract and Figures

During prenatal and postnatal development of the mammalian brain, new neurons are generated by precursor cells that are located in the germinal zones. Subsequently newborn neurons migrate to their destined location in the brain. On the migrational route immature neurons interact via a series of recognition molecules with a plethora of extracellular cues. Stimuli that are conveyed by extracellular cues are translated into complex intracellular signaling networks that eventually enable neuronal migration. In this Focused Review we discuss signaling networks underlying neuronal migration emphasizing molecules and pathways that appear to be neuron-specific.
This content is subject to copyright.
Frontiers in Neuroscience www.frontiersin.org March 2011 | Volume 5 | Article 28 | 1
FOCUSED REVIEW
published: 28 March 2011
doi: 10.3389/fnins.2011.00028
Signaling in migrating neurons: from
molecules to networks
Konstantin Khodosevich1,2* and Hannah Monyer1,2*
1 Department of Clinical Neurobiology, German Cancer Research Center (DKFZ), Heidelberg, Germany
2 Department of Clinical Neurobiology, Heidelberg University Medical Center, Heidelberg, Germany
During prenatal and postnatal development of the mammalian brain, new neurons are generated
by precursor cells that are located in the germinal zones. Subsequently newborn neurons migrate
to their destined location in the brain. On the migrational route immature neurons interact via a
series of recognition molecules with a plethora of extracellular cues. Stimuli that are conveyed
by extracellular cues are translated into complex intracellular signaling networks that eventually
enable neuronal migration. In this Focused Review we discuss signaling networks underlying
neuronal migration emphasizing molecules and pathways that appear to be neuron-specific.
Keywords: neuroblasts, signaling networks, neuronal-specific pathways
IntroductIon
In mammals neuronal migration is a fundamental
process in the development of nervous system.
The peripheral and central nervous system (CNS)
comprise numerous neurons that are born in vari-
ous locations during development and migrate
shorter or longer distances to their destination
site. Precise coordination of neuronal migration
is a prerequisite for the correct positioning and
subsequent wiring of neurons into functional
circuits. Impairment in migration results in
structural defects that are accompanied by severe
mental abnormalities.
In the peripheral nervous system, neuronal
precursors originate from undifferentiated neu-
ral crest cells and migrate during embryonic
development to their final locations (Francis and
Landis, 1999; Glebova and Ginty, 2005). In the
CNS, most neurons derive from precursor cells
that reside in the ventricular zone. Projection
neurons are born in the ventricular zone of the
dorsal telencephalon and migrate radially toward
the pia (Marin and Rubenstein, 2003). On the
contrary, interneurons are born in the ventral
telencephalon, i.e., in the ventricular zone of
the medial, lateral, and caudal ganglionic emi-
nences from where they migrate tangentially to
the dorsal telencephalon (Corbin et al., 2001;
Marin and Rubenstein, 2003). Postnatally new
neurons continue to be generated in two brain
regions, namely in the subgranular zone (SGZ)
of the dentate gyrus in the hippocampus and
the subventricular zone (SVZ) of the lateral
ventricles (Lledo et al., 2006; Zhao et al., 2008).
Neuroblasts (immature neurons) in the hip-
pocampal SGZ migrate a short distance into the
granule cell layer of the dentate gyrus and inte-
grate into previously established neural circuits
(Kempermann et al., 2004). Neuroblasts originat-
ing in the SVZ migrate over long distances via the
rostral migratory stream to the olfactory bulb,
where they mature into granule or periglomeru-
lar neurons (Lledo and Saghatelyan, 2005; Alonso
et al., 2006). Under pathological conditions, neu-
roblasts generated in the SVZ migrate also into
injured cortex and striatum (Parent et al., 2002;
Kreuzberg et al., 2010). On the migratory route,
neuroblasts receive different stimuli from extra-
cellular cues that are paramount in migration
guiding. As a result, a number of intracellular
signaling molecules are activated. Ultimately, all
signaling pathways are integrated in a “master
network” that controls the final intracellular out-
put in response to all extracellular inputs and
modifies the cytoskeleton machinery accordingly
resulting in neuroblast migration.
Edited by:
Seth G. N. Grant, The Wellcome Trust
Sanger Institute, UK
Reviewed by:
Seth G. N. Grant, The Wellcome Trust
Sanger Institute, UK
Peter C. Kind, University of Edinburgh,
UK
*Correspondence:
Konstantin Khodosevich graduated from
Lomonosov Moscow State University and
completed his PhD in human genetics at
the Institute of Bioorganic Chemistry,
Moscow. After his move to the lab of
Hannah Monyer, he started work on the
role of interneurons in higher brain
function. He is currently a senior scientist
in Hannah Monyer’s lab where he focuses
on signaling networks in the nervous
system during postnatal and prenatal
development.
k.khodosevich@dkfz-heidelberg.de;
Khodosevich and Monyer Signaling in migrating neuroblasts
Frontiers in Neuroscience www.frontiersin.org March 2011 | Volume 5 | Article 28 | 2
SpecIfIc SIgnalIng for neuronal
mIgratIon
An ever-growing number of studies on neuronal
migration employing by and large anatomical
techniques goes back to the 1960s. Starting in the
1980s, the function of particular genes/proteins in
neuronal migration became the focus of numer-
ous experimental investigations. Whilst these
valuable studies have identified important players
in migration, many of which were cytoskeleton
proteins, during the last decade the interest grew
in identifying whole pathways and understanding
their complex interactions.
In a recent study (Khodosevich et al., 2009)
using microarray gene expression analysis we
identified several intracellular signaling pathways
underlying neuroblast migration. The identi-
fied pathways were corroborated and further
expanded using bioinformatics analysis derived
from mining of public data that also included
information obtained in other cell-types and
species (see Data mining for signaling pathway
analysis). This powerful approach allows filling
in of gaps and proposing whole pathways that
would be difficult to identify if one used experi-
mental data from one system only. Needless to
say, the signaling components resulting from
bioinformatics analysis can and should be tested
experimentally to provide functional proof for
their involvement in a specific system (in our
study it was murine neuroblasts migrating in
the rostral migratory stream). For this Focused
Review we used only data obtained in studies on
mammalian neuronal migration to generate a sig-
naling network whose components are discussed
in more detail. The ultimate goal is to eventually
arrive at a unique intracellular network under-
lying signaling in migrating neuroblasts. Here
we collected published data for all subtypes of
migrating neuroblasts regardless of their birth-
place (prenatal ganglionic eminences, postnatal
SVZ, precursors of peripheral nervous system,
etc.), mode of migration (radial and tangential
migration; migration in cell chains and individual
cell migration). We also used data obtained in
vitro including neurite outgrowth studies. Clearly,
each subtype of migrating neuroblasts has its
own signaling components that are “tuned” to
the microenvironment (i.e., available extracellu-
lar stimuli) of migration. Furthermore, different
modes of neuroblast migration might rely more
on one type of molecular cues in microenviron-
ment than the other, e.g., soluble factors, mem-
brane-bound receptors, or extracellular matrix.
However, subtypes of migrating neuroblasts share
the majority of intracellular signaling compo-
nents that integrate external stimuli and result
in appropriate output. With the increasing avail-
ability of experimental data it will be eventually
possible to perform a similar analysis focusing on
distinct types of migration and investigate shared
and specific molecules and their connections.
HubS In a SIgnalIng network
controllIng neuronal mIgratIon
In Figure 1 we summarize the results derived from
several hundred studies focusing on some aspects
of signaling that control neuronal migration/neu-
rite outgrowth (the names of the individual com-
ponents are legible upon downloading of Figure
S1 in Supplementary Material). The main feature
of the signaling network is the uneven distribu-
tion of connections between individual molecules
resulting in clustering of connections. Seven key
“hubs” (shown in yellow) of intracellular sign-
aling involve 2/3 of the connections within the
network (Figures 1A,B). Such network clustering
is typical for signaling networks and was shown
in many proteomic studies (see, e.g., Giot et al.,
2003; Pocklington et al., 2006). These signaling
hubs control the input and output of the network:
cell division protein kinase 5 (Cdk5), disabled
homolog 1 (Dab1), ras-related C3 botulinum
toxin substrate 1 (Rac1), focal adhesion kinase
(FAK), rat sarcoma (Ras), Rous sarcoma onco-
gene (Src), and phosphatidylinositol 3 kinase
(PI3K). Based on their connectivity resulting from
our analysis, seven hubs can be further subdivided
into two groups: Cdk5, Dab1, and Rac1 having
each 13–14 connections, while FAK, Ras, Src, and
PI3K having 7–9 connections.
While the small GTPase Rac1 and to a much
lesser extent Cdk5 kinase are involved in migra-
tion of non-neuronal cell-types, Dab1 is a spe-
cific component of neuronal migration signaling
(Bielas et al., 2004; Ayala et al., 2007). Dab1 is
a cytoplasmic adaptor molecule that was first
described as a binding partner of the Src family
kinases Src and Fyn (Howell et al., 1997). Later its
action was also linked to Reelin signaling (Bielas
et al., 2004). However, Dab1 is involved in neuro-
nal migration not only as a target in Reelin signal-
ing, but also in amyloid precursor protein (APP;
Young-Pearse et al., 2010) and integrin signaling
(Dulabon et al., 2000; Figure 2A). Furthermore,
Dab1 is connected to other pathways via Cdk5
and Src kinases (Keshvara et al., 2002; Bock et al.,
2003; Kuo et al., 2005; Figure 2A). Since Dab1 also
directly binds to several microtubule-associated
proteins – Lis1, DISC1, and CRMP (Assadi et al.,
2003; Yamashita et al., 2006; Young-Pearse et al.,
2010) – it is a strong candidate that “tunes” com-
mon mechanisms of cell migration to neuron-
specific migration (Figure 2A).
*Correspondence:
Hannah Monyer got her MD at the
University of Heidelberg, Germany. She
was a postdoctoral fellow at Stanford
University, USA and ZMBH, Germany.
Since 1999, she is Professor and Head of
the Department of Clinical Neurobiology
at the Medical Center Heidelberg
University, and since 2010 also Professor
at the German Cancer Research Center,
Heidelberg. The studies of Hannah
Monyer’s lab aim at understanding
molecular and cellular mechanisms
underlying synchronous network
oscillations, learning, and memory with
particular focus on the functional
characterization of GABAergic
interneurons.
monyer@urz.uni-hd.de
Subventricular zone
Germinal zone in the postnatal brain in
many mammals. Spans a few
intermixed cell layers located in the
lateral wall of the paired lateral
ventricles. Contains slow-dividing
neural stem cells, fast-dividing
transit-amplifying precursors,
neuroblasts, and a few other support
cell-types.
Neuroblasts
Immature neurons with a small cell
body and usually one leading neurite.
Neuroblasts migrate by protruding
neurite and then pulling cell body in the
direction of migration.
Rostral migratory stream
Stream of migrating neuroblasts from
the subventricular zone to the olfactory
bulb. Identified in many mammals
including rodents, sheep, and primates.
Small GTPases
GTP/GDP-binding proteins. They are
active when bound to GTP and inactive
when bound to GDP. Exchange of GDP
with GTP (=activation) is catalyzed by
proteins called “guanine nucleotide
exchange factors” (GEFs). GTP
hydrolysis (=inactivation) is catalyzed
by “GTPase-activating proteins”
(GAPs).
Kinases
Enzymes that add phosphate groups
(=phosphorylate) to other proteins or
to themselves. Phosphorylation can
result in activation or inhibition of the
recipient protein.
Khodosevich and Monyer Signaling in migrating neuroblasts
Frontiers in Neuroscience www.frontiersin.org March 2011 | Volume 5 | Article 28 | 3
PI3K
A B
Dab1
Ras
Src FAK
Cdk5
Rac1
Microtubules
PI3K Dab1
Ras
Src FAK
Cdk5
Rac1
Microtubules
Actin filaments Actin filaments
FIGURE 1 | Signaling network controlling neuronal migration – the scheme
is based on experimental data derived from several hundred publications.
The only legible names denote network components that constitute “signaling
hubs.The scheme is legible upon downloading Figure S1 in Supplementary
Material. (A) Seven hubs in the neuronal migration signaling network (shown in
yellow). (B) More than 2/3 of the network connections (shown by red lines)
involve hubs. Color code for molecules: yellow – signaling hubs, red –
extracellular ligands/matrix components, green – transmembrane receptors/
channels/transporters, etc., blue – intracellular signaling molecules, magenta –
microtubule/actin-associated proteins, orange – cell nucleus components.
Cullin 5
Fbxw7
Fbxw7
SOCS
CIN85
Dab 1
Dab 1
Dab 1
Nova2
PI3K
splicing
Dab1
AAAA
AAAA
PI3K
PI3K
Src
Src
Eph
TrkB
Src
Crk/CrkL
Notch-ICD
Microtubules
DISC1
DCX
Fyn
APP
Fyn
CDKL5
PKCζ
Cdk5 Cdk5
FAK
FAK
FAK
EGFR receptors,
cell adhesion
molecules
CRMP1/2
Pctaire-1
p27kip 1 Pak1
c-Abl
Neurabin-1
Dixdc1Ndel1
DCX Tau
p35
p39
ERK1/2
MEKK
(DSLK,MEKK4)
Microtubules
F-actin
Rac1
Ras
Raf
(R-Ras, Rap-1)
(R-Ras, Rap-1)
Ras
Rnd1 Notch-ICD
(R-Ras, Rap-1)
Vav2/3
Vav2/3
Trio
Trio
Trio
Cdk5
Paxillin
P-Rex1
FARP2
Pak1
C3G
Rnd1
IQGAP1
Eph TrkB Met
receptors A/B
p27kip1
microtubule/actin
filaments-associated
proteins
G-protein
other
kinases
guanine nucleotide
exchange factors (GEFs)
ubiquitin ligases
transcriptional
regulators
degraded
proteins
chemicals
small GTPases
Wave1
srGAP
Cortactin
PIP3
Dab 1
Lis1
Notch1
degraded
Dab1
degraded
FAK PIP2 PIP3
Dab1
degraded
Met
TrkB
receptors A/B
N-syndecan
EGFR ApoER2
CB1R
Integrins DCC
L1/ CHL1
ApoER2 VLDLR Integrins
GFRα1
NCAM Plexin-A/B Trkc
Plexin-A/B
TrkA
A B C
D E F G
Dab1
Ras
VLDLR
FIGURE 2 | Signaling hubs in migrating neurons. (A) Dab1 signaling. (B) Cdk5 signaling. (C) Rac1 signaling. (D) FAK signaling. (E) Src signaling. (F) PI3K signaling.
(G) Ras signaling. Arrow – activation, circle – interaction, T-shaped end – inhibition. Color code of molecules is the same as in Figure 1. Note that some links are
indirect, for full scheme see Figure S1 in Supplementary Material.
Other central hubs in the signaling net-
work of neuronal migration are Cdk5 and
Rac1 (Figures 2B,C, respectively). Although
Cdk5 was mentioned also in the context of
non- neuronal cell migration, most evidence
for its role in migration has been derived from
Khodosevich and Monyer Signaling in migrating neuroblasts
Frontiers in Neuroscience www.frontiersin.org March 2011 | Volume 5 | Article 28 | 4
they receive input from many receptors, such as
DCC, L1, integrins, Reelin receptors ApoER2
and VLDLR, EGFR, TrkB, etc. (see Figure S1 in
Supplementary Material). Independent of their
individual activation, these kinases can interact
with each other resulting in FAK activation by Src
(Kuo et al., 2005). FAK and Src operate upstream
of the major cytoskeleton-linked hubs, Cdk5 and
Rac1. There is only limited evidence for a direct
connection between Src and the cytoskeleton in
migrating neurons. It involves the phosphoryla-
tion of cortactin, an actin-binding protein, by Src
(Hienola et al., 2006).
Interestingly, two other hubs, namely PI3K and
Ras, that are importantly involved in migration
of other cell-types are not that heavily connected
within the signaling network in migrating neu-
rons (Figures 2F,G, respectively). PI3K is a kinase
involved in numerous cell processes. However, in
neuronal migration signaling PI3K is activated
only by growth factor receptors and it is also a
downstream effector of Dab1 (Figure 2F). The
same holds true for Ras, with an addition of
semaphorin signaling (Figure 2G), in particular
if one takes into account that in our scheme Ras
stands for the whole protein family comprising
about 30 members. Direct involvement in neu-
ronal migration was demonstrated for two Ras
family members, R-Ras and Rap1 (Oinuma et al.,
2004; Toyofuku et al., 2005; Voss et al., 2008). Such
limited involvement of PI3K and Ras activity in
migrating neurons can hardly be explained by
a scarcity of study – these are well-known sig-
naling molecules, which have been studied for
decades. Most likely, PI3K and Ras have major
roles in growth factor and semaphorin signal-
ing while in other signaling cascades they play a
minor role (Figures 2F,G, see also Figure S1 in
Supplementary Material for full scheme).
neuronal-SpecIfIc SIgnalIng patHwayS
Signaling network in migrating neuroblasts is
complex and consists of hundreds interconnected
molecules that have been identified already and
yet more to be added to this list (Figure 1 and
Figure S1 in Supplementary Material). However,
the complexity can be reduced when focusing on a
specific stimulus (e.g., HGF or hepatocyte growth
factor; Figure 3A) or a group of similar stimuli
(e.g., all four members of neurotrophin protein
family; Figure 3B). Distinct extracellular stimuli
activate only one or a few subsets/pathways of the
signaling network in migrating neuroblasts and
some signaling pathways are specific for migrating
neurons. Most notable neuronal-specific exam-
ples are those that involve calcium, neurotrophin,
and Notch signaling (Figure 3C).
studies focusing on neuronal migration. Thus,
this signaling hub can be considered by and
large neuron-specific (Gupta and Tsai, 2003;
Nikolic, 2004). Conversely, Rac1 is a signal-
ing component of many migratory cell-types
(Fukata et al., 2003). In neurons almost all
extracellular migratory stimuli, such as Slit,
integrins, laminins, semaphorins, Reelin, GDNF,
EGF, neurotrophin-3 (NT-3), neurotrophin-4
(NT-4), brain-derived neurotrophic factor
(BDNF), netrin, calcium, etc. (see Figure S1 in
Supplementary Material), eventually converge
on these two intracellular proteins. Both hubs
are directly connected to microtubules and actin
filaments (Figures 2B,C). Thus, the convergence
of activating and inhibitory signals onto Cdk5
and Rac1 confers them a pivotal role in deter-
mining the direction and speed of neuronal
migration. Cdk5/Rac1 are also key players in
controlling the initiation and stop of migration.
Cdk5 and Rac1 activation is modulated by
other signaling molecules. Cdk5 is phosphor-
ylated and thus activated by p35, p39, and c-Abl
(Zukerberg et al., 2000; Keshvara et al., 2002;
Beffert et al., 2004; Zhao et al., 2009). However,
the regulation of these proteins that modulate
Cdk5 activity in migrating neurons has not been
explored much so far. Whilst it was shown that
PKCδ and Pctaire-1 regulate p35 activity (Cheng
et al., 2002; Zhao et al., 2009), there is no informa-
tion regarding the identity of molecules operating
upstream of p39. c-Abl tyrosine kinase, known
to be involved in migration of other cell-types
as well, exerts its action in neuronal migration
downstream of Slit/Robo signaling (Rhee et al.,
2002). Thus, modulation of Cdk5 in migrating
neurons remains to be explored.
Rac1 is a small GTPase whose activity is trig-
gered by guanine nucleotide exchange factors
(GEFs) and inhibited by GTPase-activating pro-
teins (GAPs; Figure 2C). Four GEFs activate Rac1
in migrating neurons – Vav2/3, FARP2, Trio, and
P-Rex1 (Schmid et al., 2004; Toyofuku et al., 2005;
Yoshizawa et al., 2005; Khodosevich et al., 2009;
Peng et al., 2010), but only one GAP inactivates it
– Slit–Robo GAP (srGAP; Wong et al., 2001). The
discrepancy in inhibitory and activating inputs
can be explained to some extent that activating
signaling is easier to detect. However, it is very
likely that there are other, still to be identified
GAPs that inactivate Rac1 in migrating neurob-
lasts. Interestingly, Rac1 activity is also modu-
lated by PKCζ, a kinase critically involved in the
neuronal polarization (Khodosevich et al., 2009;
Khodosevich and Monyer, 2010).
Amongst the less connected hubs, FAK and Src
are noteworthy (Figures 2D,E, respectively) since
Data mining for signaling pathway
analysis
Analysis of published data to connect
many molecules, usually obtained after
microarray search, in signaling
pathways. Data mining is performed
with the help of bioinformatics software
searching in published literature for
pairs of genes/proteins in the same
article/abstract/sentence, with/without
linking words, such as activation/
inhibition, etc.
Khodosevich and Monyer Signaling in migrating neuroblasts
Frontiers in Neuroscience www.frontiersin.org March 2011 | Volume 5 | Article 28 | 5
RhoA
RhoA
Cdc42
Rac1
Ras
NT-3
BDNF
Somatostatin
NT-3
NT-3
NGF
NT-3
NT-4
NMDA
VGCC
NMDA
AMPA
Gluk5
NKCC1
CaMKIV
Notch-ICD
Notch-ICD
Notch-ICD
Fbxw7
Fbxw7
IQGAP1
ADAM10
CaM
IQGAP1
presenilin
Wave1
N-WASP
Ndel 1
Ndel 1
CaMKIV
CDKL5
CaMKK2
p35
PKCδ
CaMKII
ERK1/2
MEK
CaMKIV
Raf
Cdk5
PI3K
BDNF
NT-3
NT-3
BDNF
CAPS2
Notch1
degraded
F-actin
BDNF
PI3K
A
C
B
Dab1
Ras
Src FAK
Cdk5
Rac1
Microtubules
PI3K Dab1
Ras
Src FAK
Cdk5
Rac1
Microtubules
Dixdc1
NT-3
Ca2+Ca2+ Notch1
degraded
degraded
proteins
TrkB
secreted ligands
NT-3
BarhI1
Notch-ICD
RBP-J
Hes1,Hes5 BDNF
CREB
small GTPases
ion channels kinases
ubiquitin ligases
other
microtubule/actin
filaments-associated
proteins
transcriptional
regulators
receptors,
cell adhesion
molecules
AA A AA A AAA
PIP3
K+
Ca2+
Ca2+
Ca2+
Ca2+
Ca2+
CI-
CI-
CI-CI-
Ca2+ K+K+
PIP2
Delta-like/Jagged
GABA Glutamate
CI-
CI-
CI-CI-Ca2+ Ca2+
Ca2+
Ca2+
Ca2+ Ca2+
Ca2+
K+K+K+
K+
K+
K+
L, N-Ca2+
channels
K+
channels
Notch1 SSTR TrkC TrkA TrkB
Microtubules
Tau Dynein/Dynactin DCX
GABAR
chemicals
FIGURE 3 | Examples of signaling pathways in migrating neurons. (A) Scheme of
HGF signaling (highlighted by red). (B) Scheme of signaling for neurotrophin protein
family (NT-3, NT-4, NGF, BDNF; highlighted in red). (C) Calcium, neurotrophin, and
Notch signaling are shown together. Arrow activation, circle interaction, T-shaped
end inhibition. Color code of molecules is the same as in Figure 1. Note that some
links are indirect, for full scheme see Figure S1 in Supplementary Material.
Khodosevich and Monyer Signaling in migrating neuroblasts
Frontiers in Neuroscience www.frontiersin.org March 2011 | Volume 5 | Article 28 | 6
granule-associated protein that constitutes a
further link between calcium and neurotrophin
signaling (Sadakata et al., 2007).
In addition to its major and well-studied func-
tion in determining cell fate choice and survival,
Notch1 also plays a role in neuronal migration
(Figure 3C). Upon ligand binding and stimula-
tion, the Notch receptor gets cleaved by ADAM10
and subsequently by the presenilin 1-containing
γ-secretase complex (Louvi et al., 2004; Jorissen
et al., 2010). Notch-ICD (intracellular domain)
translocates into the nucleus to interact with
RBP-J and activates Notch-dependent transcrip-
tion of Hes1 and Hes5 (Hashimoto-Torii et al.,
2008). It is of note that the signaling hub Dab1
controls Notch-ICD degradation by inhibiting
Fbxw7, a component of the ubiquitin protein
ligase complex (Hashimoto-Torii et al., 2008).
tranScrIptIon actIvatIon In mIgratIng
neuroblaStS
Although migrating neuroblasts are still able to
proliferate, a major switch in the transcription
program between precursor cells and neurob-
lasts must take place to account for proliferation
and migration. Thus, it stands to reason that
genes involved in precursor cell proliferation are
downregulated with a concomitant upregulation
of genes responsible for migration, leading to a
dramatic change in the gene expression profile.
This is supported by our previous results dem-
onstrating that around 1000 genes changed their
expression in neuroblasts that had migrated some
distance from the site of origin, the postnatal SVZ
(Khodosevich et al., 2009).
One of the most prominent transcription fac-
tors involved in the switch from precursor cells
to neuroblasts is distal-less homeobox protein
Dlx1/2 (see Figure S1 in Supplementary Material
for full scheme of transcriptional control). Dlx1/2
knockout animals exhibit strong neuronal migra-
tion defects (Cobos et al., 2007). Regulation by
Dlx1/2 include expression of a number of note-
worthy genes that are known to mediate several
fundamental processes in neuronal migration,
including repulsion/attraction, stop signal for
migration, motility, and cytoskeleton reorgani-
zation (Ghashghaei et al., 2007; Tran et al., 2007).
Whilst expression of Sema3A, Reelin, and Arx
is upregulated by Dlx1/2, that of Pak3, MAP2,
Tau, Robo1, GAP43, and Npn2 is downregulated
(Cobos et al., 2007; Le et al., 2007).
Another transcription factor, neuregulin 2
(Ngn2), downregulates the expression of the small
GTPase RhoA and upregulates the expression of
Dcx, p35, and small GTPase Rnd2 (Ge et al., 2006;
Heng et al., 2008). Dcx ( doublecortin) is a neuro-
nal-specific microtubule-associated protein that,
Studies describing intracellular signaling
mediating calcium responses in migrating neu-
rons are by and large limited to the effect of
calcium per se and calcium-dependent kinases
(Figure 3C). Calcium was shown to activate RhoA
(Guan et al., 2007) and the Rac1-activating pro-
tein IQGAP1 (Kholmanskikh et al., 2006), most
likely also through some intermediate partners.
Interestingly, while RhoA inhibits neuronal
migration, Rac1 stimulates it, clearly showing
that, depending on the upstream stimulus,
calcium-signaling can have opposite effects. The
first calcium-signaling intermediate protein is
calmodulin (CaM) that is activated directly by
calcium binding to it. CaM in turn activates
CaMKII (Ca/calmodulin dependent kinase II)
and CaMKK2 (Ca/calmodulin dependent kinase
kinase 2). As CaMKII can bind F-actin filaments,
it constitutes the link between calcium-signaling
and cytoskeleton remodeling (Fink et al., 2003).
CaMKK2 activates another Ca/calmodulin
dependent kinase – CaMKIV that upon activa-
tion translocates into the nucleus and upregulates
BDNF expression (Kokubo et al., 2009). Indeed,
we could recently demonstrate the involvement
of CaMKIV in the migration of neuroblasts
in the rostral migratory stream (Khodosevich
et al., 2009). These are the only intracellular
calcium-signaling components for which there
is experimental evidence that they are involved
in neuronal migration. Missing players in the
calcium-signaling network can be proposed,
but such an educated guess would rely on data
from other cell-types or model organisms. Of
the signaling hubs only Rac1 has been linked to
calcium-signaling via IQGAP1 (Kholmanskikh
et al., 2006).
The neurotrophin protein family comprises
four members, NT-3, NT-4, BDNF, and nerve
growth factor (NGF), which regulate neuronal
migration via tropomyosin-receptor-kinases
(Trk) A, B, and C (Figure 3C). All neurotrophins
activate hubs of intracellular signaling, such as
PI3K, Ras, Rac1, and Cdk5 (Yamauchi et al.,
2003; Berghuis et al., 2005; Yoshizawa et al., 2005;
Chiaramello et al., 2007), that, as discussed above,
will eventually lead to cytoskeleton remodeling.
Interestingly, already the expression and release
of the neurotrophins themselves is tightly con-
trolled during neuronal migration. As we men-
tioned above, calcium-signaling can activate
BDNF expression via CaMKIV (Kokubo et al.,
2009). The expression of neurotrophin NT-3
on the other hand is activated by the homeobox
protein Barhl1 (Li et al., 2004). On the next stage,
the release of neurotrophin-containing secretory
granules is mediated by Ca-dependent activa-
tor protein for secretion 2 (CAPS2), a secretory
Khodosevich and Monyer Signaling in migrating neuroblasts
Frontiers in Neuroscience www.frontiersin.org March 2011 | Volume 5 | Article 28 | 7
The morphological features of migrating neu-
roblasts differ from those of many other types of
migratory cells. During migrational “stepneurob-
last extends leading process in the direction con-
trolled by external stimuli, then nucleus moves into
the leading process and finally the trailing process is
retracted. The majority of cytoskeleton-associated
components are concentrated at the very tip of the
leading process – the growth cone. Hence neuro-
nal migration differs from the classical one known
in fibroblasts. It is not surprising that the unique
neuroblast morphology is also reflected in idi-
osyncratic intracellular signaling. Thus, although
migrating neuroblasts share a significant number
of hubs and pathways involved in the migration
of other cell-types, they are equipped with neu-
ronal-specific actors in migration that most likely
adjust the common “migratory machinery” to the
specific neuronal needs. Furthermore the weights
between certain components or even whole path-
ways within a network exhibit neuronal-specificity.
It may be that these neuronal-specific pathways in
migration are not only critical for neuroblasts get-
ting to the right place but are already a prerequisite
for neuronal connectivity.
acknowledgmentS
Images were made with the use of vector graphics
from DragonArtz Designs (www.dragonartz.net).
The work was supported in part by the Schilling
Foundation, DFG (SFB488 grant), and BMBF
(RUS 09/B38).
Supplementary materIal
The Supplementary Material for this article can
be found online at http://www.frontiersin.org/
Neuroscience/10.3389/fnins.2011.00028/abstract
Figure S1 | Full scheme of signaling network
controlling neuronal migration. The scheme was
generated by using published data regarding neu-
ronal migration/neurite outgrowth in mammals
only (i.e., excluding other cell-types and model
organisms). See full list of literature in the main
manuscript and supplementary references. Note
that we excluded some molecules that were shown
to be involved in neuronal migration/neurite out-
growth, but their intracellular partners are not
known in migrating neuroblasts. Color code of
molecules: yellow – signaling hubs, red extracel-
lular ligands/matrix components, green – trans-
membrane receptors/channels/transporters, etc.,
blue – intracellular signaling molecules, magenta
– microtubule/actin-associated proteins, orange
cell nucleus components. Arrow – activation,
circle interaction, T-shaped end inhibition.
Some links are indirect due to scarcity of pub-
lished data.
together with Dcx-like kinase (Dcxl), is required
for proper microtubule remodeling in response to
migratory stimuli (Friocourt et al., 2007). Rnd2
and p35 activate RhoA and Cdk5, respectively
(Keshvara et al., 2002; Beffert et al., 2004; Tanaka
et al., 2006; Heng et al., 2008). Since RhoA and
Cdk5 have opposite functions in the regulation of
neuronal migration, precise control of their sign-
aling by Ngn2 is very important (also taking into
account that RhoA expression per se is regulated
by Ngn2). Which other factors may interact with
Ngn2 and specify stimulation of either RhoA or
Cdk5 signaling remains to be determined.
Cooperation between transcription factor
E2F3 and retinoblastoma (Rb) results in the
inhibited expression of several genes, including
Reelin receptor VLDLR (McClellan et al., 2007).
Reelin is one of the major extracellular ligands
controlling several stages of neuronal migra-
tion. Knockout of Reelin or any of its receptors
cause various defects in brain formation due to
impaired neuronal migration during embryonic
development (Assadi et al., 2003; Hack et al.,
2007).
Finally, other mechanisms than conventional
transcription factors that regulate gene expression
in migrating neuroblasts should be mentioned.
Thus, actin per se controls its own expression via
a negative feedback loop. There are two types
of actin in migrating neurons polymerized,
F-actin, and depolymerized or free, G-actin.
When in excess, free G-actin translocates into
the nucleus and downregulates the expression of
actin by inhibiting the expression of the transcrip-
tion factor MRTF (myocardin family transcrip-
tion factor) that together with serum response
factor (SRF) controls actin expression (Stern
et al., 2009).
Posttranscriptional RNA processing is also
subject to regulation in migrating neuroblasts.
For instance, alternative splicing of the key sign-
aling hub Dab1 is regulated by the RNA-binding
protein Nova2 preventing the inclusion of two
additional exons (Yano et al., 2010), which is nec-
essary for correct migration of cortical neurons
during development.
Last but not least, as already mentioned above,
calcium and Notch signaling are involved in regu-
lation of transcription in neuroblasts.
concluSIon
This study and others (see, e.g., Lee and Megeney,
2005; Pocklington et al., 2006) using similar
approaches clearly highlight how complex sign-
aling in different systems (e.g., migrating neurons,
synapse signaling, yeast kinase signaling) can be
reduced to major components eventually leading
to the identification of general network principles.
Khodosevich and Monyer Signaling in migrating neuroblasts
Frontiers in Neuroscience www.frontiersin.org March 2011 | Volume 5 | Article 28 | 8
Guan, C. B., Xu, H. T., Jin, M., Yuan, X. B.,
and Poo, M. M. (2007). Long-range
Ca2+ signaling from growth cone to
soma mediates reversal of neuronal
migration induced by slit-2. Cell 129,
385–395.
Gupta, A ., and Tsai, L. H. (20 03).
Cyclin-dependent kinase 5 and neu-
ronal migration in the neocortex.
Neurosignals 12, 173–179.
Hack , I., Hellwig, S., Jung hans, D.,
Brunne, B., Bock, H. H., Zhao, S., and
Frotscher, M. (2007). Divergent roles
of ApoER2 and Vldlr in the migration
of cortical neurons. Development 134,
3883–3891.
Hashimoto-Torii, K., Torii, M., Sarkisian,
M. R., Bartley, C. M., Shen, J., Radtke,
F., Gridley, T., Sestan, N., and Rakic,
P. (2008). Interaction between reelin
and notch signaling regulates neuro-
nal migration in the cerebral cortex.
Neuron 60, 273–284.
Heng, J. I., Nguyen, L., Castro, D. S.,
Zimmer, C., Wildner, H., Armant, O.,
Skowronska-Krawczyk, D., Bedogni,
F., Matter, J. M., Hevner, R., and
Guillemot, F. (2008). Neurogenin 2
controls cortical neuron migration
through regulation of Rnd2. Nature
455, 114–118.
Hienola, A., Tumova, S., Kulesskiy, E.,
and Rauvala, H. (2006). N-syndecan
deficiency impairs neural migration in
brain. J. Cell Biol. 174, 569–580.
Howell, B. W., Gertler, F. B., and Cooper,
J. A. (1997). Mouse disabled (mDab1):
a Src binding protein implicated in
neuronal development. EMBO J. 16,
121–132.
Jorissen, E., Prox, J., Bernreuther, C.,
Weber, S., Schwanbeck, R., Serneels,
L., Snellinx, A., Craes sae rts, K. ,
Thathiah, A., Tesseur, I., Bartsch, U.,
Weskamp, G., Blobel, C. P., Glatzel, M.,
De Strooper, B., and Saftig, P. (2010).
The disintegrin/metalloproteinase
ADAM10 is essential for the establish-
ment of the brain cortex. J. Neurosci.
30, 4833–4844.
Kempermann, G., Wiskott, L., and Gage,
F. H. (2004). Functional significance
of adult neurogenesis. Curr. Opin.
Neurobiol. 14, 186–191.
Keshvara, L., Magdaleno, S., Benhayon,
D., and Curran, T. (2002). Cyclin-
dependent kinase 5 phosphorylates
disabled 1 independently of reelin
signaling. J. Neurosci. 22, 4869–4877.
Khodo sevi ch, K., and Monyer, H.
(2010). Signaling involved in neu-
rite outgrowth of postnatally born
sub ventricular zone neuron s in
vitro. BMC Neurosci . 11, 18. doi:
10.1186/1471-2202-11-18
Khodosevich, K., Seeburg, P. H., and
Monyer, H. (2009). Major signaling
pathways in migrating neuroblasts.
Front. Mol. Neurosci. 2:7. doi: 10.3389/
neuro.02.007.2009
Kholmanskikh, S. S., Koeller, H. B.,
Wynshaw -Bori s, A., Go mez, T.,
Letourneau, P. C., and Ross, M. E.
(2006). Calcium-dependent interac-
tion of Lis1 with IQGAP1 and Cdc42
promotes neuron al motil ity. Nat.
Neurosci. 9, 50–57.
Kokubo, M., Nishio, M., Ribar, T. J.,
Anderson, K. A., West, A. E., and
Means, A. R. (2009). BDNF-mediated
cerebellar granule cell development is
impaired in mice null for CaMKK2 or
CaMKIV. J. Neurosci. 29, 8901–8913.
Kreuzberg, M., Kanov, E., Timofeev, O.,
Schwaninger, M., Monyer, H., and
Khodosev ich, K. (2010). Increased
subventricular zone-derived cortical
neurogenesis after ischemic lesion.
Exp. Neurol. 226, 90–99.
Kuo, G., Arnaud, L., Kronstad-O’Brien,
P., and Cooper, J. A. (2005). Absence
of Fyn and Src causes a reeler-like
phenotype. J. Neurosci. 25, 8578–8586.
Le, T. N., Du, G., Fonseca, M., Zhou, Q. P.,
Wigle, J. T., and Eisenstat, D. D. (2007).
Dlx homeobox genes promote cortical
interneuron migration from the basal
forebrain by direct repression of the
semaphorin receptor neuropilin-2. J.
Biol. Chem. 282, 19071–19081.
Lee, R. E., and Megeney, L. A. (2005).
The yeast kinome displays scale free
topology with functional hub clus-
ters. BMC Bioinformatics 6, 271. doi:
10.1186/1471-2105-6-271
Li, S., Qiu, F., Xu, A., Price, S. M., and
Xiang, M. (2004). Barhl1 regulates
migration and survival of cerebellar
granule cells by controlling expres-
sion of the neurotrophin-3 gene. J.
Neurosci. 24, 3104–3114.
Lledo, P. M., and Saghatelyan, A. (2005).
Integrating new neurons into the adult
olfactory bulb: joining the network, life-
death decisions, and the effects of sensory
experience. Trends Neurosci. 28, 248–254.
Lledo, P. M., Alonso, M., and Grubb, M. S.
(2006). Adult neurogenesis and func-
tional plasticity in neuronal circuits.
Nat. Rev. Neurosci. 7, 179–193.
Louvi, A., Sisodia, S. S., and Grove, E.
A. (2004). Presenilin 1 in migration
and morphogenesis i n the central
nervous system. Development 131,
3093–3105.
Marin, O., and Rubenstein, J. L. (2003).
Cell migration in the forebrain. Annu.
Rev. Neurosci. 26, 441–483.
McClellan, K. A., Ruzhynsky, V. A., Douda,
D. N., Vanderluit, J. L., Ferguson, K.
L., Chen, D., Bremner, R., Park, D.
S., Leone, G., and Slack, R. S. (2007).
Unique requirement for Rb/E2F3 in
neuronal migration: evidence for cell
cycle-independent functions. Mol. Cell
Biol. 27, 4825–4843.
mammalian forebrain. Nat. Neurosci.
4(Suppl.), 1177–1182.
Dulabon, L., Olson, E. C., Taglienti, M.
G., Eisenhuth, S., McGrath, B., Walsh,
C. A., Kreidberg, J. A., and Anton, E.
S. (2000). Reelin binds alpha3beta1
integrin and inhibits neuronal migra-
tion. Neuron 27, 33–44.
Fink, C. C., Bayer, K. U., Myers, J. W.,
Ferrell, J. E. Jr., Schulman, H., and
Meyer, T. (2003). Selective regula-
tion of neurite extension and synapse
formation by the beta but not the
alpha isoform of CaMKII. Neuron 39,
283–297.
Francis, N. J., and Landis, S. C. (1999).
Cellular and molecular determinants
of sympathetic neuron development.
Annu. Rev. Neurosci. 22, 541–566.
Friocourt, G., Liu, J. S., Antypa, M., Rakic,
S., Walsh, C. A., and Parnavelas, J.
G. (2007). Both doublecortin and
doublecortin-like kinase play a role
in cortical interneuron migration. J.
Neurosci. 27, 3875–3883.
Fukata, M., Nakagawa, M., and Kaibuchi,
K. (2003). Roles of Rho-family
GTPases in cell polarisation and direc-
tional migration. Curr. Opin. Cell Biol.
15, 590–597.
Ge, W., He, F., Kim, K. J., Blanchi, B.,
Coskun, V., Nguyen, L., Wu, X., Zhao,
J., Heng, J. I., Martinowich, K., Tao,
J., Wu, H., Castro, D., Sobeih, M. M.,
Corfas, G., Gleeson, J. G., Greenberg,
M. E., Guillemot, F., and Sun, Y. E.
(2006). Coupling of cell migration
with neurogenesis by proneural bHLH
factors. Proc. Natl. Acad. Sci. U.S.A.
103, 1319–1324.
Ghashghaei, H. T., Lai, C., and Anton, E.
S. (2007). Neuronal migration in the
adult brain: are we there yet? Nat. Rev.
Neurosci. 8, 141–151.
Gio t, L ., B ade r, J. S., Br ouwer, C. ,
Chaudhuri, A., Kuang, B., Li, Y., Hao,
Y. L., Ooi, C. E., Godwin, B., Vitols,
E., Vijayadamodar, G., Pochart, P.,
Machineni, H., Welsh, M., Kong, Y.,
Zerhusen, B., Malcolm, R., Varrone,
Z., Collis, A., Minto, M., Burgess, S.,
McDaniel, L., Stimpson, E., Spriggs,
F., Williams, J., Neurath, K., Ioime,
N., Agee, M., Voss, E., Furtak, K.,
Renzulli, R., Aanensen, N., Carrolla, S.,
Bickelhaupt, E., Lazovatsky, Y., DaSilva,
A., Zhong, J., Stanyon, C. A., Finley,
R. L., Jr., White, K. P., Braverman, M.,
Jarvie, T., Gold, S., Leach, M., Knight,
J., Shimkets, R. A., McKenna, M. P.,
Chant, J., and Rothberg, J. M. (2003). A
protein interaction map of Drosophila
melanogaster. Science 302, 1727–1736.
Glebova, N. O., and Ginty, D. D. (2005).
Growth and survival signals control-
ling sympathetic nervous system
development. Annu. Rev. Neurosci.
28, 191–222.
referenceS
Alonso, M., Viollet, C., Gabellec, M. M.,
Meas-Yedid, V., Olivo-Marin, J. C., and
Lledo, P. M. (2006). Olfactory discrim-
ination learning increases the survival
of adult-born neurons in the olfactory
bulb. J. Neurosci. 26, 10508–10513.
Assadi, A. H., Zhang, G., Beffert, U., McNeil,
R. S., Renfro, A. L., Niu, S., Quattrocchi,
C. C., Antalffy, B. A., Sheldon, M.,
Armstrong, D. D., Wynshaw-Boris, A.,
Herz, J., D’Arcangelo, G., and Clark, G.
D. (2003). Interaction of reelin sign-
aling and Lis1 in brain development.
Nat. Genet. 35, 270–276.
Ayala, R., Shu, T., and Tsai, L. H. (2007).
Trekking across the brain: the journey
of neuronal migration. Cell 128, 29–43.
Beffert, U., Weeber, E. J., Morfini, G., Ko,
J., Brady, S. T., Tsai, L. H., Sweatt, J. D.,
and Herz, J. (2004). Reelin and cyclin-
dependent kinase 5-dependent signals
coope rate in regulating neu ronal
migration and synaptic transmission.
J. Neurosci. 24, 1897–1906.
Berghuis, P., Dobszay, M. B., Wang, X.,
Spano, S., Ledda, F., Sousa, K. M.,
Schulte, G., Ernfors, P., Mackie, K.,
Paratcha, G., Hurd, Y. L., and Harkany,
T. (2005). Endocannabinoids regulate
interneuron migration and morpho-
genesis by transactivating the TrkB
receptor. Proc. Natl. Acad. Sci. U.S.A.
102, 19115–19120.
Bielas, S., Higginbotham, H., Koizumi, H.,
Tanaka, T., and Gleeson, J. G. (2004).
Cortical neuronal migration mutants
sugg est se parate but inter secting
pathways. Annu. Rev. Cell Dev. Biol.
20, 593–618.
Bock, H. H., Jossin, Y., Liu, P., Forster,
E., May, P., Goffinet, A. M., and
Herz, J. (2003). Phosphatidylinositol
3-kinase interacts with the adaptor
protein Dab1 in response to reelin
signaling and is required for normal
cortical lamination. J. Biol. Chem. 278,
38772–38779.
Cheng, K., Li, Z., Fu, W. Y., Wang, J. H.,
Fu, A. K., and Ip, N. Y. (2002). Pctaire1
interacts with p35 and is a novel sub-
strate for Cdk5/p35. J. Biol. Chem. 277,
31988–31993.
Chiaramello, S., Dalmasso, G., Bezin, L.,
Marcel, D., Jourdan, F., Peretto, P.,
Fasolo, A., and De Marchis, S. (2007).
BDNF/TrkB interaction regul ates
migration of SVZ precursor cells via
PI3-K and MAP-K signalling path-
ways. Eur. J. Neurosci. 26, 1780–1790.
Cobos, I., Borello, U., and Rubenstein, J. L.
(2007). Dlx transcription factors pro-
mote migration through repression of
axon and dendrite growth. Neuron 54,
873–888.
Corbin, J. G., Nery, S., and Fishell, G.
(2001). Telencephalic cells take a
tangent: non-radial migration in the
Khodosevich and Monyer Signaling in migrating neuroblasts
Frontiers in Neuroscience www.frontiersin.org March 2011 | Volume 5 | Article 28 | 9
tions of adult neurogenesis. Cell 132,
645–660.
Zhao, C. T., Li, K., Li, J. T., Zheng, W.,
Liang, X. J., Geng, A. Q., Li, N., and
Yuan, X. B. (2009). PKCdelta regulates
cortical radial migration by stabiliz-
ing the Cdk5 activator p35. Proc. Natl.
Acad. Sci. U.S.A. 106, 21353–21358.
Zukerberg, L. R., Patrick, G. N., Nikolic,
M., Humbert, S., Wu, C. L., Lanier, L.
M., Gertler, F. B., Vidal, M., Van Etten,
R. A., and Tsai, L. H. (2000). Cables
links Cdk5 and c-Abl and facilitates
Cdk5 tyrosine phosphorylation,
kinase upregulation, and neurite out-
growth. Neuron 26, 633–646.
Confl ict of Interes t S tatement: T he
authors declare that the research
was conducted in the absence of any
commercial or financial relationships that
could be construed as a potential conflict
of interest.
Received: 17 November 2010; paper pend-
ing published: 19 January 2011; accepted:
23 February 2011; published online: 28
March 2011.
Citat ion: Khodo sev ich K and Monyer
H (2011) Signaling in migrating neu-
rons: from molecules to networks.
Fron t. Neurosci . 5:28. doi: 10.3389/
fnins.2011.00028
Copy right © 20 11 Khodose vich and
Monyer. This is an open-access article
subject to an exclusive license agreement
between the authors and Frontiers Media
SA, which permits unrestricted use, distri-
bution, and reproduction in any medium,
provided the original authors and source
are credited.
ing proteins and the small GTPase
Cdc42 in the slit-robo pathway. Cell
107, 209–221.
Yamashita, N., Uchida, Y., Ohshima, T.,
Hirai, S., Nakamura, F., Taniguchi,
M., Mikoshib a, K., Honn orat, J.,
Kolattukudy, P., Thomasset, N., Takei,
K., Takahashi, T., and Goshima, Y.
(2006). Collapsin response media-
tor protein 1 mediates reelin signal-
ing in cortical neuronal migration.
J. Neurosci. 26, 13357–13362.
Yamauchi, J., Chan, J. R., and Shooter,
E. M. (2003). Neurotrophin 3 acti-
vation of TrkC induce s Sc hwan n
cell migration through the c-Jun
N-terminal kinase pathway.
Proc . Natl. Acad. Sci . U.S.A. 100,
14421–14426.
Yano, M., Hayakawa-Yano, Y., Mele, A.,
and Darnell, R. B. (2010). Nova2 regu-
lates neuronal migration through an
RNA switch in disabled-1 signaling.
Neuron 66, 848–858.
Yoshizawa, M., Kawauchi, T., Sone, M.,
Nishimura, Y. V., Terao, M., Chihama,
K., Nabeshima, Y., and Hoshino, M.
(2005). Involvement of a Rac activa-
tor, P-Rex1, in neurotrophin-derived
signaling and neuronal migration. J.
Neurosci. 25, 4406–4419.
Young-Pearse , T. L., Suth , S., Luth,
E. S., Sawa, A., and Selko e, D. J.
(2010). Biochemical and functional
interaction of disrupted-in-schiz-
ophrenia 1 and amyloid precursor
protein regulates neuronal migration
during mammalian cortical develop-
ment. J. Neurosci. 30, 10431–10440.
Zhao, C., Deng, W., and Gage, F. H. (2008).
Mechanisms and functional implica-
Furuichi, T. (2007). Impaired cerebel-
lar development and function in mice
lacking CAPS2, a protein involved in
neurotrophin release. J. Neurosci. 27,
2472–2482.
Schmid, R. S., Midkiff, B. R., Kedar, V. P.,
and Maness, P. F. (2004). Adhesion
molecule L1 stimulates neuronal
migration through Vav2-Pak1 signal-
ing. Neuroreport 15, 2791–2794.
Stern, S., Debre, E., Stritt, C., Berger,
J., Posern, G., and Knoll, B. (2009).
A nuclear actin function regulates
neuronal motility by serum response
factor-dependent gene transcription.
J. Neurosci. 29, 4512–4518.
Tanaka, H., Katoh, H., and Negishi, M.
(2006). Pragmin, a novel effector of
Rnd2 GTPase, stimulates RhoA activ-
ity. J. Biol. Chem. 281, 10355–10364.
Toyofuku, T., Yoshida, J., Sugimoto, T.,
Zhang, H., Kumanogoh, A., Hori, M.,
and Kikutani, H. (2005). FARP2 triggers
signals for Sema3A-mediated axonal
repulsion. Nat. Neurosci. 8, 1712–1719.
Tran, T. S., Kolodkin, A. L., and Bharadwaj,
R. (2007). Semaphorin regulation of
cellular morphology. Annu. Rev. Cell
Dev. Biol. 23, 263–292.
Voss, A. K., Britto, J. M., Dixon, M. P.,
Sheikh, B. N., Collin, C., Tan, S. S., and
Thomas, T. (2008). C3G regulates cor-
tical neuron migration, preplate split-
ting and radial glial cell attachment.
Development 135, 2139–2149.
Wong, K., Ren, X. R., Huang, Y. Z., Xie,
Y., Liu, G., Saito, H., Tang, H., Wen,
L., Brady-Kalnay, S. M., Mei, L., Wu,
J. Y., Xiong, W. C., and Rao, Y. (2001).
Signal transduction in neuron al
migration: roles of GTPase activat-
Nikolic, M. (2004). The molecular mys-
tery of neuronal migration: FAK and
Cdk5. Trends Cell Biol. 14, 1–5.
Oinuma, I., Ishikawa, Y., Katoh, H., and
Negishi, M. (2004). The semaphorin
4D receptor plexin-B1 is a GTPase
activating protein for R-Ras. Science
305, 862–865.
Parent, J. M., Vexler, Z. S., Gong, C.,
Derugin, N., and Ferriero, D. M.
(2002). Rat forebrain neurogenesis
and striatal neuron replacement after
focal stroke. Ann. Neurol. 52, 802–813.
Peng, Y. J., He, W. Q., Tang, J., Tao, T., Chen,
C., Gao, Y. Q., Zhang, W. C., He, X. Y.,
Dai, Y. Y., Zhu, N. C., Lv, N., Zhang, C.
H., Qiao, Y. N., Zhao, L. P., Gao, X., and
Zhu, M. S. (2010). Trio is a key guanine
nucleotide exchange factor coordinat-
ing regulation of the migration and
morphogenesis of granule cells in the
developing cerebellum. J. Biol. Chem.
285, 24834–24844.
Pockli ngton, A. J., Cumisk ey, M .,
Armstrong, J. D., and Grant, S. G.
(2006). The proteomes of neuro -
transmitter receptor complexes form
modular networks with distributed
functionality underlying plasticity and
behaviour. Mol. Syst. Biol. 2, 2006. 0023.
Rhee, J., Mahfooz, N. S., Arregui, C.,
Lilien, J., Balsamo, J., and VanBerkum,
M. F. (2002). Activation of the repul-
sive receptor roundabout inhibits
N-cadherin-mediated cell adhesion.
Nat. Cell Biol. 4, 798–805.
Sadakata, T., Kakegawa, W., Mizoguchi,
A., Washida, M., Katoh-Semba, R.,
Shutoh, F., Okamoto, T., Nakashima,
H., Kimura, K., Tanaka, M., Sekine, Y.,
Itohara, S., Yuzaki, M., Nagao, S., and
... Notch signaling is a master regulator of brain development (42,43) and critical for such as neurite arborization, maturation, and migration of postmitotic neurons in neuronal development (44)(45)(46). We were moved to investigate whether Notch signaling is altered in Nbs1 knockout neurons. ...
... We show that Nbs1 is a regulatory component of the NICD-RBPJ-mediated transcriptional activity of Notch signaling. Nbs1 deletion upregulates the NICD protein level, as well as Notch activity in neurons and also other cell types tested--which may function to repress neurite outgrowth and neuronal migration, consistent with the instrumental function of Notch in neuronal differentiation, maturation, and migration (44)(45)(46). Nbs1 negatively regulates the Notch pathway in neuronal homeostasis; most likely achieved by its direct interaction with NICD. It is plausible that Nbs1 destabilizes or induces degradation of NICD once Nbs1 interacts with NICD; therefore, Nbs1 depletion results in an increase of NICD, which may also explain a limited amount of co-IP signals. ...
Article
Full-text available
NBS1 is a critical component of the MRN (MRE11/RAD50/NBS1) complex, which regulates ATM-and ATR-mediated DNA damage response (DDR) pathways. Mutations in NBS1 cause the human genomic instability syndrome Nijmegen Breakage Syndrome (NBS), of which neuronal deficits, including microcephaly and intellectual disability, are classical hallmarks. Given its function in the DDR to ensure proper proliferation and prevent death of replicating cells, NBS1 is essential for life. Here we show that, unexpectedly, Nbs1 deletion is dis-pensable for postmitotic neurons, but compromises their arborization and migration due to dysregulated Notch signaling. We find that Nbs1 interacts with NICD-RBPJ, the effector of Notch signaling, and inhibits Notch activity. Genetic ablation or pharmaceutical inhibition of Notch signaling rescues the matu-ration and migration defects of Nbs1-deficient neu-rons in vitro and in vivo. Upregulation of Notch by Nbs1 deletion is independent of the key DDR downstream effector p53 and inactivation of each MRN component produces a different pattern of Notch activity and distinct neuronal defects. These data indicate that neuronal defects and aberrant Notch activity in Nbs1-deficient cells are unlikely to be a direct consequence of loss of MRN-mediated DDR function. This study discloses a novel function of NBS1 in crosstalk with the Notch pathway in neuron development .
... Neuronal migration is a complex process, which involves the coordination of neuronal branching and extension with cellular movement, and is guided by a number of vital signaling molecules and stimuli (Khodosevich and Monyer, 2011;Cooper, 2013;Buchsbaum and Cappello, 2019). Neurons migrate via two distinct mechanisms, radial and tangential, which are predominantly utilized by cortical projection neurons and GABAergic interneurons, respectively. ...
Article
Full-text available
The adult brain is a complex structure with distinct functional sub-regions, which are generated from an initial pool of neural epithelial cells within the embryo. This transition requires a number of highly coordinated processes, including neurogenesis, i.e., the generation of neurons, and neuronal migration. These take place during a critical period of development, during which the brain is particularly susceptible to environmental insults. Neurogenesis defects have been associated with the pathogenesis of neurodevelopmental disorders (NDDs), such as autism spectrum disorder and schizophrenia. However, these disorders have highly complex multifactorial etiologies, and hence the underlying mechanisms leading to aberrant neurogenesis continue to be the focus of a significant research effort and have yet to be established. Evidence from epidemiological studies suggests that exposure to maternal infection in utero is a critical risk factor for NDDs. To establish the biological mechanisms linking maternal immune activation (MIA) and altered neurodevelopment, animal models have been developed that allow experimental manipulation and investigation of different developmental stages of brain development following exposure to MIA. Here, we review the changes to embryonic brain development focusing on neurogenesis, neuronal migration and cortical lamination, following MIA. Across published studies, we found evidence for an acute proliferation defect in the embryonic MIA brain, which, in most cases, is linked to an acceleration in neurogenesis, demonstrated by an increased proportion of neurogenic to proliferative divisions. This is accompanied by disrupted cortical lamination, particularly in the density of deep layer neurons, which may be a consequence of the premature neurogenic shift. Although many aspects of the underlying pathways remain unclear, an altered epigenome and mitochondrial dysfunction are likely mechanisms underpinning disrupted neurogenesis in the MIA model. Further research is necessary to delineate the causative pathways responsible for the variation in neurogenesis phenotype following MIA, which are likely due to differences in timing of MIA induction as well as sex-dependent variation. This will help to better understand the underlying pathogenesis of NDDs, and establish therapeutic targets.
... In mouse cerebellar GCs, the amplitude and frequency of calcium transients, via VGCCs, dictate the rate of neuronal migration, suggesting calcium acts as a speedometer to integrate various intrinsic/extrinsic cues that drives neuronal migration 56 . This can occur through calmodulin, a calcium binding protein, that interacts with a complex network of signaling molecules including calcium/calmodulin dependent kinase II (CaMKII) 57 , Ras/Raf/MEK/ERK pathway 58 , and ultimately gene expression programs. Calcium represents a convergent signal where multiple modalities have the potential to regulate the process of abGC maturation. ...
Article
Full-text available
In the mammalian hippocampus, adult-born granule cells (abGCs) contribute to the function of the dentate gyrus (DG). Disruption of the DG circuitry causes spontaneous recurrent seizures (SRS), which can lead to epilepsy. Although abGCs contribute to local inhibitory feedback circuitry, whether they are involved in epileptogenesis remains elusive. Here, we identify a critical window of activity associated with the aberrant maturation of abGCs characterized by abnormal dendrite morphology, ectopic migration, and SRS. Importantly, in a mouse model of temporal lobe epilepsy, silencing aberrant abGCs during this critical period reduces abnormal dendrite morphology, cell migration, and SRS. Using mono-synaptic tracers, we show silencing aberrant abGCs decreases recurrent CA3 back-projections and restores proper cortical connections to the hippocampus. Furthermore, we show that GABA-mediated amplification of intracellular calcium regulates the early critical period of activity. Our results demonstrate that aberrant neurogenesis rewires hippocampal circuitry aggravating epilepsy in mice.
... These contractions result in the cycles of retraction of lamellipodia and the strengthening of paxillin-based focal adhesions (Tsai and Meyer, 2012). Extracellular Ca 2+ influxes also maintain PI3K activity, which is proposed to be a major signaling hub in migrating cells (Khodosevich and Monyer, 2011) and, at the same time, promote F-actin polymerization and trigger the recruitment of Ca 2+ -sensitive enzyme protein kinase Cα (PKCα) to the plasma membrane of the leading edge of migrating cells (Evans and Falke, 2007). Ca 2+ transients also enable the translocation of Rac1 to the plasma membrane, leading to the formation of lamellipodia (Price et al., 2003). ...
Article
Full-text available
Neuronal migration is a fundamental brain development process that allows cells to move from their birthplaces to their sites of integration. Although neuronal migration largely ceases during embryonic and early postnatal development, neuroblasts continue to be produced and to migrate to a few regions of the adult brain such as the dentate gyrus and the subventricular zone (SVZ). In the SVZ, a large number of neuroblasts migrate into the olfactory bulb (OB) along the rostral migratory stream (RMS). Neuroblasts migrate in chains in a tightly organized micro-environment composed of astrocytes that ensheath the chains of neuroblasts and regulate their migration; the blood vessels that are used by neuroblasts as a physical scaffold and a source of molecular factors; and axons that modulate neuronal migration. In addition to diverse sets of extrinsic micro-environmental cues, long-distance neuronal migration involves a number of intrinsic mechanisms, including membrane and cytoskeleton remodeling, Ca2+ signaling, mitochondria dynamics, energy consumption, and autophagy. All these mechanisms are required to cope with the different micro-environment signals and maintain cellular homeostasis in order to sustain the proper dynamics of migrating neuroblasts and their faithful arrival in the target regions. Neuroblasts in the postnatal brain not only migrate into the OB but may also deviate from their normal path to migrate to a site of injury induced by a stroke or by certain neurodegenerative disorders. In this review, we will focus on the intrinsic mechanisms that regulate long-distance neuroblast migration in the adult brain and on how these pathways may be modulated to control the recruitment of neuroblasts to damaged/diseased brain areas.
... To this end, we colabeled transplanted H9 hNSCs with Ki-67, a marker that was expressed only in cells that reentered the cell cycle. Specificity of Ki-67 labeling was confirmed by strong labeling of cycling neural precursor cells in the subventricular zone (SVZ) (Figure 3, A and B), a site of postnatal neurogenesis in mice (20,21). By 1 month posttransplantation, only very few Ki-67 + cells were found in the transplants ( Figure 3A). ...
Article
Cell therapy raises high hopes for better treatment of brain disorders. However, the majority of transplanted cells often die soon after transplantation and those that survive initially continue to die in the subacute phase, diminishing the impact of transplantations. In this study, we genetically modified transplanted human neural stem cells (hNSCs), from two distant embryonic SCs lines (H9 and RC17) to express one of four prosurvival factors - Hif1a, Akt1, Bcl-2, or Bcl-xl - and studied how these modifications improve short- and long-term survival of transplanted hNSCs. All genetic modifications dramatically increased survival of the transplanted hNSCs. Importantly, three out of four modifications also enhanced the exit of hNSCs from the cell cycle, thus avoiding aberrant growth of the transplants. Bcl-xl expression provided the strongest protection of transplanted cells, reducing both immediate and delayed cell death, and stimulated hNSC differentiation towards neuronal and oligodendroglial lineages. By designing hNSCs with drug-controlled expression of Bcl-xl, we demonstrated that short-term expression of a prosurvival factor can ensure the long-term survival of transplanted cells. Importantly, transplantation of Bcl-xl expressing hNSCs into mice suffering from stroke improved behavioral outcome and recovery of motor activity in mice.
... Importantly, the neuronal migration defects were reversed by stimulating the brain with Ca 2+ and cGMP or inhibiting cAMP signaling both in vitro and in vivo (Kumada et al., 2006). Rescuing neuronal migration through changing Ca 2+ and cyclic nucleotide signaling is mediated by activating the downstream targets that are essential for neuronal migration, such as protein kinase C (PKC), Ca 2+ /calmodulin-dependent protein kinase II (CaMKII), protein phosphatase 1 (PP1), Rho GTPase, mitogen-activated protein kinase (MAPK) and phosphoinositide 3-kinase (PI 3 K) (Huang et al., 2004;Shmueli et al., 2006;Zhao et al., 2009;Khodosevich and Monyer, 2011). ...
Article
Full-text available
Migration of neurons starts in the prenatal period and continues into infancy. This developmental process is crucial for forming a proper neuronal network, and the disturbance of this process results in dysfunction of the brain such as epilepsy. Prenatal exposure to environmental stress, including alcohol, drugs, and inflammation, disrupts neuronal migration and causes neuronal migration disorders (NMDs). In this review, we summarize recent findings on this topic and specifically focusing on two different modes of migration, radial, and tangential migration during cortical development. The shared mechanisms underlying the NMDs are discussed by comparing the molecular changes in impaired neuronal migration under exposure to different types of prenatal environmental stress.
... TOR signaling has also been reported as a key molecular pathway in nervous system development [41]. The calcium signaling pathway is a neuronalspecific signaling pathway [42], and several DE genes and lncGenes annotated in the MAPK/apoptosis pathways are also annotated in GO functional analysis of the calcium signaling pathway. Many studies have observed the effects of calcium on neurons; Duchen reported that neuronal cell death was mediated by calcium signaling [43]. ...
Article
Full-text available
Background: Differential gene expression analysis using RNA-seq data is a popular approach for discovering specific regulation mechanisms under certain environmental settings. Both gene ontology (GO) and KEGG pathway enrichment analysis are major processes for investigating gene groups that participate in common biological responses or possess related functions. However, traditional approaches based on differentially expressed genes only detect a few significant GO terms and pathways, which are frequently insufficient to explain all-inclusive gene regulation mechanisms. Methods: Transcriptomes of survivin (birc5) gene knock-down experimental and wild-type control zebrafish embryos were sequenced and assembled, and a differential expression (DE) gene list was obtained for traditional functional enrichment analysis. In addition to including DE genes with significant fold-change levels, we considered additional associated genes near or overlapped with differentially expressed long noncoding RNAs (DE lncRNAs), which may directly or indirectly activate or inhibit target genes and play important roles in regulation networks. Both the original DE gene list and the additional DE lncRNA-associated genes were combined to perform a comprehensive overrepresentation analysis. Results: In this study, a total of 638 DE genes and 616 DE lncRNA-associated genes (lncGenes) were leveraged simultaneously in searching for significant GO terms and KEGG pathways. Compared to the traditional approach of only using a differential expression gene list, the proposed method of employing DE lncRNA-associated genes identified several additional important GO terms and KEGG pathways. In GO enrichment analysis, 60% more GO terms were obtained, and several neuron development functional terms were retrieved as complete annotations. We also observed that additional important pathways such as the FoxO and MAPK signaling pathways were retrieved, which were shown in previous reports to play important roles in apoptosis and neuron development functions regulated by the survivin gene. Conclusions: We demonstrated that incorporating genes near or overlapped with DE lncRNAs into the DE gene list outperformed the traditional enrichment analysis method for effective biological functional interpretations. These hidden interactions between lncRNAs and target genes could facilitate more comprehensive analyses.
Article
A major challenge in regulatory developmental neurotoxicity (DNT) assessment is lack of toxicological information for many compounds. Therefore, the Test Guidelines programme of the Organisation for Economic Cooperation and Development (OECD) took the initiative to coordinate an international collaboration between diverse stakeholders to consider integration of alternative approaches towards improving the current chemical DNT testing. During the past few years, a series of workshops was organized during which a consensus was reached that incorporation of a DNT testing battery that relies on in vitro assays anchored to key neurodevelopmental processes should be developed. These key developmental processes include neural progenitor cell proliferation, neuronal and oligodendrocyte differentiation, neural cell migration, neurite outgrowth, synaptogenesis and neuronal network formation, as well key events identified in the existing Adverse Outcome Pathways (AOPs). AOPs deliver mechanistic information on the causal links between molecular initiating event, intermediate key events and an adverse outcome of regulatory concern, providing the biological context to facilitate development of Integrated Approaches to Testing and Assessment (IATA) for various regulatory purposes. Developing IATA case studies, using mechanistic information derived from AOPs, is expected to increase scientific confidence for the use of in vitro methods within an IATA, thereby facilitating regulatory uptake. This manuscript summarizes the current state of international efforts to enhance DNT testing by using an in vitro battery of assays focusing on the role of AOPs in informing the development of IATA for different regulatory purposes, aiming to deliver an OECD guidance document on use of in vitro DNT battery of assays that include in vitro data interpretation.
Article
Full-text available
Objective Maternal unbalanced nutritional habits during embryonic development and perinatal stages perturb hypothalamic neuronal programming of the offspring, thus increasing obesity-associated diabetes risk. However, the underlying molecular mechanisms remain largely unknown. In this study we sought to determine the translatomic signatures associated with POMC neuron malprogramming in maternal obesogenic conditions. Methods We used the Ribotag mouse model to specifically profile the translatome of POMC neurons during neonatal (P0) and perinatal (P21) life and its neuroanatomical, functional and physiological consequences. Results Maternal HFD exposure did not interfere with offspring’s hypothalamic POMC neuron specification, but significantly impaired their spatial distribution and axonal extension to target areas. Importantly, we established POMC neuron-specific translatome signatures accounting for aberrant neuronal development and axonal growth. These anatomical and molecular alterations caused metabolic dysfunction in early-life and adulthood. Conclusions Our study provides fundamental insights on the molecular mechanisms underlying POMC neuron malprogramming in obesogenic contexts.
Article
Full-text available
El desarrollo neurológico humano requiere una serie de pasos que permitan orientar, regular y diferenciar los diversos componentes cerebrales, para así garantizar, de una manera bastante precisa, la correcta organización y funcionamiento de las estructuras neuronales. La neurogénesis está clásicamente dividida en cuatro etapas consecutivas: proliferación, migración, diferenciación y maduración. En los humanos,estas ocurren desde la tercera semana de gestación hasta la vida adulta y precisan de un complejo grupo de paquetes genéticos, así como de algunos factores asociados, que se han ido descubriendo gracias a los avances en la biología molecular. El artículo es una revisión acerca del desarrollo neuroembriológico humano y los componentes genéticos más relevantes encontrados en la literatura.
Article
Full-text available
Although clinically distinct, schizophrenia and Alzheimer's disease are common and devastating disorders that profoundly impair cognitive function. For Alzheimer's disease, key mechanistic insights have emerged from genetic studies that identified causative mutations in amyloid precursor protein (APP) and presenilin. Several genes have been associated with schizophrenia and other major psychoses, and understanding their normal functions will help elucidate the underlying causes of these disorders. One such gene is disrupted-in-schizophrenia 1 (DISC1). DISC1 and APP have been implicated separately in cortical development, with each having roles in both neuronal migration and neurite outgrowth. Here, we report a previously unrecognized biochemical and functional interaction between DISC1 and APP. Using in utero electroporation in the living rat brain, we show that DISC1 acts downstream of APP and Disabled-1 to regulate cortical precursor cell migration. Specifically, overexpression of DISC1 rescues the migration defect caused by a loss of APP expression. Moreover, knockdown of APP in cultured embryonic neurons results in altered subcellular localization of DISC1. Using transfected cells and normal brain tissue, we show that APP and DISC1 coimmunoprecipitate and that the intracellular domain of APP interacts with the N-terminal domain of DISC1. Based on these findings, we hypothesize that the APP cytoplasmic region transiently interacts with DISC1 to help regulate the translocation of DISC1 to the centrosome, where it plays a key role in controlling neuronal migration during cortical development.
Article
Full-text available
Orchestrated regulation of neuronal migration and morphogenesis is critical for neuronal development and establishment of functional circuits, but its regulatory mechanism is incompletely defined. We established and analyzed mice with neural-specific knock-out of Trio, a guanine nucleotide exchange factor with multiple guanine nucleotide exchange factor domains. Knock-out mice showed defective cerebella and severe signs of ataxia. Mutant cerebella had no granule cells in the internal granule cell layer due to aberrant granule cell migration as well as abnormal neurite growth. Trio-deficient granule cells showed reduced extension of neurites and highly branched and misguided processes with perturbed stabilization of actin and microtubules. Trio deletion caused down-regulation of the activation of Rac1, RhoA, and Cdc42, and mutant granule cells appeared to be unresponsive to neurite growth-promoting molecules such as Netrin-1 and Semaphorin 6A. These results suggest that Trio may be a key signal module for the orchestrated regulation of neuronal migration and morphogenesis during cerebellar development. Trio may serve as a signal integrator decoding extrinsic signals to Rho GTPases for cytoskeleton organization.
Article
Full-text available
The metalloproteinase and major amyloid precursor protein (APP) alpha-secretase candidate ADAM10 is responsible for the shedding of proteins important for brain development, such as cadherins, ephrins, and Notch receptors. Adam10(-/-) mice die at embryonic day 9.5, due to major defects in development of somites and vasculogenesis. To investigate the function of ADAM10 in brain, we generated Adam10 conditional knock-out (cKO) mice using a Nestin-Cre promotor, limiting ADAM10 inactivation to neural progenitor cells (NPCs) and NPC-derived neurons and glial cells. The cKO mice die perinatally with a disrupted neocortex and a severely reduced ganglionic eminence, due to precocious neuronal differentiation resulting in an early depletion of progenitor cells. Premature neuronal differentiation is associated with aberrant neuronal migration and a disorganized laminar architecture in the neocortex. Neurospheres derived from Adam10 cKO mice have a disrupted sphere organization and segregated more neurons at the expense of astrocytes. We found that Notch-1 processing was affected, leading to downregulation of several Notch-regulated genes in Adam10 cKO brains, in accordance with the central role of ADAM10 in this signaling pathway and explaining the neurogenic phenotype. Finally, we found that alpha-secretase-mediated processing of APP was largely reduced in these neurons, demonstrating that ADAM10 represents the most important APP alpha-secretase in brain. Our study reveals that ADAM10 plays a central role in the developing brain by controlling mainly Notch-dependent pathways but likely also by reducing surface shedding of other neuronal membrane proteins including APP.
Article
Full-text available
Neurite outgrowth is a key process during neuronal migration and differentiation. Complex intracellular signaling is involved in the initiation of neurite protrusion and subsequent elongation. Although, in general many constituents of the machinery involved in this multi-stage process are common for neurons in distinct brain areas, there are notable differences between specific neuronal subtypes. We analyzed key intracellular components of neurite outgrowth signaling in postnatally born subventricular zone (SVZ) neurons in vitro. We showed that inhibitors of PI3K, Akt1, PKCzeta and small GTPases significantly reduced neurite outgrowth. Transfection of SVZ-derived neurons with inactive forms of Rac1 or Cdc42 also decreased neurite length whereas transfection with constitutively active forms of Rac1, Cdc42 or Akt1 as well as with full-length PI3K or PKCzeta increased neurite length. PI3K, Akt1 and PKCzeta acted upstream of the small GTPases Rac1 and Cdc42, which in turn modulate lamellipodia formation of SVZ-derived neurons. We showed the involvement of PI3K/Akt1/PKCzeta/Rac1/Cdc42 pathway in neurite outgrowth of postnatally born SVZ neurons.
Article
Full-text available
Cyclin-dependent kinase 5 (Cdk5) and its activator p35 are critical for radial migration and lamination of cortical neurons. However, how this kinase is regulated by extracellular and intracellular signals during cortical morphogenesis remains unclear. Here, we show that PKCdelta, a member of novel PKC expressing in cortical neurons, could stabilize p35 by direct phosphorylation. PKCdelta attenuated the degradation of p35 but not its mutant derivative, which could not be phosphorylated by PKCdelta. Down-regulation of PKCdelta by in utero electroporation of specific small interference RNA (siRNA) severely impaired the radial migration of cortical neurons. This migration defect was similar to that caused by down-regulation of p35 and could be prevented by cotransfection with the wild-type but not the mutant p35. Furthermore, PKCdelta could be activated by the promigratory factor brain-derived neurotrophic factor (BDNF) and was required for the activation of Cdk5 by BDNF. Both PKCdelta and p35 were required for the promigratory effect of BDNF on cultured newborn neurons. Thus, PKCdelta may promote cortical radial migration through maintaining the proper level of p35 in newborn neurons.
Article
Neurite extension and branching are important neuronal plasticity mechanisms that can lead to the addition of synaptic contacts in developing neurons and changes in the number of synapses in mature neurons. Here we show that Ca2+/calmodulin-dependent protein kinase II (CaMKII) regulates movement, extension, and branching of filopodia and fine dendrites as well as the number of synapses in hippocampal neurons. Only CaMKIIβ, which peaks in expression early in development, but not CaMKIIα, has this morphogenic activity. A small insert in CaMKIIβ, which is absent in CaMKIIα, confers regulated F-actin localization to the enzyme and enables selective upregulation of dendritic motility. These results show that the two main neuronal CaMKII isoforms have markedly different roles in neuronal plasticity, with CaMKIIα regulating synaptic strength and CaMKIIβ controlling the dendritic morphology and number of synapses.
Article
Mice that are mutant for Reelin or Dab1, or doubly mutant for the VLDL receptor (VLDLR) and ApoE receptor 2 (ApoER2), show disorders of cerebral cortical lamination. How Reelin and its receptors regulate laminar organization of cerebral cortex is unknown. We show that Reelin inhibits migration of cortical neurons and enables detachment of neurons from radial glia. Recombinant and native Reelin associate with α3β1 integrin, which regulates neuron–glia interactions and is required to achieve proper laminar organization. The effect of Reelin on cortical neuronal migration in vitro and in vivo depends on interactions between Reelin and α3β1 integrin. Absence of α3β1 leads to a reduction of Dab1, a signaling protein acting downstream of Reelin. Thus, Reelin may arrest neuronal migration and promote normal cortical lamination by binding α3β1 integrin and modulating integrin-mediated cellular adhesion.
Article
The Slit protein guides neuronal and leukocyte migration through the transmembrane receptor Roundabout (Robo). We report here that the intracellular domain of Robo interacts with a novel family of Rho GTPase activating proteins (GAPs). Two of the Slit-Robo GAPs (srGAPs) are expressed in regions responsive to Slit. Slit increased srGAP1-Robo1 interaction and inactivated Cdc42. A dominant negative srGAP1 blocked Slit inactivation of Cdc42 and Slit repulsion of migratory cells from the anterior subventricular zone (SVZa) of the forebrain. A constitutively active Cdc42 blocked the repulsive effect of Slit. These results have demonstrated important roles for GAPs and Cdc42 in neuronal migration. We propose a signal transduction pathway from the extracellular guidance cue to intracellular actin polymerization.
Article
In adult rodents stroke enhances neurogenesis resulting in the addition of neurons to forebrain regions such as striatum or cortex where postnatal neurogenesis under normal conditions plays a negligible role. In the cortex, new neurons are generated either from local cortical precursors that are activated by stroke or from precursors residing in the subventricular zone (SVZ) of lateral ventricles that under normal conditions supply neuroblasts by and large only for the olfactory bulb. In this study we used 5HT3A-EGFP transgenic mice in which all neuroblasts originating in the SVZ are EGFP-labeled. We induced stroke in these mice and by combination of EGFP detection with BrdU injections we labeled all post-stroke-generated SVZ-derived neuroblasts. We showed an increase in SVZ-derived neuroblasts 14 and 35 days after stroke in the ipsilateral hemisphere. Post-stroke-generated SVZ-derived neuroblasts migrated to the cortex and survived for at least 35 days representing 2% of BrdU-positive cells in peri-infarct area where they differentiate into mature neurons. Thus, stroke enhances SVZ neurogenesis and attracts newborn neurons to the injury zone.
Article
Neuronal migration leads to a highly organized laminar structure in the mammalian brain, and its misregulation causes lissencephaly and behavioral and cognitive defects. Reelin signaling, which is mediated in part by a key adaptor, disabled-1 (Dab1), plays a critical but incompletely understood role in this process. We found that the neuron-specific RNA-binding protein Nova2 regulates neuronal migration in late-generated cortical and Purkinje neurons. An unbiased HITS-CLIP and exon junction array search for Nova-dependent reelin-pathway RNAs at E14.5 revealed only one candidate-an alternatively spliced isoform of Dab1 (Dab1.7bc). In utero electroporation demonstrated that Dab1.7bc was sufficient to induce neuronal migration defects in wild-type mice and exacerbate defects when Dab1 levels were reduced, whereas Dab1 overexpression mitigates defects in Nova2 null mice. Thus, Nova2 regulates an RNA switch controlling the ability of Dab1 to mediate neuronal responsiveness to reelin signaling and neuronal migration, suggesting new links between splicing regulation, brain disease, and development.