Sie sind auf Seite 1von 19

Neuron

Review

Transcriptional Regulation of Neuronal Polarity


and Morphogenesis in the Mammalian Brain
Luis de la Torre-Ubieta1 and Azad Bonni1,*
1Department of Neurobiology, Harvard Medical School, 77 Avenue Louis Pasteur, Boston, MA 02115, USA

*Correspondence: azad_bonni@hms.harvard.edu
DOI 10.1016/j.neuron.2011.09.018

The highly specialized morphology of a neuron, typically consisting of a long axon and multiple branching
dendrites, lies at the core of the principle of dynamic polarization, whereby information flows from dendrites
toward the soma and to the axon. For more than a century, neuroscientists have been fascinated by how
shape is important for neuronal function and how neurons acquire their characteristic morphology. During
the past decade, substantial progress has been made in our understanding of the molecular underpinnings
of neuronal polarity and morphogenesis. In these studies, transcription factors have emerged as key players
governing multiple aspects of neuronal morphogenesis from neuronal polarization and migration to axon
growth and pathfinding to dendrite growth and branching to synaptogenesis. In this review, we will highlight
the role of transcription factors in shaping neuronal morphology with emphasis on recent literature in
mammalian systems.

Introduction: The Life of a Neuron Is a Transcriptional mentally inherited pathways that operate largely independently
Smorgasbord of cellular environments, orchestrate neuronal responses to
To integrate into neuronal circuits, newly generated neurons extrinsic cues and in turn may be influenced by these cues.
engage in a series of stereotypical developmental events. After Invertebrate model organisms have been invaluable to the study
exit from the cell-cycle, postmitotic neurons first undergo axo- of the cell-intrinsic mechanisms that orchestrate neuronal
dendritic polarization, a process that encompasses the initial morphogenesis. Elegant studies in Drosophila have spear-
specification of axons and dendrites and their coordinate growth headed the discovery of in vivo functions for transcription factors
giving rise to the unique neuronal shape. Concurrently, many in diverse aspects of neuronal morphogenesis. In particular,
neurons undergo extensive migration to reach their final destina- studies of the da sensory neurons in the fly peripheral nervous
tions in the brain. Axons grow to their appropriate targets, system have defined roles for different transcription factors in
dendrites arborize and prune to cover the demands of their distinct aspects of dendrite development, from growth and
receptive field, and synapses form and are refined to ensure branching to tiling (Jan and Jan, 2003, 2010).
proper connectivity. How neurons accomplish all these tasks Several observations also highlight the importance of cell-
has been the subject of intense scrutiny during the past few intrinsic mechanisms in the control of neuronal morphogenesis
decades. A large body of work has established that these funda- and connectivity in mammalian neurons. For example, the in vivo
mental developmental events are regulated by extrinsic cues developmental programs of polarization, migration, axon and
including secreted polypeptide growth factors, adhesion mole- dendrite growth, and synapse formation are recapitulated in
cules, extracellular matrix components, and neuronal activity distinct populations of neurons dissociated in primary culture
(Dijkhuizen and Ghosh, 2005b; Huber et al., 2003; Katz and (Banker and Goslin, 1991; Powell et al., 1997). Of course,
Shatz, 1996; Markus et al., 2002a; McAllister, 2002; Tessier-Lav- extrinsic cues and cell-intrinsic mechanisms do not operate in
igne and Goodman, 1996). Extrinsic cues are thought to regulate isolation. Isolated primary Purkinje neurons polarize and extend
both the overall design of neuronal shape as well as their fine axons, but the proper formation of their dendrites and dendritic
structural elements such as axon branch points and dendritic spines requires signals from granule neurons (Baptista et al.,
spines. Growth factors, guidance proteins, and other extrinsic 1994). Nevertheless, although extrinsic signals influence neu-
cues act via specific cell surface receptor proteins, which in ronal morphogenesis, neurons often seem to carry a memory
turn regulate intracellular signaling proteins that directly influ- or intrinsic potential that is not altered by a new and different
ence cytoskeletal elements. Members of the Rho GTPase family environment. Transplantation studies have suggested that
of proteins and protein kinases have emerged as key signaling neuronal precursors of the cerebral cortex that give rise to
intermediaries that couple the effects of extrinsic cues to the later-born upper layer neurons are restricted in their develop-
control of actin and microtubule dynamics (Dhavan and Tsai, mental potential and do not give rise to earlier-born deep-layer
2001; Dickson, 2002; Govek et al., 2005; Hur and Zhou, 2010; neurons when placed in the subventricular zone (SVZ) of younger
Luo, 2000; O’Donnell et al., 2009; Wayman et al., 2008b). hosts undergoing deep layer neurogenesis (Desai and McCon-
Accumulating evidence also supports the concept that cell- nell, 2000; Frantz and McConnell, 1996). Likewise, transplanta-
intrinsic mechanisms have major roles in neuronal morphogen- tion studies have revealed that dendrite morphology and laminar
esis and connectivity. These mechanisms comprise develop- specificity of granule neurons in the rat olfactory bulb appear to

22 Neuron 72, October 6, 2011 ª2011 Elsevier Inc.


Neuron

Review

be specified at the time of birth in the SVZ (Kelsch et al., 2007). axons and then dendrites in vivo is closely reproduced when
These studies are consistent with the idea that cell intrinsic mech- these cells are grown in primary culture or in organotypic cere-
anisms specify a developmental template for different popula- bellar explants (Kawaji et al., 2004; Powell et al., 1997). These
tions of neurons that is retained in new environments. This studies suggest that the basic set of instructions to shape
intrinsic identity may also influence how neurons respond to granule neurons is intrinsically encoded. In light of the high abun-
extrinsic cues. Application of the same neurotrophic factor to dance of granule neurons in the cerebellum, the existence of
neurons located in distinct cerebral cortical layers elicits differen- methods to obtain a highly homogeneous population of granule
tial effects on dendrite morphology (McAllister et al., 1995, 1997), neurons from the rat or mouse brain (Bilimoria and Bonni, 2008),
suggesting that neurons inherit distinct developmental programs a relatively simple circuit architecture and accessibility for in vivo
that dictate their responses to extrinsic signals. Purified rat studies, the cerebellar cortex has become an excellent system to
embryonic retinal ganglion neurons cultured in a variety of condi- study intrinsic determinants of neuronal morphogenesis.
tions grow axons much faster than ganglion neurons from post- Transcription factors play critical roles in all major stages of
natal animals (Goldberg et al., 2002b). In addition, with matura- the life of a granule neuron in the cerebellar cortex (Figure 1).
tion retinal granule neurons undergo a switch from preferential These will be briefly described here and examined in depth in
axon growth to preferential dendrite growth (Goldberg et al., subsequent dedicated sections. Axon growth in granule neurons
2002b). Collectively, these observations suggest that neurons is controlled by the transcriptional regulators SnoN and Id2, both
harbor developmentally inherited cell-intrinsic mechanisms that of which are subject to degradation by the ubiquitin proteasome
determine in large part neuronal morphogenesis. system (Konishi et al., 2004; Lasorella et al., 2006; Stegmüller
Transcriptional control of gene expression represents a major et al., 2006). Cdh1-anaphase promoting complex (Cdh1-APC),
mode of cell-intrinsic regulation of neuronal development. Tran- an E3 ubiquitin ligase, targets SnoN and Id2 for degradation
scription factors can govern entire developmental programs, and in turn restricts axon growth (Konishi et al., 2004; Lasorella
directing distinct stages of neuronal development as well as et al., 2006; Stegmüller et al., 2006). Interestingly, a recent study
altering the competency and response of cells to extrinsic has revealed that SnoN also regulates in an isoform-specific
cues. Accordingly, often the expression of one or a set of tran- manner granule neuron migration and positioning by controlling
scription factors is sufficient to direct the subtype specification the expression of the microtubule-binding protein doublecortin
of distinct neuronal populations and thus their morphology and (Dcx) (Huynh et al., 2011).
projection patterns (Arlotta et al., 2005; Chen et al., 2005b; Following parallel fiber axon growth, establishment of synaptic
Hand et al., 2005; Lai et al., 2008; Liodis et al., 2007; Molyneaux connections in the molecular layer occurs through complex
et al., 2005, 2007; Polleux et al., 2007). The current challenge is interactions between pre-synaptic sites in parallel fiber axons
to understand the extent of intrinsic regulation by identifying the and dendritic spines in Purkinje neurons. The development of
transcription factors responsible in different aspects of neuronal parallel fiber presynaptic sites has recently been discovered to
morphogenesis, their direct targets, and the interplay with be under the purview of transcription factor regulation as well,
extrinsic cues. with the basic helix-loop-helix (bHLH) family member NeuroD2
Studies of the mammalian cerebellar cortex have highlighted inhibiting the formation of presynaptic sites in newly generated
the importance of transcription factors in distinct aspects of granule neurons (Yang et al., 2009). Analogous to SnoN-and
neuronal morphogenesis and connectivity (Figure 1). The rodent Id2-control of axon growth, NeuroD2 is also regulated by the
cerebellar cortex provides an excellent model system for the ubiquitin-proteasome pathway where the Cdh1-APC-related
study of mechanisms that shape neurons (Altman and Bayer, ligase Cdc20-APC triggers NeuroD2 degradation in mature
1997; Hatten, 1999; Palay and Chan-Palay, 1974; Ramón y Cajal, neurons and thereby promotes presynaptic differentiation
1995). Postmitotic granule neurons are generated after division of (Yang et al., 2009). Thus, different aspects of axon development,
progenitors located in the external granule layer (EGL). A newly growth and presynaptic development are regulated by the APC
generated granule neuron first extends a single process along acting on different transcription factors.
the molecular layer (ML). A second process is then generated Dendrite development in granule neurons consists of a series
at the opposite pole of the neuron, giving it a bipolar morphology. of events beginning with the initiation of growth and branching,
A phase of tangential migration follows as the bipolar processes leading to the formation of an exuberant arbor, followed by
continue to grow before the neuron generates a third leading pruning, and culminating in the formation of postsynaptic struc-
process perpendicular to the plane of the ML that directs somal tures termed dendritic claws at the ends of the remaining few
migration radially toward the internal granule layer (IGL). As the dendrites. Dendritic claws house sites of connectivity with
soma migrates inward in the cerebellar cortex, the two processes mossy fiber terminals and Golgi neuron axons (Altman and
in the ML fuse while the neuron continues to extend a trailing Bayer, 1997; Palay and Chan-Palay, 1974; Ramón y Cajal,
process perpendicular to the plane of the ML. The intersection 1995). As with the axons, dendrite growth and maturation
of these orthogonally oriented processes gives rise to the charac- are also under transcriptional control in granule neurons. Intrigu-
teristic T-shaped parallel fiber axon of granule neurons. Once ingly, transcription factors in these developmental steps are
granule neurons reach the IGL, they begin to extend dendrites, strongly influenced by neuronal activity and calcium signaling.
which following pruning and maturation establish synaptic The bHLH transcription factor NeuroD promotes dendrite
connections (Altman and Bayer, 1997; Ramón y Cajal, 1995). growth in response to activation of L-type voltage sensitive
The stereotyped sequence of events by which granule neu- calcium channels (VSCCs) (Gaudillière et al., 2004). In a later
rons acquire a polarized morphology by sequentially generating phase of development, the sumoylated repressor form of the

Neuron 72, October 6, 2011 ª2011 Elsevier Inc. 23


Neuron

Review

P1

Axon growth Polarization


FOXO
Pak1

p300 EGL
SnoN1/2
N1 2
Ccd1
Id2

E47 ML
NogoR, Sema3F, Unc5A,
Notch1, Jagged2

Developmental Time
Barhl1
NT-3

Migration and Positioning


CREB IGL
BDNF

NFIA,B,X
EphrinB1, N-Cadherin

SnoN1 FOXO1
Dcx
Dendritic claw formation
Dendrite pruning
P8 Dendrite growth Presynaptic
res naptic site formation
ormation
Developmental Time P12
NeuroD
N r Cdc20 -
? APC
NeuroD2
r
Cplx2

Sp4
Rat cerebellum NT-3

SUMO
MEF2A
Nur-77

Figure 1. Transcription Factors Orchestrate Distinct Stages of Neuronal Morphogenesis in the Cerebellar Cortex
The morphogenesis of granule neurons in the cerebellar cortex proceeds in discreet stages governed by distinct transcription factors. After exit from the cell
cycle, the FOXO transcription factors trigger granule neuron polarization by regulating the expression of the kinase Pak1. Axon growth is promoted by at least two
transcriptional regulators, SnoN1 and SnoN2, acting as transcriptional coactivators in association with p300, and the bHLH inhibitor protein Id2. The tran-
scriptional regulator SnoN2 promotes migration from the IGL into the EGL by inhibiting the transcriptional repressor complex SnoN1/FOXO1. The transcription
factors Barhl1, NFIA, B, X, and CREB also promote migration into the IGL. The SnoN1/FOXO1 transcriptional complex directly represses Dcx and thereby
controls the positioning of granule neurons within the IGL. Dendrite morphogenesis includes the stages of growth, pruning and maturation. The transcription
factor NeuroD promotes the initiation of dendrite growth and branching, while Sp4 regulates dendrite pruning, and the sumoylated repressor form of the
transcription factor MEF2A drives the differentiation of postsynaptic dendritic claws. Concurrent with dendrite pruning and maturation, development of
presynaptic structures in parallel fiber axons is regulated by the transcription factor NeuroD2, which is regulated by the ubiquitin ligase Cdc20-APC. Image
depicts a coronal or parlobular section of the rat cerebellar cortex at different stages of development as drawn by Ramón y Cajal (1995). See text for details.

transcription factor myocyte enhancer factor 2A (MEF2A) drives orchestrating programs of gene expression to shape axons
postsynaptic dendritic claw differentiation in a manner that is and dendrites and ultimately synapses with other neurons.
also regulated by VSCC activation (Shalizi et al., 2006). These
studies suggest that activity-dependent calcium signaling regu- FOXO Transcription Factors Regulate Neuronal
lates dendrite growth and maturation at least in part through Polarization and Positioning
changes in gene expression governed by transcription factors. The polarization of neurons leading to the generation of axons
The rather ubiquitous presence of transcription factor regula- and dendrites represents an essential step in the establishment
tion in different aspects of neuronal morphogenesis has been of neuronal circuits in the developing brain. Mature axons and
extended to the earliest step of neuronal polarization. Accord- dendrites are morphologically, biochemically, and functionally
ingly, the FOXO transcription factors (Forkhead domain type O) distinct (Craig and Banker, 1994; Falnikar and Baas, 2009).
have been discovered to trigger neuronal polarization in the Understanding the mechanisms by which neurons acquire and
mammalian brain (de la Torre-Ubieta et al., 2010). Thus, as maintain a polarized morphology is a fundamental question in
soon as neurons are born, transcription factors go to work neurobiology.

24 Neuron 72, October 6, 2011 ª2011 Elsevier Inc.


Neuron

Review

The study of the molecular basis of neuronal polarization is a postnatal rat pups. FOXO knockdown triggers the formation of
relatively recent endeavor. Within this growing field, the majority aberrant processes in the IGL and the loss of associated parallel
of the molecular players regulating neuronal polarity have been fiber axons (de la Torre-Ubieta et al., 2010). Expression of an
characterized in studies of primary hippocampal neurons (Dotti RNAi-resistant form of FOXO6 in the background of FOXO
et al., 1988). After plating, dissociated hippocampal neurons first RNAi reverses the polarity phenotype in primary neurons and
issue several undifferentiated neurites (stage 2). Afterwards, one in postnatal rat pups. These findings suggest that the FOXO tran-
of the neurites is selected by an apparent stochastic process to scription factors, and in particular the brain-enriched protein
become an axon, displaying accelerated growth with concomi- FOXO6, play a critical role in the regulation of neuronal polarity.
tant expression of axon markers (stage 3) (Craig and Banker, How might transcription factors drive neuronal polarization, an
1994). Axon specification, which occurs during the transition event that is specified locally within neuronal processes? A plau-
from stage 2 to stage 3, represents a critical step in neuronal sible model would be that they trigger the expression of polarity-
polarization. An array of proteins including molecular scaffolds, associated proteins and thereby establish the competency of
Rho-GTPases and their regulators, protein kinases, kinesin neurons to undergo polarization. Consistent with this model,
motors, and microtubule-associated proteins (MAPs) converge analysis of an array of genes implicated in neuronal polarity
at the nascent axon to regulate cytoskeletal dynamics and suggests that the FOXO transcription factors regulate the
promote axon specification and growth (Arimura and Kaibuchi, expression of the polarity complex protein mPar6, the Ras-
2007; Barnes and Polleux, 2009). Other studies using the cerebral GTPase R-Ras, the Rac1-GEF STEF, the MAPs adenomatous
cortex as a model system are beginning to implicate extracellular polyposis coli (APC) and collapsing response mediator protein
signals in axon formation. The neurotrophin BDNF and the growth 2 (CRMP-2), the kinesin family member KIF5A, and the protein
factor TGF-b act via the protein kinases SAD-A/B and the Par kinase Pak1 (Figure 2; de la Torre-Ubieta et al., 2010). Within
complex, respectively, to promote axonogenesis (Barnes et al., this set of genes, Pak1 is the most robustly downregulated
2007; Shelly et al., 2007; Yi et al., 2010). Extrinsic cues may also gene in FOXO-knockdown neurons. The FOXO proteins occupy
regulate neuronal polarization by preventing axon differentiation the Pak1 gene and thereby directly activate Pak1 transcription
in favor of dendrite morphogenesis. The guidance cue Sema- in neurons. Knockdown of Pak1 in granule neurons phenocopies
phorin 3A (Sema 3A) repels axons and attracts apical dendrites the polarity phenotype induced by FOXO knockdown, and ex-
in cortical neurons (Polleux et al., 2000). Two recent studies pression of Pak1 partly reverses the polarity phenotype triggered
have expanded upon these findings, suggesting that Sema 3A by FOXO RNAi (de la Torre-Ubieta et al., 2010). These findings
signaling in diverse populations of neurons suppresses axon suggest that the protein kinase Pak1 is a direct and physiologi-
specification and instead promotes dendrite formation (Nish- cally relevant transcriptional target of the FOXO proteins in the
iyama et al., 2011; Shelly et al., 2011). Sema 3A suppresses control of neuronal polarity, though additional targets mediating
axon differentiation by inducing cGMP/PKG signaling and FOXO-dependent neuronal polarity remain to be identified.
concomitantly reducing cAMP levels and inhibiting PKA activity, Pak1 activity is regulated by the Rho-GTPases Cdc42 and
thus leading to decreased activity of the axon-promoting kinases Rac1 (Bokoch, 2003), which interact with the Par polarity
LKB1 and SAD-A/B and increased activity of GSK3b (Shelly et al., complex (Joberty et al., 2000; Lin et al., 2000), suggesting that
2011). However, Sema 3A knockout as well as BDNF knockout Pak1 may be activated locally at the nascent axon downstream
mice do not display overt defects of neuronal polarity, suggesting of the Par complex. Thus, the FOXO transcription factors may
that alternative compensatory mechanisms are at play (Behar control both the expression of Pak1 and its upstream regulators
et al., 1996; Ernfors et al., 1994; Jones et al., 1994; Polleux (Figure 2). The FOXO proteins regulate the expression of the
et al., 1998, 2000). Other studies suggest that the plane of the microtubule-associated protein APC (de la Torre-Ubieta et al.,
last mitotic division and the position of the centrosome provide 2010), which localizes mPar3 to the nascent axon (Shi et al.,
spatial cues that establish the site of axon generation in both 2004), and expression of the kinesin KIF5A, which is important
primary hippocampal and cortical neurons in vivo (de Anda for the transport of CRMP-2 to the axon (Kimura et al., 2005).
et al., 2005, 2010). Although these studies have begun to elucidate Therefore, the FOXO transcription factors may act as critical
the local mechanisms responsible for axon specification and regulators of polarity by triggering the expression of several
polarization, the cell-intrinsic regulatory mechanisms that might components of the local machinery controlling neuronal polarity.
orchestrate neuronal polarization have been largely unexplored. The discovery of FOXO proteins as key determinants of polarity
Recently, the FOXO transcription factors have been identified should pave the way for future studies aimed at identifying addi-
as key regulators of neuronal polarity (Figure 2). The FOXO pro- tional potential transcriptional regulators in neuronal polarity.
teins are expressed in developing neurons in the brain, including The FOXO transcription factors are tightly controlled by post-
in hippocampal and cerebellar granule neurons at a time when translational modifications, raising the question of how their
they undergo neuronal polarization and morphogenesis. Knock- function in neuronal polarity might be regulated. Growth factors
down of FOXO1, FOXO3, and FOXO6 by RNA interference inhibit FOXO-dependent transcription via the PI3K-Akt signaling
(RNAi) in primary granule or hippocampal neurons leads to pathway (Biggs et al., 1999; Brunet et al., 1999; Gan et al., 2005;
profound impairment of neuronal polarity (de la Torre-Ubieta Guo et al., 1999; Kops et al., 1999; Nakae et al., 2000; Zheng
et al., 2010). FOXO knockdown neurons extend several unspec- et al., 2002). Interestingly, IGF-1 signaling and localized PI3K
ified, morphologically similar processes that express both activity at the nascent axon promote axon specification in hippo-
axonal and dendritic markers. This phenotype is recapitulated campal neurons (Jiang et al., 2005; Ménager et al., 2004; Shi
in the cerebellar cortex in vivo upon induction of FOXO RNAi in et al., 2003; Sosa et al., 2006; Yoshimura et al., 2006), raising

Neuron 72, October 6, 2011 ª2011 Elsevier Inc. 25


Neuron

Review

FOXO
Pak1

Par6
APC
CRMP-2
Kif5A

aPKC
Par6
APC
Par3
P r

Regulation of Actin and


Microtubule Dynamics
Kif5A

CRMP-2

Pak1

Figure 2. FOXO Transcription Factors Drive Neuronal Polarization


FOXO transcription factors occupy the Pak1 gene promoter and induce its expression to promote neuronal polarization. The kinase Pak1 regulates actin and
microtubules dynamics by distinct mechanisms at the nascent axon and is required for neuronal polarization. A number of additional FOXO putative tran-
scriptional targets, including mPar6, APC, Kif5A, and CRMP-2 regulate polarization by acting locally at the nascent axon. The Par complex protein mPar3 is
targeted to the nascent axon by APC. The microtubule-associated protein CRMP-2 is transported to the axon by Kif5A.

a potential paradox of how the FOXO transcription factors, which suppressors in hematopoietic stem cells in vivo (Paik et al.,
are inhibited by the PI3K-Akt signaling pathway, promote 2007; Tothova et al., 2007). However, genetic ablation of different
neuronal polarization. It remains unclear, however, whether FOXO family members in mice results in distinct phenotypes
localized Akt signaling in the axon influences the activity of the in vivo (Castrillon et al., 2003; Furuyama et al., 2004; Hosaka
FOXO transcription factors in the nucleus. Notably, growth factor et al., 2004; Kitamura et al., 2002; Lin et al., 2004; Nakae et al.,
inhibition of FOXO proteins can be countered in cellular contexts 2002; Polter et al., 2009; Renault et al., 2009), suggesting specific
whereby the protein kinases MST1, JNK, and AMPK promote the roles for individual family members. The FOXO proteins FOXO1,
nuclear accumulation of FOXO proteins and thereby induce FOXO3, and FOXO6 appear to operate redundantly in driving
FOXO-dependent transcription (Essers et al., 2004; Greer neuronal polarization (de la Torre-Ubieta et al., 2010). However,
et al., 2007; Lehtinen et al., 2006). It will be interesting to in rescue experiments in the background of FOXO RNAi, expres-
determine if these or other signals stimulate FOXO-dependent sion of FOXO1 or FOXO3 only partially restores polarity, whereas
transcription in neuronal polarization. expression of FOXO6 substantially restores polarity. Therefore,
There has been much interest in the specific biological roles FOXO6 may have some nonoverlapping functions in neuronal
of different FOXO family members. The FOXO proteins are ex- polarity. It will be important in the future to characterize the tran-
pressed in overlapping patterns in the brain and other tissues scriptional targets of individual FOXO family members to under-
and appear to bind to similar sites within responsive genes stand the contribution of each FOXO protein to neuronal polarity.
(Furuyama et al., 2000; Hoekman et al., 2006). Accordingly, the Neuronal polarization temporally overlaps with radial migration
FOXO transcription factors have redundant roles as tumor in certain populations of neurons in the mammalian brain. In the

26 Neuron 72, October 6, 2011 ª2011 Elsevier Inc.


Neuron

Review

A Figure 3. Isoform-Specific Functions of SnoN in the


Control of Migration and Axon Growth
Nucleus (A) The two spliced isoforms of the Sno gene, SnoN1 and
EGL
SnoN2, have distinct roles in granule neuron migration. SnoN2
promotes migration from the EGL into the IGL, while SnoN1
SnoN2

regulates positioning within the IGL. SnoN1 associates with


SnoN2
the transcription factor FOXO1 and thereby represses Dcx
Regulation of migration gene expression. SnoN2 inhibits SnoN1 function.
from EGL to IGL and positioning (B) SnoN associates with the transcriptional coactivator p300
SnoN1

and promotes axon growth by inducing the expression of


SnoN1 FOXO1 in the IGL
Ccd1. Extrinsic cues impinge on SnoN function during
Dcx
development. TGF-b induces Smad2 phosphorylation and
thereby stimulates Cdh1-APC-dependent degradation of
SnoN and consequent inhibition of axon growth.
IGL

TGF-β
SnoN1 and SnoN2, which differ by a 46 amino acid
B region present only in SnoN1 (Pearson-White and
Cytoplasm
Crittenden, 1997; Pelzer et al., 1996). SnoN1 has
P
Smad2
an essential function in limiting the extent of migra-
tion of granule neurons within the IGL and thus in
Nucleus
the correct positioning of granule neurons within
the IGL. Specific knockdown of SnoN1 in granule
p300 neurons in vivo results in abnormal accumulation
SnoN1/2
2 of granule neurons within the deep IGL close to
Ccd1 Axon growth
the white matter (Huynh et al., 2011). By contrast,
P SnoN2 promotes the migration of granule neurons
1 - Smad2
Cdh1 from the EGL to the IGL. Accordingly, SnoN2
APC
SnoN1/2
knockdown impairs migration into the IGL, leading
Inhibition of axon growth
Ccd1 to the accumulation of granule neurons in the EGL
(Huynh et al., 2011). Therefore, SnoN1 and SnoN2
have opposing functions in the control of granule
neuron migration (Figure 3).
developing cerebral cortex, cortical neurons undergo a transition The SnoN isoforms control migration in part by regulating the
from a multipolar to bipolar morphology as they leave the inter- expression of the X-linked mental retardation and epilepsy gene
mediate zone (IZ) and move toward the cortical plate, and this encoding doublecortin (Dcx). Dcx promotes microtubule stability
morphological transition is regarded as polarization in cortical and polymerization and is thought to be critical for the dynamic
neurons (Calderon de Anda et al., 2008; Noctor et al., 2004; coupling between the nucleus and the centrosome during nucle-
Tabata and Nakajima, 2003). Interestingly, genes implicated in okinesis (Gleeson et al., 1999; Horesh et al., 1999; Koizumi et al.,
axo-dendritic polarization including the mPar complex protein 2006). SnoN1 forms a transcriptional complex with FOXO1 that
Par6, the kinases LKB1, MARK2, GSK3b, and Pak1, and the occupies the Dcx gene and thereby represses its expression in
Rho GTPases Rac1 and Cdc42 are also required for migration neurons (Figure 3; Huynh et al., 2011). Consistent with these find-
(Asada and Sanada, 2010; Asada et al., 2007; Barnes and ings, knockdown of the SnoN1-FOXO1 complex derepresses
Polleux, 2009; Causeret et al., 2009; Konno et al., 2005; Sapir Dcx expression and hence stimulates excessive migration of
et al., 2008; Solecki et al., 2004). The migration phenotypes granule neurons within the IGL in the cerebellar cortex (Huynh
associated with misregulation or inhibition of these genes often et al., 2011). SnoN2 antagonizes SnoN1 function by associating
coincide with a multipolar or aberrant morphology in the stalled with SnoN1 via a coiled-coil domain interaction and inhibiting
neurons. In view of these observations, in some instances it is the ability of SnoN1 to repress FOXO1-dependent transcription
challenging to determine whether the failure of neurons to (Figure 3; Huynh et al., 2011). Thus, these findings both define
polarize precedes the migration defects, or whether the inverse a transcriptional repressor complex for FOXO proteins and
relationship holds. Notably, postmitotic granule neurons of the illustrate how control of abundance of two splicing isoforms of
cerebellum undergo axo-dendritic polarization before the onset the same transcriptional regulator leads to opposing cellular
of radial migration. In this sense, cerebellar granule neurons responses during development.
provide a simpler system for the study of signaling pathways The study of neuronal polarization is relevant beyond the con-
specific for migration or polarity. text of brain development. Spinal cord injury presents a scenario
Taking advantage of this experimental system, a recent study where neurons have to regrow axons from an axonal stump.
has uncovered that FOXO1 and the transcriptional regulator Axon transection can lead to the respecification of a dendrite
SnoN play key roles in the migration and positioning of granule into an axon (Bradke and Dotti, 2000; Gomis-Rüth et al., 2008;
neurons in the cerebellar cortex (Figure 3; Huynh et al., 2011). Goslin and Banker, 1989; Takahashi et al., 2007). In particular,
Alternative splicing generates two isoforms of the SnoN protein, depending on the proximity of the injury to the soma, neurons

Neuron 72, October 6, 2011 ª2011 Elsevier Inc. 27


Neuron

Review

either extend an axon from the original stump or from a dendrite findings suggest that cell-intrinsic mechanisms orchestrate
(Gomis-Rüth et al., 2008). These studies suggest that neuronal responses to neurotrophins in the control of axon growth.
polarity is plastic, and conversely the polarized state is actively Several lines of evidence support the concept that the
maintained by dedicated mechanisms (Bisbal et al., 2008; capacity of a neuron to extend axons and project to the appro-
Hedstrom et al., 2008; Jiang et al., 2005; Kobayashi et al., priate targets is intrinsically encoded. Neurons of the peripheral
1992; Nakada et al., 2003; Winckler et al., 1999; Yin et al., nervous system (PNS), but not the central nervous system (CNS),
2008). Thus, regulators of neuronal polarity might influence have the capacity to regenerate axons after injury (Aguayo et al.,
axon regeneration by directing axon re-specification and exten- 1991). The axon growth-inhibiting environment of the adult CNS,
sion. In this regard, it will be important to determine if FOXO- chiefly generated by myelin proteins, contributes to this differen-
dependent transcription is required for axon regeneration and tial response (Filbin, 2003; He and Koprivica, 2004; Schwab,
in particular whether activators of FOXO proteins can accelerate 2004). However, the observation that embryonic CNS or adult
axon growth after injury. Along these lines, increased SIRT1 PNS neurons can extend axons on top of adult white matter
activity is associated with protection of dorsal root ganglion suggests that an intrinsic property of neurons in the adult CNS
(DRG) axons from Wallerian degeneration (Araki et al., 2004). contributes to the failure of axon regeneration after injury (Davies
In light of the observation that SIRT1-induced deacetylation et al., 1997, 1999; Schwab and Bartholdi, 1996). Consistently,
of FOXO proteins stimulates FOXO-dependent transcription embryonic RGCs have a higher capacity to extend axons than
(Brunet et al., 2004; Daitoku et al., 2004), the FOXO proteins postnatal RGCs, and this change in the capacity of axon growth
might mediate the protective effect of SIRT1 against axon requires new gene transcription (Moore et al., 2009). Importantly,
degeneration. Because the FOXO proteins are regulated by emerging evidence suggests that the intrinsic axonal growth
distinct signaling pathways in response to cellular stress, capacity is regulated by transcription factors, both during devel-
including the protein kinase JNK which stimulates axon regener- opment and in the context of injury.
ation after injury (Lindwall et al., 2004), the FOXO proteins are Evidence for a cell-intrinsic mechanism regulating axon growth
ideally positioned to promote axon regeneration after injury. has also emerged from studies of granule neurons of the devel-
oping cerebellar cortex. The ubiquitin ligase Cdh1-APC plays
Transcription Factors Regulate Axon Growth a critical role in the control of axon growth and patterning in the
and Guidance: From Development to Regeneration rodent cerebellar cortex (Konishi et al., 2004). Knockdown of
Several classes of neurons, including projection neurons in the Cdh1 in primary granule neurons stimulates axon growth even
cerebral cortex must extend axons over very long distances in the presence of the growth-inhibiting environment of myelin.
in a stereotyped path to innervate specific targets. Beyond Localization of Cdh1 in the nucleus is required for Cdh1-APC-
the fundamental question of how neurons accomplish this inhibition of axon growth (Stegmüller et al., 2006). As briefly
monumental task during development, understanding the mech- described above, subsequent studies have identified the tran-
anisms that promote axon growth may form the basis of treat- scriptional modulator SnoN and the HLH protein Id2 as
ments aimed at recovery in the central nervous system following substrates of neuronal Cdh1-APC in the control of axon growth
injury or disease. (Lasorella et al., 2006; Stegmüller et al., 2006, 2008). Expression
The role of extrinsic cues, including neurotrophic factors, in of SnoN alone can overcome myelin-dependent growth inhibi-
promoting axon elongation is compelling. Exposure of distinct tion, suggesting that SnoN drives a genetic program that pro-
populations of primary neurons, including retinal ganglion cells, motes axon growth under different extrinsic stimuli (Stegmüller
DRG neurons, and hippocampal neurons to NGF, BDNF, or NT-3 et al., 2006). Interestingly, in contrast to the opposing functions
promotes axon growth robustly (Goldberg et al., 2002a; Lentz of SnoN1 and SnoN2 in the control of granule neuron migration
et al., 1999; Markus et al., 2002b; Shinoda et al., 2007). Impor- and positioning, the two isoforms of SnoN collaborate to promote
tantly, a requirement for neurotrophin signaling in normal axon axon growth (Huynh et al., 2011; Stegmüller et al., 2006). Although
development has been validated in vivo (Glebova and Ginty, SnoN is widely considered to have transcriptional repressive
2004; Kuruvilla et al., 2004; Patel et al., 2000, 2003; Tucker functions (Luo, 2004), including in the control of neuronal posi-
et al., 2001). Neurotrophins act through the distinct Trk receptors tioning (Huynh et al., 2011), SnoN functions as a transcriptional
activating signaling cascades relayed by the PI3K-Akt and coactivator in the control of axon growth (Figure 3; Ikeuchi
Ras-MAPK signaling pathways, which in turn directly regulate et al., 2009). In particular, SnoN associates with the histone
cytoskeletal elements modulating actin and microtubule polymer- acetyltrasferase p300 and thereby induces the expression of
ization at the growth cone (Huber et al., 2003; Zhou and Snider, a large set of genes in neurons (Ikeuchi et al., 2009). These find-
2006). However, neurotrophins also induce changes in transcrip- ings support the concept that SnoN acts in a dual transcriptional
tion that are thought to play critical roles in axon growth (Segal and activating or repressive manner in a cell-or target-specific
Greenberg, 1996). Accordingly, neurotrophin signaling regulates manner (Pot and Bonni, 2008; Pot et al., 2010). In promoting
the transcription factors CREB and NFAT to stimulate axon axon growth, the cytoskeletal scaffold protein Ccd1 represents
growth (Graef et al., 2003; Lonze et al., 2002). Conversely, tran- a critical downstream target of SnoN (Ikeuchi et al., 2009). Ccd1
scription factors regulate the expression of neurotrophin recep- localizes to the actin cytoskeleton at growth cones and activates
tors to specify neuronal subtypes and promote axon growth. the protein kinase c-Jun kinase (JNK) (Ikeuchi et al., 2009), which
For example, the transcription factor Runx1 induces the timely has been implicated in axon growth (Oliva et al., 2006).
expression of TrkA to promote the specification of nociceptive Whereas SnoN drives axon growth by triggering the expres-
neurons and growth of their axons (Marmigère et al., 2006). These sion of regulators of the actin cytoskeleton, Id2 is thought to

28 Neuron 72, October 6, 2011 ª2011 Elsevier Inc.


Neuron

Review

promote axon growth by antagonizing the function of the bHLH expression pattern of a number of KLF proteins from embryonic
transcription factor E47, which induces the expression of a to postnatal stages correlates with their ability to suppress or
number of genes involved in axon repulsion including NogoR, promote axon growth. Overexpression of KLF4, which is upregu-
Sema3F, and Unc5A (Lasorella et al., 2006). Thus, Id2 stimulates lated postnatally in retinal ganglion neurons, suppresses axon
axon growth by modulating the response of neurons to guidance growth. Conversely, retinal ganglion neurons from KLF4 knock-
cues. Interestingly, TGFb signaling through the protein Smad2 out mice exhibit increased axon growth in culture and regenerate
regulates the abundance of SnoN protein and consequently after optic nerve crush injury (Moore et al., 2009). These findings
axon growth (Stegmüller et al., 2008), thus highlighting how support the idea that the regulated expression of transcription
intrinsic determinants integrate signals from extrinsic cues for factors during development controls the intrinsic potential for
proper development. axon growth in neurons.
Although transcriptional regulators such as NFAT, SnoN, and Studies of axon regeneration in dorsal root ganglion (DRG)
Id2 appear to regulate axon growth in postmitotic neurons, tran- neurons have also supported an important role for transcription
scription factors that primarily regulate neurogenesis may also factors in the control of axon growth. Transection of the periph-
coordinate axon growth in differentiated neurons. In studies of eral branch, but not the central branch, in DRG neurons triggers
retinotectal projection neurons and spinal cord motor neurons, axon growth and promotes axon regeneration of central
several transcription factors including Vax2, Zic2, Lim1, and branches following a later spinal cord injury (Neumann and
Lmx1b have been reported to regulate the timely and cell- Woolf, 1999; Richardson and Issa, 1984; Smith and Skene,
specific expression of proteins involved in axon guidance, 1997). Comparing the transcriptional profiles of DRG neurons
including Ephrins A and B and their receptors (Barbieri et al., with transected central versus peripheral branches reveals that
2002; Dufour et al., 2003; Herrera et al., 2003; Kania and Jessell, approximately 10% of the genes with altered expression 12 hr
2003; Kania et al., 2000; Mui et al., 2002; Schulte et al., 1999; after the procedure are transcription factors (Zou et al., 2009).
Williams et al., 2003). Neurons residing in distinct cerebral The transcriptional regulator Smad1 represents one of the genes
cortical layers display specific projection patterns, suggesting upregulated in DRGs with transected peripheral branches rela-
that subtype specification is linked to aspects of axon morpho- tive to central branches. Smad1 promotes axon growth in DRG
genesis. Functional characterization of transcription factors neurons following injury, an effect that is potentiated by BMP
exhibiting layer-specific expression in the cerebral cortex of signaling. Similar studies have identified a role for the transcrip-
knockout mice has uncovered a requirement for the transcrip- tion factors STAT3, ATF3, CREB, and c-Jun in promoting axon
tion factors FEZL and CTIP2 in the generation of proper layer growth after injury (Gao et al., 2004; Lindwall et al., 2004; Qiu
V neuron projections to subcortical targets (Chen et al., 2005a; et al., 2005; Raivich et al., 2004; Seijffers et al., 2007; Tsujino
Molyneaux et al., 2005). The transcription factors Tbr1 and et al., 2000). Changes in the expression of transcription factors
Sox5 are required for the specification of layer VI neurons and have also been identified in other models of neuronal injury,
their projections to the thalamus (Bedogni et al., 2010; Lai including stroke. A number of these transcription factors,
et al., 2008; McKenna et al., 2011), and Sat2b is required for including STAT3 and KLF7 may play a role in axon sprouting after
callosal projections (Alcamo et al., 2008; Britanova et al., stroke (Li et al., 2010b). Thus, there might be shared transcrip-
2008). Expression of Fezl or Ctip2 in vivo forces neurons to tional responses following stroke with those promoting axon
acquire a deep-layer projection pattern while suppressing the regeneration after neuronal injury. Taken together, these studies
expression of Tbr1 and Sat2b (Chen et al., 2008; Molyneaux highlight the importance of transcriptional responses in axon
et al., 2005). Conversely, Tbr1 and Sat2b are thought to directly regeneration and offer the prospect that cell-intrinsic responses
repress Fezl and Ctip2, respectively (Alcamo et al., 2008; might provide a target for development of new therapeutic possi-
McKenna et al., 2011). Thus, a complex transcriptional network bilities in neurological diseases.
specifies a particular cortical subtype, in part by suppressing the A major focus in the study of the role of transcription factors in
expression of factors that drive alternate subtypes. Identification axon growth and regeneration is the identity of the relevant target
of the downstream mechanisms mediating the effects of these genes. Axon guidance molecules including members of the
transcription factors on cortical projection patterns will reveal ephrin and semaphorin families of proteins have been identified
whether they are directly linked to the machinery controlling as key targets (Polleux et al., 2007). Fewer studies have identified
axon growth and guidance or if they act primarily in neuron direct cytoskeletal regulators that might act at the growth cone
specification. or in axon protein transport. The transcription factor COUP-TFI
Considering the evidence indicating that neurons have a devel- (NR2F1) plays a critical role in neurogenesis, differentiation,
opmentally regulated intrinsic axon growth capacity (Blackmore migration, and formation of commissural projections. Primary
and Letourneau, 2006; Bouslama-Oueghlani et al., 2003; Gold- hippocampal neurons from COUP-TFI knockout mice initially
berg et al., 2002b), several groups have sought to characterize grow short abnormal axons but later grow to the same extent
the transcriptome of neurons exhibiting different axon growth as wild-type cells (Armentano et al., 2006). The expression of
capabilities in development and in the context of injury (Costigan the cytoskeletal regulators MAP1B and RND2 is altered in
et al., 2002; Méchaly et al., 2006; Moore et al., 2009; Zou et al., COUP-TFI knockout brains in microarray analyses (Armentano
2009). Transcriptional profiling of retinal ganglion neurons from et al., 2006). The tumor suppressor p53 has also been reported
embryonic to postnatal stages has revealed that several mem- to promote axon growth by regulating the expression of cGKI,
bers of the Krupel-like family (KLF) of transcription factors are a kinase that counteracts growth cone collapse induced by
regulated throughout development (Moore et al., 2009). The semaphorin 3A signaling (Tedeschi et al., 2009b) or by inducing

Neuron 72, October 6, 2011 ª2011 Elsevier Inc. 29


Neuron

Review

the expression of cytoskeletal regulators including GAP-43, phases. Just as with axon specification and neuron migration,
Coronin1, and the GTPase Rab13 following axonal injury (Di granule neurons of the rodent cerebellar cortex provide a robust
Giovanni et al., 2006; Tedeschi et al., 2009a). The case of p53 model system for the study of dendrite development including
is more complex as other studies suggest that p53 can promote their distinct stages of growth, pruning, and postsynaptic
axon growth independently of transcription by inhibiting Rho maturation (Figure 1). In recent years, a number of transcription
kinase (ROCK) at the axon (Qin et al., 2009, 2010). Collectively, factors have been discovered to regulate distinct stages of
these studies support the idea that transcription factors can dendrite development in granule neurons. As part of the
independently regulate two different aspects of axon develop- process of establishing neuronal polarity, the FOXO transcrip-
ment, growth and guidance, by inducing different target genes tion factors, and in particular the brain-enriched protein
according to the developmental requirements of the cell. FOXO6, inhibit the growth of dendrites while simultaneously
Is axon growth regulated by epigenetic mechanisms? promoting the growth of axons (de la Torre-Ubieta et al.,
Compelling evidence on epigenetic mechanisms selectively 2010). Thus, even as neurons migrate and their axons grow,
regulating axon growth in the mammalian brain is scarce. Epige- transcriptional mechanisms are at play to inhibit the formation
netic regulators including the histone acetyltransferase CBP and of dendrites. In this capacity, the FOXO proteins may inhibit
the chromatin modifier Sat2b influence cortical and motor a cell-intrinsic switch from axon to dendrite growth in the
neuron projection patterns, but this is also linked to a role in brain. The bHLH protein NeuroD plays a critical role in the initi-
neuronal subtype specification (Alcamo et al., 2008; Britanova ation of dendrite growth as well as the branching of granule
et al., 2008; Lee et al., 2009). Loss of function of the methyl- neuron dendrite arbors in the cerebellar cortex (Gaudillière
CpG-binding transcriptional repressor MeCP2 has been associ- et al., 2004). While NeuroD promotes the initiation of dendrite
ated with several abnormalities in neuronal morphogenesis growth and elaboration, the zinc-finger transcription factor
including disrupted axon projections (Belichenko et al., 2009; Sp4 promotes the pruning of the granule neuron dendrite arbor
Degano et al., 2009). Axonal targeting defects observed in (Ramos et al., 2007, 2009), and the MADS domain transcription
MeCP2 knockout mice are attributed to changes in the expres- factor MEF2A triggers the morphogenesis of the postsynaptic
sion of the guidance factor Semaphorin3F, albeit in a non-cell- dendritic claws (Shalizi et al., 2006, 2007). Collectively, these
autonomous fashion (Degano et al., 2009). Among the genes studies support the concept that different transcription factors
identified in a screen for axonal sprouting after stroke is ATRX are dedicated to distinct aspects of dendrite development
(a-thalassemia/mental retardation syndrome X-linked) (Li et al., (Figure 1). Whether and how these transcription factors might
2010b), a chromatin remodeling enzyme linked to mental retar- regulate each other in the control of dendrite morphogenesis
dation that has also been implicated in dendrite development is an unanswered question.
and neuronal survival (Bérubé et al., 2005; Shioda et al., 2011). An interesting feature of the role of transcription factors in
ATRX appears to be upregulated in sprouting neurons relative the regulation of dendrite development is that they are robustly
to nonsprouting neurons. Knockdown of ATRX by RNAi reduces influenced by calcium signaling and consequently neuronal
basal axon growth of cultured DRG neurons and prevents axonal activity (Figure 4). Membrane depolarization is critical for the
sprouting after stroke in vivo (Li et al., 2010b). Interestingly, ATRX development of dendrite growth and branching, including in
and MeCP2 can interact in vitro and in cells, and in MeCP2 granule neurons of the cerebellar cortex (Gaudillière et al.,
knockout cells ATRX fails to localize to heterochromatin, display- 2004; Okazawa et al., 2009). Calcium influx via L-type calcium
ing instead a diffuse expression pattern (Nan et al., 2007). Thus, channels triggers the activation of the protein kinase CaMKIIa
some of the neuronal defects observed in MeCP2 mutants might (Hudmon and Schulman, 2002; Wayman et al., 2008b). Once
be due to abnormal ATRX activity. Future studies will be needed activated, CaMKIIa induces the phosphorylation of NeuroD at
to understand the extent of epigenetic mechanisms in axon Serine 336 (Gaudillière et al., 2004). Structure-function analyses
growth. of NeuroD in the background of NeuroD RNAi indicate that the
CaMKIIa-induced phosphorylation of NeuroD, including at
Transcription Factors Direct Distinct Stages of Dendrite Serine 336, is essential for the ability of NeuroD to mediate mem-
Morphogenesis brane depolarization-dependent dendrite growth (Gaudillière
As the receptive limbs of neurotransmission in the brain, et al., 2004). How the CaMKIIa-induced phosphorylation acti-
dendrites have evolved to display immense variety of shape vates the transcriptional function of NeuroD remains to be
and size. Dendrite architecture strongly influences the process- determined. Since depolarization-induced NeuroD-dependent
ing of information (Spruston, 2008), suggesting that the morpho- transcription in transient reporter assays is not affected by muta-
genesis of dendrite arbors directly impacts the flow of informa- tion of Serine 336 (Gaudillière et al., 2004), the possibility that
tion across the brain. Although we will focus on the role of the CaMKIIa-induced phosphorylation triggers changes in
transcription factors on dendrite morphology in mammalian NeuroD-recruitment of chromatin remodeling enzymes is an
systems, significant contributions in this field have also come intriguing possibility that remains to be tested. The NeuroD
from studies in the fly nervous system. We refer the reader to target genes that couple calcium signaling to the growth of
excellent reviews on this topic (Corty et al., 2009; Jan and Jan, dendrites also remain unknown. Interestingly, the role of NeuroD
2003, 2010). in dendrite morphogenesis seems to extend beyond early post-
Dendrites come in greater variety of shapes and size than natal development into the regulation of dendrites in adult-born
axons, and the morphogenesis of dendrites in different popula- neurons. Adult-born granule neurons of the hippocampus in
tions of mammalian neurons proceeds in distinct stereotypical NeuroD null mice display shorter dendrites as compared to

30 Neuron 72, October 6, 2011 ª2011 Elsevier Inc.


Neuron

Review

Ca2+

VSCC or NMDAR

CaMKK
Cytoplasm

Neuronal Activity
CaMKIγ CaMKIV CaMKIIα

P P
CREB NeuroD
Nucleus

CREST

Other -BDNF
TR -Wnt-2
-cpg15
BAF57 BRD7,9
BAF180 -Other genes regulating
ARID1, BAF47 dendrite morphogenesis
ARID2A,B
BAF60A,C BRG1,BRM
Actin P CBP P
BAF45B,C BAF155 BAF170 BAF53B
CREB NeuroD

Figure 4. Neuronal Activity Regulates Transcription-Dependent Dendrite Growth


Activity-dependent gene expression is regulated at multiple levels. Neuronal activity leading to calcium influx via voltage-sensitive calcium channels or NMDA
receptors leads to the activation of CaMKK and downstream kinases CaMKIg, CaMKIV, or CaMKIIa to regulate transcription factors that control dendrite growth.
Activation of CaMKIg or CaMKIV leads to phosphorylation and activation of CREB, whereas CaMKIIa phosphorylates NeuroD and induces NeuroD-dependent
transcription. A number of transcriptional regulators (TR) including CBP, CREST, TORC1, and CRTC1 associate and regulate CREB-dependent transcription. In
another layer of regulation, epigenetic mechanisms, including chromatin modification by the chromatin remodeling complex nBAF have a critical role in activity-
dependent dendrite growth. CREST associates with the nBAF complex and regulates the expression of a number of genes important for dendrite growth. The
complex interplay between transcription factors, transcriptional regulators and chromatin modifying enzymes that regulate gene expression in response to
neuronal activity remains to be elucidated.

wild-type neurons (Gao et al., 2009). Whether calcium signaling MEF2A Lysine 408 from sumoylation to acetylation, thereby
is relevant to NeuroD-dependent dendrite morphogenesis in converting MEF2A from a transcriptional repressor form to an
adult-born neurons remains an open question. activator, and leading to the inhibition of postsynaptic dendritic
Calcium signaling also regulates the function of the transcrip- claw differentiation (Shalizi et al., 2006). Consistent with these
tion factor MEF2A in postsynaptic dendritic differentiation. A findings, activation of MEF2-dependent transcription triggers
calcium-regulated sumoylated transcriptionally repressive form elimination of postsynaptic sites in other populations of brain
of MEF2A drives the differentiation of dendritic claws in the cere- neurons (Barbosa et al., 2008; Flavell et al., 2006, 2008; Pfeiffer
bellar cortex (Shalizi et al., 2006, 2007). Sumoylation of MEF2A et al., 2010; Pulipparacharuvil et al., 2008). What might be the
at Lysine 408, which converts MEF2A into a transcriptional purpose of calcium influx through L-type VSCCs inhibiting the
repressor, is dependent on the status of phosphorylation of a function of sumoylated MEF2A in postsynaptic dendritic claw
nearby site, Serine 403, which in turn is regulated by the cal- differentiation? A plausible explanation is that calcium influx in
cium-regulated phosphatase calcineurin (Shalizi et al., 2006). membrane depolarized granule neurons during earlier phases
The phosphorylation of MEF2A at Serine 403 is required for the of dendrite development might coordinately promote dendrite
sumoylation of MEF2A at Lysine 408, owing to increasing the growth and branching via NeuroD and concomitantly inhibit
catalytic efficiency of the SUMO E2 enzyme Ubc9 acting on the premature formation of postsynaptic dendrite sites. Alterna-
MEF2A as a substrate (Mohideen et al., 2009; Shalizi et al., tively, with neuronal maturation, calcium influx induced by trans-
2006). Strikingly, calcineurin-induced dephosphorylation of synaptic signaling might induce the refinement of postsynaptic
MEF2A at Serine 403 triggers a switch in the modification of dendritic structures.

Neuron 72, October 6, 2011 ª2011 Elsevier Inc. 31


Neuron

Review

The idea that calcium signaling and VSCCs induce the early Just as in the cerebellar cortex, studies of dendrite morpho-
growth of dendrites and simultaneously inhibit the later steps genesis in the cerebral cortex and hippocampus have high-
of dendrite development has been further evaluated in recent lighted the regulation of transcription factors by neuronal activity
studies. Interestingly, the resting membrane potential of newly and calcium influx (Figure 4). Prominent among these is the
generated granule neurons in the EGL is depolarized, and it is transcription factor cAMP-responsive element binding protein
hyperpolarized with maturation in the IGL (Okazawa et al., (CREB), which is modulated by a variety of extrinsic cues and
2009). Hyperpolarization of granule neurons in cerebellar slices regulates neuronal survival, dendrite growth, and synaptic
triggers dendritic pruning and differentiation, including the function (Flavell and Greenberg, 2008; Lonze and Ginty, 2002;
formation of dendritic claws (Okazawa et al., 2009). Switching Shaywitz and Greenberg, 1999). Neuronal activity stimulates
between these stages of dendrite morphogenesis coincides CaMKIV-dependent phosphorylation and activation of CREB in
with changes in the expression of a large number of genes, cortical neurons and thereby induces dendrite growth and arbor-
including the transcription factors Etv1, Math2, Tle1, and Hey1, ization (Redmond et al., 2002). In more recent studies, CaMKIg
suggesting that these proteins might regulate dendrite matura- has been found to mediate neuronal activity-dependent phos-
tion (Okazawa et al., 2009; Sato et al., 2005). Collectively, studies phorylation and activation of CREB in hippocampal neurons,
of dendrite morphogenesis in the cerebellar cortex support the leading to increased dendritic arborization (Wayman et al.,
idea that both the early phases of dendrite growth and activity- 2006). The CREB coactivator CBP also participates in neuronal
dependent remodeling are under the purview of transcription activity-induced dendrite morphogenesis (Redmond et al.,
factor regulation. 2002). Another calcium-regulated transcriptional coactivator
Although studies in the cerebellar cortex have provided com- termed CREST, which also associates with CBP, is required
pelling evidence for cell-intrinsic regulation of stage-dependent for activity-dependent dendrite growth development in the cere-
dendrite morphogenesis that is widely relevant to diverse bral cortex (Aizawa et al., 2004). Recent studies have identified
populations of neurons in the brain, transcription factors can additional CREB binding partners that act as coactivators
also shape the development of dendritic arbors characteristic required for CREB-dependent dendrite growth, including
of a particular neuronal subtype. Transcription factors set up TORC1 (transducer of regulated CREB activity) and CRTC1
complex dendrite morphologies in a neuron-specific manner (CREB-regulated transcription co-activator), which operate
in Drosophila (Corty et al., 2009; Jan and Jan, 2003, 2010). Tran- downstream of activity-dependent signaling and BDNF, respec-
scriptional mechanisms specifying dendrite arbors in the tively (Finsterwald et al., 2010; Li et al., 2009). These studies
mammalian brain are also beginning to be described. Tempo- highlight the complexity of CREB-dependent transcription. It
rally specific or layer-specific expression of transcription factors will be important to elucidate the context and signaling mecha-
in the cerebral cortex may define the morphological identity nisms controlling the association of CREB with different coregu-
of neurons (Arlotta et al., 2005; Molyneaux et al., 2009; Moly- lators and the consequences on CREB-dependent transcription.
neaux et al., 2007). The zinc finger transcription factor Fezf2 is Although a role for these transcriptional regulators in dendrite
required for dendritic arbor complexity in layer V/VI neurons development is compelling, the downstream mechanisms are
specifically (Chen et al., 2005b). The mammalian homologs of incompletely understood. BDNF represents a potential relevant
the Drosophila transcription factor Cut, Cux1 and Cux2, have target of CREB and associated proteins in the control of dendrite
been implicated in layer II/III pyramidal neuron dendrite devel- development and branching (Cheung et al., 2007; Dijkhuizen and
opment by two different groups, though with seemingly conflict- Ghosh, 2005a; Horch and Katz, 2002; McAllister et al., 1997; Tao
ing conclusions (Cubelos et al., 2010; Li et al., 2010a). Using et al., 1998). The secreted signaling protein Wnt-2, which pro-
a combination of knockout mice and in vivo RNAi to generate motes dendritic arborization, is also induced by CREB down-
Cux1-and Cux2-deficient cortical neurons in the intact cerebral stream of neuronal activity (Wayman et al., 2006). Likewise, the
cortex, Cubelos and colleagues have found that Cux1 and cpg15 gene, which encodes a GPI-anchored protein, is induced
Cux2 additively promote dendrite growth and branching as by CREB downstream of calcium signaling and promotes
well as dendritic spine formation. Cux1 and Cux2 directly repress dendrite arbor elaboration in Xenopus tectal neurons (Fujino
the putative chromatin modifying proteins Xlr3b and Xlr4b, et al., 2003; Nedivi et al., 1998). Interestingly, the microRNA
which couple Cux1 and Cux2 to regulation of dendritic spine miR-132 is also induced by CREB in an activity-dependent
morphogenesis, while the transcriptional targets involved in manner and promotes the elaboration of dendrite arbors in
dendrite arbor formation remain to be identified (Cubelos et al., hippocampal neurons (Magill et al., 2010; Wayman et al.,
2010). In contrast, using cortical cultures Li and colleagues 2008a). Taking into account that CREB mediates several aspects
have found that overexpression of Cux1, but not Cux2, de- of neuronal development including neuronal survival (Bonni
creases dendrite complexity, and conversely that knockdown et al., 1999; Lonze et al., 2002; Riccio et al., 1999), identifying
of Cux1 leads to excessive dendritic arbor size in cortical neu- the specific direct targets involved in dendrite growth will clarify
rons. Li and colleagues have also reported that Cux1 directly the role of CREB in neuronal morphogenesis.
represses the cell-cycle regulator p27kip1 and thereby inhibits The complexity of transcriptional regulation in activity-depen-
dendrite growth through RhoA (Li et al., 2010a). The findings dent dendrite growth is further highlighted by evidence demon-
from Cubelos and colleagues whereby Cux1 promotes dendritic strating that the nBAF chromatin remodeling complex is required
complexity are consistent with the function of the fly homolog for dendrite development (Figure 4; Wu et al., 2007). The multi-
Cut, suggesting functional evolutionary conservation of this tran- meric nBAF complex is assembled from several homologous
scription factor. proteins in a developmental-specific manner. In the context of

32 Neuron 72, October 6, 2011 ª2011 Elsevier Inc.


Neuron

Review

this combinatorial assembly, the BAF53a subunit, which is tion. In addition, the functions of different transcription factors
present in neuronal progenitors, is replaced by the BAF53b may overlap temporally to control a specific feature of neuronal
subunit, which is specific for differentiated neurons (Lessard morphology and connectivity.
et al., 2007). Genetic ablation of BAF53b in mice leads to An important goal of future research in the study of transcrip-
abnormalities in basal and activity-dependent dendrite growth. tional regulation of neuronal morphogenesis will be to define the
Interestingly, the nBAF complex associates with CREST and relationship between different transcription factors regulating
modulates the expression of a large number of genes involved distinct phases of neuronal development. For example, it will
in neurite growth (Wu et al., 2007). This is of particular interest be interesting to determine whether and how the functions of
in light of the observation that at least two other epigenetic FOXO6, NeuroD, Sp4, and sumoylated MEF2A intersect in the
regulators, the histone demethylase SMCX and the DNA course of orchestrating granule neuron dendrite arbor develop-
methyl-binding transcriptional repressor MeCP2, which are ment in the cerebellar cortex. Do any of these transcriptional
mutated in cases of X-linked mental retardation (XLMR) and factors regulate the expression of another factor acting in a
Rett syndrome also control dendrite growth (Ballas et al., 2009; subsequent or preceding step of dendrite development? Do
Iwase et al., 2007; Zhou et al., 2006). These studies suggest any of these factors interact with other transcription factors
that epigenetic mechanisms altering chromatin structure, which and thereby regulate their activity? Finally, do upstream signals
can drive longer lasting transcriptional changes or provide addi- impinging on a specific transcription factor, such as CaMKIIa
tional levels of regulation, contribute to dendrite development. or calcineurin that control NeuroD and MEF2A activity respec-
Elucidation of the interplay between these epigenetic regulators tively, influence the activity of another transcription factor acting
and transcription factors in the context of dendrite development on a different stage of dendrite development?
should advance our understanding of these disorders. Another important goal of future studies will be to determine
The few transcription factors that have been characterized in the extent of programs of gene expression regulated by different
dendrite development in the mammalian brain to date likely transcription factors acting at distinct stages of neuronal devel-
only represent the tip of the iceberg. Further, the targets of opment. Advances in genomic technologies will facilitate these
many of these transcription factors are largely unknown. Regula- studies and yield large datasets for analysis of transcription
tors of cytoskeletal components, including Rho-GTPases and factor-dependent networks of genes at distinct developmental
microtubule-binding proteins, have been identified as targets stages. Comparisons of these data sets as well as analyses of
of transcription factor regulation in the context of dendrite devel- transcription factor occupancies at target genes should also
opment (Cobos et al., 2007; Hand et al., 2005; Li et al., 2010a; Wu provide novel insights into cooperative mechanisms between
et al., 2007). It will be interesting to determine whether additional different transcriptional regulators in neuronal morphogenesis
mechanisms, including contact-mediated signaling and secre- and connectivity. Several transcription factors that direct
tory function through the Golgi apparatus, also operate down- neuronal morphogenesis in postmitotic neurons also have roles
stream of specific transcriptional regulators in the control of in neuron specification. Although dissociating such distinct
dendrite morphogenesis. roles may not always be a simple task, transcriptional profiling
coupled with ChIP-Seq analyses may allow for the characteriza-
Concluding Remarks tion of targetomes associated with specific developmental
Transcription factors play a prominent role in all facets of programs.
neuronal development from neuronal polarization and migration The complexity of transcriptional regulation is vast. Transcrip-
to axon and dendrite morphogenesis to synapse differentiation. tion factors are controlled by posttranslational modifications,
A flurry of studies during the past decade has unraveled many which lead to changes in protein stability, localization, activity,
functions of transcription factors and regulators in neuronal or interaction partners. These modifications may not simply
development in the mammalian brain. Prior to these studies, stimulate or inhibit transcriptional activity of the factor but may
transcription factors were generally considered to govern the induce a switch in the mode of a transcription factor’s function
transition from precursor cells to postmitotic neurons, and this between activator and repressor. Additionally, association with
transition was thought to unleash a differentiation program, re- epigenetic regulators, including chromatin remodeling com-
sulting in the mature morphology of neurons. A major conclusion plexes, may induce longer lasting or widespread changes in
of studies of the past decade is that transcription factors con- gene expression. Finally, transcription factors often regulate
tinue to play key regulatory roles in postmitotic neurons to the expression of other transcription factors creating complex
specify and regulate the development of distinct morphological cascades. How and to what extent these cascades may be
compartments. Another related key conclusion is the concept involved in other aspects of neuronal morphogenesis is a task
that different transcription factors are dedicated to distinct for future studies.
phases of neuronal morphogenesis and connectivity. This, Finally, studies of transcriptional regulation offer the basis for
however, is an oversimplification. Although some transcription elucidation of key mechanisms of brain development as well as
factors have a restricted expression pattern and orchestrate serve the foundation for a better understanding of the molecular
specific aspects of development, others operate in a pleiotropic basis of developmental disorders of the brain in which deregula-
manner to regulate several steps of development. In some tion of neuronal morphogenesis and connectivity plays a promi-
cases, transcription factors operate as nodes to coordinate nent role (Kaufmann and Moser, 2000; McManus and Golden,
two different aspects of neuronal development, such as neuronal 2005; Penzes et al., 2011; Schwartzkroin and Walsh, 2000; Siso-
branching and migration or dendrite growth and synapse forma- diya, 2004). Mutations in several transcriptional regulators have

Neuron 72, October 6, 2011 ª2011 Elsevier Inc. 33


Neuron

Review

been implicated in diverse array of neurodevelopmental disor- Barbosa, A.C., Kim, M.S., Ertunc, M., Adachi, M., Nelson, E.D., McAnally, J.,
Richardson, J.A., Kavalali, E.T., Monteggia, L.M., Bassel-Duby, R., and Olson,
ders from mental retardation and autism spectrum disorders to E.N. (2008). MEF2C, a transcription factor that facilitates learning and memory
inherited ataxias to epilepsy syndromes (Grinberg and Millen, by negative regulation of synapse numbers and function. Proc. Natl. Acad. Sci.
2005; Gutierrez-Delicado and Serratosa, 2004; Helmlinger USA 105, 9391–9396.
et al., 2006; Hong et al., 2005; Orr, 2010). Understanding the Barnes, A.P., and Polleux, F. (2009). Establishment of axon-dendrite polarity in
normal functions of these transcriptional regulators in neuronal developing neurons. Annu. Rev. Neurosci. 32, 347–381.
morphogenesis and connectivity will be a major first step toward
Barnes, A.P., Lilley, B.N., Pan, Y.A., Plummer, L.J., Powell, A.W., Raines, A.N.,
understanding the pathogenesis of these disorders. Sanes, J.R., and Polleux, F. (2007). LKB1 and SAD kinases define a pathway
required for the polarization of cortical neurons. Cell 129, 549–563.
ACKNOWLEDGMENTS
Bedogni, F., Hodge, R.D., Elsen, G.E., Nelson, B.R., Daza, R.A., Beyer, R.P.,
Bammler, T.K., Rubenstein, J.L., and Hevner, R.F. (2010). Tbr1 regulates
We thank members of the Bonni laboratory, in particular Luis Mejı́a, Yoshiho regional and laminar identity of postmitotic neurons in developing neocortex.
Ikeuchi, and Chi Zhang, for helpful discussions and critical reading of the Proc. Natl. Acad. Sci. USA 107, 13129–13134.
manuscript. The authors are supported by NIH grant NS041021 (A.B.) and
the Albert J. Ryan Foundation (L.T.U.). Behar, O., Golden, J.A., Mashimo, H., Schoen, F.J., and Fishman, M.C. (1996).
Semaphorin III is needed for normal patterning and growth of nerves, bones
and heart. Nature 383, 525–528.
REFERENCES
Belichenko, P.V., Wright, E.E., Belichenko, N.P., Masliah, E., Li, H.H., Mobley,
Aguayo, A.J., Rasminsky, M., Bray, G.M., Carbonetto, S., McKerracher, L., W.C., and Francke, U. (2009). Widespread changes in dendritic and axonal
Villegas-Pérez, M.P., Vidal-Sanz, M., and Carter, D.A. (1991). Degenerative morphology in Mecp2-mutant mouse models of Rett syndrome: evidence for
and regenerative responses of injured neurons in the central nervous system disruption of neuronal networks. J. Comp. Neurol. 514, 240–258.
of adult mammals. Philos. Trans. R. Soc. Lond. B Biol. Sci. 331, 337–343.
Bérubé, N.G., Mangelsdorf, M., Jagla, M., Vanderluit, J., Garrick, D., Gibbons,
Aizawa, H., Hu, S.C., Bobb, K., Balakrishnan, K., Ince, G., Gurevich, I., Cowan, R.J., Higgs, D.R., Slack, R.S., and Picketts, D.J. (2005). The chromatin-remod-
M., and Ghosh, A. (2004). Dendrite development regulated by CREST, eling protein ATRX is critical for neuronal survival during corticogenesis. J. Clin.
a calcium-regulated transcriptional activator. Science 303, 197–202. Invest. 115, 258–267.

Alcamo, E.A., Chirivella, L., Dautzenberg, M., Dobreva, G., Fariñas, I., Biggs, W.H., 3rd, Meisenhelder, J., Hunter, T., Cavenee, W.K., and Arden, K.C.
Grosschedl, R., and McConnell, S.K. (2008). Satb2 regulates callosal projec- (1999). Protein kinase B/Akt-mediated phosphorylation promotes nuclear
tion neuron identity in the developing cerebral cortex. Neuron 57, 364–377. exclusion of the winged helix transcription factor FKHR1. Proc. Natl. Acad.
Sci. USA 96, 7421–7426.
Altman, J., and Bayer, S.A. (1997). Development of the Cerebellar System: In
Relation to Its Evolution, Structure, and Functions (Boca Raton, FL: CRC Bilimoria, P.M., and Bonni, A. (2008). Cultures of cerebellar granule neurons.
Press). CSH Protoc. 2008. 10.1101/pdb.prot5107.

Araki, T., Sasaki, Y., and Milbrandt, J. (2004). Increased nuclear NAD biosyn- Bisbal, M., Conde, C., Donoso, M., Bollati, F., Sesma, J., Quiroga, S.,
thesis and SIRT1 activation prevent axonal degeneration. Science 305, 1010– Dı́az Añel, A., Malhotra, V., Marzolo, M.P., and Cáceres, A. (2008). Protein
1013. kinase d regulates trafficking of dendritic membrane proteins in developing
neurons. J. Neurosci. 28, 9297–9308.
Arimura, N., and Kaibuchi, K. (2007). Neuronal polarity: from extracellular
signals to intracellular mechanisms. Nat. Rev. Neurosci. 8, 194–205. Blackmore, M., and Letourneau, P.C. (2006). Changes within maturing
neurons limit axonal regeneration in the developing spinal cord. J. Neurobiol.
Arlotta, P., Molyneaux, B.J., Chen, J., Inoue, J., Kominami, R., and Macklis, 66, 348–360.
J.D. (2005). Neuronal subtype-specific genes that control corticospinal motor
neuron development in vivo. Neuron 45, 207–221. Bokoch, G.M. (2003). Biology of the p21-activated kinases. Annu. Rev. Bio-
chem. 72, 743–781.
Armentano, M., Filosa, A., Andolfi, G., and Studer, M. (2006). COUP-TFI is
required for the formation of commissural projections in the forebrain by Bonni, A., Brunet, A., West, A.E., Datta, S.R., Takasu, M.A., and Greenberg,
regulating axonal growth. Development 133, 4151–4162. M.E. (1999). Cell survival promoted by the Ras-MAPK signaling pathway by
transcription-dependent and -independent mechanisms. Science 286,
Asada, N., and Sanada, K. (2010). LKB1-mediated spatial control of GSK3beta
1358–1362.
and adenomatous polyposis coli contributes to centrosomal forward move-
ment and neuronal migration in the developing neocortex. J. Neurosci. 30,
Bouslama-Oueghlani, L., Wehrlé, R., Sotelo, C., and Dusart, I. (2003). The
8852–8865.
developmental loss of the ability of Purkinje cells to regenerate their axons
Asada, N., Sanada, K., and Fukada, Y. (2007). LKB1 regulates neuronal occurs in the absence of myelin: an in vitro model to prevent myelination. J.
migration and neuronal differentiation in the developing neocortex through Neurosci. 23, 8318–8329.
centrosomal positioning. J. Neurosci. 27, 11769–11775.
Bradke, F., and Dotti, C.G. (2000). Differentiated neurons retain the capacity to
Ballas, N., Lioy, D.T., Grunseich, C., and Mandel, G. (2009). Non-cell autono- generate axons from dendrites. Curr. Biol. 10, 1467–1470.
mous influence of MeCP2-deficient glia on neuronal dendritic morphology.
Nat. Neurosci. 12, 311–317. Britanova, O., de Juan Romero, C., Cheung, A., Kwan, K.Y., Schwark, M.,
Gyorgy, A., Vogel, T., Akopov, S., Mitkovski, M., Agoston, D., et al. (2008).
Banker, G., and Goslin, K. (1991). Culturing Nerve Cells (Cambridge, MA: MIT Satb2 is a postmitotic determinant for upper-layer neuron specification in
Press). the neocortex. Neuron 57, 378–392.

Baptista, C.A., Hatten, M.E., Blazeski, R., and Mason, C.A. (1994). Cell-cell Brunet, A., Bonni, A., Zigmond, M.J., Lin, M.Z., Juo, P., Hu, L.S., Anderson,
interactions influence survival and differentiation of purified Purkinje cells M.J., Arden, K.C., Blenis, J., and Greenberg, M.E. (1999). Akt promotes cell
in vitro. Neuron 12, 243–260. survival by phosphorylating and inhibiting a Forkhead transcription factor.
Cell 96, 857–868.
Barbieri, A.M., Broccoli, V., Bovolenta, P., Alfano, G., Marchitiello, A.,
Mocchetti, C., Crippa, L., Bulfone, A., Marigo, V., Ballabio, A., and Banfi, S. Brunet, A., Sweeney, L.B., Sturgill, J.F., Chua, K.F., Greer, P.L., Lin, Y., Tran,
(2002). Vax2 inactivation in mouse determines alteration of the eye dorsal- H., Ross, S.E., Mostoslavsky, R., Cohen, H.Y., et al. (2004). Stress-dependent
ventral axis, misrouting of the optic fibres and eye coloboma. Development regulation of FOXO transcription factors by the SIRT1 deacetylase. Science
129, 805–813. 303, 2011–2015.

34 Neuron 72, October 6, 2011 ª2011 Elsevier Inc.


Neuron

Review
Calderon de Anda, F., Gärtner, A., Tsai, L.H., and Dotti, C.G. (2008). Pyramidal Desai, A.R., and McConnell, S.K. (2000). Progressive restriction in fate poten-
neuron polarity axis is defined at the bipolar stage. J. Cell Sci. 121, 178–185. tial by neural progenitors during cerebral cortical development. Development
127, 2863–2872.
Castrillon, D.H., Miao, L., Kollipara, R., Horner, J.W., and DePinho, R.A. (2003).
Suppression of ovarian follicle activation in mice by the transcription factor Dhavan, R., and Tsai, L.H. (2001). A decade of CDK5. Nat. Rev. Mol. Cell Biol.
Foxo3a. Science 301, 215–218. 2, 749–759.

Causeret, F., Terao, M., Jacobs, T., Nishimura, Y.V., Yanagawa, Y., Obata, K., Di Giovanni, S., Knights, C.D., Rao, M., Yakovlev, A., Beers, J., Catania, J.,
Hoshino, M., and Nikolic, M. (2009). The p21-activated kinase is required for Avantaggiati, M.L., and Faden, A.I. (2006). The tumor suppressor protein
neuronal migration in the cerebral cortex. Cereb. Cortex 19, 861–875. p53 is required for neurite outgrowth and axon regeneration. EMBO J. 25,
4084–4096.
Chen, B., Schaevitz, L.R., and McConnell, S.K. (2005a). Fezl regulates the
differentiation and axon targeting of layer 5 subcortical projection neurons in Dickson, B.J. (2002). Molecular mechanisms of axon guidance. Science 298,
cerebral cortex. Proc. Natl. Acad. Sci. USA 102, 17184–17189. 1959–1964.

Chen, J.G., Rasin, M.R., Kwan, K.Y., and Sestan, N. (2005b). Zfp312 is Dijkhuizen, P.A., and Ghosh, A. (2005a). BDNF regulates primary dendrite
required for subcortical axonal projections and dendritic morphology of formation in cortical neurons via the PI3-kinase and MAP kinase signaling
deep-layer pyramidal neurons of the cerebral cortex. Proc. Natl. Acad. Sci. pathways. J. Neurobiol. 62, 278–288.
USA 102, 17792–17797.
Dijkhuizen, P.A., and Ghosh, A. (2005b). Regulation of dendritic growth by
calcium and neurotrophin signaling. Prog. Brain Res. 147, 17–27.
Chen, B., Wang, S.S., Hattox, A.M., Rayburn, H., Nelson, S.B., and McConnell,
S.K. (2008). The Fezf2-Ctip2 genetic pathway regulates the fate choice of Dotti, C.G., Sullivan, C.A., and Banker, G.A. (1988). The establishment of
subcortical projection neurons in the developing cerebral cortex. Proc. Natl. polarity by hippocampal neurons in culture. J. Neurosci. 8, 1454–1468.
Acad. Sci. USA 105, 11382–11387.
Dufour, A., Seibt, J., Passante, L., Depaepe, V., Ciossek, T., Frisén, J.,
Cheung, Z.H., Chin, W.H., Chen, Y., Ng, Y.P., and Ip, N.Y. (2007). Cdk5 is Kullander, K., Flanagan, J.G., Polleux, F., and Vanderhaeghen, P. (2003).
involved in BDNF-stimulated dendritic growth in hippocampal neurons. Area specificity and topography of thalamocortical projections are controlled
PLoS Biol. 5, e63. 10.1371/journal.pbio.0050063. by ephrin/Eph genes. Neuron 39, 453–465.
Cobos, I., Borello, U., and Rubenstein, J.L. (2007). Dlx transcription factors Ernfors, P., Lee, K.F., and Jaenisch, R. (1994). Mice lacking brain-derived
promote migration through repression of axon and dendrite growth. Neuron neurotrophic factor develop with sensory deficits. Nature 368, 147–150.
54, 873–888.
Essers, M.A., Weijzen, S., de Vries-Smits, A.M., Saarloos, I., de Ruiter, N.D.,
Corty, M.M., Matthews, B.J., and Grueber, W.B. (2009). Molecules and mech- Bos, J.L., and Burgering, B.M. (2004). FOXO transcription factor activation
anisms of dendrite development in Drosophila. Development 136, 1049–1061. by oxidative stress mediated by the small GTPase Ral and JNK. EMBO J.
23, 4802–4812.
Costigan, M., Befort, K., Karchewski, L., Griffin, R.S., D’Urso, D., Allchorne, A.,
Sitarski, J., Mannion, J.W., Pratt, R.E., and Woolf, C.J. (2002). Replicate high- Falnikar, A., and Baas, P.W. (2009). Critical roles for microtubules in axonal
density rat genome oligonucleotide microarrays reveal hundreds of regulated development and disease. Results Probl. Cell Differ. 48, 47–64.
genes in the dorsal root ganglion after peripheral nerve injury. BMC Neurosci.
3, 16. Filbin, M.T. (2003). Myelin-associated inhibitors of axonal regeneration in the
adult mammalian CNS. Nat. Rev. Neurosci. 4, 703–713.
Craig, A.M., and Banker, G. (1994). Neuronal polarity. Annu. Rev. Neurosci. 17,
267–310. Finsterwald, C., Fiumelli, H., Cardinaux, J.R., and Martin, J.L. (2010).
Regulation of dendritic development by BDNF requires activation of CRTC1
Cubelos, B., Sebastián-Serrano, A., Beccari, L., Calcagnotto, M.E., Cisneros, by glutamate. J. Biol. Chem. 285, 28587–28595.
E., Kim, S., Dopazo, A., Alvarez-Dolado, M., Redondo, J.M., Bovolenta, P.,
et al. (2010). Cux1 and Cux2 regulate dendritic branching, spine morphology, Flavell, S.W., and Greenberg, M.E. (2008). Signaling mechanisms linking
and synapses of the upper layer neurons of the cortex. Neuron 66, 523–535. neuronal activity to gene expression and plasticity of the nervous system.
Annu. Rev. Neurosci. 31, 563–590.
Daitoku, H., Hatta, M., Matsuzaki, H., Aratani, S., Ohshima, T., Miyagishi, M.,
Nakajima, T., and Fukamizu, A. (2004). Silent information regulator 2 potenti- Flavell, S.W., Cowan, C.W., Kim, T.K., Greer, P.L., Lin, Y., Paradis, S., Griffith,
ates Foxo1-mediated transcription through its deacetylase activity. Proc. E.C., Hu, L.S., Chen, C., and Greenberg, M.E. (2006). Activity-dependent
Natl. Acad. Sci. USA 101, 10042–10047. regulation of MEF2 transcription factors suppresses excitatory synapse
number. Science 311, 1008–1012.
Davies, S.J., Fitch, M.T., Memberg, S.P., Hall, A.K., Raisman, G., and Silver, J.
Flavell, S.W., Kim, T.K., Gray, J.M., Harmin, D.A., Hemberg, M., Hong, E.J.,
(1997). Regeneration of adult axons in white matter tracts of the central
Markenscoff-Papadimitriou, E., Bear, D.M., and Greenberg, M.E. (2008).
nervous system. Nature 390, 680–683.
Genome-wide analysis of MEF2 transcriptional program reveals synaptic
target genes and neuronal activity-dependent polyadenylation site selection.
Davies, S.J., Goucher, D.R., Doller, C., and Silver, J. (1999). Robust regenera-
Neuron 60, 1022–1038.
tion of adult sensory axons in degenerating white matter of the adult rat spinal
cord. J. Neurosci. 19, 5810–5822. Frantz, G.D., and McConnell, S.K. (1996). Restriction of late cerebral cortical
progenitors to an upper-layer fate. Neuron 17, 55–61.
de Anda, F.C., Pollarolo, G., Da Silva, J.S., Camoletto, P.G., Feiguin, F., and
Dotti, C.G. (2005). Centrosome localization determines neuronal polarity. Fujino, T., Lee, W.C., and Nedivi, E. (2003). Regulation of cpg15 by signaling
Nature 436, 704–708. pathways that mediate synaptic plasticity. Mol. Cell. Neurosci. 24, 538–554.
de Anda, F.C., Meletis, K., Ge, X., Rei, D., and Tsai, L.H. (2010). Centrosome Furuyama, T., Nakazawa, T., Nakano, I., and Mori, N. (2000). Identification of
motility is essential for initial axon formation in the neocortex. J. Neurosci. the differential distribution patterns of mRNAs and consensus binding
30, 10391–10406. sequences for mouse DAF-16 homologues. Biochem. J. 349, 629–634.

de la Torre-Ubieta, L., Gaudillière, B., Yang, Y., Ikeuchi, Y., Yamada, T., Furuyama, T., Kitayama, K., Shimoda, Y., Ogawa, M., Sone, K., Yoshida-Araki,
DiBacco, S., Stegmüller, J., Schüller, U., Salih, D.A., Rowitch, D., et al. K., Hisatsune, H., Nishikawa, S., Nakayama, K., Nakayama, K., et al. (2004).
(2010). A FOXO-Pak1 transcriptional pathway controls neuronal polarity. Abnormal angiogenesis in Foxo1 (Fkhr)-deficient mice. J. Biol. Chem. 279,
Genes Dev. 24, 799–813. 34741–34749.

Degano, A.L., Pasterkamp, R.J., and Ronnett, G.V. (2009). MeCP2 deficiency Gan, L., Zheng, W., Chabot, J.G., Unterman, T.G., and Quirion, R. (2005).
disrupts axonal guidance, fasciculation, and targeting by altering Semaphorin Nuclear/cytoplasmic shuttling of the transcription factor FoxO1 is regulated
3F function. Mol. Cell. Neurosci. 42, 243–254. by neurotrophic factors. J. Neurochem. 93, 1209–1219.

Neuron 72, October 6, 2011 ª2011 Elsevier Inc. 35


Neuron

Review
Gao, Y., Deng, K., Hou, J., Bryson, J.B., Barco, A., Nikulina, E., Spencer, T., Hoekman, M.F., Jacobs, F.M., Smidt, M.P., and Burbach, J.P. (2006). Spatial
Mellado, W., Kandel, E.R., and Filbin, M.T. (2004). Activated CREB is sufficient and temporal expression of FoxO transcription factors in the developing and
to overcome inhibitors in myelin and promote spinal axon regeneration in vivo. adult murine brain. Gene Expr. Patterns 6, 134–140.
Neuron 44, 609–621.
Hong, E.J., West, A.E., and Greenberg, M.E. (2005). Transcriptional control of
Gao, Z., Ure, K., Ables, J.L., Lagace, D.C., Nave, K.A., Goebbels, S., Eisch, cognitive development. Curr. Opin. Neurobiol. 15, 21–28.
A.J., and Hsieh, J. (2009). Neurod1 is essential for the survival and maturation
of adult-born neurons. Nat. Neurosci. 12, 1090–1092. Horch, H.W., and Katz, L.C. (2002). BDNF release from single cells elicits local
dendritic growth in nearby neurons. Nat. Neurosci. 5, 1177–1184.
Gaudillière, B., Konishi, Y., de la Iglesia, N., Yao, G., and Bonni, A. (2004).
A CaMKII-NeuroD signaling pathway specifies dendritic morphogenesis. Horesh, D., Sapir, T., Francis, F., Wolf, S.G., Caspi, M., Elbaum, M., Chelly, J.,
Neuron 41, 229–241. and Reiner, O. (1999). Doublecortin, a stabilizer of microtubules. Hum. Mol.
Genet. 8, 1599–1610.
Glebova, N.O., and Ginty, D.D. (2004). Heterogeneous requirement of NGF for
sympathetic target innervation in vivo. J. Neurosci. 24, 743–751. Hosaka, T., Biggs, W.H., 3rd, Tieu, D., Boyer, A.D., Varki, N.M., Cavenee, W.K.,
and Arden, K.C. (2004). Disruption of forkhead transcription factor (FOXO)
Gleeson, J.G., Lin, P.T., Flanagan, L.A., and Walsh, C.A. (1999). Doublecortin family members in mice reveals their functional diversification. Proc. Natl.
is a microtubule-associated protein and is expressed widely by migrating Acad. Sci. USA 101, 2975–2980.
neurons. Neuron 23, 257–271.
Huber, A.B., Kolodkin, A.L., Ginty, D.D., and Cloutier, J.F. (2003). Signaling at
Goldberg, J.L., Espinosa, J.S., Xu, Y., Davidson, N., Kovacs, G.T., and Barres,
the growth cone: ligand-receptor complexes and the control of axon growth
B.A. (2002a). Retinal ganglion cells do not extend axons by default: promotion
and guidance. Annu. Rev. Neurosci. 26, 509–563.
by neurotrophic signaling and electrical activity. Neuron 33, 689–702.

Goldberg, J.L., Klassen, M.P., Hua, Y., and Barres, B.A. (2002b). Amacrine- Hudmon, A., and Schulman, H. (2002). Neuronal CA2+/calmodulin-dependent
signaled loss of intrinsic axon growth ability by retinal ganglion cells. Science protein kinase II: the role of structure and autoregulation in cellular function.
296, 1860–1864. Annu. Rev. Biochem. 71, 473–510.

Gomis-Rüth, S., Wierenga, C.J., and Bradke, F. (2008). Plasticity of polariza- Hur, E.M., and Zhou, F.Q. (2010). GSK3 signalling in neural development. Nat.
tion: changing dendrites into axons in neurons integrated in neuronal circuits. Rev. Neurosci. 11, 539–551.
Curr. Biol. 18, 992–1000.
Huynh, M.A., Ikeuchi, Y., Netherton, S., de la Torre-Ubieta, L., Kanadia, R.,
Goslin, K., and Banker, G. (1989). Experimental observations on the develop- Stegmüller, J., Cepko, C., Bonni, S., and Bonni, A. (2011). An isoform-specific
ment of polarity by hippocampal neurons in culture. J. Cell Biol. 108, 1507– SnoN1-FOXO1 repressor complex controls neuronal morphogenesis and
1516. positioning in the mammalian brain. Neuron 69, 930–944.

Govek, E.E., Newey, S.E., and Van Aelst, L. (2005). The role of the Rho Ikeuchi, Y., Stegmüller, J., Netherton, S., Huynh, M.A., Masu, M., Frank, D.,
GTPases in neuronal development. Genes Dev. 19, 1–49. Bonni, S., and Bonni, A. (2009). A SnoN-Ccd1 pathway promotes axonal
morphogenesis in the mammalian brain. J. Neurosci. 29, 4312–4321.
Graef, I.A., Wang, F., Charron, F., Chen, L., Neilson, J., Tessier-Lavigne, M.,
and Crabtree, G.R. (2003). Neurotrophins and netrins require calcineurin/ Iwase, S., Lan, F., Bayliss, P., de la Torre-Ubieta, L., Huarte, M., Qi, H.H.,
NFAT signaling to stimulate outgrowth of embryonic axons. Cell 113, 657–670. Whetstine, J.R., Bonni, A., Roberts, T.M., and Shi, Y. (2007). The X-linked
mental retardation gene SMCX/JARID1C defines a family of histone H3 lysine
Greer, E.L., Oskoui, P.R., Banko, M.R., Maniar, J.M., Gygi, M.P., Gygi, S.P., 4 demethylases. Cell 128, 1077–1088.
and Brunet, A. (2007). The energy sensor AMP-activated protein kinase
directly regulates the mammalian FOXO3 transcription factor. J. Biol. Chem. Jan, Y.N., and Jan, L.Y. (2003). The control of dendrite development. Neuron
282, 30107–30119. 40, 229–242.

Grinberg, I., and Millen, K.J. (2005). The ZIC gene family in development and Jan, Y.N., and Jan, L.Y. (2010). Branching out: mechanisms of dendritic arbor-
disease. Clin. Genet. 67, 290–296. ization. Nat. Rev. Neurosci. 11, 316–328.
Guo, S., Rena, G., Cichy, S., He, X., Cohen, P., and Unterman, T. (1999). Jiang, H., Guo, W., Liang, X., and Rao, Y. (2005). Both the establishment and
Phosphorylation of serine 256 by protein kinase B disrupts transactivation the maintenance of neuronal polarity require active mechanisms: critical roles
by FKHR and mediates effects of insulin on insulin-like growth factor-binding of GSK-3beta and its upstream regulators. Cell 120, 123–135.
protein-1 promoter activity through a conserved insulin response sequence.
J. Biol. Chem. 274, 17184–17192. Joberty, G., Petersen, C., Gao, L., and Macara, I.G. (2000). The cell-polarity
protein Par6 links Par3 and atypical protein kinase C to Cdc42. Nat. Cell
Gutierrez-Delicado, E., and Serratosa, J.M. (2004). Genetics of the epilepsies. Biol. 2, 531–539.
Curr. Opin. Neurol. 17, 147–153.
Jones, K.R., Fariñas, I., Backus, C., and Reichardt, L.F. (1994). Targeted
Hand, R., Bortone, D., Mattar, P., Nguyen, L., Heng, J.I., Guerrier, S., Boutt, E.,
disruption of the BDNF gene perturbs brain and sensory neuron development
Peters, E., Barnes, A.P., Parras, C., et al. (2005). Phosphorylation of Neuroge-
but not motor neuron development. Cell 76, 989–999.
nin2 specifies the migration properties and the dendritic morphology of pyra-
midal neurons in the neocortex. Neuron 48, 45–62.
Kania, A., and Jessell, T.M. (2003). Topographic motor projections in the limb
Hatten, M.E. (1999). Central nervous system neuronal migration. Annu. Rev. imposed by LIM homeodomain protein regulation of ephrin-A:EphA interac-
Neurosci. 22, 511–539. tions. Neuron 38, 581–596.

He, Z., and Koprivica, V. (2004). The Nogo signaling pathway for regeneration Kania, A., Johnson, R.L., and Jessell, T.M. (2000). Coordinate roles for LIM
block. Annu. Rev. Neurosci. 27, 341–368. homeobox genes in directing the dorsoventral trajectory of motor axons in
the vertebrate limb. Cell 102, 161–173.
Hedstrom, K.L., Ogawa, Y., and Rasband, M.N. (2008). AnkyrinG is required
for maintenance of the axon initial segment and neuronal polarity. J. Cell Katz, L.C., and Shatz, C.J. (1996). Synaptic activity and the construction of
Biol. 183, 635–640. cortical circuits. Science 274, 1133–1138.

Helmlinger, D., Tora, L., and Devys, D. (2006). Transcriptional alterations and Kaufmann, W.E., and Moser, H.W. (2000). Dendritic anomalies in disorders
chromatin remodeling in polyglutamine diseases. Trends Genet. 22, 562–570. associated with mental retardation. Cereb. Cortex 10, 981–991.

Herrera, E., Brown, L., Aruga, J., Rachel, R.A., Dolen, G., Mikoshiba, K., Kawaji, K., Umeshima, H., Eiraku, M., Hirano, T., and Kengaku, M. (2004). Dual
Brown, S., and Mason, C.A. (2003). Zic2 patterns binocular vision by speci- phases of migration of cerebellar granule cells guided by axonal and dendritic
fying the uncrossed retinal projection. Cell 114, 545–557. leading processes. Mol. Cell. Neurosci. 25, 228–240.

36 Neuron 72, October 6, 2011 ª2011 Elsevier Inc.


Neuron

Review
Kelsch, W., Mosley, C.P., Lin, C.W., and Lois, C. (2007). Distinct mammalian Lin, L., Hron, J.D., and Peng, S.L. (2004). Regulation of NF-kappaB, Th activa-
precursors are committed to generate neurons with defined dendritic projec- tion, and autoinflammation by the forkhead transcription factor Foxo3a. Immu-
tion patterns. PLoS Biol. 5, e300. 10.1371/journal.pbio.0050300. nity 21, 203–213.

Kimura, T., Watanabe, H., Iwamatsu, A., and Kaibuchi, K. (2005). Tubulin and Lindwall, C., Dahlin, L., Lundborg, G., and Kanje, M. (2004). Inhibition of c-Jun
CRMP-2 complex is transported via Kinesin-1. J. Neurochem. 93, 1371–1382. phosphorylation reduces axonal outgrowth of adult rat nodose ganglia and
dorsal root ganglia sensory neurons. Mol. Cell. Neurosci. 27, 267–279.
Kitamura, T., Nakae, J., Kitamura, Y., Kido, Y., Biggs, W.H., 3rd, Wright, C.V.,
White, M.F., Arden, K.C., and Accili, D. (2002). The forkhead transcription Liodis, P., Denaxa, M., Grigoriou, M., Akufo-Addo, C., Yanagawa, Y., and
factor Foxo1 links insulin signaling to Pdx1 regulation of pancreatic beta cell Pachnis, V. (2007). Lhx6 activity is required for the normal migration and spec-
growth. J. Clin. Invest. 110, 1839–1847. ification of cortical interneuron subtypes. J. Neurosci. 27, 3078–3089.

Kobayashi, T., Storrie, B., Simons, K., and Dotti, C.G. (1992). A functional Lonze, B.E., and Ginty, D.D. (2002). Function and regulation of CREB family
barrier to movement of lipids in polarized neurons. Nature 359, 647–650. transcription factors in the nervous system. Neuron 35, 605–623.

Koizumi, H., Higginbotham, H., Poon, T., Tanaka, T., Brinkman, B.C., and Lonze, B.E., Riccio, A., Cohen, S., and Ginty, D.D. (2002). Apoptosis, axonal
Gleeson, J.G. (2006). Doublecortin maintains bipolar shape and nuclear trans- growth defects, and degeneration of peripheral neurons in mice lacking
location during migration in the adult forebrain. Nat. Neurosci. 9, 779–786. CREB. Neuron 34, 371–385.

Konishi, Y., Stegmüller, J., Matsuda, T., Bonni, S., and Bonni, A. (2004). Cdh1- Luo, L. (2000). Rho GTPases in neuronal morphogenesis. Nat. Rev. Neurosci.
APC controls axonal growth and patterning in the mammalian brain. Science 1, 173–180.
303, 1026–1030.
Luo, K. (2004). Ski and SnoN: negative regulators of TGF-beta signaling. Curr.
Konno, D., Yoshimura, S., Hori, K., Maruoka, H., and Sobue, K. (2005). Involve- Opin. Genet. Dev. 14, 65–70.
ment of the phosphatidylinositol 3-kinase/rac1 and cdc42 pathways in radial
migration of cortical neurons. J. Biol. Chem. 280, 5082–5088. Magill, S.T., Cambronne, X.A., Luikart, B.W., Lioy, D.T., Leighton, B.H., West-
brook, G.L., Mandel, G., and Goodman, R.H. (2010). . microRNA-132 regulates
Kops, G.J., de Ruiter, N.D., De Vries-Smits, A.M., Powell, D.R., Bos, J.L., and dendritic growth and arborization of newborn neurons in the adult hippo-
Burgering, B.M. (1999). Direct control of the Forkhead transcription factor AFX campus. Proc. Natl. Acad. Sci. USA 107, 20382–20387.
by protein kinase B. Nature 398, 630–634.
Markus, A., Patel, T.D., and Snider, W.D. (2002a). Neurotrophic factors and
Kuruvilla, R., Zweifel, L.S., Glebova, N.O., Lonze, B.E., Valdez, G., Ye, H., and axonal growth. Curr. Opin. Neurobiol. 12, 523–531.
Ginty, D.D. (2004). A neurotrophin signaling cascade coordinates sympathetic
neuron development through differential control of TrkA trafficking and retro- Markus, A., Zhong, J., and Snider, W.D. (2002b). Raf and akt mediate distinct
grade signaling. Cell 118, 243–255. aspects of sensory axon growth. Neuron 35, 65–76.

Marmigère, F., Montelius, A., Wegner, M., Groner, Y., Reichardt, L.F., and
Lai, T., Jabaudon, D., Molyneaux, B.J., Azim, E., Arlotta, P., Menezes, J.R.,
Ernfors, P. (2006). The Runx1/AML1 transcription factor selectively regulates
and Macklis, J.D. (2008). SOX5 controls the sequential generation of distinct
development and survival of TrkA nociceptive sensory neurons. Nat. Neurosci.
corticofugal neuron subtypes. Neuron 57, 232–247.
9, 180–187.
Lasorella, A., Stegmüller, J., Guardavaccaro, D., Liu, G., Carro, M.S.,
McAllister, A.K. (2002). Neurotrophins and cortical development. Results
Rothschild, G., de la Torre-Ubieta, L., Pagano, M., Bonni, A., and Iavarone,
Probl. Cell Differ. 39, 89–112.
A. (2006). Degradation of Id2 by the anaphase-promoting complex couples
cell cycle exit and axonal growth. Nature 442, 471–474.
McAllister, A.K., Lo, D.C., and Katz, L.C. (1995). Neurotrophins regulate
dendritic growth in developing visual cortex. Neuron 15, 791–803.
Lee, S., Lee, B., Lee, J.W., and Lee, S.K. (2009). Retinoid signaling and neuro-
genin2 function are coupled for the specification of spinal motor neurons McAllister, A.K., Katz, L.C., and Lo, D.C. (1997). Opposing roles for endoge-
through a chromatin modifier CBP. Neuron 62, 641–654. nous BDNF and NT-3 in regulating cortical dendritic growth. Neuron 18,
767–778.
Lehtinen, M.K., Yuan, Z., Boag, P.R., Yang, Y., Villén, J., Becker, E.B.,
DiBacco, S., de la Iglesia, N., Gygi, S., Blackwell, T.K., and Bonni, A. (2006). McKenna, W.L., Betancourt, J., Larkin, K.A., Abrams, B., Guo, C., Rubenstein,
A conserved MST-FOXO signaling pathway mediates oxidative-stress J.L., and Chen, B. (2011). Tbr1 and Fezf2 regulate alternate corticofugal
responses and extends life span. Cell 125, 987–1001. neuronal identities during neocortical development. J. Neurosci. 31, 549–564.
Lentz, S.I., Knudson, C.M., Korsmeyer, S.J., and Snider, W.D. (1999). Neuro- McManus, M.F., and Golden, J.A. (2005). Neuronal migration in developmental
trophins support the development of diverse sensory axon morphologies. disorders. J. Child Neurol. 20, 280–286.
J. Neurosci. 19, 1038–1048.
Méchaly, I., Bourane, S., Piquemal, D., Al-Jumaily, M., Ventéo, S., Puech, S.,
Lessard, J., Wu, J.I., Ranish, J.A., Wan, M., Winslow, M.M., Staahl, B.T., Wu, Scamps, F., Valmier, J., and Carroll, P. (2006). Gene profiling during develop-
H., Aebersold, R., Graef, I.A., and Crabtree, G.R. (2007). An essential switch ment and after a peripheral nerve traumatism reveals genes specifically
in subunit composition of a chromatin remodeling complex during neural induced by injury in dorsal root ganglia. Mol. Cell. Neurosci. 32, 217–229.
development. Neuron 55, 201–215.
Ménager, C., Arimura, N., Fukata, Y., and Kaibuchi, K. (2004). PIP3 is involved
Li, S., Zhang, C., Takemori, H., Zhou, Y., and Xiong, Z.Q. (2009). TORC1 regu- in neuronal polarization and axon formation. J. Neurochem. 89, 109–118.
lates activity-dependent CREB-target gene transcription and dendritic growth
of developing cortical neurons. J. Neurosci. 29, 2334–2343. Mohideen, F., Capili, A.D., Bilimoria, P.M., Yamada, T., Bonni, A., and Lima,
C.D. (2009). A molecular basis for phosphorylation-dependent SUMO conju-
Li, N., Zhao, C.T., Wang, Y., and Yuan, X.B. (2010a). The transcription factor gation by the E2 UBC9. Nat. Struct. Mol. Biol. 16, 945–952.
Cux1 regulates dendritic morphology of cortical pyramidal neurons. PLoS
ONE 5, e10596. 10.1371/journal.pone.0010596. Molyneaux, B.J., Arlotta, P., Hirata, T., Hibi, M., and Macklis, J.D. (2005). Fezl
is required for the birth and specification of corticospinal motor neurons.
Li, S., Overman, J.J., Katsman, D., Kozlov, S.V., Donnelly, C.J., Twiss, J.L., Neuron 47, 817–831.
Giger, R.J., Coppola, G., Geschwind, D.H., and Carmichael, S.T. (2010b). An
age-related sprouting transcriptome provides molecular control of axonal Molyneaux, B.J., Arlotta, P., Menezes, J.R., and Macklis, J.D. (2007). Neuronal
sprouting after stroke. Nat. Neurosci. 13, 1496–1504. subtype specification in the cerebral cortex. Nat. Rev. Neurosci. 8, 427–437.

Lin, D., Edwards, A.S., Fawcett, J.P., Mbamalu, G., Scott, J.D., and Pawson, T. Molyneaux, B.J., Arlotta, P., Fame, R.M., MacDonald, J.L., MacQuarrie, K.L.,
(2000). A mammalian PAR-3-PAR-6 complex implicated in Cdc42/Rac1 and and Macklis, J.D. (2009). Novel subtype-specific genes identify distinct
aPKC signalling and cell polarity. Nat. Cell Biol. 2, 540–547. subpopulations of callosal projection neurons. J. Neurosci. 29, 12343–12354.

Neuron 72, October 6, 2011 ª2011 Elsevier Inc. 37


Neuron

Review
Moore, D.L., Blackmore, M.G., Hu, Y., Kaestner, K.H., Bixby, J.L., Lemmon, Penzes, P., Cahill, M.E., Jones, K.A., VanLeeuwen, J.E., and Woolfrey, K.M.
V.P., and Goldberg, J.L. (2009). KLF family members regulate intrinsic axon (2011). Dendritic spine pathology in neuropsychiatric disorders. Nat. Neurosci.
regeneration ability. Science 326, 298–301. 14, 285–293.

Mui, S.H., Hindges, R., O’Leary, D.D., Lemke, G., and Bertuzzi, S. (2002). The Pfeiffer, B.E., Zang, T., Wilkerson, J.R., Taniguchi, M., Maksimova, M.A.,
homeodomain protein Vax2 patterns the dorsoventral and nasotemporal axes Smith, L.N., Cowan, C.W., and Huber, K.M. (2010). Fragile X mental retardation
of the eye. Development 129, 797–804. protein is required for synapse elimination by the activity-dependent transcrip-
tion factor MEF2. Neuron 66, 191–197.
Nakada, C., Ritchie, K., Oba, Y., Nakamura, M., Hotta, Y., Iino, R., Kasai, R.S.,
Yamaguchi, K., Fujiwara, T., and Kusumi, A. (2003). Accumulation of anchored Polleux, F., Giger, R.J., Ginty, D.D., Kolodkin, A.L., and Ghosh, A. (1998).
proteins forms membrane diffusion barriers during neuronal polarization. Nat. Patterning of cortical efferent projections by semaphorin-neuropilin interac-
Cell Biol. 5, 626–632. tions. Science 282, 1904–1906.

Nakae, J., Barr, V., and Accili, D. (2000). Differential regulation of gene expres- Polleux, F., Ince-Dunn, G., and Ghosh, A. (2007). Transcriptional regulation
sion by insulin and IGF-1 receptors correlates with phosphorylation of a single of vertebrate axon guidance and synapse formation. Nat. Rev. Neurosci. 8,
amino acid residue in the forkhead transcription factor FKHR. EMBO J. 19, 331–340.
989–996.
Polleux, F., Morrow, T., and Ghosh, A. (2000). Semaphorin 3A is a chemoat-
Nakae, J., Biggs, W.H., 3rd, Kitamura, T., Cavenee, W.K., Wright, C.V., Arden, tractant for cortical apical dendrites. Nature 404, 567–573.
K.C., and Accili, D. (2002). Regulation of insulin action and pancreatic beta-cell
function by mutated alleles of the gene encoding forkhead transcription factor Polter, A., Yang, S., Zmijewska, A.A., van Groen, T., Paik, J.H., Depinho, R.A.,
Foxo1. Nat. Genet. 32, 245–253. Peng, S.L., Jope, R.S., and Li, X. (2009). Forkhead box, class O transcription
factors in brain: regulation and behavioral manifestation. Biol. Psychiatry 65,
Nan, X., Hou, J., Maclean, A., Nasir, J., Lafuente, M.J., Shu, X., Kriaucionis, S., 150–159.
and Bird, A. (2007). Interaction between chromatin proteins MECP2 and ATRX
is disrupted by mutations that cause inherited mental retardation. Proc. Natl. Pot, I., and Bonni, S. (2008). SnoN in TGF-beta signaling and cancer biology.
Acad. Sci. USA 104, 2709–2714. Curr. Mol. Med. 8, 319–328.

Nedivi, E., Wu, G.Y., and Cline, H.T. (1998). Promotion of dendritic growth by Pot, I., Ikeuchi, Y., Bonni, A., and Bonni, S. (2010). SnoN: bridging neurobi-
CPG15, an activity-induced signaling molecule. Science 281, 1863–1866. ology and cancer biology. Curr. Mol. Med. 10, 667–673.

Neumann, S., and Woolf, C.J. (1999). Regeneration of dorsal column fibers into Powell, S.K., Rivas, R.J., Rodriguez-Boulan, E., and Hatten, M.E. (1997).
and beyond the lesion site following adult spinal cord injury. Neuron 23, 83–91. Development of polarity in cerebellar granule neurons. J. Neurobiol. 32,
223–236.
Nishiyama, M., Togashi, K., von Schimmelmann, M.J., Lim, C.S., Maeda, S.,
Yamashita, N., Goshima, Y., Ishii, S., and Hong, K. (2011). Semaphorin 3A Pulipparacharuvil, S., Renthal, W., Hale, C.F., Taniguchi, M., Xiao, G., Kumar,
induces CaV2.3 channel-dependent conversion of axons to dendrites. Nat. A., Russo, S.J., Sikder, D., Dewey, C.M., Davis, M.M., et al. (2008). Cocaine
Cell Biol. 13, 676–685. regulates MEF2 to control synaptic and behavioral plasticity. Neuron 59,
621–633.
Noctor, S.C., Martı́nez-Cerdeño, V., Ivic, L., and Kriegstein, A.R. (2004).
Cortical neurons arise in symmetric and asymmetric division zones and Qin, Q., Baudry, M., Liao, G., Noniyev, A., Galeano, J., and Bi, X. (2009).
migrate through specific phases. Nat. Neurosci. 7, 136–144. A novel function for p53: regulation of growth cone motility through interaction
with Rho kinase. J. Neurosci. 29, 5183–5192.
O’Donnell, M., Chance, R.K., and Bashaw, G.J. (2009). Axon growth and guid-
ance: receptor regulation and signal transduction. Annu. Rev. Neurosci. 32, Qin, Q., Liao, G., Baudry, M., and Bi, X. (2010). Role of calpain-mediated p53
383–412. truncation in semaphorin 3A-induced axonal growth regulation. Proc. Natl.
Acad. Sci. USA 107, 13883–13887.
Okazawa, M., Abe, H., Katsukawa, M., Iijima, K., Kiwada, T., and Nakanishi, S.
(2009). Role of calcineurin signaling in membrane potential-regulated matura- Qiu, J., Cafferty, W.B., McMahon, S.B., and Thompson, S.W. (2005). Condi-
tion of cerebellar granule cells. J. Neurosci. 29, 2938–2947. tioning injury-induced spinal axon regeneration requires signal transducer
and activator of transcription 3 activation. J. Neurosci. 25, 1645–1653.
Oliva, A.A., Jr., Atkins, C.M., Copenagle, L., and Banker, G.A. (2006). Activated
c-Jun N-terminal kinase is required for axon formation. J. Neurosci. 26, 9462– Raivich, G., Bohatschek, M., Da Costa, C., Iwata, O., Galiano, M., Hristova, M.,
9470. Nateri, A.S., Makwana, M., Riera-Sans, L., Wolfer, D.P., et al. (2004). The AP-1
transcription factor c-Jun is required for efficient axonal regeneration. Neuron
Orr, H.T. (2010). Nuclear ataxias. Cold Spring Harb. Perspect. Biol. 2, a000786. 43, 57–67.

Paik, J.H., Kollipara, R., Chu, G., Ji, H., Xiao, Y., Ding, Z., Miao, L., Tothova, Z., Ramón y Cajal, S. (1995). Histology of the Nervous System of Man and
Horner, J.W., Carrasco, D.R., et al. (2007). FoxOs are lineage-restricted redun- Vertebrates (New York: Oxford University Press).
dant tumor suppressors and regulate endothelial cell homeostasis. Cell 128,
309–323. Ramos, B., Gaudillière, B., Bonni, A., and Gill, G. (2007). Transcription factor
Sp4 regulates dendritic patterning during cerebellar maturation. Proc. Natl.
Palay, S.L., and Chan-Palay, V. (1974). Cerebellar Cortex: Cytology and Acad. Sci. USA 104, 9882–9887.
Organization (Berlin, Heidelberg, New York: Springer).
Ramos, B., Valı́n, A., Sun, X., and Gill, G. (2009). Sp4-dependent repression of
Patel, T.D., Jackman, A., Rice, F.L., Kucera, J., and Snider, W.D. (2000). neurotrophin-3 limits dendritic branching. Mol. Cell. Neurosci. 42, 152–159.
Development of sensory neurons in the absence of NGF/TrkA signaling in vivo.
Neuron 25, 345–357. Redmond, L., Kashani, A.H., and Ghosh, A. (2002). Calcium regulation of
dendritic growth via CaM kinase IV and CREB-mediated transcription. Neuron
Patel, T.D., Kramer, I., Kucera, J., Niederkofler, V., Jessell, T.M., Arber, S., and 34, 999–1010.
Snider, W.D. (2003). Peripheral NT3 signaling is required for ETS protein
expression and central patterning of proprioceptive sensory afferents. Neuron Renault, V.M., Rafalski, V.A., Morgan, A.A., Salih, D.A., Brett, J.O., Webb, A.E.,
38, 403–416. Villeda, S.A., Thekkat, P.U., Guillerey, C., Denko, N.C., et al. (2009). FoxO3
regulates neural stem cell homeostasis. Cell Stem Cell 5, 527–539.
Pearson-White, S., and Crittenden, R. (1997). Proto-oncogene Sno expres-
sion, alternative isoforms and immediate early serum response. Nucleic Acids Riccio, A., Ahn, S., Davenport, C.M., Blendy, J.A., and Ginty, D.D. (1999).
Res. 25, 2930–2937. Mediation by a CREB family transcription factor of NGF-dependent survival
of sympathetic neurons. Science 286, 2358–2361.
Pelzer, T., Lyons, G.E., Kim, S., and Moreadith, R.W. (1996). Cloning and
characterization of the murine homolog of the sno proto-oncogene reveals Richardson, P.M., and Issa, V.M. (1984). Peripheral injury enhances central
a novel splice variant. Dev. Dyn. 205, 114–125. regeneration of primary sensory neurones. Nature 309, 791–793.

38 Neuron 72, October 6, 2011 ª2011 Elsevier Inc.


Neuron

Review
Sapir, T., Sapoznik, S., Levy, T., Finkelshtein, D., Shmueli, A., Timm, T., Spruston, N. (2008). Pyramidal neurons: dendritic structure and synaptic inte-
Mandelkow, E.M., and Reiner, O. (2008). Accurate balance of the polarity gration. Nat. Rev. Neurosci. 9, 206–221.
kinase MARK2/Par-1 is required for proper cortical neuronal migration.
J. Neurosci. 28, 5710–5720. Stegmüller, J., Konishi, Y., Huynh, M.A., Yuan, Z., Dibacco, S., and Bonni, A.
(2006). Cell-intrinsic regulation of axonal morphogenesis by the Cdh1-APC
Sato, M., Suzuki, K., Yamazaki, H., and Nakanishi, S. (2005). A pivotal role of target SnoN. Neuron 50, 389–400.
calcineurin signaling in development and maturation of postnatal cerebellar
granule cells. Proc. Natl. Acad. Sci. USA 102, 5874–5879. Stegmüller, J., Huynh, M.A., Yuan, Z., Konishi, Y., and Bonni, A. (2008).
TGFbeta-Smad2 signaling regulates the Cdh1-APC/SnoN pathway of axonal
Schulte, D., Furukawa, T., Peters, M.A., Kozak, C.A., and Cepko, C.L. (1999). morphogenesis. J. Neurosci. 28, 1961–1969.
Misexpression of the Emx-related homeobox genes cVax and mVax2 ventral-
izes the retina and perturbs the retinotectal map. Neuron 24, 541–553. Tabata, H., and Nakajima, K. (2003). Multipolar migration: the third mode of
radial neuronal migration in the developing cerebral cortex. J. Neurosci. 23,
Schwab, M.E. (2004). Nogo and axon regeneration. Curr. Opin. Neurobiol. 14, 9996–10001.
118–124.
Takahashi, D., Yu, W., Baas, P.W., Kawai-Hirai, R., and Hayashi, K. (2007).
Schwab, M.E., and Bartholdi, D. (1996). Degeneration and regeneration of Rearrangement of microtubule polarity orientation during conversion of
axons in the lesioned spinal cord. Physiol. Rev. 76, 319–370. dendrites to axons in cultured pyramidal neurons. Cell Motil. Cytoskeleton
64, 347–359.
Schwartzkroin, P.A., and Walsh, C.A. (2000). Cortical malformations and
epilepsy. Ment. Retard. Dev. Disabil. Res. Rev. 6, 268–280. Tao, X., Finkbeiner, S., Arnold, D.B., Shaywitz, A.J., and Greenberg, M.E.
(1998). Ca2+ influx regulates BDNF transcription by a CREB family transcrip-
Segal, R.A., and Greenberg, M.E. (1996). Intracellular signaling pathways tion factor-dependent mechanism. Neuron 20, 709–726.
activated by neurotrophic factors. Annu. Rev. Neurosci. 19, 463–489.
Tedeschi, A., Nguyen, T., Puttagunta, R., Gaub, P., and Di Giovanni, S. (2009a).
Seijffers, R., Mills, C.D., and Woolf, C.J. (2007). ATF3 increases the intrinsic A p53-CBP/p300 transcription module is required for GAP-43 expression,
growth state of DRG neurons to enhance peripheral nerve regeneration. axon outgrowth, and regeneration. Cell Death Differ. 16, 543–554.
J. Neurosci. 27, 7911–7920.
Tedeschi, A., Nguyen, T., Steele, S.U., Feil, S., Naumann, U., Feil, R., and
Shalizi, A., Gaudillière, B., Yuan, Z., Stegmüller, J., Shirogane, T., Ge, Q., Tan, Di Giovanni, S. (2009b). The tumor suppressor p53 transcriptionally regulates
Y., Schulman, B., Harper, J.W., and Bonni, A. (2006). A calcium-regulated cGKI expression during neuronal maturation and is required for cGMP-depen-
MEF2 sumoylation switch controls postsynaptic differentiation. Science 311, dent growth cone collapse. J. Neurosci. 29, 15155–15160.
1012–1017.
Tessier-Lavigne, M., and Goodman, C.S. (1996). The molecular biology of
Shalizi, A., Bilimoria, P.M., Stegmüller, J., Gaudillière, B., Yang, Y., Shuai, K.,
axon guidance. Science 274, 1123–1133.
and Bonni, A. (2007). PIASx is a MEF2 SUMO E3 ligase that promotes postsyn-
aptic dendritic morphogenesis. J. Neurosci. 27, 10037–10046.
Tothova, Z., Kollipara, R., Huntly, B.J., Lee, B.H., Castrillon, D.H., Cullen, D.E.,
McDowell, E.P., Lazo-Kallanian, S., Williams, I.R., Sears, C., et al. (2007).
Shaywitz, A.J., and Greenberg, M.E. (1999). CREB: a stimulus-induced tran-
FoxOs are critical mediators of hematopoietic stem cell resistance to physio-
scription factor activated by a diverse array of extracellular signals. Annu.
logic oxidative stress. Cell 128, 325–339.
Rev. Biochem. 68, 821–861.
Tsujino, H., Kondo, E., Fukuoka, T., Dai, Y., Tokunaga, A., Miki, K., Yonenobu,
Shelly, M., Cancedda, L., Heilshorn, S., Sumbre, G., and Poo, M.M. (2007).
LKB1/STRAD promotes axon initiation during neuronal polarization. Cell K., Ochi, T., and Noguchi, K. (2000). Activating transcription factor 3 (ATF3)
induction by axotomy in sensory and motoneurons: A novel neuronal marker
129, 565–577.
of nerve injury. Mol. Cell. Neurosci. 15, 170–182.
Shelly, M., Cancedda, L., Lim, B.K., Popescu, A.T., Cheng, P.L., Gao, H., and
Poo, M.M. (2011). Semaphorin3a regulates neuronal polarization by suppress- Tucker, K.L., Meyer, M., and Barde, Y.A. (2001). Neurotrophins are required for
ing axon formation and promoting dendrite growth. Neuron 71, 433–446. nerve growth during development. Nat. Neurosci. 4, 29–37.

Shi, S.H., Jan, L.Y., and Jan, Y.N. (2003). Hippocampal neuronal polarity Wayman, G.A., Impey, S., Marks, D., Saneyoshi, T., Grant, W.F., Derkach, V.,
specified by spatially localized mPar3/mPar6 and PI 3-kinase activity. Cell and Soderling, T.R. (2006). Activity-dependent dendritic arborization mediated
112, 63–75. by CaM-kinase I activation and enhanced CREB-dependent transcription of
Wnt-2. Neuron 50, 897–909.
Shi, S.H., Cheng, T., Jan, L.Y., and Jan, Y.N. (2004). APC and GSK-3beta are
involved in mPar3 targeting to the nascent axon and establishment of neuronal Wayman, G.A., Davare, M., Ando, H., Fortin, D., Varlamova, O., Cheng, H.Y.,
polarity. Curr. Biol. 14, 2025–2032. Marks, D., Obrietan, K., Soderling, T.R., Goodman, R.H., and Impey, S.
(2008a). An activity-regulated microRNA controls dendritic plasticity by
Shinoda, T., Taya, S., Tsuboi, D., Hikita, T., Matsuzawa, R., Kuroda, S., Iwa- down-regulating p250GAP. Proc. Natl. Acad. Sci. USA 105, 9093–9098.
matsu, A., and Kaibuchi, K. (2007). DISC1 regulates neurotrophin-induced
axon elongation via interaction with Grb2. J. Neurosci. 27, 4–14. Wayman, G.A., Lee, Y.S., Tokumitsu, H., Silva, A.J., and Soderling, T.R.
(2008b). Calmodulin-kinases: modulators of neuronal development and plas-
Shioda, N., Beppu, H., Fukuda, T., Li, E., Kitajima, I., and Fukunaga, K. (2011). ticity. Neuron 59, 914–931.
Aberrant calcium/calmodulin-dependent protein kinase II (CaMKII) activity is
associated with abnormal dendritic spine morphology in the ATRX mutant Williams, S.E., Mann, F., Erskine, L., Sakurai, T., Wei, S., Rossi, D.J., Gale, N.W.,
mouse brain. J. Neurosci. 31, 346–358. Holt, C.E., Mason, C.A., and Henkemeyer, M. (2003). Ephrin-B2 and EphB1
mediate retinal axon divergence at the optic chiasm. Neuron 39, 919–935.
Sisodiya, S.M. (2004). Malformations of cortical development: burdens and
insights from important causes of human epilepsy. Lancet Neurol. 3, 29–38. Winckler, B., Forscher, P., and Mellman, I. (1999). A diffusion barrier maintains
distribution of membrane proteins in polarized neurons. Nature 397, 698–701.
Smith, D.S., and Skene, J.H. (1997). A transcription-dependent switch controls
competence of adult neurons for distinct modes of axon growth. J. Neurosci. Wu, J.I., Lessard, J., Olave, I.A., Qiu, Z., Ghosh, A., Graef, I.A., and Crabtree,
17, 646–658. G.R. (2007). Regulation of dendritic development by neuron-specific chro-
matin remodeling complexes. Neuron 56, 94–108.
Solecki, D.J., Model, L., Gaetz, J., Kapoor, T.M., and Hatten, M.E. (2004).
Par6alpha signaling controls glial-guided neuronal migration. Nat. Neurosci. Yang, Y., Kim, A.H., Yamada, T., Wu, B., Bilimoria, P.M., Ikeuchi, Y., de la Igle-
7, 1195–1203. sia, N., Shen, J., and Bonni, A. (2009). A Cdc20-APC ubiquitin signaling
pathway regulates presynaptic differentiation. Science 326, 575–578.
Sosa, L., Dupraz, S., Laurino, L., Bollati, F., Bisbal, M., Cáceres, A., Pfenninger,
K.H., and Quiroga, S. (2006). IGF-1 receptor is essential for the establishment Yi, J.J., Barnes, A.P., Hand, R., Polleux, F., and Ehlers, M.D. (2010). TGF-beta
of hippocampal neuronal polarity. Nat. Neurosci. 9, 993–995. signaling specifies axons during brain development. Cell 142, 144–157.

Neuron 72, October 6, 2011 ª2011 Elsevier Inc. 39


Neuron

Review
Yin, D.M., Huang, Y.H., Zhu, Y.B., and Wang, Y. (2008). Both the establishment Zhou, F.Q., and Snider, W.D. (2006). Intracellular control of developmental
and maintenance of neuronal polarity require the activity of protein kinase D in and regenerative axon growth. Philos. Trans. R. Soc. Lond. B Biol. Sci. 361,
the Golgi apparatus. J. Neurosci. 28, 8832–8843. 1575–1592.

Yoshimura, T., Arimura, N., Kawano, Y., Kawabata, S., Wang, S., and Zhou, Z., Hong, E.J., Cohen, S., Zhao, W.N., Ho, H.Y., Schmidt, L., Chen,
Kaibuchi, K. (2006). Ras regulates neuronal polarity via the PI3-kinase/Akt/ W.G., Lin, Y., Savner, E., Griffith, E.C., et al. (2006). Brain-specific phosphor-
GSK-3beta/CRMP-2 pathway. Biochem. Biophys. Res. Commun. 340, ylation of MeCP2 regulates activity-dependent Bdnf transcription, dendritic
62–68. growth, and spine maturation. Neuron 52, 255–269.

Zheng, W.H., Kar, S., and Quirion, R. (2002). FKHRL1 and its homologs are Zou, H., Ho, C., Wong, K., and Tessier-Lavigne, M. (2009). Axotomy-induced
new targets of nerve growth factor Trk receptor signaling. J. Neurochem. 80, Smad1 activation promotes axonal growth in adult sensory neurons. J. Neuro-
1049–1061. sci. 29, 7116–7123.

40 Neuron 72, October 6, 2011 ª2011 Elsevier Inc.

Das könnte Ihnen auch gefallen