ArticlePDF AvailableLiterature Review

The Involvement of Neuron-Specific Factors in Dendritic Spinogenesis: Molecular Regulation and Association with Neurological Disorders

Wiley
Neural Plasticity
Authors:

Abstract and Figures

Dendritic spines are the location of excitatory synapses in the mammalian nervous system and are neuron-specific subcellular structures essential for neural circuitry and function. Dendritic spine morphology is determined by the F-actin cytoskeleton. F-actin remodeling must coordinate with different stages of dendritic spinogenesis, starting from dendritic filopodia formation to the filopodia-spines transition and dendritic spine maturation and maintenance. Hundreds of genes, including F-actin cytoskeleton regulators, membrane proteins, adaptor proteins, and signaling molecules, are known to be involved in regulating synapse formation. Many of these genes are not neuron-specific, but how they specifically control dendritic spine formation in neurons is an intriguing question. Here, we summarize how ubiquitously expressed genes, including syndecan-2, NF1 (encoding neurofibromin protein), VCP, and CASK, and the neuron-specific gene CTTNBP2 coordinate with neurotransmission, transsynaptic signaling, and cytoskeleton rearrangement to control dendritic filopodia formation, filopodia-spines transition, and dendritic spine maturation and maintenance. The aforementioned genes have been associated with neurological disorders, such as autism spectrum disorders (ASDs), mental retardation, learning difficulty, and frontotemporal dementia. We also summarize the corresponding disorders in this report.
This content is subject to copyright. Terms and conditions apply.
Review Article
The Involvement of Neuron-Specific Factors in
Dendritic Spinogenesis: Molecular Regulation and
Association with Neurological Disorders
Hsiao-Tang Hu, Pu-Yun Shih, Yu-Tzu Shih, and Yi-Ping Hsueh
Institute of Molecular Biology, Academia Sinica, Taipei 11529, Taiwan
Correspondence should be addressed to Yi-Ping Hsueh; yph@gate.sinica.edu.tw
Received  June ; Accepted  July 
AcademicEditor:DeepakP.Srivastava
Copyright ©  Hsiao-Tang Hu et al. is is an open access article distributed under the Creative Commons Attribution License,
which permits unrestricted use, distribution, and reproduction in any medium, provided the original work is properly cited.
Dendritic spines are the location of excitatory synapses in the mammalian nervous system and are neuron-specic subcellular
structures essential for neural circuitry and function. Dendritic spine morphology is determined by the F-actin cytoskeleton. F-
actin remodeling must coordinate with dierent stages of dendritic spinogenesis, starting from dendritic lopodia formation to the
lopodia-spines transition and dendritic spine maturation and maintenance. Hundreds of genes, including F-actin cytoskeleton
regulators, membrane proteins, adaptor proteins, and signaling molecules, are known to be involved in regulating synapse
formation. Many of these genes are not neuron-specic, but how they specically control dendritic spine formation in neurons is an
intriguing question. Here, we summarize how ubiquitously expressed genes, including syndecan-, NF (encoding neurobromin
protein), VCP, and CASK, and the neuron-specic gene CTTNBP coordinate with neurotransmission, transsynaptic signaling, and
cytoskeleton rearrangement to control dendritic lopodia formation, lopodia-spines transition, and dendritic spine maturation
and maintenance. e aforementioned genes have been associated with neurological disorders, such as autism spectrum disorders
(ASDs), mental retardation, learning diculty, and frontotemporal dementia. We also summarize the corresponding disorders in
this report.
1. Introduction
e tiny protrusions emerging from dendrites known as
dendritic spines are the primary subcellular locations of exci-
tatory synapses in the mammalian central nervous system [].
Dendritic spines are typically - 𝜇m in length and .– 𝜇m
in width of the spine head, with diverse morphologies, such as
mushroomlike, stubby, and thin spines. ese structures are
mainly supported by the F-actin cytoskeleton. us, F-actin
cytoskeletal proteins and regulators are important factors
for generating dendritic spines. Many membrane proteins
and adaptor and signaling molecules are also involved in
controlling dendritic spine formation and maintenance [].
Several mechanisms have been described to form dendritic
spines []. e most popular mechanism is that dendritic
lopodia serve as precursors for dendritic spine formation.
Interestingly, lopodia are ubiquitously found on various cell
types. In contrast, dendritic spines are neuron-specic struc-
tures. us, the transition from lopodia to spines should
be controlled by neuron-specic factors.
Neuron-specic factors controlling dendritic spinogene-
sisfallintotwocategories.erstgroupisproteinsspecif-
ically expressed in neurons. e second group is neuron-
specic cellular responses or processes. ese proteins or
responses directly or indirectly regulate F-actin rearrange-
ment and dynamics to promote dendritic spine formation.
Studies of cytoskeleton-associated cortactin-binding protein
 (CTTNBP) and heparan sulfate transmembrane prot-
eoglycan (HSPG) syndecan- serve as examples for these two
categories, respectively. CTTNBP is a neuron-specic cyto-
skeleton-associated protein and that is enriched at dendritic
spines of mature neurons. Although syndecan- is widely
expressed in many cell types, it is highly concentrated at
Hindawi Publishing Corporation
Neural Plasticity
Volume 2016, Article ID 5136286, 10 pages
http://dx.doi.org/10.1155/2016/5136286
Neural Plasticity
CytoTMEctodomain CN
a.a. sequence
C1 C2
V
SDC4
RMKKKDEGSY
SDC1
SDC2
SDC3
RMKKKDEGSY
RMKKKDEGSY
RMKKKDEGSY
EFYA
EFYA
EFYA
EFYA
Neurobromin
Dendritic
lopodia formation
Dendritic
spine formation
CASK
DLGERK - P S SAAYQKAPTK - -
DLG-KK-P-I--YKKAPT--N
SLEEPKQANGGAYQK-PTKQE
TLEEPKQA- SVTYQK - PDKQE
F : Schematic structure and amino acid sequences of syndecans. C, conserved domain ; C, conserved domain ; Cyto, cytoplasmic
domain; SDC, syndecan-; SDC, syndecan-; SDC, syndecan-; SDC, syndecan-; TM, transmembrane; V, variable region.
synapses in neurons. Syndecan- cooperates with other pro-
teins to trigger neurotransmission through a neuron-specic
signal to induce dendritic spine formation. Genomic analyses
of patients with autism spectrum disorders (ASDs) indicated
that both CTTNBP and syndecan- were associated with
ASDs [, ]. Additionally, neurobromin, CASK, and VCP
coordinate with syndecan- to control dendritic spinogenesis
and were also associated with neurological disorders. ese
ndings suggest that these genes are critical for neuronal
function, likely through their regulation of dendritic spine
formation. In this review, we will summarize the functions
of these proteins in dendritic spinogenesis and use these pro-
teins as examples to discuss how neuron-specic molecules
coordinate with ubiquitously expressed proteins to generate
neuron-specic signals for dendritic spine formation.
2. The HSPG Syndecan-2 Triggers
Dendritic Spine Formation
2.1. Syndecan-2 Is Enriched at Dendritic Spines and Is Required
for Dendritic Spine Formation. Syndecan- is a type I mem-
brane protein with a heparan sulfate modication at its
ectodomain (Figure ) []. In mammals, the syndecan pro-
tein family contains four members, syndecan-, syndecan-,
syndecan-, and syndecan- []. In rodent brains, syndecan-
 and syndecan- are the two major syndecans expressed in
neurons with dierential distribution; syndecan- is highly
concentrated at synapses, while syndecan- is distributed
along the axonal sha []. Syndecan- is involved in cell-
cell and cell-matrix interactions through its heparan sul-
fate modication. It can also bind growth factors, such as
broblast growth factor (FGF) and epidermal cell growth
factor, and it acts as a coreceptor for these growth factors [].
Syndecan- is broadly and dynamically expressed in several
tissues and cell types [, ]. During neural development,
its expression gradually increases concurrent with synapse
formation [, ]. In mature neurons, such as cultured rat
hippocampal neurons at  days aer plating in vitro (DIV)
or later, syndecan- is highly enriched at dendritic spines
[, ]. More importantly, overexpression of syndecan- in
immature rat hippocampal cultured neurons, such as - DIV,
when endogenous syndecan- is not yet expressed, dendritic
lopodia are massively induced at - DIV and dendritic
lopodia are then transformed to dendritic spines at - DIV
[, ]. ose dendritic spines are expected to be functional,
as they are adjacent to the presynaptic marker synaptophysin
based on confocal microscopy [, ]. Syndecan--induced
dendritic spinogenesis serves as a model to explore the
mechanisms underlying the initiation of dendritic spinogen-
esis (namely, dendritic lopodia formation), the transition
from lopodia to spines, and dendritic spine maturation and
maintenance.
2.2. e C1 and C2 Motifs of Syndecan-2 Work Sequen-
tially to Promote Dendritic Spinogenesis. e ectodomain
of syndecan- heparan sulfate modication is involved in
cell-cell and cell-matrix interactions []. Its transmembrane
domain is required for homodimerization or oligomerization
[], which is critical for the protein-protein interactions
of syndecan- []. e cytoplasmic domain of syndecan-
contains only  amino acid residues (Figure ). Although it
is short, it is divided into three motifs, conserved domain 
(C), the variable region (V), and conserved domain  (C).
e C and C motifs are conserved among dierent synde-
cans, while the sequences of the V regions vary (Figure ).
e C motif is essential for syndecan--induced dendritic
lopodia formation of rat hippocampal cultured neurons,
as the syndecan-ΔC mutant completely loses the ability
to promote lopodia formation and spine formation at  as
well as  DIV [, ]. e C is required for the dendritic
lopodia-spines transition and dendritic spine maintenance
[, ]. Expression of the C deletion mutant syndecan-
ΔC at  DIV promotes dendritic lopodia formation at 
DIV. However, those lopodia are unable to transform into
dendritic spine at  DIV [, , ]. ese analyses indicate
that the function of syndecan- in dendritic spinogenesis can
be separated into two sequential steps, namely, lopodia and
spine formation, which are controlled by two distinct motifs
in syndecan-.
Neural Plasticity
NF1 GRD
1 2818 aa
LRD CTD
Adenylate cyclase
Ras VCP
Jn Pn SDC 2
2260
11681530 1545195 0
(a)
Neurobromin
PKA p
Ena
VAS P
cAMP
VCP
Spine formation
Actin Filopodia formation
SDC2
(b)
F : Function of neurobromin in neurons. (a) Neurobromin-interacting proteins. e Jn and Pn fragments interact with syndecan-.
e leucine-rich domain (LRD) binds VCP. e GAP-related domain (GRD) downregulates the Ras pathway. Both GRD and the C-terminal
half of neurobromin are involved in adenylate cyclase activity regulation. CTD, C-terminal region. (b) Neurobromin controls dendritic
lopodia and spine formation through the PKA-Ena/VASP and VCP pathways, respectively.
Because both C and C motifs are short and lack rec-
ognizable enzymatic domains, syndecan- binding partners
have been identied to determine its molecular mechanism
underlying dendritic spine formation. Several direct binding
partners (summarized in Table ) have been identied for
the C domains of syndecan-, including neurobromin []
and ezrin []. e C motif directly interacts with syntenin
[], CASK [], and synbindin []. Among these, the inter-
actions between syndecan- and neurobromin and CASK
have been shown to be relevant in dendritic spine formation.
Because the cytoplasmic tail of syndecan- is very short, it
isunlikelythatasinglesyndecan-moleculecansimultane-
ously interact with all of its binding partners. Because the C
and C motifs are involved in two sequential processes, it
is likely that neurobromin and CASK sequentially interact
with syndecan-. Alternatively, it is possible that because
syndecan- forms at least a dimer through its transmem-
brane domain, dierent syndecan- molecules in dimers or
oligomers separately interact with neurobromin and CASK.
is would suggest that syndecan-, neurobromin, and
CASK form a single large complex. Further investigation,
including coimmunoprecipitation experiments, is required to
address this question.
2.3. Neurobromin Interacts with the C1 Motif of Syndecan-
2 and Promotes Syndecan-2-Induced Dendritic Filopodia For-
mation. Neurobromin encoded by the neurobromato-
sistypeI(NF1) gene is characterized by its RasGAP-
(Ras GTPase activating protein-) related domain (GRD)
T : SDC interacting proteins.
Binding site
in SDC
Binding site
for SDC Function
NF C LRD Filopodia formation
Ezrin C N-ter. Links to actin
cytoskeleton
Syntenin C PDZ Cell adhesion and
migration
CASK C PDZ Dendritic spine
formation
Synbindin C PDZ-like Vesicle transport
(Figure (a)) [–]. Similar to syndecan-, neurobromin is
widely expressed in dierent cell types, though its expression
levelismuchhigherinthenervoussystem[].NFisone
of the most common human inherited disorders featured
bychangesinskinpigmentation,benigntumorgrowth,and
learning diculty [, ]. Neurobromin suppresses tumor
growth through its ability to downregulate the RAS pathway
[]. In addition to its RAS activity, neurobromin can
increase cAMP concentration by activating adenylate cyclase
[]. Although the molecular mechanisms are less clear, the
GRD and C-terminal region of neurobromin are required
for cAMP pathway activation (Figure (a)) []. Both Gs-
dependent and Gs-independent pathways are involved in
neurobromin-regulated adenylate cyclase activation [].
ecAMPpathwayhasbeenshowntobeinvolvedinlearning
Neural Plasticity
and memory in Drosophila [] and dendritic spine formation
in the mammalian nervous system [].
In a yeast two-hybrid screen using dierent fragments of
neurobromin as baits, syndecan- was identied as a neu-
robromin binding partner []. Notably, neurobromin has
two independent interacting domains for the C motif of
syndecan-. One is the Jn fragment corresponding to amino
acid residues – in the GRD of human neurobromin;
the other is the Pn fragment containing amino acid residues
– (Figure (a)) []. e Jn and Pn compete for bind-
ing to the C motif of syndecan-. In addition to biochem-
ical studies demonstrating the direct interaction between
syndecan- and neurobromin, uorescence immunostain-
ing further demonstrated the colocalization of syndecan-
 and neurobromin at synapses in cultured hippocampal
neurons []. Moreover, both Nf knockdown and haploin-
suciency reduce the density of dendritic spines in both rat
hippocampal and mouse cortical cultured neurons and in
brains [, ], consistent with a function of neurobromin
in regulating dendritic spine formation.
e next question is how the syndecan--neurobromin
complex regulates dendritic spine formation. One study
examined syndecan- downstream signaling for triggering
lopodia formation. Using a panel of inhibitors to sup-
press various kinase activities, protein kinase A (PKA) was
identied to be required for syndecan--induced lopodia
formation []. Combined with the analysis using dierent
motif deletion mutants of syndecan-, we found that the C
motif of syndecan- is essential for PKA-dependent lopodia
formation []. Because neurobromin interacts with the
C motif and also activates the cAMP pathway, cultured
hippocampal neurons were then used to investigate whether
neurobromin mediates syndecan--induced lopodia for-
mation. Both Nf knockdown and Jn fragment expression,
which acts as a dominant-negative to disrupt the interac-
tion between endogenous neurobromin and syndecan-,
suppress syndecan--induced dendritic lopodia formation
of rat hippocampal cultured neurons at  DIV []. us,
neurobromin mediates the signal from syndecan- to the
cAMP pathway to initiate dendritic spinogenesis.
Because lopodia are supported by F-actin bundles, the
syndecan--neurobromin-cAMP pathway has to induce
F-actin polymerization and bundle formation to promote
dendritic lopodia formation. e Ena (Enabled)/VASP
(Vasodilator-Stimulated Phosphoprotein) protein family is
a group of F-actin regulators that initiate actin polymer-
ization and bundling []. PKA phosphorylation promotes
Ena/VASP protein activity to regulate the F-actin cytoskele-
ton []. Upon syndecan- overexpression, Ena/VASP phos-
phorylation increases, consistent with cAMP pathway acti-
vation. Moreover, disruption of Ena/VASP activity impairs
syndecan--induced dendritic lopodia formation []. In
summary, these studies indicate that syndecan- overexpres-
sion enhances the ability of neurobromin to activate the
PKA pathway, which then induces the Ena/VASP activity to
promote F-actin bundling and lopodia formation.
Although the PKA pathway is required for dendritic
lopodia formation, increased intracellular cAMP concen-
trations alone cannot induce dendritic lopodia formation
[], suggesting that other factor(s) are involved. From
an immunoprecipitation-mass spectrometry study, valosin-
containing protein (VCP, also known as P) was identied
as a neurobromin-binding protein []. e entire D and
D ATPase domains of VCP are required for the interac-
tion with the leucine-rich domain (LRD) of neurobromin
[]. VCP is a causative gene of inclusion body myopathy
associated with Paget’s disease of bone and frontotemporal
dementia (IBMPFD) []. IBMPFD patients frequently suer
from dementia. In addition, VCP mutations are associated
with ASDs and amyotrophic lateral sclerosis [, ]. ese
evidences suggest that VCP mutations impair brain function.
A combination of human genetic studies, mouse genetic
models, and cultured hippocampal and cortical neurons have
indicated that neurobromin interacts with VCP and guides
VCP to promote dendritic spinogenesis []. e roles of
VCP and neurobromin in dendritic spine formation may
account for the neural phenotypes in patients with mutations
in the NF and VCP genes. However, it is still unclear how
VCP regulates dendritic spine formation. To fully address the
molecular regulation of neurobromin and VCP in dendritic
spinogenesis, further studies are required.
e function of the syndecan--neurobromin interac-
tion in dendritic spine formation is summarized in
Figure (b).
2.4. CASK and Syndecan-2 Interactions Regulate Dendritic
Spine Maturation. CASK is a ubiquitously expressed gene
and is critical for brain development and function [].
MutationsinthehumanCASKgeneresultinX-linked
mental retardation and microcephaly with pontine and cere-
bellar hypoplasia [–]. CASK belongs to the membrane-
associated guanylate kinase (MAGUK) family and functions
as a scaold protein to interact with more than two dozen
cellular proteins []. It is widely distributed in neurons,
including synapses, dendrites, axons, and soma []. At
synapses, it localizes to both pre- and postsynaptic sites [].
In mouse pontine explants and rat hippocampal cultured
neurons, CASK knockdown impairs synapse formation at the
pre- and postsynapse, respectively [, ]. At presynaptic
sites, it binds the membrane protein neurexin and other
scaold proteins, such as Mint, mLin, and liprin, to control
presynaptic button formation [–]. CASK uses its PDZ
domain at the postsynaptic site to interact with the C
motif of syndecan- []. In cultured hippocampal neurons,
expression of the PDZ alone of CASK or the C-terminal
tail of syndecan- that disrupts the interaction between
endogenous CASK and syndecan- reduces dendritic spine
density, narrows spine heads, and shortens spine length at
 DIV, suggesting that the CASK-syndecan- interaction is
critical for dendritic spine formation [].
To investigate whether CASK is involved in dendritic
spinogenesis initiation or dendritic spine stabilization, a
time course study using a knockdown approach in cultured
hippocampal neurons has been performed []. e time
window of – DIV covering the initiation toward matura-
tion of dendritic spinogenesis was used for analysis. At  DIV,
Neural Plasticity
Filopodia-spines
transitionSpine maintenance
LIN7
NMDAR
SDC2
C2 C2
GSN
P4.1
SUMO
CASK
CASK
Ca2+
F : Syndecan- coordinates with calcium inux to control
dendritic spine formation and maturation. Syndecan- links the
CASK-mLIN-NMDAR complex through its C motif and directs
this complex to target to lopodial tips. It increases the accessibility
of postsynaptic lopodia to presynaptic stimulation, which is critical
for calcium inux to induce the lopodia-spines transition. In
addition to linking mLIN and NMDAR, CASK interacts with
the protein .-F-actin cytoskeleton. is interaction provides a
physical link between the membrane and cytoskeleton to stabilize
the dendritic spine structure. GSN, gelsolin.
wild-type dendritic spines are immature, long, and thin, and
they are present at a low density. As they mature at  DIV,
dendritic spine density increases, spine length decreases, and
spine width increases. Compared to control neurons, CASK
knockdown does not aect spine density, length, or size at
 DIV, suggesting that CASK is not critical for dendritic
spinogenesis initiation. At  DIV, CASK knockdown induces
dendritic spines withdraw and the spine heads fail to enlarge.
e spine density is decreased compared to control neurons
[]. e data indicate that CASK is important for dendritic
spine maturation, likely by linking the membrane protein
syndecan- to the F-actin cytoskeleton via protein . to
stabilize dendritic spines (Figure ) [].
2.5. Neurotransmission-Induced Calcium Inux Is Critical
for the Syndecan-2-Induced Filopodia-Spines Transition. In
human embryonic kidney HEK cells, syndecan- over-
expression induced numerous lopodia on the cell surface
[]. However, these lopodia cannot mature into spines.
Because neurobromin and CASK are also expressed in
HEK cells, the aforementioned studies cannot explain
why syndecan--induced dendritic spines are only present in
neurons. A neuron-specic factor must be present to control
dendritic spine formation. Because neurotransmission is a
neuron-specic event and because dendritic lopodia are
able to receive neurotransmission signals from presynaptic
buttons [], neurotransmission seems a likely factor that
triggers the lopodia-spines transformation in a neuron-
specic manner. Indeed, EGTA treatment to chelate extra-
cellular calcium or AP treatment to block NMDAR activity,
a major neurotransmitter gated calcium channel, impairs
the endogenous lopodia-spines transition at – DIV
and syndecan--induced lopodia-spines transition at –
DIV []. In syndecan--overexpressing neurons, intracellu-
lar calcium concentration is increased compared to control
neuronsatDIV.isincreaseisduetoNMDAR-regulated
calcium inux because AP treatment eectively reduced the
intracellular calcium concentration induced by syndecan-
[]. e C motif of syndecan- is required for syndecan-
overexpression-induced calcium inux [], suggesting that
the interaction with CASK is involved in calcium inux.
Previous studies have shown that CASK interacts with mLIN
via the L domains in both proteins [–] and that mLIN
interacts with the C-terminal tail of NMDAR subunit b
(NMDARb) through its PDZ domain []. us, the CASK-
mLIN complex links NMDAR to syndecan-. e interac-
tion between syndecan-, CASK, mLIN, and NMDARb
facilitates NMDAR localization to the tips of dendritic
lopodia, where NMDAR may be activated by presynaptic
stimulation, namely, glutamate, and induce calcium inux.
Disruption of the syndecan-, CASK, mLIN, and NMDAR
complex by overexpressing the interacting domains impairs
NMDAR lopodial distribution, calcium inux, and the
lopodia-spines transition [], suggesting that syndecan-
triggers calcium inux via the CASK-mLIN-NMDAR com-
plex and induces the lopodia-spines transition (Figure ).
e morphological feature of the lopodia-spines tran-
sitionisdendriticspineheadenlargementandspinelength
shortening. e F-actin cytoskeleton must be rearranged to
allow for this morphological change. Calcium is known to
regulate F-actin dynamics in dendritic spines [–], and
gelsolin is a calcium-activated F-actin regulator. It acts as
an F-actin severing and capping protein [–]. Gelsolin
deciency impairs lopodial retraction of developing neu-
rons [] and inhibits activity-dependent F-actin remodel-
ing in mature dendritic spines []. It is also critical for
the lopodia-spines transition induced by the syndecan-
-CASK-mLIN-NMDAR complex, as gelsolin knockdown
maintains syndecan--induced protrusions at the lopodial
stage []. It is possible that other calcium regulated F-actin
regulators also act downstream of syndecan- to control the
lopodia-spines transition. More investigations are required
to further elucidate the regulation.
2.6. Conclusion of the Role of Syndecan-2 Signaling in Den-
dritic Spine Formation. rough its interactions with intra-
cellular binding partners, the ubiquitously expressing protein
syndecan- modulates the F-actin cytoskeleton, triggers neu-
rotransmission, and promotes neuron-specic synapse for-
mation. From dendritic lopodia formation, lopodia-spines
transition to dendritic spine maturation, syndecan- interacts
with dierent binding partners to control F-actin behaviors.
Syndecan- rst activates the PKA pathway via neuro-
bromin to promote F-actin polymerization and bundling for
Neural Plasticity
dendritic lopodia formation []. It recruits NMDAR to
lopodial tips through its interaction with the CASK-mLIN
complex and increases the postsynaptic responsiveness to
presynaptic stimulation []. Calcium inux induces F-actin
cytoskeleton rearrangement to allow for the morphological
change from lopodia to spines []. To further promote
dendritic spine maturation and maintenance, syndecan-
binds to the protein . through interactions with CASK [].
roughout the entire process, neuron specicity falls within
NMDAR-mediated calcium inux, which induces F-actin
cytoskeleton remodeling to result in morphological changes
to the dendritic spine. ese studies provide a comprehensive
example of how a neuron-specic ion channel coordinates
with other adhesion molecules and synaptic proteins to
control dendritic spine formation.
3. The Neuron-Specific Cytoskeleton
Regulator CTTNBP2 Is Highly Associated
with Autism Spectrum Disorders
To identify a neuron-specic F-actin regulator involved in
dendritic spinogenesis, we searched the database and litera-
ture and focused on cortactin-binding protein  (CTTNBP).
CTTNBP gene encodes a brain-specic protein that inter-
acts with the SH domain of cortactin through its proline-
rich domain []. Cortactin promotes and stabilizes F-
actin branching [, ] and thus plays a critical role for
dendritic spine morphological maintenance []. Because
cortactin is a ubiquitously expressed protein, its function in
controlling dendritic spinogenesis must be regulated by a
neuron-specic factor. e specic expression of CTTNBP
in the brain makes it a good candidate to control cortactin
in dendritic spinogenesis. Furthermore, de novo mutations
in the CTTNBP gene have been repeatedly identied in
ASDpatients[,,].Inagenomicanalysiscovering
ASD patients, results indicated that CTTNBP is a high-
condence risk factor for ASDs with a false discovery rate less
than .% []. ese genetic data support a critical role for
CTTNBP in brain development and function.
3.1. CTTNBP2 Variant Transcripts and ASD Mutations. In the
expression tag sequence (EST) database (http://www.ncbi
.nlm.nih.gov/), three variants have been identied as
CTTNBP transcripts, namely,CTTNBP-Short (CT TNBP-
S), CTTNBP-Intron (CTTNBP-I), and CTTNBP-Long
(CTTNBP-L). Based on the nucleotide sequence, the rst
 predicted amino acid residues are shared among all
variants []. Using an antibody against the common region
of the CTTNBP variants, immunoblotting revealed that the
Short form of CTTNBP is the predominant protein product
in brains. e protein products of the Intron and Long forms
are undetectable in adult brains []. us, the following
studies of CTTNBP in neurons focused on CTTNBP-S.
It is still unclear whether the CTTNBP-I and CTTNBP-L
variants play any role in neurons. erefore, mutation
analysis of ASD patients is meaningful when the mutation
was located within the CTTNBP-S variant sequences.
Seven de novo ASD mutations in the CTTNBP gene have
been identied in the exons encoding CTTNBP-S []. To
further explore the association of CTTNBP with ASD,
these mutations should be investigated in detail to determine
their eects on CTTNBP molecular function and neuronal
morphogenesis.
Analysis of the amino acid sequence of CTTNBP-S
predicts a coiled-coil domain at the N-terminal region and
proline-rich domain at the C-terminus. e middle region
does not contain any recognizable protein structure []. e
N-terminal coiled-coil domain mediates CTTNBP-S
homooligomerization and heterooligomerization of
CTTNBP-S and the striatin family [, ]. e C-termi-
nal proline-rich domain interacts with cortactin []. e
middle region is required for the protein’s association with the
microtubule cytoskeleton []. e functions of these inter-
actions are discussed below (Figure ).
3.2. CTTNBP2-S Controls Cortactin Mobility and Regulates
Dendritic Spine Formation and Maintenance. CTTNBP-
S localizes to dendritic spines to control the cortactin-
F-actin cytoskeleton. Both endogenous CTTNBP-S and
overexpressed Myc-tagged CTTNBP-S were found to be
highly concentrated at dendritic spines in mature cultured
hippocampal neurons. Immunouorescence analysis of adult
brains also indicated that CTTNBP-S colocalized with F-
actin puncta in vivo, presumably to dendritic spines [].
CTTNBP-S is critical for dendritic spine formation, as
CTTNBP knockdown right before dendritic spinogenesis
at  DIV reduces spine density and spine head width mea-
sured at  DIV. Consistent with the morphological changes,
the frequency of mEPSC (miniature excitatory postsynaptic
synaptic current) is lower in CTTNBP knockdown neurons
at  DIV []. In addition to dendritic spine formation,
CTTNBP-S is involved in dendritic spine maintenance, as
CTTNBP-S knockdown in mature neurons, such as  DIV,
still reduces dendritic spine density at  DIV []. Cor-
tactin is required for CTTNBP-S’s regulation of dendritic
spinogenesis, as a CTTNBP-S mutant that cannot interact
with cortactin cannot rescue CTTNBP knockdown-induced
spine deciency []. Moreover, uorescence recovery aer
photobleaching (FRAP) analysis indicates that CTTNBP-S
regulates cortactin mobility in mature dendritic spines. In the
presence of CTTNBP-S, cortactin more stably localizes to
dendritic spines. e data suggest that CTTNBP-S retains
cortactin in dendritic spines and controls dendritic spine
formation and maintenance [].
CTTNBP-S also controls distribution of striatin family
proteins in dendritic spines []. e striatin protein fam-
ily contains three mammalian members, namely, striatin,
zinedin, and SGNA. ey function as B-type regulatory
subunits of protein phosphatase A (PPA) to control PPA
subcellular location and substrate specicity [, ]. All
three striatin family members are highly enriched in den-
dritic spines []. Striatin protein distribution to synapses
is mediated by its interactions with CTTNBP-S through
the N-terminal coiled-coil domains of both CTTNBP-S
and striatin family members. Similar to cortactin, CTTNBP
Neural Plasticity
NC
1 118 258 523 630
CC Mid P-rich
Striatin
Self-oligomerization Microtubule Cortactin
(a)
Axonal outgrowth Dendrite development Synapse formation
and maintenance
CTTNBP2
Stable microtubule
Unstable microtubule
Actin lament
Cortactin
Striatin/PP2A complex
CTTNBP2 KD CTTNBP2 KD
(b)
F : CTTNBP and neuronal dierentiation. (a) Schematic domain structure and CTTNBP-S-interacting proteins. CC, coiled-coil
domain; Mid, middle region; P-rich, proline-rich domain. (b) e function of CTTNBP-S in neuronal morphogenesis. CTTNBP-S controls
microtubule stability in the dendritic shas and cortactin mobility in dendritic spines. Upon CTTNBP knockdown during dendritic
extension, dendritic complexity decreases. During synaptogenesis, CTTNBP-S helps maintain cortactin in dendritic spines and promotes
dendritic spine formation and maintenance.
knockdown impairs dendritic spine targeting of striatins [].
In conclusion, CTTNBP-S regulates F-actin dynamics and
PPA signaling at dendritic spines.
3.3. CTTNBP2-S Modulates Microtubule Stability and
Regulates Dendritic Arborization. In COS cells, exogenous
CTTNBP-S was unexpectedly associated with the micro-
tubule cytoskeleton in addition to the cortactin-F-actin cyto-
skeleton []. Cell-matrix interactions inuence the cyto-
skeleton association of CTTNBP-S. In COS cells,
CTTNBP-S preferentially associates with the F-actin cyto-
skeleton within one hour aer plating. CTTNBP-S gradually
shis its preference to the microtubule cytoskeleton when
establishing cell-matrix interactions []. CTTNBP-S cyto
skeletal associations also change in neurons. CTTNBP-S is
highly concentrated at dendritic spines in mature neurons
where CTTNBP-S interacts with F-actin cytoskeletons.
In the premature stages when dendritic spines have not
yet formed, CTTNBP-S is already expressed and forms
puncta attached on microtubule bundles along the dendritic
sha []. e association of CTTNBP-S with microtubules
increases microtubule stability by bundling the microtubules.
Two CTTNBP-S domains are required for microtubule
bundling. e Mid domain is required for the association
of microtubule, and the N-terminal coiled-coil domain is
involved in CTTNBP-S oligomerization. Oligomerization
allows the CTTNBP-S oligomer to contain multiple micro-
tubule binding sites to induce microtubule bundling [].
During the dendritic extension stage, CTTNBP-S knock-
down or disruption of microtubule bundling by overexpres-
sion of the N-terminal coiled-coil domain or Mid domain
impairs dendritic arborization []. e studies suggest that,
in addition to controlling the F-actin cytoskeleton,
CTTNBP-S regulates microtubule stability to inuence
dendrite morphology.
3.4. Outstanding Questions about CTTNBP2. e dual roles
of CTTNBP-S in controlling F-actin and the microtubule
cytoskeletons require further investigation. As a neuron-
specic morphology regulator and a high-condence risk
factor for ASDs, CTTNBP deserves further study. Several
questions remain to be addressed. For instance, what is the
molecular mechanism regulating the association between
CTTNBP-S and F-actin and microtubules? Are the associa-
tions of CTTNBP-S with F-actin and microtubules mutually
exclusive? Alternatively, can CTTNBP-S act as a bridge to
link F-actin and microtubules? Only cultured hippocam-
pal neurons have been examined in functional studies of
CTTNBP-S. In the future, in vivo studies should be con-
sidered. Particularly, to address the association of CTTNBP
with ASDs, a mouse genetic model is required. e impact
of CTTNBP ASD mutations on the molecular function
Neural Plasticity
of CTTNBP-S, brain development, and cognition must be
studied to further understand the biological signicance of
CTTNBP.
4. Conclusions
Although hundreds of genes are involved in dendritic spine
formation, they should be either neuron-specic or directly
or indirectly controlled by or linked to neuron-specic
signaling or proteins to specically regulate dendritic spine
formation in neurons. In this review, syndecan--induced
dendritic spine formation and the role of CTTNBP-S in con-
trolling neuronal morphology provide two distinct examples
of how neuronal morphology can be regulated in a neuron-
specic manner. e regulation of neuronal morphology
is critical for normal brain function. Understanding these
regulations is crucial for basic research and for understanding
neurological disorder etiology, which could contribute to
potential therapeutic treatments of the diseases.
Conflict of Interests
e authors declare no competing nancial interests.
Acknowledgments
is work was supported by grants from Academia Sinica
(AS--TP-B) and the Ministry of Science and Technology
(MOST --B--, --B--, and -
-B--) to Yi-Ping Hsueh. Hsiao-Tang Hu is sup-
ported by the Postdoctoral Fellowship of Academia Sinica.
References
[] K. M. Harris and J. K. Stevens, “Dendritic spines of CA pyra-
midal cells in the rat hippocampus: serial electron microscopy
with reference to their biophysical characteristics,Journal of
Neuroscience, vol. , no. , pp. –, .
[] C. Sala and M. Segal, “Dendritic spines: the locus of structural
and functional plasticity,Physiological Reviews,vol.,no.,
pp. –, .
[] I.M.EthellandE.B.Pasquale,“Molecularmechanismsofden-
dritic spine development and remodeling,Progress in Neurobi-
ology,vol.,no.,pp.,.
[] Y. Ishikawa-Brush, J. F. Powell, P. Bolton et al., “Autism and
multipleexostosesassociatedwithanX;translocationoccur-
ring within the GRPR gene and 󸀠to the SDC gene,Human
Molecular Genetics,vol.,no.,pp.,.
[] S.DeRubeis,X.He,A.P.Goldbergetal.,“Synaptic,transcrip-
tional and chromatin genes disrupted in autism,Nature,vol.
,no.,pp.,.
[] V. Lories, J.-J. Cassiman, H. D. Van Berghe, and G. David, “Dif-
ferential expressionof cell surface heparan sulfate proteoglycans
in human mammary epithelial cells and lung broblasts,e
Journal of Biological Chemistr y, vol. , no. , pp. –,
.
[] M.Berneld,M.G
¨
otte,P.W.Parketal.,“Functionsofcellsur-
face heparan sulfate proteoglycans,Annual Review of Biochem-
istry,vol.,pp.,.
[] Y.-P. Hsueh and M. Sheng, “Regulated expression and subcellu-
lar localization of syndecan heparan sulfate proteoglycans and
the syndecan-binding protein CASK/LIN- during rat brain
development,e Journal of Neuroscience,vol.,no.,pp.
–, .
[] I. M. Ethell and Y. Yamaguchi, “Cell surface heparan sulfate
proteoglycan syndecan- induces the maturation of dendritic
spines in rat hippocampal neurons,eJournalofCellBiology,
vol. , no. , pp. –, .
[] Y.-P.Hsueh,F.-C.Yang,V.Kharaziaetal.,“Directinteractionof
CASK/LIN- and syndecan heparan sulfate proteoglycan and
their overlapping distribution in neuronal synapses,e Jour-
nal of Cell Biology,vol.,no.,pp.,.
[] Y.-L. Lin, Y.-T. Lei, C.-J. Hong, and Y.-P. Hsueh, “Syndecan-
induces lopodia and dendritic spine formation via the neu-
robromin-PKA-Ena/VASP pathway,e Journal of Cell Biol-
ogy,vol.,no.,pp.,.
[] H. T. Hu, H. Umemori, and Y. P. Hsueh, “Postsynaptic syn-
decan- induces transsynaptic signaling via broblast growth
factor- for bidirectional synaptic dierentiation,” Under
Review.
[] S. Choi, E. Lee, S. Kwon et al., “Transmembrane domain-
induced oligomerization is crucial for the functions of synde-
can- and syndecan-,e Journal of Biological Chemistry,vol.
, no. , pp. –, .
[] M. Volta, S. Calza, A. M. Roberts, and R. G. Roberts, “Character-
isation of the interaction between syndecan-, neurobromin
and CASK: dependence of interaction on syndecan dimeriza-
tion,Biochemical and Biophysical Research Communications,
vol. , no. , pp. –, .
[] H.-T. Hu and Y.-P. Hsueh, “Calcium inux and postsynaptic
proteins coordinate the dendritic lopodium-spine transition,
Developmental Neurobiology,vol.,no.,pp.,.
[] H.-W. Chao, C.-J. Hong, T.-N. Huang, Y.-G. Lin, and Y.-P.
Hsueh, “SUMOylation of the MAGUK protein CASK regulates
dendritic spinogenesis,e Journal of Cell Biology,vol.,no.
,pp.,.
[]Y.-P.Hsueh,A.M.Roberts,M.Volta,M.Sheng,andR.G.
Roberts, “Bipartite interaction between neurobromatosis type
I protein (neurobromin) and syndecan transmembrane hep-
aran sulfate proteoglycans,Journal of Neuroscience,vol.,no.
, pp. –, .
[] F. Gran´
es, C. Berndt, C. Roy, P. Mangeat, M. Reina, and S.
Vilar ´
o, “Identicationof a novel Ezrin-binding site in syndecan-
 cytoplasmic domain,FEBS Letters,vol.,no.,pp.
, .
[] J. J. Grootjans, P. Zimmermann, G. Reekmans et al., “Syntenin,
a PDZ protein that binds syndecan cytoplasmic domains,
Proceedings of the National Academy of Sciences of the United
States of America, vol. , no. , pp. –, .
[] I. M. Ethell, K. Hagihara, Y. Miura, F. Irie, and Y. Yamaguchi,
“Synbindin, a novel syndecan--binding protein in neuronal
dendritic spines,e Journal of Cell Biology,vol.,no.,pp.
–, .
[] G. F. Xu, B. Lin, K. Tanaka et al., “e catalytic domain of the
neurobromatosis type  gene product stimulates ras GTPase
and complements ira mutants of S. cerevisiae,” Cell,vol.,no.
, pp. –, .
[] G. Xu, P. O’Connell, D. Viskochil et al., “e neurobromatosis
type  gene encodes a protein related to GAP,” Cell,vol.,no.
, pp. –, .
Neural Plasticity
[] R. M. Cawthon, R. Weiss, G. Xu et al., “A major segment of
the neurobromatosis type  gene: cDNA sequence, genomic
structure, and point mutations,Cell,vol.,no.,pp.,
.
[] R. Ballester, D. Marchuk, M. Boguski et al., “e NF locus
encodes a protein functionally related to mammalian GAP and
yeast IRA proteins,” Cell,vol.,no.,pp.,.
[] M. M. Daston and N. Ratner, “Neurobromin, a predominantly
neuronal GTPase activating protein in the adult, is ubiquitously
expressed during development,Developmental Dynamics,vol.
, no. , pp. –, .
[] D. H. Gutmann and F. S. Collins, “Recent progress toward
understanding the molecular biology of von Recklinghausen
neurobromatosis,Annals of Neurology,vol.,no.,pp.
, .
[] B. A. J. Ponder, “Neurobromatosis: from gene to phenotype,
Seminars in Cancer Biology,vol.,no.,pp.,.
[] T.Jacks,T.S.Shih,E.M.Schmitt,R.T.Bronson,A.Bernards,
and R. A. Weinberg, “Tumour predisposition in mice heterozy-
gous for a targeted mutation in Nf,Nature Genetics,vol.,no.
, pp. –, .
[] H.-F. Guo, I. e, F. Hannan, A. Bernards, and Y. Zhong,
“Requirement of Drosophila NF for activation of adenylyl
cyclasebyPACAP-likeneuropeptides,Science,vol.,no.
, pp. –, .
[] J.Tong,F.Hannan,Y.Zhu,A.Bernards,andY.Zhong,“Neu-
robromin regulates G protein-stimulated adenylyl cyclase
activity,Nature Neuroscience,vol.,no.,pp.,.
[] H.-F.Guo,J.Tong,F.Hannan,L.Luo,andY.Zhong,“Aneuro-
bromatosis--regulated pathway is required for learning in
Drosophila,” Nature,vol.,no.,pp.,.
[] H.-F. Wang, Y.-T. Shih, C.-Y. Chen, H.-W. Chao, M.-J. Lee, and
Y.-P. Hsueh, “Valosin-containing protein and neurobromin
interact to regulate dendritic spine density,e Journal of
Clinical Investigation,vol.,no.,pp.,.
[] M.Krause,E.W.Dent,J.E.Bear,J.J.Loureiro,andF.B.Gertler,
“Ena/VASP proteins: regulators of the actin cytoskeleton and
cell migration,Annual Review of Cell and Developmental
Biology,vol.,pp.,.
[] C.Lebrand,E.W.Dent,G.A.Strasseretal.,“CriticalroleofEna/
VASP proteins for lopodia formation in neurons and in
function downstream of netrin-,Neuron,vol.,no.,pp.
, .
[] G. D. J. Watts, J. Wymer, M. J. Kovach et al., “Inclusion body
myopathy associated with Paget disease of bone and frontotem-
poral dementia is caused by mutant valosin-containing protein,
Nature Genetics,vol.,no.,pp.,.
[] J. O. Johnson, J. Mandrioli, M. Benatar et al., “Exome sequenc-
ing reveals VCP mutations as a cause of familial ALS,Neuron,
vol. , no. , pp. –, .
[] I. Iossifov, M. Ronemus, D. Levy et al., “De novo gene disrup-
tions in children on the autistic spectrum,Neuron,vol.,no.
, pp. –, .
[] Y.-P. Hsueh, “Calcium/calmodulin-dependent serine protein
kinase and mental retardation,Annals of Neurology,vol.,no.
, pp. –, .
[] J. Najm, D. Horn, I. Wimplinger et al., “Mutations of CASK
cause an X-linked brain malformation phenotype with micro-
cephaly and hypoplasia of the brainstem and cerebellum,
Nature Genetics,vol.,no.,pp.,.
[] G. Piluso, M. Carella, M. D’Avanzo et al., “Genetic heterogeneity
of FG syndrome: a fourth locus (FGS) maps to Xp.-p. in
an Italian family,Human Genetics,vol.,no.,pp.,
.
[] P. S. Tarpey, R. Smith, E. Pleasance et al., “A systematic, large-
scale resequencing screen of X-chromosome coding exons in
mental retardation,Nature Genetics,vol.,no.,pp.,
.
[] M. J. Lyons, J. M. Graham Jr., G. Neri et al., “Clinical experience
in the evaluation of  patients with a prior diagnosis of FG
syndrome,Journal of Medical Genetics,vol.,no.,pp.,
.
[] S. Hayashi, N. Okamoto, Y. Chinen et al., “Novel intragenic
duplications and mutations of CASK in patients with mental
retardation and microcephaly with pontine and cerebellar
hypoplasia (MICPCH),Human Genetics,vol.,no.,pp.
, .
[] Y.-P. Hsueh, “e role of the MAGUK protein CASK in
neural development and synaptic function,Current Medicinal
Chemistry,vol.,no.,pp.,.
[] B. A. Samuels, Y.-P. Hsueh, T. Shu et al., “Cdk promotes synap-
togenesis by regulating the subcellular distribution of the
MAGUK family member CASK,Neuron,vol.,no.,pp.
, .
[] Y.Hata,S.Butz,andT.C.S
¨
udhof, “CASK: a novel dlg/PSD
homolog with an N-terminal calmodulin-dependent protein
kinase domain identied by interaction with neurexins,Journal
of Neuroscience, vol. , no. , pp. –, .
[] O. Olsen, K. A. Moore, M. Fukata et al., “Neurotransmitter
release regulated by a MALS-liprin-alpha presynaptic complex,
Journal of Cell Biology,vol.,no.,pp.,.
[] S. A. Spangler, S. K. Schmitz, J. T. Kevenaar et al., “Liprin-alpha
promotes the presynaptic recruitment and turnover of RIM/
CASK to facilitate synaptic transmission,e Journal of Cell
Biology,vol.,no.,pp.,.
[] C. Lohmann and T. Bonhoeer, “A role for local calcium signal-
ing in rapid synaptic partner selection by dendritic lopodia,
Neuron,vol.,no.,pp.,.
[] J.-P. Borg, S. W. Straight, S. M. Kaech et al., “Identication of an
evolutionarily conserved heterotrimeric protein complex
involved in protein targeting,Journal of Biological Chemistry,
vol.,no.,pp.,.
[] S. Butz, M. Okamoto, and T. C. S¨
udhof, “A tripartite protein
complex with the potential to couple synaptic vesicle exocytosis
to cell adhesion in brain,Cell, vol. , no. , pp. –, .
[] S. M. Kaech, C. W. Whiteld, and S. K. Kim, “e LIN-/LIN-
/LIN- complex mediates basolateral membrane localization
of the C. elegans EGF receptor LET- in vulval epithelial cells,
Cell, vol. , no. , pp. –, .
[] K. Jo, R. Derin, M. Li, and D. S. Bredt, “Characterization
of MALS/Velis-, -, and -: a family of mammalian LIN-
 homologs enriched at brain synapses in association with
thepostsynapticdensity-/NMDAreceptorpostsynapticcom-
plex,e Journal of Neuroscience,vol.,no.,pp.,
.
[] P. Hotulainen and C. C. Hoogenraad, “Actin in dendritic spines:
connecting dynamics to function,e Journal of Cell Biology,
vol. , no. , pp. – , .
[]K.F.Tolias,J.G.Duman,andK.Um,“Controlofsynapse
development and plasticity byR ho GTPase regulatory proteins,
Progress in Neurobiology, vol. , no. , pp. –, .
 Neural Plasticity
[] T. Saneyoshi and Y. Hayashi, “e Ca2+ and rho gtpase signaling
pathways underlying activity-dependent actin remodeling at
dendritic spines,Cytoskeleton,vol.,no.,pp.,.
[] H. Q. Sun, M. Yamamoto, M. Mejillano, and H. L. Yin, “Gelsolin,
a multifunctional actin regulatory protein,e Journal of
Biological Chemistry,vol.,no.,pp.,.
[] A. M. McGough, C. J. Staiger, J.-K. Min, and K. D. Simonetti,
“e gelsolin familyof actin regulatory proteins: modular struc-
tures, versatile functions,FEBS Letters, vol. , no. -, pp. –
, .
[] P. Silacci, L. Mazzolai, C. Gauci, N. Stergiopulos, H. L. Yin, and
D. Hayoz, “Gelsolin superfamily proteins: key regulators of cel-
lular functions,Cellular and Molecular Life Sciences,vol.,no.
-, pp. –, .
[] M.Lu,W.Witke,D.J.Kwiatkowski,andK.S.Kosik,“Delayed
retraction of lopodia in gelsolin null mice,e Journal of Cell
Biology,vol.,no.,pp.,.
[] E. N. Star, D. J. Kwiatkowski, and V. N. Murthy, “Rapid turnover
of actin in dendritic spines and its regulation by activity,Nature
Neuroscience,vol.,no.,pp.,.
[] Y. Ohoka and Y. Takai, “Isolation and characterization of cor-
tactin isoforms and a novel cortactin-binding protein, CBP,
Genes to Cells,vol.,no.,pp.,.
[] T. Uruno, J. Liu, P. Zhang et al., “Activation of Arp/ complex-
mediated actin polymerization by cortactin,Nature Cell Biol-
ogy,vol.,no.,pp.,.
[] A. M. Weaver, A. V. Karginov, A. W. Kinley et al., “Cortactin pro-
motes and stabilizes Arp/-induced actin lament network
formation,Current Biology, vol. , no. , pp. –, .
[] H. Hering and M. Sheng, “Activity-dependent redistribution
and essential role of cortactin in dendritic spine morphogene-
sis,e Journal of Neuroscience,vol.,no.,pp.,
.
[] J.Cheung,E.Petek,K.Nakabayashi,L.-C.Tsui,J.B.Vincent,
and S. W. Scherer, “Identication of the human cortactin-
binding protein- gene from the autism candidate region at
q,Genomics,vol.,no.-,pp.,.
[] Y.-K. Chen and Y.-P. Hsueh, “Cortactin-binding protein  mod-
ulates the mobility of cortactin and regulates dendritic spine
formation and maintenance,e Journal of Neuroscience,vol.
,no.,pp.,.
[] Y.-K. Chen, C.-Y. Chen, H.-T. Hu, and Y.-P. Hsueh, “CTTNBP,
but not CTTNBPNL, regulates dendritic spinogenesis and
synaptic distribution of the striatin-PPA complex,Molecular
Biology of the Cell, vol. , no. , pp. –, .
[] P.-Y. Shih, S.-P. Lee, Y.-K. Chen, and Y.-P. Hsueh, “Cortactin-
binding protein  increases microtubule stability and regulates
dendritic arborization,Journal of Cell Science,vol.,no.,
pp. –, .
[] J. D. Arroyo and W. C. Hahn, “Involvement of PPA in viral and
cellular transformation,Oncogene,vol.,no.,pp.
, .
[] M. Benoist, S. Gaillard, and F. Castets, “e striatin family: a
new signaling platform in dendritic spines,Journal of Physiol-
ogy Paris,vol.,no.-,pp.,.
[] S. Gaillard, Y. Bailly, M. Benoist et al., “Targeting of proteins
of the striatin family to dendritic spines: role of the coiled-coil
domain,Tra c,vol.,no.,pp.,.
... The excitatory synapses in the mammalian nervous system are located in dendritic spines, which are the neuron-specific subcellular structures necessary for neural circuitry and function [27,28]. Recent evidence suggests that an alteration in dendritic spines may be involved in the pathogenesis of ASD [29,30]. ...
... In the mammalian nervous system, dendritic spines are located at excitatory synaptic sites; they are neuron-specific subcellular structures that are necessary for neural circuitry and function. Hundreds of genes are known to be involved in the regulation of dendritic spine formation; most of these genes are non-neuron specific, including ANXA1, syndecan-2, NF1 (encoding neurofibromin), VCP (valosin-containing protein), CASK, and CTTNBP2 [29,62]. CTTNBP2 is a neuron-specific gene that regulates dendritic spine formation [29]. ...
... Hundreds of genes are known to be involved in the regulation of dendritic spine formation; most of these genes are non-neuron specific, including ANXA1, syndecan-2, NF1 (encoding neurofibromin), VCP (valosin-containing protein), CASK, and CTTNBP2 [29,62]. CTTNBP2 is a neuron-specific gene that regulates dendritic spine formation [29]. CTTNBP2 is located in a region associated with ASD, namely, the autism candidate region at 7q31 [54]. ...
Article
Full-text available
Autism spectrum disorder (ASD) affects around 1% of children with no effective blood test or cure. Recent studies have suggested that these are neurological disorders with a strong genetic basis and that they are associated with the abnormal formation of dendritic spines. Chromosome microarray (CMA) together with high-throughput sequencing technology has been used as a powerful tool to identify new candidate genes for ASD. In the present study, CMA was first used to scan for genome-wide copy number variants in a proband, and no clinically significant copy number variants were found. Whole-exome sequencing (WES) was used further for genetic testing of the whole quad family affected by ASD, including the proband, his non-autistic sister, and his parents. Sanger sequencing and MassARRAY-based validation were used to identify and confirm variants associated with ASD. WES yielded a 151-fold coverage depth for each sample. A total of 98.65% of the targeted whole-exome region was covered at >20-fold depth. A de novo variant in CTTNBP2, p.M115T, was identified. The CTTNBP2 gene belongs to a family of ankyrin repeat domain-containing proteins associated with dendritic spine formation. Although CTTNBP2 has been associated with ASD, limited studies have been developed to identify clinically relevant de novo mutations of CTTNBP2 in children with ASD; family-based WES successfully identified a clinically relevant mutation in the CTTNBP2 gene in a quad family affected by ASD. Considering the neuron-specific expression of CTTNBP2 and its role in dendritic spine formation, our results suggest a correlation between the CTTNBP2 mutation and ASD, providing genetic evidence for ASD spine pathology. Although the present study is currently insufficient to support the assertion that the de novo mutation M115T in CTTNBP2 directly causes the autism phenotype, our study provides support for the assertion that this mutation is a candidate clinically relevant variant in autism.
... The actin bundle is surrounded by high negative curvature plasma membrane that is stabilised by proteins that link curved plasma membrane to the cytoskeleton such as IRSp53 (Nakagawa et al. 2003;Mattila et al. 2007;Tsai et al. 2018). How the process of filopodial protrusion is controlled is important for understanding the mechanistic and genetic basis of intellectual disabilities and autism amongst other conditions (Truesdell et al. 2015;Hu et al. 2016;Gouder et al. 2019;Wit & Hiesinger 2022). As an established axon guidance model, here we used ex vivo dissected primary Xenopus retinal ganglion cells to elucidate the mechanisms of filopodial extension. ...
Article
Full-text available
Filopodia are narrow actin-rich protrusions with important roles in neuronal development where membrane-binding adaptor proteins have emerged as upstream regulators that link membrane interactions to actin regulators, for example I-BAR and F-BAR domain-containing proteins interacting with Ena/VASP and formins. To explore the significance of the F-BAR neuronal membrane adaptor TOCA-1 in filopodia we used quantitative analysis of TOCA-1 and filopodial dynamics in Xenopus retinal ganglion cells, where Ena/VASP proteins have a native role in filopodial extension. Both the adaptors and their binding partners are part of diverse and redundant protein networks that can functionally compensate for each other. Increasing density of TOCA-1 enhances Ena/VASP binding in vitro and an accumulation of TOCA-1, and its coincidence with Ena, correlates with filopodial protrusion in vivo. Two-colour single molecule localisation microscopy of TOCA-1 and Ena supports their nanoscale association. TOCA-1 clusters promote filopodial protrusion depending on a functional SH3 domain and activation of Cdc42, which we perturbed using small molecule inhibitor CASIN. We propose that TOCA-1 clusters act independently of membrane curvature to recruit and promote Ena activity for filopodial protrusion.
... PKA-enabled/vasodilator-stimulated phosphoprotein (PKA-Ena/VASP). Neurofibromin modulates PKA-Ena/VASP pathway, implicated in the formation of filopodia and dendritic spine 6 , axonal outgrowth 122 , actin polymerization along bundle formation 123 , and probably for proper differentiation of nerve cells 6 . In part due to neurofibromin association with microtubular and microfilamentous cytoskeleton 91 , FAK 90 and syndecan-2 76 , the normal development of the cerebral cortex may be affected 74 . ...
Article
Full-text available
Neurofibromin controls many cell processes, such as growth, learning, and memory. If neurofibromin is not working properly, it can lead to health problems, including issues with the nervous, skeletal, and cardiovascular systems and cancer. This review examines neurofibromin's binding partners, signaling pathways and potential therapeutic targets. In addition, it summarizes the different post-translational modifications that can affect neurofibromin's interactions with other molecules. It is essential to investigate the molecular mechanisms that underlie neurofibromin variants in order to provide with functional connections between neurofibromin and its associated proteins for possible therapeutic targets based on its biological function.
... The NLS domain is necessary for the nuclear localization of neurofibromin, while the function of SBR is to bind syndecans. The interaction between neurofibromin and syndecan is important for cell differentiation and proliferation, and for synaptic plasticity [61]. Functional studies are needed to confirm the possible role of C-terminal NF1 mutations in the development of the spinal form of the disease. ...
Article
Full-text available
Simple Summary At present, no systematic study of the clinical spectrum and molecular characteristics of NF1 patients with spinal neurofibromatosis (SNF), a phenotypic subclass of neurofibromatosis 1 (NF1), has been carried out. Here, we provide evidence that SNF patients are at high risk of problematic neurofibromas, presenting not only bilateral neurofibromas involving all spinal roots, but also a higher incidence of internal neurofibromas and nerve root swelling. From a histopathological view, not only neurofibromas, but also neurogangliomas are present in SNF. The analysis of 19 families with at least 1 member affected by SNF showed a high phenotypic variability within the SNF families. Furthermore, we discovered a higher prevalence of missense mutations in SNF compared to classical NF1. Both clinical features and genetic testing can help in identifying cases at risk of SNF, and that are more likely to benefit from a spinal MRI scan. Abstract Spinal neurofibromatosis (SNF), a phenotypic subclass of neurofibromatosis 1 (NF1), is characterized by bilateral neurofibromas involving all spinal roots. In order to deepen the understanding of SNF’s clinical and genetic features, we identified 81 patients with SNF, 55 from unrelated families, and 26 belonging to 19 families with at least 1 member affected by SNF, and 106 NF1 patients aged >30 years without spinal tumors. A comprehensive NF1 mutation screening was performed using NGS panels, including NF1 and several RAS pathway genes. The main features of the SNF subjects were a higher number of internal neurofibromas (p < 0.001), nerve root swelling (p < 0.001), and subcutaneous neurofibromas (p = 0.03), while hyperpigmentation signs were significantly less frequent compared with the classical NF1-affected cohorts (p = 0.012). Fifteen patients underwent neurosurgical intervention. The histological findings revealed neurofibromas in 13 patients and ganglioneuromas in 2 patients. Phenotypic variability within SNF families was observed. The proportion of missense mutations was higher in the SNF cases than in the classical NF1 group (21.40% vs. 7.5%, p = 0.007), conferring an odds ratio (OR) of 3.34 (CI = 1.33–10.78). Two unrelated familial SNF cases harbored in trans double NF1 mutations that seemed to have a subclinical worsening effect on the clinical phenotype. Our study, with the largest series of SNF patients reported to date, better defines the clinical and genetic features of SNF, which could improve the management and genetic counseling of NF1.
... Some CTTNBP2-regulated proteins, such as SHANK3, SHANK2, STRN (striatin) and RAC3, physically associate with CTTNBP2 35 . Thus, CTTNBP2 acts as a multidomain adaptor molecule to control F-actin and microtubule cytoskeletons and multiple synaptic molecules 37 . However, it remains unclear how CTTNBP2 interacts with and controls the distribution of many different synaptic proteins. ...
Article
Full-text available
Many synaptic proteins form biological condensates via liquid-liquid phase separation (LLPS). Synaptopathy, a key feature of autism spectrum disorders (ASD), is likely relevant to the impaired phase separation and/or transition of ASD-linked synaptic proteins. Here, we report that LLPS and zinc-induced liquid-to-gel phase transition regulate the synaptic distribution and protein-protein interaction of cortactin-binding protein 2 (CTTNBP2), an ASD-linked protein. CTTNBP2 forms self-assembled condensates through its C-terminal intrinsically disordered region and facilitates SHANK3 co-condensation at dendritic spines. Zinc binds the N-terminal coiled-coil region of CTTNBP2, promoting higher-order assemblies. Consequently, it leads to reduce CTTNBP2 mobility and enhance the stability and synaptic retention of CTTNBP2 condensates. Moreover, ASD-linked mutations alter condensate formation and synaptic retention of CTTNBP2 and impair mouse social behaviors, which are all ameliorated by zinc supplementation. Our study suggests the relevance of condensate formation and zinc-induced phase transition to the synaptic distribution and function of ASD-linked proteins.
... Firstly, it is involved in the formation of synapses and the interaction between synapses [46]. For example, CASK regulates axon growth and branch by interacting with Bcl11A [24]; Interaction between CASK and syndecan-2 regulates maturation of dendritic protein [25]. At presynaptic sites, CASK forms compound with MALS/Mint-1/Liprinα through its CaMK and L27A domains. ...
Article
Full-text available
Background The pathogenic variation of CASK gene can cause CASK related mental disorders. The main clinical manifestations are microcephaly with pontine and cerebellar hypoplasia, X-linked mental disorders with or without nystagmus and FG syndrome. The main pathogenic mechanism is the loss of function of related protein caused by variant. We reported a Chinese male newborn with a de novo variant in CASK gene. Case presentation We present an 18-day-old baby with growth retardation and brain hypoplasia. Whole-exome sequencing was performed, which detected a hemizygous missense variant c.764G > A of CASK gene. The variant changed the 255th amino acid from Arg to His. Software based bioinformatics analyses were conducted to infer its functional effect. Conclusions In this paper, a de novo variant of CASK gene was reported. Moreover, a detailed description of all the cases described in the literature is reported. CASK variants cause a variety of clinical phenotypes. Its diagnosis is difficult due to the lack of typical clinical symptoms. Genetic testing should be performed as early as possible if this disease is suspected. This case provides an important reference for the diagnosis and treatment of future cases.
Article
Full-text available
The AAA+ ATPase valosin containing protein (VCP) is essential for cell and organ homeostasis, especially in cells of the nervous system. As part of a large network, VCP collaborates with many cofactors to ensure proteostasis under normal, stress, and disease conditions. A large number of mutations have revealed the importance of VCP for human health. In particular, VCP facilitates the dismantling of protein aggregates and the removal of dysfunctional organelles. These are critical events to prevent malfunction of the brain and other parts of the nervous system. In line with this idea, VCP mutants are linked to the onset and progression of neurodegeneration and other diseases. The intricate molecular mechanisms that connect VCP mutations to distinct brain pathologies continue to be uncovered. Emerging evidence supports the model that VCP controls cellular functions on multiple levels and in a cell type specific fashion. Accordingly, VCP mutants derail cellular homeostasis through several mechanisms that can instigate disease. Our review focuses on the association between VCP malfunction and neurodegeneration. We discuss the latest insights in the field, emphasize open questions, and speculate on the potential of VCP as a drug target for some of the most devastating forms of neurodegeneration.
Article
Syndecans are transmembrane heparan sulfate proteoglycans present on most mammalian cell surfaces. They have a long evolutionary history, a single syndecan gene being expressed in bilaterian invertebrates. Syndecans have attracted interest because of their potential roles in development and disease, including vascular diseases, inflammation and various cancers. Recent structural data is providing important insights into their functions, which are complex, involving both intrinsic signaling through cytoplasmic binding partners and co-operative mechanisms where syndecans form a signaling nexus with other receptors such as integrins and tyrosine kinase growth factor receptors. While the cytoplasmic domain of syndecan-4 has a well-defined dimeric structure, the syndecan ectodomains are intrinsically disordered, which is linked to a capacity to interact with multiple partners. However, it remains to fully establish the impact of glycanation and partner proteins on syndecan core protein conformations. Genetic models indicate that a conserved property of syndecans links the cytoskeleton to calcium channels of the transient receptor potential class, compatible with roles as mechanosensors. In turn, syndecans influence actin cytoskeleton organization to impact motility, adhesion and the extracellular matrix environment. Syndecan clustering with other cell surface receptors into signaling microdomains has relevance to tissue differentiation in development, for example in stem cells, but also in disease where syndecan expression can be markedly up-regulated. Since syndecans have potential as diagnostic and prognostic markers as well as possible targets in some forms of cancer, it remains important to unravel structure/function relationships in the four mammalian syndecans.
Article
Spinal neurofibromatosis (SNF) is a form of neurofibromatosis type 1 (NF1) characterized by bilateral neurofibromas involving all spinal roots. The pathogenic mechanisms determining the SNF form are currently unknown. To verify the presence of genetic variants possibly related to SNF or classic NF1, we studied 106 sporadic NF1 and 75 SNF patients using an NGS panel of 286 genes encoding RAS pathway effectors and neurofibromin interactors and evaluated the expression of syndecans (SDC1, SDC2, SDC3, SDC4), the NF1 3' tertile interactors, by quantitative real-time PCR. We previously identified 75 and 106 NF1 variants in SNF and NF1 cohorts, respectively. The analysis of the distribution of pathogenic NF1 variants in the three NF1 tertiles showed a significantly higher prevalence of NF1 3' tertile mutations in SNF than in the NF1 cohort. We hypothesized a potential pathogenic significance of the 3' tertile NF1 variants in SNF. The analysis of syndecan expression on PBMCs RNAs from 16 SNF, 16 classic NF1 patients and 16 healthy controls showed that the expression levels of SDC2 and SDC3 were higher in SNF and NF1 patients than in controls; moreover, SDC2, SDC3 and SDC4 were significantly over expressed in patients mutated in the 3' tertile compared to controls. Two different mutational NF1 spectra seem to characterize SNF and classic NF1, suggesting a pathogenic role of NF1 3' tertile and its interactors, syndecans, in SNF. Our study, providing new insights on a possible role of neurofibromin C-terminal in SNF, could address effective personalized patient management and treatments.
Article
Full-text available
Neurexins are neuronal cell surface proteins with hundreds of isoforms. In yeast two-hybrid screens for intracellular molecules interacting with different neurexins, we identified a single interacting protein called CASK. CASK is composed of an N-terminal Ca2+, calmodulin-dependent protein kinase sequence and a C-terminal region that is similar to the intercellular junction proteins dlg-A, PSD95/SAP90, SAP97, Z01, and Z02 and that contains DHR-, SH3-, and guanylate kinase domains. CASK is enriched in brain in synaptic plasma membranes but is also detectable at low levels in all tissues tested. The cytoplasmic domains of all three neurexins bind CASK in a salt-labile interaction. In neurexin I, this interaction is dependent on the C-terminal three residues. Thus, CASK is a membrane-associated protein that combines domains found in Ca2+ - activated protein kinases and in proteins specific for intercellular junctions, suggesting that it may be a signaling molecule operating at the plasma membrane, possibly in conjunction with neurexins.
Article
Full-text available
The neurofibromatosis type 1 (NF1) gene encodes a large tumor suppressor protein (neurofibromin). Although it is known to possess Ras GTPase-activating protein (GAP) activity, the cellular role of neurofibromin remains unclear. Here we used yeast two-hybrid screening to identify neurofibromin-interacting proteins. Syndecan-2, a transmembrane heparan sulfate proteoglycan (HSPG), was isolated as a binding partner for two distinct regions of the neurofibromin protein. We subsequently found that neurofibromin can bind all four mammalian syndecans. NF1 interaction requires the transmembrane domain and a membrane-proximal region of the cytoplasmic tail of syndecan, but not the C terminus of syndecan known to bind to CASK, a membrane-associated guanylate kinase (MAGUK). Neurofibromin, syndecans, and CASK have overlapping subcellular distributions in axons and synapses of neurons, as shown by biochemical fractionation and immunostaining. Moreover, neurofibromin exists in a complex with syndecan and CASK in vivo , as evidenced by their coimmunoprecipitation from rat brain. Our findings suggest that interaction with different members of the syndecan family may be a mechanism for localizing neurofibromin to specialized domains of the plasma membrane.
Article
Full-text available
Functional synapse formation requires tight coordination between pre- and post-synaptic termini. Previous studies have shown that postsynaptic expression of heparan sulfate proteoglycan syndecan-2 (SDC2) induces dendritic spinogenesis. Those SDC2-induced dendritic spines are frequently associated with presynaptic termini. However, how postsynaptic SDC2 accelerates maturation of corresponding presynaptic termini is unknown. Because fibroblast growth factor 22 (FGF22), a heparan sulfate binding growth factor, has been shown to act as a presynaptic organizer released from the postsynaptic site, it seems possible that postsynaptic SDC2 presents FGF22 to the presynaptic FGF receptor to promote presynaptic differentiation. Here, we show that postsynaptic SDC2 uses its ectodomain to interact with and facilitate dendritic filopodial targeting of FGF22, triggering presynaptic maturation. Since SDC2 also enhances filopodial targeting of NMDAR via interaction with the CASK-mLIN7-MINT1 adaptor complex, presynaptic maturation promoted by FGF22 further feeds back to activate NMDAR at corresponding postsynaptic sites through increased neurotransmitter release and, consequently, promotes the dendritic filopodia-spines (F-S) transition. Meanwhile, via regulation of the KIF17 motor, CaMKII (activated by the NMDAR pathway) may further facilitate FGF22 targeting to dendritic filopodia that receive presynaptic stimulation. Our study suggests a positive feedback that promotes the coordination of postsynaptic and presynaptic differentiation.
Article
Full-text available
The genetic architecture of autism spectrum disorder involves the interplay of common and rare variants and their impact on hundreds of genes. Using exome sequencing, here we show that analysis of rare coding variation in 3,871 autism cases and 9,937 ancestry-matched or parental controls implicates 22 autosomal genes at a false discovery rate (FDR) < 0.05, plus a set of 107 autosomal genes strongly enriched for those likely to affect risk (FDR < 0.30). These 107 genes, which show unusual evolutionary constraint against mutations, incur de novo loss-of-function mutations in over 5% of autistic subjects. Many of the genes implicated encode proteins for synaptic formation, transcriptional regulation and chromatin-remodelling pathways. These include voltage-gated ion channels regulating the propagation of action potentials, pacemaking and excitability-transcription coupling, as well as histone-modifying enzymes and chromatin remodellers-most prominently those that mediate post-translational lysine methylation/demethylation modifications of histones.
Article
Full-text available
Neurons are characterized by subcellular compartments such as axons, dendrites and synapses that have highly specialized morphologies and biochemical specificities. Cortactin binding protein 2 (CTTNBP2), a neuron-specific F-actin regulator, has been shown to play a role in the regulation of dendritic spine formation and maintenance. Here, we show that in addition to F-actin, CTTNBP2 also associates with microtubules before mature dendritic spines formed. The association of CTTNBP2 and microtubules induced the formation of microtubule bundles. Although the middle (Mid) region of CTTNBP2 was sufficient for association with microtubules, for microtubule bundling, the N-terminal region containing the coiled coil motifs (NCC) mediating the dimerization or oligomerization of CTTNBP2 is also required. Our study indicates that CTTNBP2 proteins form a dimer or oligomer and bring multiple microtubule filaments together to form bundles. In cultured hippocampal neurons, knockdown of CTTNBP2 or expression of the Mid or NCC domain alone reduced the acetylation levels of microtubules and impaired dendritic arborization. The study suggests that CTTNBP2 influences both the F-actin and microtubule cytoskeletons and regulates dendritic spine formation and dendritic arborization.
Article
Full-text available
The introduction of high-resolution time lapse imaging and molecular biological tools has changed dramatically the rate of progress towards the understanding of the complex structure-function relations in synapses of central spiny neurons. Standing issues, including the sequence of molecular and structural processes leading to formation, morphological change, and longevity of dendritic spines, as well as the functions of dendritic spines in neurological/psychiatric diseases are being addressed in a growing number of recent studies. There are still unsettled issues with respect to spine formation and plasticity: Are spines formed first, followed by synapse formation, or are synapses formed first, followed by emergence of a spine? What are the immediate and long-lasting changes in spine properties following exposure to plasticity-producing stimulation? Is spine volume/shape indicative of its function? These and other issues are addressed in this review, which highlights the complexity of molecular pathways involved in regulation of spine structure and function, and which contributes to the understanding of central synaptic interactions in health and disease.
Article
Full-text available
The presynaptic active zone mediates synaptic vesicle exocytosis, and modulation of its molecular composition is important for many types of synaptic plasticity. Here, we identify synaptic scaffold protein liprin-α2 as a key organizer in this process. We show that liprin-α2 levels were regulated by synaptic activity and the ubiquitin-proteasome system. Furthermore, liprin-α2 organized presynaptic ultrastructure and controlled synaptic output by regulating synaptic vesicle pool size. The presence of liprin-α2 at presynaptic sites did not depend on other active zone scaffolding proteins but was critical for recruitment of several components of the release machinery, including RIM1 and CASK. Fluorescence recovery after photobleaching showed that depletion of liprin-α2 resulted in reduced turnover of RIM1 and CASK at presynaptic terminals, suggesting that liprin-α2 promotes dynamic scaffolding for molecular complexes that facilitate synaptic vesicle release. Therefore, liprin-α2 plays an important role in maintaining active zone dynamics to modulate synaptic efficacy in response to changes in network activity.
Article
Full-text available
CASK, the rat homolog of a gene (LIN-2) required for vulval differentiation in Caenorhabditis elegans, is expressed in mammalian brain, but its function in neurons is unknown. CASK is distributed in a punctate somatodendritic pattern in neurons. By immunogold EM, CASK protein is concentrated in synapses, but is also present at nonsynaptic membranes and in intracellular compartments. This immunolocalization is consistent with biochemical studies showing the presence of CASK in soluble and synaptosomal membrane fractions and its enrichment in postsynaptic density fractions of rat brain. By yeast two-hybrid screening, a specific interaction was identified between the PDZ domain of CASK and the COOH terminal tail of syndecan-2, a cell surface heparan sulfate proteoglycan (HSPG). The interaction was confirmed by coimmunoprecipitation from heterologous cells. In brain, syndecan-2 localizes specifically at synaptic junctions where it shows overlapping distribution with CASK, consistent with an interaction between these proteins in synapses. Cell surface HSPGs can bind to extracellular matrix proteins, and are required for the action of various heparin-binding polypeptide growth/differentiation factors. The synaptic localization of CASK and syndecan suggests a potential role for these proteins in adhesion and signaling at neuronal synapses.
Article
Dendritic spines are the major locations of excitatory synapses in the mammalian central nervous system. The transformation from dendritic filopodia to dendritic spines has been recognized as one type of spinogenesis. For instance, syndecan-2 (SDC2), a synaptic heparan sulfate proteoglycan, is highly concentrated at dendritic spines and required for spinogenesis. It induces dendritic filopodia formation, followed by spine formation. However, the molecular regulation of the filopodium-spine transition induced by SDC2 is still unclear. In this report, we show that calcium is an important signal downstream of SDC2 in regulation of filopodium-spine transition but not filopodia formation. SDC2 interacted with the postsynaptic proteins CASK and LIN7 and further recruited NMDAR to the tips of filopodia induced by SDC2. Calcium influx via NMDAR promoted spine maturation because addition of EDTA or AP5 to the culture medium effectively prevented morphological change from dendritic filopodia to dendritic spines. Our data also indicated that F-actin rearrangement regulated by calcium influx is involved in the morphological change, because the knockdown of gelsolin, a calcium-activated F-actin severing molecule, impaired the filopodium-spine transition induced by SDC2. In conclusion, our study demonstrates that postsynaptic proteins coordinate to trigger calcium signalling and cytoskeleton rearrangement and consequently control filopodium-spine transition. © 2014 Wiley Periodicals, Inc. Develop Neurobiol, 2014
Article
The human neurofibromatosis type 1 (NF1) tumor suppressor protein functions as a Ras-specific guanosine triphosphatase–activating protein, but the identity of Ras- mediated pathways modulated by NF1 remains unknown. A study of Drosophila NF1 mutants revealed that NF1 is essential for the cellular response to the neuropeptide PACAP38 (pituitary adenylyl cyclase–activating polypeptide) at the neuromuscular junction. The peptide induced a 100-fold enhancement of potassium currents by activating the Ras-Raf and adenylyl cyclase–adenosine 3’,5’-monophosphate (cAMP) pathways. This response was eliminated in NF1 mutants. NF1 appears to regulate the rutabaga-encoded adenylyl cyclase rather than the Ras-Raf pathway. Moreover, the NF1 defect was rescued by the exposure of cells to pharmacological treatment that increased concentrations of cAMP.