SlideShare a Scribd company logo
1 of 12
Download to read offline
Cerebral Cortex Advance Access published August 1, 2011
Cerebral Cortex
doi:10.1093/cercor/bhr199


WIP Is a Negative Regulator of Neuronal Maturation and Synaptic Activity
A. Franco1, S. Knafo2,3, I. Banon-Rodriguez1, P. Merino-Serrais2, I. Fernaud-Espinosa2, M. Nieto1, J.J. Garrido2,3,4, J.A. Esteban3,
F. Wandosell3,4 and I.M. Anton1,4
1
 Centro Nacional de Biotecnologı´ a (CNB-CSIC), 28049 Madrid, Spain, 2Instituto Cajal (CSIC), 28002 Madrid, Spain, 3Centro de
Biologı´ a Molecular ‘‘Severo Ochoa’’ (CSIC-UAM), Universidad Autonoma de Madrid, 28049 Madrid, Spain and 4CIBERNED, Centro
                                                                  ´
Investigacion Biomedica en Red de Enfermedades Neurodegenerativas, 28031 Spain
             ´        ´
A. Franco and S. Knafo contributed equally to this work
Address correspondence to Dr Ines M. Anton, Cellular and Molecular Department, Centro Nacional de Biotecnologı´ a (CNB-CSIC), Darwin 3, 28049
                                 ´        ´
Madrid, Spain. Email: ianton@cnb.csic.es.


Wiskott--Aldrich syndrome protein (WASP) --interacting protein                  factors (NPF), such as neural Wiskott-Aldrich syndrome protein
(WIP) is an actin-binding protein involved in the regulation of actin           (N-WASP). The Arp2/3 complex nucleates actin, inducing
polymerization in cells, such as fibroblasts and lymphocytes.                    branching and elongation, and with N-WASP, it mediates
Despite its recognized function in non-neuronal cells, the role of              neurite elongation (Suetsugu, Hattori, et al. 2002; Pinyol et al.
WIP in the central nervous system has not been examined                         2007) and neurite branching (Kakimoto et al. 2004). N-WASP




                                                                                                                                                           Downloaded from cercor.oxfordjournals.org at Universidad Autonoma de Madrid. Facultad de Medicina on August 2, 2011
previously. We used WIP-deficient mice to examine WIP function                   interacts with WASP-interacting protein (WIP), a broadly
both in vivo and in vitro. We report here that WIP2/2 hippocampal               expressed proline-rich protein that regulates N-WASP function
neurons exhibit enlargement of somas as well as overgrowth of                   as NPF and whose deficiency modifies actin polymerization
neuritic and dendritic branches that are more evident in early                  kinetics and the density of the subcortical actin network
developmental stages. Dendritic arborization and synaptogenesis,                (Anton et al. 2007). Through WASP/N-WASP--dependent or
which includes generation of postsynaptic dendritic spines, are                 --independent mechanisms, WIP participates in a wide variety
actin-dependent processes that occur in parallel at later stages.               of cellular functions, including signaling, endocytosis, and actin
WIP deficiency also increases the amplitude and frequency of                     cytoskeleton remodeling (Anton et al. 2007). WIP deficiency in
miniature excitatory postsynaptic currents, suggesting that WIP2/2              mice alters the immune response, reducing T and mast cell
neurons have more mature synapses than wild-type neurons. These                 activity and increasing B cell function (Anton et al. 2002;
findings reveal WIP as a previously unreported regulator of                      Kettner et al. 2004). Moreover, WIP null mice have a pro-
neuronal maturation and synaptic activity.                                      gressive immunological disorder of autoimmune nature, with
                                                                                evident ulcerative colitis, interstitial pneumonitis, glomerular
Keywords: dendritic spine, electrophysiology, neuritogenesis, N-WASP,           nephropathy with IgA deposits, autoantibodies, and joint
synapse                                                                         inflammation that lead, all together, to premature death
                                                                                (Curcio et al. 2007). Although molecular details of WIP-
                                                                                WASP/N-WASP inter-action have been studied extensively
                                                                                (Volkman et al. 2002; Ho et al. 2004; Dong et al. 2007; Peterson
Introduction                                                                    et al. 2007), few data are available on its functional impact and
Neuronal cytoarchitecture is first established through neurito-                  even fewer regarding the central nervous system, where the
genesis, a process in which neurons extend their neurites to                    role of WIP has not been previously addressed.
form a functional network during neuronal development (de                           Using the WIP knockout mouse as a tool, here, we describe
Curtis 2007). Neuron morphology greatly determines the final                     that loss of this protein impacts neurite and dendrite dynamics
complexity of the nervous system and is essential for the signal                and morphology, both in early and in late developmental stages,
flow that underlies information integration and processing. It is                in vitro and in vivo. Gross examination of WIP–/– brain revealed
therefore important that neuritogenesis occurs at the right                     changes in forebrain and hippocampal size. Extensive analysis
place and time for correct establishment of synaptic contacts                   of WIP–/– hippocampal neuron development showed premature
with proper targets (de Curtis 2007). Several environmental                     neuritogenesis. Finally, electrophysiological and immunocyto-
cues converge on common coordinated intracellular pathways to                   chemical analyses demonstrated modified synaptic activity of
modulate neuritogenesis. Such intracellular events involve sig-                 WIP–/– mature neurons. These studies show that WIP is an
naling transduction, exocytic and endocytic mechanisms related                  essential negative regulator in the control of the cytoskeletal
to membrane trafficking and cytoskeletal rearrangements.                         events that underlie neuronal and synaptic development.
   Neurite initiation and outgrowth are based on the capacity
of the neuronal cytoskeleton, constituted mainly of actin                       Materials and Methods
microfilaments (MF) and tubulin microtubules (MT), to
assemble and disassemble in response to extracellular signals                   Mice
(Luo 2002; Conde and Caceres 2009). The polarized growth of                     Wild-type (WT) and WIP KO SV129/BL6 mice (Anton et al. 2002) were
neurites requires the initial depolymerization of actin MF                      housed in specific pathogen-free conditions at the animal facility of the
(Bradke and Dotti 1999), stabilization of MT (Ferreira and                      Centro de Biologı´ a Molecular ‘‘Severo Ochoa,’’ Madrid, Spain. The
                                                                                mouse colony was maintained by continuous mating of heterozygous
Caceres 1989), and accumulation of a number of specific                          females with heterozygous males for more than 20 generations. To
proteins (Wiggin et al. 2005). Actin polymerization is con-                     obtain control or WIP–/– embryos/litters, we mate control male and
trolled by the actin-related protein (Arp2/3) complex and by                    female or WIP–/– male and female mice. Handling of mice and all
the action of actin-binding proteins and nucleation-promoting                   manipulations were carried out in accordance with national and

Ó The Author 2011. Published by Oxford University Press. All rights reserved.
For permissions, please e-mail: journals.permissions@oup.com
European Community guidelines and were reviewed and approved by                 a 403 oil-immersion lens (NA, 1.3). Metamorph 6.2r6 (Universal
the institutional committee for animal welfare. All quantification was           Imaging) software was used to process the time-lapse captured images.
conducted in a genotype-blind manner.
                                                                                Morphometry
                                                                                Image stacks (physical size 76.9 3 76.9 lm) were imported to the
Brain Lysates and Western Blot
                                                                                confocal module of Neurolucida 7.1 (MicroBrightfield, Inc., Williston,
Control or WIP–/– brains were homogenized in lysis buffer (20 mM 4-(2-
                                                                                VT), and neuronal dendritic trees were traced by drawing the dendrites
hydroxyethyl)-1-piperazineethanesulfonic acid, pH 7.4, 100 mM NaCl, 5
                                                                                and the bifurcation points. Sholl analysis was performed for each traced
mM ethylenediaminetetraacetic acid, 1% Triton X-100, 100 mM NaF, 1
                                                                                neuron by automatically calculating the number of dendritic intersec-
mM Na3VO4, and the Complete Protease Inhibitor Cocktail, Roche
                                                                                tions and the dendritic length at 10-lm interval starting from the soma.
Diagnostics), and soluble extracts were resolved by sodium dodecyl
                                                                                Total dendritic length and total number of intersections and branches for
sulfate--polyacrylamide gel electrophoresis after determination of
                                                                                each neuron were also calculated as an index of dendritic complexity.
protein concentration by Bradford analysis (BioRad). Proteins were
                                                                                Soma area was determined by drawing soma contours while tracing cells.
then transferred to nitrocellulose filters, which were blocked and
incubated with a mouse monoclonal antibody (mAb) specific to WIP
(1/1000, 3D10; a generous gift of Prof. R. Geha, Children’s Hospital,           Miniature Excitatory Postsynaptic Currents
Boston, MA). After exposure to a specific secondary antibody, antibody           Miniature excitatory postsynaptic currents (mEPSC) were recorded
binding was visualized by enhanced chemiluminiscence substrate                  from dissociated hippocampal neurons bathed in artificial cerebro-
(Amersham Biosciences).                                                         spinal fluid (containing 119 mM NaCl, 2.5 mM KCl, 1 mM NaH2PO4, 11
                                                                                mM glucose, 26 mM NaHCO3, 2.5 mM CaCl2, and 1.3 mM MgCl2) in the
                                                                                presence of 1 lM tetrodotoxin and 100 lM picrotoxin (at 29 °C).
Primary Hippocampal Cultures                                                    Spontaneous activity was recorded for 3 min for each cell. mEPSC were




                                                                                                                                                             Downloaded from cercor.oxfordjournals.org at Universidad Autonoma de Madrid. Facultad de Medicina on August 2, 2011
                                                                                identified using pClamp software and corrected by eye on the basis of
Neurons                                                                         their kinetics.
Primary hippocampal cultures were prepared as described (Dotti et al.
1988; Kaech and Banker 2006). Briefly, hippocampi from E18 mouse
embryos (WT and WIP–/–) were washed and digested with 0.25% trypsin             Morphology and Unbiased Stereology
(15 min, 37 °C). The tissue was then dissociated, resuspended in
minimun essential medium with 10% horse serum, and plated on poly-              Perfusion
L-lysine--coated coverslips (1 mg/ml) at a density of 6 3 103 cells/cm2         Three-month-old male mice (WT, n = 6; WIP–/–, n = 6) were
for imaging at early times (up to 24 h after plating) and at a density of 4 3   anesthetized with pentobarbital (0.04 mg/kg) and perfused trans-
103 cells/cm2 for electrophysiological recordings. In some experiments,         cardially with PBS (20 ml) followed by 100 ml 4% PFA (pH 7.4) in the
neurons were allowed to adhere to the substrate (1 h) and then                  same buffer. Brains were postfixed in the same fixative (24 h).
incubated with 2 or 5 lM wiskostatin (BIOMOL International) in
dimethyl sulfoxide (DMSO). In all cases, after 3 h, plating medium was          Volumetric Analyses
replaced with neurobasal medium supplemented with B27 (Gibco). For              Brains were sectioned coronally at a thickness of 50 lm to facilitate
long-term culture, at this time, neuron-including coverslips were               measurements. Strict morphological criteria were used in all mice to
transferred into dishes containing an astrocyte monolayer, with                 determine the boundaries of these brain regions. Briefly, hippocampal
neurons oriented facing the glia but without contacting them.                   outlines encompassed the CA1--3 fields of Ammon’s horn and the
                                                                                subiculum but not the presubiculum or fimbria hippocampus. The
DNA Constructs and Transient Transfection                                       dentate gyrus was measured separately. Starting with one of the
pLVWIP-GFP was obtained by digestion of pcDNA3WIP-GFP (kindly                   sections, randomly selected across brains, 1 of every 6 sections was
provided by Prof. N. Ramesh, Children’s Hospital, Boston, MA) and by            analyzed through the extent of a hemisphere of the brain. One
cloning the insert into pLV; control pLVGFP was generated in a similar          hemisphere of each brain was analyzed. Using this sampling strategy,
manner. Suspended WT (cortical and hippocampal) or WIP–/– (cortical)            7--10 histological sections per brain were analyzed. All volumetric
neurons were nucleofected using a 6 lg DNA/100 ll suspension                    quantifications were performed with an Olympus BX51 with a 1.253
(Amaxa pulser; Lonza, Germany). Cells were maintained in suspension             objective, a motorized XYZ axis computer-controlled stage (Prior
(4 h) to permit exogenous gene expression and then plated for 24 h              Scientific, Houston, TX), a digital video camera (JVC), and Stereo-
before fixation for immunofluorescence analysis.                                  Investigator, a stereology software package (version 8.03, MicroBright-
                                                                                Field). When calculating hippocampus volume, the boundaries were
Immunofluorescence                                                               defined and the volumes determined with Stereo-Investigator software.
At 3 h, 1 and 22 DIV (days in vitro) postplating, cells were fixed in 4%
paraformaldehyde (PFA) in phosphate-buffered saline (PBS) (pH 7.4; 20           Intracellular Injection of Lucifer Yellow
min, room temperature). Cells were then permeablilized with 0.1%                Coronal sections (150 lm) were cut on a vibratome. Sections were
Triton X-100 and labeled with phalloidin-tetramethylrhodamine iso-              prelabeled with 4,6-diamidino-2-phenylindole, and a continuous cur-
thiocyanate (Sigma) and antibodies to tyrosinated a-tubulin (1/400;             rent used to inject individual cells with lucifer yellow (8% in 0.1 M Tris
T9028, Sigma), MAP2 (MT-associated protein, 1/400; 514, Sanchez                 buffer, pH 7.4). Dye was injected into neurons in the dentate gyrus until
Martin et al. 1998), PSD-95 (5 lg/ml; 75-028, NeuroMab), or GFP (1/             the individual dendrites of each cell could be traced to an abrupt end at
100, 11814460; Roche); secondary fluo-rescent antibody or biotinylated           the distal tips and the dendritic spines were readily visible, indicating
antibody and labeled streptavidin were then added. Cells were                   that the dendrites were completely filled. The sections were then
excluded from analysis if they showed obvious features of toxicity,             processed, first with rabbit anti-lucifer yellow (Cajal Institute,
such as neurite fragmentation/blebbing or vacuoles in the cell body.            1/400 000 in stock solution of 2% Bovine serum albumin, 1% Triton
                                                                                X-100, 5% sucrose in PBS) and then with Alexa 488-conjugated
Imaging                                                                         secondary antibody (Molecular Probes, 1/1000, 4 h). Sections were
Confocal images were acquired digitally on a confocal LSM510 Meta               mounted on a glass slide in fresh ProLong Gold antifade reagent
microscope (Zeiss) coupled to an inverted Axiovert 200 microscope               (Invitrogen, Eugene, OR; 24 h, room temperature in the dark) and
(Zeiss). Image stacks (logical size 1024 3 1024 pixels) consisted of 10         sealed with nail polish.
image planes acquired through a 403 (numerical aperture (NA), 1.3) or
a 633 oil-immersion lens (NA 1.4).                                              Confocal Microscopy
   Time-lapse images of phase-contrast fields were captured on an                Imaging was performed on a Leica laser scanning multispectral
inverted Axiovert 200 microscope (Zeiss) equipped with a mono-                  confocal microscope (TCS SP5) using an argon laser. Image stacks
chrome CCD camera and ultrafast filter change. Image stacks (logical             (physical size 76.9 3 76.9 lm, logical size 1024 3 1024 pixels) consisted
size 1024 3 1024 pixels) consisted of 6 image planes acquired through           of 100--350 image planes acquired through a 633 glycerol-immersion


Page 2 of 12 WIP Regulates Neuronal Maturation   d
                                                     Franco et al.
lens (NA, 1.3; working distance, 280 lm; refraction index, 1.45) with                    murine brain and corroborate previous mRNA studies (Ramesh
a calculated optimal zoom factor of 3.2 and a z-step of 0.14 lm (voxel                   et al. 1997; Tsuboi 2006) as well as the data included in the Allen
size, 75.1 3 75.1 3 136.4 nm). These settings and optics represent the                   atlas (http://www.brain-map.org/).
highest resolution currently possible with confocal microscopy. For
each neuron (5 neurons per mouse, 55 neurons total), 1--5 randomly
selected dendrites were scanned from soma to tip, and stacks were                        Brain Hypertrophy in WIP-Deficient Mice
processed with a 3D blind deconvolution algorithm (Autodeblur;                           To assess general effects of WIP deficiency on the mouse brain,
Autoquant, Media Cybernetics) for 10 iterations to reduce the out-of-                    we determined volumes of the hippocampus and of the rest of
focus light.                                                                             the murine forebrain (3-month-old male mice). Contours of the
                                                                                         structures of interest were drawn on Nissl-stained serial sec-
Statistical Analysis                                                                     tions with the aid of Stereo-Investigator software to yield the
Quantification was performed in a blind fashion by independent                            volume of each. There was a significant increase (30%) in
researchers. For statistical analyses, we used the two-tailed Student t-                 forebrain volume in WIP-deficient brains (n = 5) compared
test to compare means, the chi-square test to compare nominal                            with WT littermates (n = 6, Fig. 1B--D and Supplementary Fig.
variables, and the two-way analysis of variance to compare Sholl analysis
                                                                                         1). The hippocampus proper (without the dentate gyrus) and
or the Kolmogorov--Smirnov test to compare cumulative frequency
analysis. Results are shown as mean ± standard error of the mean.                        the dentate gyrus itself were also hypertrophied in WIP–/– brain
                                                                                         (Fig. 1E,F). WIP deficiency thus causes general macrocephaly
                                                                                         that includes hippocampal hypertrophy.
Results




                                                                                                                                                                               Downloaded from cercor.oxfordjournals.org at Universidad Autonoma de Madrid. Facultad de Medicina on August 2, 2011
                                                                                         Enhanced Early Neuronal Development in WIP –/– Neurons
WIP Is Expressed in the WT Mouse Brain                                                   The enlarged brain observed in WIP–/– mice relative to their WT
The expression pattern of WIP messenger RNA (mRNA) suggests                              littermates raised the possibility that the greater brain volume
that WIP is ubiquitously expressed in all mouse tissues, including                       could be due to increased neuritic branching since dendrites
brain (Ramesh et al. 1997; Tsuboi 2006). To date, however, the                           and axon collaterals account for most of the brain volume
levels of endogenous WIP protein have only been analyzed in                              (Acebes and Ferrus 2000). To test this hypothesis at the cellular
                                                                                                           ´
fibroblasts, myoblasts, and hematopoietic-derived cells (reviewed                         level, we examined the effect of WIP deficiency on early
in Anton et al. 2007). To study WIP expression in brain, we probed                       neuronal development. Primary hippocampal neurons from
western blots of cell lysates from 3 regions of the adult mouse brain                    control and WIP–/– embryos were grown at very low density so
(cortex, hippocampus, and olfactory bulb), using the WIP-specific                         that their neurite arbors did not overlap to avoid confusion
mAb 3D10 (Koduru et al. 2007). This mAb recognized 2 protein                             about which neurites protruded from each cell body. Cells
bands in cell lysates from each of the regions examined (Fig. 1A),                       were labeled with anti-tyrosinated a-tubulin and fluorescent
which could represent 2 WIP isoforms and/or be the result of                             phalloidin (Fig. 2A,B). As a measure of neuronal development,
posttranslational modifications of WIP. Neither band was present                          we quantified the fraction of neurons at each developmental
in WIP-deficient tissue. These data demonstrate WIP expression in                         stage at 3 h postplating. At stage 1, the MT-containing soma is




Figure 1. Brain hypertrophy in WIP-deficient mice. (A) Western blot indicates WIP expression in WT mouse brain (cortex, hippocampus, and olfactory bulb; 80 lg total protein/
lane) but not in WIPÀ/À mouse brain. Glyceraldehyde-3-phosphate dehydrogenase labeling confirmed equivalent protein loading in cortical samples. (B) Representative WT (left)
and WIPÀ/À brain (right) at 3 months of age. Note: the enlargement of the WIPÀ/À brain. Scale bar, 4 mm. (C) Representative Nissl staining showing hippocampus enlargement
in WIPÀ/À mice. Scale bar, 500 lm. (D--F) Average volumes of forebrain (D), hippocampus (E), and dentate gyrus (F) are increased in WIPÀ/À mice. P values were determined
with Student’s t-test; *P  0.05; **P  0.01.


                                                                                                                                               Cerebral Cortex Page 3 of 12
Downloaded from cercor.oxfordjournals.org at Universidad Autonoma de Madrid. Facultad de Medicina on August 2, 2011




Figure 2. WIP deficiency accelerates development of dissociated hippocampal neurons. (A) Representative confocal images of WT- and WIPÀ/À-dissociated hippocampal
neurons fixed at 3 h postplating and stained for F-actin (phalloidin-tetramethylrhodamine isothiocyanate, red) and MT (Alexa488-anti-tyrosinated tubulin [TT], green). At this time
point, the WIPÀ/À neuron population showed more advanced development compared with WT neurons. Scale bar, 10 lm. (B) High magnification images of the inset in A. Scale
bar, 5 lm. (C) Neuron classification by developmental stage (3 h postplating) showed significantly lower frequency of WIPÀ/À at developmental stage 1 compared with WT



Page 4 of 12 WIP Regulates Neuronal Maturation     d
                                                       Franco et al.
surrounded by flattened actin-rich lamellipodia, while in stage                         (pLVGFP; Fig. 3C). At 24 h postplating, while 31% of GFP-
2, the lamellipodia are transformed into equidistant neurites                          expressing neurons were in stage 1, the frequency of stage 1
(Dotti et al. 1988). Stage 1+ is defined as cells with incipient                        WIP-GFP--expressing neurons was 80% (Fig. 3D). The de-
tubulin-rich small protrusions. We found a clear difference in                         velopmental delay at 24 h induced by WIP overexpression was
the distribution of these populations (Fig. 2C); there was                             confirmed in cortical embryonic neurons (Supplementary Fig.
a significant decrease (35%) in the frequency of WIP–/–                                 2). This effect was sustained over time, since 48 h after plating,
neurons classified as developmental stage 1 compared with                               22.6% of neurons with increased WIP levels remained at stage 1
WT neurons (n = 4 cultures and n = 150 neurons/genotype,                               compared with none of the GFP-expressing neurons.
Fig. 2C). Conversely, there was an increase (45%) in the                                  We used rescue experiments to confirm the WIP contribution
percentage of WIP-deficient neurons at stage 2. As another                              to neuron development. WIP–/– neurons were nucleofected with
measure of neuronal development, we quantified the number                               a lentiviral-based vector coding for WIP-GFP or control GFP, and
of neurites arising from each neuron at 3 h postplating and                            24 h after plating, the fraction of cells in stage 1 or 2 was
found a significant increase (36%) in the average number of                             quantified (Fig. 3E,F; WIP-GFP--expressing WIP–/– neurons). The
neurites arising from WIP–/– (n = 172) compared with WT                                population distribution of rescued neurons was indistinguishable
neurons (n = 151, Fig. 2D). This finding was confirmed in time-                          from that of WT neurons and differed significantly from that of
lapse images made with phase-contrast microscopy for 3 h                               GFP-expressing WIP–/– neurons. WIP-GFP expression in WIP –/–
after plating (Fig. 2E and Supplementary Videos 1 and 2).                              neurons also reversed the phenotype, as the mean number of
These data suggest that, at 3 h after plating, WIP–/– neurons are                      primary neurites (Fig. 3G and Supplementary Fig. 3) and neuritic




                                                                                                                                                                             Downloaded from cercor.oxfordjournals.org at Universidad Autonoma de Madrid. Facultad de Medicina on August 2, 2011
in a more advanced developmental stage than WT neurons.                                bifurcations (Supplementary Fig. 3) were similar to those of GFP-
This implies enhanced early neuronal development in the                                positive WT neurons.
absence of WIP.                                                                           These findings indicate that WIP acts as a negative regulator
                                                                                       of neuronal differentiation and that neuronal development can
                                                                                       be modulated bidirectionally by WIP levels.
N-WASP Activity and Fine-Tuned WIP Levels Regulate Neuronal
Development
To test whether this accelerated neuronal development                                  Enhanced Dendritic Maturation in WIP –/– Neurons
continues at later developmental times, we quantified the                               To test whether the WIP regulatory role persists in differen-
fraction of neurons that remains at developmental stage 1 at 24 h                      tiated neurons, we also imaged cultured hippocampal neurons
postplating. Cultured hippocampal neurons were fixed and                                at 22 DIV (Fig. 4A). For quantitative analysis of the branching
labeled with anti-tyrosinated a-tubulin antibody and fluores-                           pattern of the neuritic or dendritic tree at the distinct neu-
cent phalloidin and imaged by confocal microscopy. As                                  ronal developmental stages, we traced neurons (1 and 22 DIV)
observed at 3 h postplating, we found a decrease of 48% in                             with Neurolucida software. Sholl analysis was used to measure
the frequency of WIP–/– neurons at developmental stage 1                               the number of neurites/dendrites crossing circles at various
(Fig. 3A left and Fig. 3B), implying that WIP–/– neurons still                         radial distances from the soma (Sholl 1953). Both neuronal
show accelerated development at this time.                                             types showed addition of dendritic branches over time (Fig.
   To determine whether this phenotype depends on the                                  4B,D), although the number of crossings was significantly
activity of N-WASP, an actin NPF with WIP-binding capacity,                            higher in WIP–/– neurons compared with WT neurons at both
we incubated cultures (23 h) with wiskostatin, a selective                             time points. This finding implies that WIP–/– neurons show
reversible N-WASP inhibitor (Peterson et al. 2004). Wiskosta-                          higher ramification of their neuritic (1 DIV) and dendritic
tin is a cell-permeable N-alkylated carbazole derivative that                          arbor (22 DIV, Fig. 4D). Nevertheless, the difference in the
selectively blocks actin filament assembly by binding to N-                             number of crossings between phenotypes, which was 82% at
WASP, stabilizing the autoinhibited conformation, and pre-                             1 DIV, decreased to 45% at 22 DIV (Supplementary Fig. 4A).
venting activation of the Arp2/3 complex (Wegner et al.                                   The increased ramification was reflected in greater total
2008). Wiskostatin produced a dose-dependent increase in                               neuritic or dendritic length of WIP–/– neurons at both time points
the fraction of stage 1 neurons, indicating impairment in                              (Fig. 4C,E), a difference that was also reduced in mature neurons
neuronal development after blockade of N-WASP-dependent                                (from 109% to 40%; Supplementary Fig. 4B). This difference was
filament assembly (Fig. 3A,B) (da Silva and Dotti 2002;                                 even smaller in neurons of adult mice (see below).
Dent et al. 2007). Inhibition of N-WASP activity with 2 lM                                Soma size determines the number of dendrites and associated
wiskostatin induced a phenotype in WIP–/– neurons resem-                               branches in neurons (Kernell and Zwaagstra 1989; Kollins and
bling that of untreated control neurons, and with 5 lM                                 Davenport 2005). To analyze whether WIP deficiency alters
wiskostatin, WIP–/– neurons no longer showed a lower                                   soma area, we quantified this parameter in WT and WIP–/–
frequency of stage 1 neurons (Fig. 3A,B). This finding suggests                         neurons. Similar to our observations for the dendritic arbor,
that N-WASP activity is essential in mediating the accelerated                         soma area was significantly larger in WIP–/– neurons (Fig. 4F,G),
development of WIP–/– neurons.                                                         but this difference attenuated from 40% to 27% with time
   To further evaluate the role of WIP in neurite sprouting, WT                        (Supplementary Fig. 4C). The effect of WIP deficiency on
neurons were nucleofected with lentiviral constructs for over-                         promoting neuritic or dendritic arborization and on soma size is
expression of WIP-GFP (pLVWIP-GFP) or GFP as control                                   thus greater in early developmental stages.


neurons, implying an average more advanced developmental stage for WIPÀ/À neurons. Chi-square test, *P 5 0.0194. (D) The average number of primary neurites arising from
WIPÀ/À somas was significantly larger compared with WT neurons (3 h postplating), again indicating the more advanced developmental stage of WIPÀ/À neurons at this time.
Data derived from quantification of 150 neurons in each experimental group (from 4 separate experiments). Student’s t-test; ***P  0.001. (E) Selected images from a phase-
contrast time-lapse series of control (WT; Supplementary Video1) and WIP-deficient (WIPÀ/À; Supplementary Video2) hippocampal embryonic neurons cultured on poly-L-lysine
over a 3-h period.

                                                                                                                                             Cerebral Cortex Page 5 of 12
Downloaded from cercor.oxfordjournals.org at Universidad Autonoma de Madrid. Facultad de Medicina on August 2, 2011
Figure 3. Inhibition of N-WASP prevents WIPÀ/À phenotype, WIP overexpression delays neurite protrusion, and WIP reexpression reverts the phenotype. (A) Representative images
of dissociated WT and WIPÀ/À hippocampal neurons, alone or wiskostatin-treated, fixed at 24 h postplating and stained for F-actin (phalloidin-tetramethylrhodamine isothiocyanate,
red) and tyrosinated MT (tyrosinated tubulin [TT], green). As at 3 h postplating (Fig. 2), a typical 24-h WIPÀ/À neuron showed more advanced development than a WT neuron. Scale
bar, 10 lm. (B) Percentage of cells at developmental stage 1, alone or wiskostatin-treated. Cells were dissociated and cultured without additives (control), with DMSO, or with
wiskostatin (2 or 5 lM). Wiskostatin (2 lM) caused a significant increase in the frequency of WIPÀ/À neurons at stage 1, equivalent to the frequency for untreated WT neurons. A
higher wiskostatin concentration (5 lM) prevented differentiation of both WT and WIPÀ/À neurons. Student’s t-test; *P  0.05; **P  0.01. (C) Images of phalloidin-stained (red)
hippocampal neurons expressing GFP (left) or GFP-WIP (right) at 24 h postplating. (D) The percentage of cells at developmental stage 1 was greater in WIP-overexpressing cells,
implying that WIP overexpression delays neurite formation. Student’s t-test; *P  0.05. (E) Representative images of cortical control WT neurons nucleofected to express GFP and of
WIPÀ/À neurons nucleofected to express GFP or WIP-GFP. Cortical neurons were fixed at 24 h postplating and stained for F-actin (phalloidin-tetramethylrhodamine isothiocyanate, red)
and anti-GFP (green). A typical WIPÀ/À neuron--expressing WIP-GFP showed similar development to that of a WT neuron. Scale bar, 10 lm. (F) Percentage of cells at developmental
stage 1 after WIP reexpression. The fraction of cells at stage 1 was quantified at 24 h postplating. Distribution of rescued WIPÀ/À neurons was similar to that of WT neurons,
confirming the role of WIP in neuronal development. Student’s t-test; **P  0.01. (G) Mean number of primary neurites per GFP-expressing WT neuron is similar to that of WIP-GFP--
expressing WIPÀ/À neurons and differs significantly from that of WIPÀ/ÀGFP-expressing neurons. Student’s t-test; ***P  0.001; ns, not significant.

Enhanced Neuronal Ramification in Adult WIP –/– Mice                                         (Fig. 5A) and studied the structure of granule cells in the
To test whether WIP deficiency produces a long-lasting effect                                dentate gyrus. Sholl analysis showed significantly more cross-
on neuronal maturation in vivo, we evaluated dendritic archi-                               ings in the distal part of the dendrite in WIP–/– mice (n = 59
tecture in adult (3-month-old) mice after injection of lucifer                              neurons from 6 mice) compared with WT mice (n = 39
yellow into hippocampal neurons of fixed brain sections                                      neurons from 5 mice, Fig. 5B). This increased number of

Page 6 of 12 WIP Regulates Neuronal Maturation     d
                                                       Franco et al.
Downloaded from cercor.oxfordjournals.org at Universidad Autonoma de Madrid. Facultad de Medicina on August 2, 2011




Figure 4. Increased ramification in WIPÀ/À neurons in vitro. (A) Representative images of dissociated WT and WIPÀ/À hippocampal neurons fixed at 1 and 22 DIV and stained
with anti-tyrosinated-a-tubulin (1 DIV) or anti-MAP2 (22 DIV) antibodies. Note the greater ramification of WIPÀ/À neurons at both 1 and 22 DIV. (B and C) Sholl analysis of traced
WT and WIPÀ/À neurons showed more ramification in WIPÀ/À neurons (B), reflected as greater total neuritic or dendritic length for these cells (C). Two-way analysis of variance;
**P  0.01; ***P  0.001. (D) Total number of crossings at 1 and 22 DIV. Note the increase with time in the total number of crossings for both genotypes. (E) Total neuritic
or dendritic length at 1 and 22 DIV. Note the increase over time in the total dendritic length for both genotypes. (F) Representative images of WT and WIPÀ/À somas
demonstrating increased soma surface in WIPÀ/À neurons. (G) WIPÀ/À neuronal somas are larger than WT neuronal somas at 1 and 22 DIV. (D, E, and G) Student’s t-test;
**P  0.01; ***P  0.001.


                                                                                                                                                   Cerebral Cortex Page 7 of 12
crossings reflects a larger number of ramifications at the outer       neuronal development. We found that, in hippocampal primary
molecular layer of the dentate gyrus. As a result of the higher      neurons, loss of WIP accelerates the onset of neuritogenesis and
dendritic ramification, the dendritic length in this layer is also    increases neuritic and dendritic branching. Moreover, lack of
increased in WIP–/– mice (Fig. 5C ), leading to greater total        WIP enhances PSD-95 accumulation in hippocampal dendritic
dendritic length (18%) in WIP–/– mice (Fig. 5D). Cumulative          spines and increases neuronal synaptic activity. Conversely, WIP
frequency analysis indicated that WIP–/– mice have a sub-            overexpression blocks the initiation of neuritogenesis, suggesting
population of highly ramified neurons not detected in WT              that WIP is a bidirectional regulator of neuronal development.
mice (Fig. 5E ). WIP deficiency thus leads to enhanced                WIP Modulates Neuritic Branching During Neuronal
dendritic ramification in the adult mouse. Nonetheless, the           Development
magnitude of change in total dendritic length of the adult
                                                                     Marked dendritic elaboration normally begins in neuronal
WIP–/– mouse is smaller than in early developmental stages.
                                                                     cultures 2--3 days after axonal outgrowth (Nowakowski and Rakic
                                                                     1979; Dotti et al. 1988). Our experiments showed that immature
Enhanced Synaptic Maturation in WIP –/– Neurons
                                                                     WIP–/– neurons begin to emit neurites starting at the first day in
During later stages of neuronal development, dendritic               culture, much earlier than control neurons. After cell cycle exit
arborization and synaptogenesis occur in parallel (Cantallops        and before neuronal polarization, cortical postmitotic neurons
et al. 2000; Cline 2001). The actin cytoskeleton has a pivotal       make a transition through a multipolar stage, when multiple
role in both processes. In the context of synaptogenesis, actin      neurites emerge rapidly from the cell body (Barnes and Polleux
dynamics control the morphogenesis and function of the den-          2009). This morphological transition is also controlled by WIP




                                                                                                                                          Downloaded from cercor.oxfordjournals.org at Universidad Autonoma de Madrid. Facultad de Medicina on August 2, 2011
dritic spines, small actin-rich protrusions from dendritic shafts    during brain development, as GFP in utero electroporation shows
(Ethell and Pasquale 2005). As we found that WIP modulates           a notable increase in the percentage of multipolar cells in WIP–/–
dendritic arborization, we examined the possibility that WIP
                                                                     embryos, in parallel to a reduction in the fraction of round and
deficiency fosters synaptic maturation. We measured the effects
                                                                     unipolar neurons (not shown). In addition, neurites in immature
of WIP deficiency on spontaneous miniature (action-potential          WIP–/– neurons are more branched than neurites of WT neurons.
independent) postsynaptic currents (mEPSC) in dissociated
                                                                     Dendritic branching is also increased in WIP–/– mature neurons,
hippocampal neurons (22 DIV) in the presence of 1 lM
                                                                     although to a lesser extent than at early neuronal developmental
tetrodotoxin (Fig. 6A). Both the amplitude and the frequency         stages (Fig. 4 and Supplementary Fig. 4). Sholl analysis demon-
of mEPSC were significantly increased in WIP–/– compared with
                                                                     strated that the initial morphological features acquired by
WT neurons (n = 17 and 15 neurons, respectively; Fig. 6B,C ).        immature WIP–/– neurons determine the pattern of dendritic
mEPSC amplitude is indicative of the postsynaptic strength of        branching of the more differentiated cells. We therefore suggest
individual functional synapses, whereas mEPSC frequency              that WIP acts as a negative modulator that controls the precise
depends on synapse number and presynaptic properties                 timing of the correct onset of dendritic development. Our results
(Turrigiano et al. 1998; Han and Stevens 2009). WIP deficiency        point to the possibility that some pathologies detected in mature
functionally increases both the strength and possibly the            neurons might have their origin, yet unknown, in altered
number of individual synapses, suggesting that at this time,         processes that occur during early neuronal development.
WIP –/– neurons show either a more mature phenotype or more
abundant and/or enlarged dendritic spines.                           WIP versus Other Negative Regulators of Dendritic
   To examine whether WIP deficiency resulted in an increased         Branching
number of structural synapses, we stained dissociated hippo-         Numerous molecules are involved in the positive control of
campal neurons at the same developmental stage (22 DIV) for the      neuritic or dendritic outgrowth and branching, many of them
postsynaptic marker PSD-95, known to drive maturation of             related to the control of cytoskeleton dynamics (McAllister 2000;
glutamatergic synapses (El-Husseini et al. 2000). WIP–/– neurons     Urbanska et al. 2008). Only a minority of these molecules, such as
showed increased fluorescence intensity of individual PSD-95          PTEN (Kwon et al. 2001; Jaworski et al. 2005) and RhoA (Negishi
puncta, indicating greater PSD-95 accumulation in spines in the      and Katoh 2002; da Silva et al. 2003), have been identified as
absence of WIP (WT, n = 117 spines from 6 neurons; WIP–/–, n =       negative regulators of these processes. Similarly to our findings,
168 spines from 7 neurons, Fig. 6D,E ). Frequency analysis           conditional adult Pten mutant mice show enlargement of cortex
showed that in WIP–/– neurons, staining of some PSD-95--positive     and hippocampus associated with dendritic hypertrophy and
spines is particularly intense, whereas such a population was not    increased soma size (Kwon et al. 2001, 2006). At difference from
found in WT neurons (Fig. 6F ). Positive spine density was           the WIP–/– phenotype observed here, PTEN is not necessary for
nonetheless similar in WIP–/– and WT neurons (Fig. 6G ). As WIP–/–   initiation of neuritogenesis (Lachyankar et al. 2000). The most
neurons are substantially more branched than WT neurons (and         striking difference between WIP- and PTEN-mediated negative
their total dendritic length is therefore increased), the total      regulation thus appears to depend on the temporal window of
number of PSD-95--positive spines is significantly increased in       neuronal development in which both proteins operate. PTEN
WIP–/– neurons (Fig. 6H). These neuronal cultures were               does not control early stages of neuritogenesis, and its influence
maintained over WT astrocytes, excluding possible astrocyte          on neuronal structure increases progressively throughout the
involvement in the synaptic modulation derived from the lack of      animal’s life. In contrast, WIP modulates neuronal development
WIP. These results indicate that WIP deficiency increases the         and synaptogenesis early in postnatal life, and its effects are
number and size of structural and functional synapses.               attenuated as neuronal maturation progresses.
                                                                        Similarly to the absence of WIP, RhoA inhibition increases
                                                                     both the number of primary neurites and total neuritic length
Discussion                                                           in immature dissociated hippocampal neurons, whereas ex-
Here, we present the first report of a role for WIP in the brain,     pression of a constitutively active form of RhoA arrests cells in
showing a specific role for WIP as a negative regulator of murine     stage 1 (da Silva et al. 2003). In mature neurons, transfection of

Page 8 of 12 WIP Regulates Neuronal Maturation   d
                                                     Franco et al.
Downloaded from cercor.oxfordjournals.org at Universidad Autonoma de Madrid. Facultad de Medicina on August 2, 2011




Figure 5. Increased ramification in WIPÀ/À neurons in vivo. (A) Representative projection images of lucifer yellow--injected granule neurons of 3-month-old WT and WIPÀ/À
mice. Scale bar, 25 lm. (B and C) Sholl analysis of traced WT and WIPÀ/À granule neurons showed greater ramification in the distal part of WIPÀ/À neurons (B). This is reflected
as greater dendritic length for these neurons (two-way analysis of variance; *P  0.05) (C). (D) Total dendritic length is greater in neurons from WIPÀ/À mice. Student’s t-test;
*P  0.05. (E) Cumulative frequency curves of total dendritic length indicating a shift toward higher values in neurons from WIPÀ/À mice. Kolmogorov--Smirnov test; *P  0.05.

a dominant-negative RhoA construct does not affect dendritic                               Possible Mechanisms for WIP-Mediated Neurite Sprouting
morphology (Nakayama et al. 2000), whereas WIP–/– neurons                                  and Branching
still show enhanced dendritic arborization. The effect of WIP                              Sprouting of primary neurites and interstitial branching from
on neuronal maturation thus appears to be longer lasting than                              neuritic or dendritic shafts follows the same sequence as
that of other negative modulators of neuritic and dendritic                                cortical cytoskeletal rearrangements (Wu et al. 1999; Dent et al.
outgrowth and branching.                                                                   2007). For the extension of a single filopodium, which is


                                                                                                                                                   Cerebral Cortex Page 9 of 12
Downloaded from cercor.oxfordjournals.org at Universidad Autonoma de Madrid. Facultad de Medicina on August 2, 2011

Figure 6. WIP deficiency increases the strength and the number of individual synapses. (A) Representative trace (calibration: 20 pA, 500 ms) and event (calibration: 10 pA, 2
ms) of mEPSC recorded from a WT and a WIPÀ/À neuron. (B and C) Cumulative frequency representation of the amplitude (B) and frequency (C) of mEPSC, showing significant
shifts to higher values in WIPÀ/À neurons. Kolmogorov--Smirnov test; ***P  0.001. (D) Representative projection confocal images of WT and WIPÀ/À dendrites, stained with
anti-MAP2 (blue) and -PSD-95 antibodies (green). Note that there are intense dendritic spines in WIPÀ/À but not in WT dendrites. (E) Average intensity of PSD-95--positive spines
in WT and WIPÀ/À neurons. Student’s t-test; ***P  0.001. (F) Cumulative frequency representation of the intensity of PSD-95--positive spines. Observe the subpopulation of
particularly intense PSD-95--positive spines in WIPÀ/À but not in WT neurons. Kolmogorov--Smirnov test; ***P  0.001. (G) Sholl analysis of PSD-95--positive spine density in WT
and WIPÀ/À neurons. (H) The number of PSD-95--positive spines was calculated as a function of the distance from soma (Sholl analysis) by multiplying spine density for each
distance by total dendritic length at the same distance. Two-way analysis of variance; ***P  0.001.


subsequently stabilized by MT invasion, F-actin and MT must                                   Our data indicate that WIP modulates neuritogenesis
undergo local depolymerization triggered by extracellular signals                          through its previously described ability to maintain N-WASP
(Acebes and Ferrus 2000; da Silva and Dotti 2002; Luo 2002).
                 ´                                                                         in its autoinhibited state (Martinez-Quiles et al. 2001). N-WASP
WIP inhibits F-actin depolymerization (Martinez-Quiles et al.                              promotes neurite outgrowth and regulates neurite branching
2001), which could explain the neuritogenesis arrest found in                              in hippocampal neurons (Suetsugu, Hattori, et al. 2002; Abe
WIP-overexpressing neurons.                                                                et al. 2003; Pinyol et al. 2007). At the molecular level, N-WASP


Page 10 of 12 WIP Regulates Neuronal Maturation    d
                                                       Franco et al.
acts as a signal integration device that can precisely target actin       Daniel Gallego for electrophysiological signal processing, Jose Ramon
                                                                                                                                         ´    ´
polymerization to membrane sites at which PI(4,5)P2 and                   Valverde for statistical assistance and Catherine Mark for editorial
activated Src kinases and Cdc42 are located (Prehoda et al.               assistance. Conflict of Interest : None declared.
2000; Suetsugu, Miki, et al. 2002). Cdc42 stimulates the actin-
polymerizing activity of N-WASP, creating free barbed ends
                                                                          References
from which actin polymerization can take place (Miki,
Suetsugu, et al. 1998), leading to filopodium formation (Miki,             Abe T, Kato M, Miki H, Takenawa T, Endo T. 2003. Small GTPase Tc10
                                                                              and its homologue RhoT induce N-WASP-mediated long process
Sasaki, et al. 1998). In the absence of WIP, N-WASP would be
                                                                              formation and neurite outgrowth. J Cell Sci. 116:155--168.
more easily released from inhibition and be hyperactivated to             Acebes A, Ferrus A. 2000. Cellular and molecular features of axon
                                                                                            ´
initiate premature filopodium formation. This interpretation is                collaterals and dendrites. Trends Neurosci. 23:557--565.
supported by pharmacological inhibition of N-WASP by wiskos-              Anton IM, de la Fuente MA, Sims TN, Freeman S, Ramesh N, Hartwig JH,
tatin, which blocked the accelerated neuritogenesis seen in WIP-              Dustin ML, Geha RS. 2002. WIP deficiency reveals a differential role
deficient neurons (Fig. 3).                                                    for WIP and the actin cytoskeleton in T and B cell activation.
                                                                              Immunity. 16:193--204.
                                                                          Anton IM, Jones GE, Wandosell F, Geha R, Ramesh N. 2007. WASP-
WIP Modulates Synaptic Activity
                                                                              interacting protein (WIP): working in polymerisation and much
We report here that mEPSC amplitude and frequency are                         more. Trends Cell Biol. 17:555--562.
increased in WIP–/– neurons, suggesting both stronger AMPA                Barnes AP, Polleux F. 2009. Establishment of axon-dendrite polarity in
(alpha-amino-3-hydroxy-5-methyl-4-isoxazole propionic acid)                   developing neurons. Annu Rev Neurosci. 32:347--381.




                                                                                                                                                         Downloaded from cercor.oxfordjournals.org at Universidad Autonoma de Madrid. Facultad de Medicina on August 2, 2011
receptor--mediated synaptic currents and more functional                  Bradke F, Dotti CG. 1999. The role of local actin instability in axon
synapses in these cells (Fig. 6). In addition, we describe an                 formation. Science. 283:1931--1934.
increase in PSD-95 accumulation at individual spines in WIP–/–            Cantallops I, Haas K, Cline HT. 2000. Postsynaptic CPG15 promotes
                                                                              synaptic maturation and presynaptic axon arbor elaboration in vivo.
neurons. These 2 events are probably linked since PSD-95                      Nat Neurosci. 3:1004--1011.
drives the maturation of excitatory synapses with the in-                 Cline HT. 2001. Dendritic arbor development and synaptogenesis. Curr
corporation of AMPA receptors (El-Husseini et al. 2000).                      Opin Neurobiol. 11:118--126.
Indeed, the contribution of AMPA receptors to synaptic                    Conde C, Caceres A. 2009. Microtubule assembly, organization and
transmission increases gradually with neuronal maturation                     dynamics in axons and dendrites. Nat Rev Neurosci. 10:319--332.
(Mammen et al. 1997; Petralia et al. 1999). The increase in               Curcio C, Pannellini T, Lanzardo S, Forni G, Musiani P, Anton IM. 2007.
PSD-95 accumulation at individual WIP–/– spines might suggest                 WIP null mice display a progressive immunological disorder that
                                                                              resembles Wiskott-Aldrich Syndrome. J Pathol. 211:67--75.
that the PSD (and therefore the synapse) is enlarged in the               da Silva JS, Dotti CG. 2002. Breaking the neuronal sphere: regulation of
absence of WIP. Given that the synaptic area correlates                       the actin cytoskeleton in neuritogenesis. Nat Rev Neurosci.
positively with the number of synaptic AMPA receptors (Nusser                 3:694--704.
et al. 1998), our electrophysiological findings are strengthened           da Silva JS, Medina M, Zuliani C, Di Nardo A, Witke W, Dotti CG. 2003.
by the morphological analysis of PSD-95 in spines.                            RhoA/ROCK regulation of neuritogenesis via profilin IIa-mediated
   In conclusion, this study sheds light on the molecular events              control of actin stability. J Cell Biol. 162:1267--1279.
that control early neuronal development by presenting WIP as              de Curtis I. 2007. Intracellular mechanisms of neuritogenesis. New
                                                                              York: Springer.
a previously undescribed regulator that prevents premature
                                                                          Dent EW, Kwiatkowski AV, Mebane LM, Philippar U, Barzik M,
dendritic and synaptic maturation.                                            Rubinson DA, Gupton S, Van Veen JE, Furman C, Zhang J, et al.
                                                                              2007. Filopodia are required for cortical neurite initiation. Nat Cell
                                                                              Biol. 9:1347--1359.
Supplementary Material
                                                                          Dong X, Patino-Lopez G, Candotti F, Shaw S. 2007. Structure-function
Supplementary material     can   be   found   at:   http://www.cercor.        analysis of the WIP role in T cell receptor-stimulated NFAT
oxfordjournals.org/.                                                          activation: evidence that WIP-WASP dissociation is not required
                                                                              and that the WIP NH2 terminus is inhibitory. J Biol Chem.
                                                                              282:30303--30310.
Funding                                                                   Dotti CG, Sullivan CA, Banker GA. 1988. The establishment of polarity
Grants from Consejo Superior de Investigaciones Cientı´ ficas-                 by hippocampal neurons in culture. J Neurosci. 8:1454--1468.
Comunidad de Madrid (CCG08-CSIC/SAL-3471), CSIC (PIE2-                    El-Husseini AE, Schnell E, Chetkovich DM, Nicoll RA, Bredt DS. 2000.
00720I002), and the Spanish Ministry of Education and Science                 PSD-95 involvement in maturation of excitatory synapses. Science.
                                                                              290:1364--1368.
(BFU2007-64144 and BFU2010-21374) to I.M.A., from Centro de
                                                                          Ethell IM, Pasquale EB. 2005. Molecular mechanisms of dendritic spine
Investigacion Biomedica en Red Enfermedades Neurodegener-
            ´        ´                                                        development and remodeling. Prog Neurobiol. 75:161--205.
ativas (Instituto de Salud Carlos III), the Plan Nacional DGCYT           Ferreira A, Caceres A. 1989. The expression of acetylated microtubules
(SAF2009-12249-C02-01) to F.W. and SAF2010-15676 to S.K.,                     during axonal and dendritic growth in cerebellar macroneurons
and by an institutional grant from the Fundacion Ramon Areces.
                                                 ´     ´                      which develop in vitro. Brain Res Dev Brain Res. 49:205--213.
A.F. was a recipient of an FPU MEC fellowship (AP2005-3405),              Han EB, Stevens CF. 2009. Development regulates a switch between
I.B. held a contract from the Comunidad Autonoma de Madrid
                                                   ´                          post- and presynaptic strengthening in response to activity
                                                                              deprivation. Proc Natl Acad Sci U S A. 106:10817--10822.
and S.K., a Ramon y Cajal contract.
                  ´
                                                                          Ho HY, Rohatgi R, Lebensohn AM, Le M, Li J, Gygi SP, Kirschner MW.
                                                                              2004. Toca-1 mediates Cdc42-dependent actin nucleation by
Notes                                                                         activating the N-WASP-WIP complex. Cell. 118:203--216.
We thank Chiara Ragazzini and Sonia Perez for their excellent technical
                                        ´                                 Jaworski J, Spangler S, Seeburg DP, Hoogenraad CC, Sheng M. 2005. Control
assistance and Javier de Felipe for his contribution to the morphomet-        of dendritic arborization by the phosphoinositide-3’-kinase-Akt-
ric analysis with NeuroLucida Software. We are grateful to Raif Geha          mammalian target of rapamycin pathway. J Neurosci. 25:11300--11312.
and Narayanaswamy Ramesh for the 3D10 mAb and to Lola Ledesma             Kaech S, Banker G. 2006. Culturing hippocampal neurons. Nat Protoc.
and Carlos Dotti for helpful advice on the manuscript. We acknowledge         1:2406--2415.


                                                                                                                         Cerebral Cortex Page 11 of 12
Kakimoto T, Katoh H, Negishi M. 2004. Identification of splicing           Peterson FC, Deng Q, Zettl M, Prehoda KE, Lim WA, Way M,
   variants of Rapostlin, a novel RND2 effector that interacts with          Volkman BF. 2007. Multiple WASP-interacting protein recognition
   neural Wiskott-Aldrich syndrome protein and induces neurite               motifs are required for a functional interaction with N-WASP. J Biol
   branching. J Biol Chem. 279:14104--14110.                                 Chem. 282:8446--8453.
Kernell D, Zwaagstra B. 1989. Dendrites of cat’s spinal motoneurones:     Peterson JR, Bickford LC, Morgan D, Kim AS, Ouerfelli O,
   relationship between stem diameter and predicted input conduc-            Kirschner MW, Rosen MK. 2004. Chemical inhibition of N-WASP
   tance. J Physiol. 413:255--269.                                           by stabilization of a native autoinhibited conformation. Nat Struct
Kettner A, Kumar L, Anton IM, Sasahara Y, de la Fuente M, Pivniouk VI,       Mol Biol. 11:747--755.
   Falet H, Hartwig JH, Geha RS. 2004. WIP regulates signaling via the    Petralia RS, Esteban JA, Wang YX, Partridge JG, Zhao HM, Wenthold RJ,
   high affinity receptor for immunoglobulin E in mast cells. J Exp Med.      Malinow R. 1999. Selective acquisition of AMPA receptors over
   199:357--368.
                                                                             postnatal development suggests a molecular basis for silent
Koduru S, Massaad M, Wilbur C, Kumar L, Geha R, Ramesh N. 2007. A
                                                                             synapses. Nat Neurosci. 2:31--36.
   novel anti-WIP monoclonal antibody detects an isoform of WIP that
                                                                          Pinyol R, Haeckel A, Ritter A, Qualmann B, Kessels MM. 2007.
   lacks the WASP binding domain. Biochem Biophys Res Commun.
                                                                             Regulation of N-WASP and the Arp2/3 complex by Abp1 controls
   353:875--881.
Kollins KM, Davenport RW. 2005. Branching morphogenesis in                   neuronal morphology. PLoS One. 2:e400.
   vertebrate neurons. Austin (TX): Landes Bioscience.                    Prehoda KE, Scott JA, Mullins RD, Lim WA. 2000. Integration of multiple
Kwon CH, Luikart BW, Powell CM, Zhou J, Matheny SA, Zhang W, Li Y,           signals through cooperative regulation of the N-WASP-Arp2/3
   Baker SJ, Parada LF. 2006. Pten regulates neuronal arborization and       complex. Science. 290:801--806.
   social interaction in mice. Neuron. 50:377--388.                       Ramesh N, Anton IM, Hartwig JH, Geha RS. 1997. WIP, a protein
Kwon CH, Zhu X, Zhang J, Knoop LL, Tharp R, Smeyne RJ,                       associated with wiskott-aldrich syndrome protein, induces actin
                                                                             polymerization and redistribution in lymphoid cells. Proc Natl Acad




                                                                                                                                                       Downloaded from cercor.oxfordjournals.org at Universidad Autonoma de Madrid. Facultad de Medicina on August 2, 2011
   Eberhart CG, Burger PC, Baker SJ. 2001. Pten regulates neuronal
   soma size: a mouse model of Lhermitte-Duclos disease. Nat Genet.          Sci U S A. 94:14671--14676.
   29:404--411.                                                           Sanchez Martin C, Diaz-Nido J, Avila J. 1998. Regulation of a site-specific
Lachyankar MB, Sultana N, Schonhoff CM, Mitra P, Poluha W,                   phosphorylation of the microtubule-associated protein 2 during the
   Lambert S, Quesenberry PJ, Litofsky NS, Recht LD, Nabi R, et al.          development of cultured neurons. Neuroscience. 87:861--870.
   2000. A role for nuclear PTEN in neuronal differentiation.             Sholl DA. 1953. Dendritic organization in the neurons of the visual and
   J Neurosci. 20:1404--1413.                                                motor cortices of the cat. J Anat. 87:387--406.
Luo L. 2002. Actin cytoskeleton regulation in neuronal morphogenesis      Suetsugu S, Hattori M, Miki H, Tezuka T, Yamamoto T, Mikoshiba K,
   and structural plasticity. Annu Rev Cell Dev Biol. 18:601--635.           Takenawa T. 2002. Sustained activation of N-WASP through
Mammen AL, Huganir RL, O’Brien RJ. 1997. Redistribution and sta-             phosphorylation is essential for neurite extension. Dev Cell.
   bilization of cell surface glutamate receptors during synapse             3:645--658.
   formation. J Neurosci. 17:7351--7358.                                  Suetsugu S, Miki H, Takenawa T. 2002. Spatial and temporal regulation
Martinez-Quiles N, Rohatgi R, Anton IM, Medina M, Saville SP, Miki H,        of actin polymerization for cytoskeleton formation through Arp2/3
   Yamaguchi H, Takenawa T, Hartwig JH, Geha RS, et al. 2001. WIP            complex and WASP/WAVE proteins. Cell Motil Cytoskeleton.
   regulates N-WASP-mediated actin polymerization and filopodium              51:113--122.
   formation. Nat Cell Biol. 3:484--491.                                  Tsuboi S. 2006. A complex of Wiskott-Aldrich syndrome protein with
McAllister AK. 2000. Cellular and molecular mechanisms of dendrite           mammalian verprolins plays an important role in monocyte
   growth. Cereb Cortex. 10:963--973.
                                                                             chemotaxis. J Immunol. 176:6576--6585.
Miki H, Sasaki T, Takai Y, Takenawa T. 1998. Induction of filopodium
                                                                          Turrigiano GG, Leslie KR, Desai NS, Rutherford LC, Nelson SB. 1998.
   formation by a WASP-related actin-depolymerizing protein N-WASP.
                                                                             Activity-dependent scaling of quantal amplitude in neocortical
   Nature. 391:93--96.
                                                                             neurons. Nature. 391:892--896.
Miki H, Suetsugu S, Takenawa T. 1998. WAVE, a novel WASP-family
                                                                          Urbanska M, Blazejczyk M, Jaworski J. 2008. Molecular basis of dendritic
   protein involved in actin reorganization induced by Rac. EMBO J.
   17:6932--6941.                                                            arborization. Acta Neurobiol Exp (Wars). 68:264--288.
Nakayama AY, Harms MB, Luo L. 2000. Small GTPases Rac and Rho in          Volkman BF, Prehoda KE, Scott JA, Peterson FC, Lim WA. 2002. Structure
   the maintenance of dendritic spines and branches in hippocampal           of the N-WASP EVH1 domain-WIP complex: insight into the
   pyramidal neurons. J Neurosci. 20:5329--5338.                             molecular basis of Wiskott-Aldrich Syndrome. Cell. 111:565--576.
Negishi M, Katoh H. 2002. Rho family GTPases as key regulators for        Wegner AM, Nebhan CA, Hu L, Majumdar D, Meier KM, Weaver AM,
   neuronal network formation. J Biochem. 132:157--166.                      Webb DJ. 2008. N-wasp and the arp2/3 complex are critical
Nowakowski RS, Rakic P. 1979. The mode of migration of neurons to            regulators of actin in the development of dendritic spines and
   the hippocampus: a Golgi and electron microscopic analysis in             synapses. J Biol Chem. 283:15912--15920.
   foetal rhesus monkey. J Neurocytol. 8:697--718.                        Wiggin GR, Fawcett JP, Pawson T. 2005. Polarity proteins in axon
Nusser Z, Lujan R, Laube G, Roberts JD, Molnar E, Somogyi P. 1998. Cell      specification and synaptogenesis. Dev Cell. 8:803--816.
   type and pathway dependence of synaptic AMPA receptor number           Wu GY, Zou DJ, Rajan I, Cline H. 1999. Dendritic dynamics in vivo
   and variability in the hippocampus. Neuron. 21:545--559.                  change during neuronal maturation. J Neurosci. 19:4472--4483.




Page 12 of 12 WIP Regulates Neuronal Maturation   d
                                                      Franco et al.

More Related Content

What's hot

Targeted foreign gene expression in spinal cord
Targeted foreign gene expression in spinal cordTargeted foreign gene expression in spinal cord
Targeted foreign gene expression in spinal cordAhmed Madni
 
Evolution of transposons, genomes, and organisms (Hertweck Fall 2014)
Evolution of transposons, genomes, and organisms (Hertweck Fall 2014)Evolution of transposons, genomes, and organisms (Hertweck Fall 2014)
Evolution of transposons, genomes, and organisms (Hertweck Fall 2014)Kate Hertweck
 
Next Generation Sequencing
Next Generation SequencingNext Generation Sequencing
Next Generation SequencingAamir Wahab
 
artificial gene synthesis
artificial gene synthesisartificial gene synthesis
artificial gene synthesisImdad Takkar
 
Fall 2016 Research Summation
Fall 2016 Research SummationFall 2016 Research Summation
Fall 2016 Research SummationAllie Poles
 
Molecular medicine presentation
Molecular medicine presentationMolecular medicine presentation
Molecular medicine presentationUmme Habiba
 
When is a genome finished?
When is a genome finished? When is a genome finished?
When is a genome finished? Keith Bradnam
 
Understanding Junk DNA
Understanding Junk DNAUnderstanding Junk DNA
Understanding Junk DNADouglas Arndt
 
seminar on new technologies of cell and molecular biology
seminar on new technologies of cell and molecular biologyseminar on new technologies of cell and molecular biology
seminar on new technologies of cell and molecular biologyBiswajit Deka
 
New insights into the human genome by ENCODE project
New insights into the human genome by ENCODE project New insights into the human genome by ENCODE project
New insights into the human genome by ENCODE project Senthil Natesan
 
Whole genome sequence.
Whole genome sequence.Whole genome sequence.
Whole genome sequence.jayalakshmi311
 
NEURAL STEM CELLS AND NEUROGENESIS
NEURAL STEM CELLS AND NEUROGENESISNEURAL STEM CELLS AND NEUROGENESIS
NEURAL STEM CELLS AND NEUROGENESISOindrillaDuttaGupta
 
Central dogma of molecular biology 20 11-2015
Central dogma of molecular biology 20 11-2015Central dogma of molecular biology 20 11-2015
Central dogma of molecular biology 20 11-2015Yahya Noori, Ph.D
 
Neural crest cell migration-Cell tracing techniques.
Neural crest cell migration-Cell tracing techniques.Neural crest cell migration-Cell tracing techniques.
Neural crest cell migration-Cell tracing techniques.sanjeev jain
 

What's hot (20)

Targeted foreign gene expression in spinal cord
Targeted foreign gene expression in spinal cordTargeted foreign gene expression in spinal cord
Targeted foreign gene expression in spinal cord
 
Evolution of transposons, genomes, and organisms (Hertweck Fall 2014)
Evolution of transposons, genomes, and organisms (Hertweck Fall 2014)Evolution of transposons, genomes, and organisms (Hertweck Fall 2014)
Evolution of transposons, genomes, and organisms (Hertweck Fall 2014)
 
Zinc finger
Zinc fingerZinc finger
Zinc finger
 
Junk DNA And The Human Genome
Junk DNA And The Human GenomeJunk DNA And The Human Genome
Junk DNA And The Human Genome
 
Next Generation Sequencing
Next Generation SequencingNext Generation Sequencing
Next Generation Sequencing
 
artificial gene synthesis
artificial gene synthesisartificial gene synthesis
artificial gene synthesis
 
Sandhya Gangaraju
Sandhya GangarajuSandhya Gangaraju
Sandhya Gangaraju
 
Fall 2016 Research Summation
Fall 2016 Research SummationFall 2016 Research Summation
Fall 2016 Research Summation
 
Molecular medicine presentation
Molecular medicine presentationMolecular medicine presentation
Molecular medicine presentation
 
Sandhya Gangaraju
Sandhya GangarajuSandhya Gangaraju
Sandhya Gangaraju
 
When is a genome finished?
When is a genome finished? When is a genome finished?
When is a genome finished?
 
Understanding Junk DNA
Understanding Junk DNAUnderstanding Junk DNA
Understanding Junk DNA
 
seminar on new technologies of cell and molecular biology
seminar on new technologies of cell and molecular biologyseminar on new technologies of cell and molecular biology
seminar on new technologies of cell and molecular biology
 
Whole genome sequencing
Whole genome sequencingWhole genome sequencing
Whole genome sequencing
 
New insights into the human genome by ENCODE project
New insights into the human genome by ENCODE project New insights into the human genome by ENCODE project
New insights into the human genome by ENCODE project
 
Genomics and Plant Genomics
Genomics and Plant GenomicsGenomics and Plant Genomics
Genomics and Plant Genomics
 
Whole genome sequence.
Whole genome sequence.Whole genome sequence.
Whole genome sequence.
 
NEURAL STEM CELLS AND NEUROGENESIS
NEURAL STEM CELLS AND NEUROGENESISNEURAL STEM CELLS AND NEUROGENESIS
NEURAL STEM CELLS AND NEUROGENESIS
 
Central dogma of molecular biology 20 11-2015
Central dogma of molecular biology 20 11-2015Central dogma of molecular biology 20 11-2015
Central dogma of molecular biology 20 11-2015
 
Neural crest cell migration-Cell tracing techniques.
Neural crest cell migration-Cell tracing techniques.Neural crest cell migration-Cell tracing techniques.
Neural crest cell migration-Cell tracing techniques.
 

Viewers also liked

Alzforum print news
Alzforum   print newsAlzforum   print news
Alzforum print newsshiraknafo
 
Knafo et al. p lo s b
Knafo et al. p lo s bKnafo et al. p lo s b
Knafo et al. p lo s bshiraknafo
 
In tech amygdala-in_alzheimer_s_disease
In tech amygdala-in_alzheimer_s_diseaseIn tech amygdala-in_alzheimer_s_disease
In tech amygdala-in_alzheimer_s_diseaseshiraknafo
 
Morphological alterations to neurons of the amygdala
Morphological alterations to neurons of the amygdalaMorphological alterations to neurons of the amygdala
Morphological alterations to neurons of the amygdalashiraknafo
 
Cereb. cortex 2009-knafo-586-92
Cereb. cortex 2009-knafo-586-92Cereb. cortex 2009-knafo-586-92
Cereb. cortex 2009-knafo-586-92shiraknafo
 
Role of CMAs under companies Act.
Role of CMAs under companies Act.Role of CMAs under companies Act.
Role of CMAs under companies Act.Robin Singh
 
Spines, plasticity, and cognition in alzheimer model mice.
Spines, plasticity, and cognition in alzheimer model mice.Spines, plasticity, and cognition in alzheimer model mice.
Spines, plasticity, and cognition in alzheimer model mice.shiraknafo
 

Viewers also liked (10)

Pten jurado
Pten juradoPten jurado
Pten jurado
 
Alzforum print news
Alzforum   print newsAlzforum   print news
Alzforum print news
 
Pip3
Pip3Pip3
Pip3
 
App ca1 paula
App ca1 paulaApp ca1 paula
App ca1 paula
 
Knafo et al. p lo s b
Knafo et al. p lo s bKnafo et al. p lo s b
Knafo et al. p lo s b
 
In tech amygdala-in_alzheimer_s_disease
In tech amygdala-in_alzheimer_s_diseaseIn tech amygdala-in_alzheimer_s_disease
In tech amygdala-in_alzheimer_s_disease
 
Morphological alterations to neurons of the amygdala
Morphological alterations to neurons of the amygdalaMorphological alterations to neurons of the amygdala
Morphological alterations to neurons of the amygdala
 
Cereb. cortex 2009-knafo-586-92
Cereb. cortex 2009-knafo-586-92Cereb. cortex 2009-knafo-586-92
Cereb. cortex 2009-knafo-586-92
 
Role of CMAs under companies Act.
Role of CMAs under companies Act.Role of CMAs under companies Act.
Role of CMAs under companies Act.
 
Spines, plasticity, and cognition in alzheimer model mice.
Spines, plasticity, and cognition in alzheimer model mice.Spines, plasticity, and cognition in alzheimer model mice.
Spines, plasticity, and cognition in alzheimer model mice.
 

Similar to Cer cor11 franco-cercor-bhr199

Katoh_et_al-2015-Brain_and_Behavior
Katoh_et_al-2015-Brain_and_BehaviorKatoh_et_al-2015-Brain_and_Behavior
Katoh_et_al-2015-Brain_and_BehaviorRhea Kimpo
 
mottin-2011-Jcbfm-nature-publishing
mottin-2011-Jcbfm-nature-publishingmottin-2011-Jcbfm-nature-publishing
mottin-2011-Jcbfm-nature-publishingStéphane MOTTIN
 
Cellvizio® LAB Fibered Fluorescence Microscope and Microdialysis Research
Cellvizio®  LAB Fibered Fluorescence Microscope and  Microdialysis Research Cellvizio®  LAB Fibered Fluorescence Microscope and  Microdialysis Research
Cellvizio® LAB Fibered Fluorescence Microscope and Microdialysis Research FUJIFILM VisualSonics Inc.
 
Peripheral neuropathyumbilicalcord
Peripheral neuropathyumbilicalcordPeripheral neuropathyumbilicalcord
Peripheral neuropathyumbilicalcordComprehensiveBiologi
 
Development and implementation of a novel interactome platform in studies on ...
Development and implementation of a novel interactome platform in studies on ...Development and implementation of a novel interactome platform in studies on ...
Development and implementation of a novel interactome platform in studies on ...Ewelina Maliszewska-Cyna, PhD
 
Mary Lin Paper
Mary Lin PaperMary Lin Paper
Mary Lin PaperMary Lin
 
Studying Epilepsy in Awake Head-Fixed Mice Using Microscopy, Electrophysiolog...
Studying Epilepsy in Awake Head-Fixed Mice Using Microscopy, Electrophysiolog...Studying Epilepsy in Awake Head-Fixed Mice Using Microscopy, Electrophysiolog...
Studying Epilepsy in Awake Head-Fixed Mice Using Microscopy, Electrophysiolog...InsideScientific
 
Heraud 2009 Neuro Image
Heraud 2009  Neuro ImageHeraud 2009  Neuro Image
Heraud 2009 Neuro Imageuvperson
 
The Role Of The Cytoskeleton During Neuronal Polarization
The Role Of The Cytoskeleton During Neuronal PolarizationThe Role Of The Cytoskeleton During Neuronal Polarization
The Role Of The Cytoskeleton During Neuronal PolarizationDalí Mb
 
Neuroscience: Transforming Visual Percepts into Memories
Neuroscience: Transforming Visual Percepts into MemoriesNeuroscience: Transforming Visual Percepts into Memories
Neuroscience: Transforming Visual Percepts into Memoriesmustafa sarac
 
Poster sot 2015_am_final
Poster sot 2015_am_finalPoster sot 2015_am_final
Poster sot 2015_am_finalPete Shuster
 
Neuronal survival and programmed cell death.pptx
Neuronal survival and programmed cell death.pptxNeuronal survival and programmed cell death.pptx
Neuronal survival and programmed cell death.pptxNitish kumar
 
Neutrophil extracellular trap
Neutrophil extracellular trapNeutrophil extracellular trap
Neutrophil extracellular trapNainshi Bhatt
 

Similar to Cer cor11 franco-cercor-bhr199 (20)

Katoh_et_al-2015-Brain_and_Behavior
Katoh_et_al-2015-Brain_and_BehaviorKatoh_et_al-2015-Brain_and_Behavior
Katoh_et_al-2015-Brain_and_Behavior
 
nikolakopoulou et al., 2010_final
nikolakopoulou et al., 2010_finalnikolakopoulou et al., 2010_final
nikolakopoulou et al., 2010_final
 
Edgardo Arroyo CV
Edgardo Arroyo CVEdgardo Arroyo CV
Edgardo Arroyo CV
 
mottin-2011-Jcbfm-nature-publishing
mottin-2011-Jcbfm-nature-publishingmottin-2011-Jcbfm-nature-publishing
mottin-2011-Jcbfm-nature-publishing
 
6926.full
6926.full6926.full
6926.full
 
Summer Poster_PDF
Summer Poster_PDF Summer Poster_PDF
Summer Poster_PDF
 
Cellvizio® LAB Fibered Fluorescence Microscope and Microdialysis Research
Cellvizio®  LAB Fibered Fluorescence Microscope and  Microdialysis Research Cellvizio®  LAB Fibered Fluorescence Microscope and  Microdialysis Research
Cellvizio® LAB Fibered Fluorescence Microscope and Microdialysis Research
 
Peripheral neuropathyumbilicalcord
Peripheral neuropathyumbilicalcordPeripheral neuropathyumbilicalcord
Peripheral neuropathyumbilicalcord
 
Development and implementation of a novel interactome platform in studies on ...
Development and implementation of a novel interactome platform in studies on ...Development and implementation of a novel interactome platform in studies on ...
Development and implementation of a novel interactome platform in studies on ...
 
Hu et al., 2005
Hu et al., 2005Hu et al., 2005
Hu et al., 2005
 
Mary Lin Paper
Mary Lin PaperMary Lin Paper
Mary Lin Paper
 
Studying Epilepsy in Awake Head-Fixed Mice Using Microscopy, Electrophysiolog...
Studying Epilepsy in Awake Head-Fixed Mice Using Microscopy, Electrophysiolog...Studying Epilepsy in Awake Head-Fixed Mice Using Microscopy, Electrophysiolog...
Studying Epilepsy in Awake Head-Fixed Mice Using Microscopy, Electrophysiolog...
 
Heraud 2009 Neuro Image
Heraud 2009  Neuro ImageHeraud 2009  Neuro Image
Heraud 2009 Neuro Image
 
The Role Of The Cytoskeleton During Neuronal Polarization
The Role Of The Cytoskeleton During Neuronal PolarizationThe Role Of The Cytoskeleton During Neuronal Polarization
The Role Of The Cytoskeleton During Neuronal Polarization
 
Optogenetics
OptogeneticsOptogenetics
Optogenetics
 
ISX9 - Karthik Gopalakrishnan
ISX9 - Karthik GopalakrishnanISX9 - Karthik Gopalakrishnan
ISX9 - Karthik Gopalakrishnan
 
Neuroscience: Transforming Visual Percepts into Memories
Neuroscience: Transforming Visual Percepts into MemoriesNeuroscience: Transforming Visual Percepts into Memories
Neuroscience: Transforming Visual Percepts into Memories
 
Poster sot 2015_am_final
Poster sot 2015_am_finalPoster sot 2015_am_final
Poster sot 2015_am_final
 
Neuronal survival and programmed cell death.pptx
Neuronal survival and programmed cell death.pptxNeuronal survival and programmed cell death.pptx
Neuronal survival and programmed cell death.pptx
 
Neutrophil extracellular trap
Neutrophil extracellular trapNeutrophil extracellular trap
Neutrophil extracellular trap
 

Cer cor11 franco-cercor-bhr199

  • 1. Cerebral Cortex Advance Access published August 1, 2011 Cerebral Cortex doi:10.1093/cercor/bhr199 WIP Is a Negative Regulator of Neuronal Maturation and Synaptic Activity A. Franco1, S. Knafo2,3, I. Banon-Rodriguez1, P. Merino-Serrais2, I. Fernaud-Espinosa2, M. Nieto1, J.J. Garrido2,3,4, J.A. Esteban3, F. Wandosell3,4 and I.M. Anton1,4 1 Centro Nacional de Biotecnologı´ a (CNB-CSIC), 28049 Madrid, Spain, 2Instituto Cajal (CSIC), 28002 Madrid, Spain, 3Centro de Biologı´ a Molecular ‘‘Severo Ochoa’’ (CSIC-UAM), Universidad Autonoma de Madrid, 28049 Madrid, Spain and 4CIBERNED, Centro ´ Investigacion Biomedica en Red de Enfermedades Neurodegenerativas, 28031 Spain ´ ´ A. Franco and S. Knafo contributed equally to this work Address correspondence to Dr Ines M. Anton, Cellular and Molecular Department, Centro Nacional de Biotecnologı´ a (CNB-CSIC), Darwin 3, 28049 ´ ´ Madrid, Spain. Email: ianton@cnb.csic.es. Wiskott--Aldrich syndrome protein (WASP) --interacting protein factors (NPF), such as neural Wiskott-Aldrich syndrome protein (WIP) is an actin-binding protein involved in the regulation of actin (N-WASP). The Arp2/3 complex nucleates actin, inducing polymerization in cells, such as fibroblasts and lymphocytes. branching and elongation, and with N-WASP, it mediates Despite its recognized function in non-neuronal cells, the role of neurite elongation (Suetsugu, Hattori, et al. 2002; Pinyol et al. WIP in the central nervous system has not been examined 2007) and neurite branching (Kakimoto et al. 2004). N-WASP Downloaded from cercor.oxfordjournals.org at Universidad Autonoma de Madrid. Facultad de Medicina on August 2, 2011 previously. We used WIP-deficient mice to examine WIP function interacts with WASP-interacting protein (WIP), a broadly both in vivo and in vitro. We report here that WIP2/2 hippocampal expressed proline-rich protein that regulates N-WASP function neurons exhibit enlargement of somas as well as overgrowth of as NPF and whose deficiency modifies actin polymerization neuritic and dendritic branches that are more evident in early kinetics and the density of the subcortical actin network developmental stages. Dendritic arborization and synaptogenesis, (Anton et al. 2007). Through WASP/N-WASP--dependent or which includes generation of postsynaptic dendritic spines, are --independent mechanisms, WIP participates in a wide variety actin-dependent processes that occur in parallel at later stages. of cellular functions, including signaling, endocytosis, and actin WIP deficiency also increases the amplitude and frequency of cytoskeleton remodeling (Anton et al. 2007). WIP deficiency in miniature excitatory postsynaptic currents, suggesting that WIP2/2 mice alters the immune response, reducing T and mast cell neurons have more mature synapses than wild-type neurons. These activity and increasing B cell function (Anton et al. 2002; findings reveal WIP as a previously unreported regulator of Kettner et al. 2004). Moreover, WIP null mice have a pro- neuronal maturation and synaptic activity. gressive immunological disorder of autoimmune nature, with evident ulcerative colitis, interstitial pneumonitis, glomerular Keywords: dendritic spine, electrophysiology, neuritogenesis, N-WASP, nephropathy with IgA deposits, autoantibodies, and joint synapse inflammation that lead, all together, to premature death (Curcio et al. 2007). Although molecular details of WIP- WASP/N-WASP inter-action have been studied extensively (Volkman et al. 2002; Ho et al. 2004; Dong et al. 2007; Peterson Introduction et al. 2007), few data are available on its functional impact and Neuronal cytoarchitecture is first established through neurito- even fewer regarding the central nervous system, where the genesis, a process in which neurons extend their neurites to role of WIP has not been previously addressed. form a functional network during neuronal development (de Using the WIP knockout mouse as a tool, here, we describe Curtis 2007). Neuron morphology greatly determines the final that loss of this protein impacts neurite and dendrite dynamics complexity of the nervous system and is essential for the signal and morphology, both in early and in late developmental stages, flow that underlies information integration and processing. It is in vitro and in vivo. Gross examination of WIP–/– brain revealed therefore important that neuritogenesis occurs at the right changes in forebrain and hippocampal size. Extensive analysis place and time for correct establishment of synaptic contacts of WIP–/– hippocampal neuron development showed premature with proper targets (de Curtis 2007). Several environmental neuritogenesis. Finally, electrophysiological and immunocyto- cues converge on common coordinated intracellular pathways to chemical analyses demonstrated modified synaptic activity of modulate neuritogenesis. Such intracellular events involve sig- WIP–/– mature neurons. These studies show that WIP is an naling transduction, exocytic and endocytic mechanisms related essential negative regulator in the control of the cytoskeletal to membrane trafficking and cytoskeletal rearrangements. events that underlie neuronal and synaptic development. Neurite initiation and outgrowth are based on the capacity of the neuronal cytoskeleton, constituted mainly of actin Materials and Methods microfilaments (MF) and tubulin microtubules (MT), to assemble and disassemble in response to extracellular signals Mice (Luo 2002; Conde and Caceres 2009). The polarized growth of Wild-type (WT) and WIP KO SV129/BL6 mice (Anton et al. 2002) were neurites requires the initial depolymerization of actin MF housed in specific pathogen-free conditions at the animal facility of the (Bradke and Dotti 1999), stabilization of MT (Ferreira and Centro de Biologı´ a Molecular ‘‘Severo Ochoa,’’ Madrid, Spain. The mouse colony was maintained by continuous mating of heterozygous Caceres 1989), and accumulation of a number of specific females with heterozygous males for more than 20 generations. To proteins (Wiggin et al. 2005). Actin polymerization is con- obtain control or WIP–/– embryos/litters, we mate control male and trolled by the actin-related protein (Arp2/3) complex and by female or WIP–/– male and female mice. Handling of mice and all the action of actin-binding proteins and nucleation-promoting manipulations were carried out in accordance with national and Ó The Author 2011. Published by Oxford University Press. All rights reserved. For permissions, please e-mail: journals.permissions@oup.com
  • 2. European Community guidelines and were reviewed and approved by a 403 oil-immersion lens (NA, 1.3). Metamorph 6.2r6 (Universal the institutional committee for animal welfare. All quantification was Imaging) software was used to process the time-lapse captured images. conducted in a genotype-blind manner. Morphometry Image stacks (physical size 76.9 3 76.9 lm) were imported to the Brain Lysates and Western Blot confocal module of Neurolucida 7.1 (MicroBrightfield, Inc., Williston, Control or WIP–/– brains were homogenized in lysis buffer (20 mM 4-(2- VT), and neuronal dendritic trees were traced by drawing the dendrites hydroxyethyl)-1-piperazineethanesulfonic acid, pH 7.4, 100 mM NaCl, 5 and the bifurcation points. Sholl analysis was performed for each traced mM ethylenediaminetetraacetic acid, 1% Triton X-100, 100 mM NaF, 1 neuron by automatically calculating the number of dendritic intersec- mM Na3VO4, and the Complete Protease Inhibitor Cocktail, Roche tions and the dendritic length at 10-lm interval starting from the soma. Diagnostics), and soluble extracts were resolved by sodium dodecyl Total dendritic length and total number of intersections and branches for sulfate--polyacrylamide gel electrophoresis after determination of each neuron were also calculated as an index of dendritic complexity. protein concentration by Bradford analysis (BioRad). Proteins were Soma area was determined by drawing soma contours while tracing cells. then transferred to nitrocellulose filters, which were blocked and incubated with a mouse monoclonal antibody (mAb) specific to WIP (1/1000, 3D10; a generous gift of Prof. R. Geha, Children’s Hospital, Miniature Excitatory Postsynaptic Currents Boston, MA). After exposure to a specific secondary antibody, antibody Miniature excitatory postsynaptic currents (mEPSC) were recorded binding was visualized by enhanced chemiluminiscence substrate from dissociated hippocampal neurons bathed in artificial cerebro- (Amersham Biosciences). spinal fluid (containing 119 mM NaCl, 2.5 mM KCl, 1 mM NaH2PO4, 11 mM glucose, 26 mM NaHCO3, 2.5 mM CaCl2, and 1.3 mM MgCl2) in the presence of 1 lM tetrodotoxin and 100 lM picrotoxin (at 29 °C). Primary Hippocampal Cultures Spontaneous activity was recorded for 3 min for each cell. mEPSC were Downloaded from cercor.oxfordjournals.org at Universidad Autonoma de Madrid. Facultad de Medicina on August 2, 2011 identified using pClamp software and corrected by eye on the basis of Neurons their kinetics. Primary hippocampal cultures were prepared as described (Dotti et al. 1988; Kaech and Banker 2006). Briefly, hippocampi from E18 mouse embryos (WT and WIP–/–) were washed and digested with 0.25% trypsin Morphology and Unbiased Stereology (15 min, 37 °C). The tissue was then dissociated, resuspended in minimun essential medium with 10% horse serum, and plated on poly- Perfusion L-lysine--coated coverslips (1 mg/ml) at a density of 6 3 103 cells/cm2 Three-month-old male mice (WT, n = 6; WIP–/–, n = 6) were for imaging at early times (up to 24 h after plating) and at a density of 4 3 anesthetized with pentobarbital (0.04 mg/kg) and perfused trans- 103 cells/cm2 for electrophysiological recordings. In some experiments, cardially with PBS (20 ml) followed by 100 ml 4% PFA (pH 7.4) in the neurons were allowed to adhere to the substrate (1 h) and then same buffer. Brains were postfixed in the same fixative (24 h). incubated with 2 or 5 lM wiskostatin (BIOMOL International) in dimethyl sulfoxide (DMSO). In all cases, after 3 h, plating medium was Volumetric Analyses replaced with neurobasal medium supplemented with B27 (Gibco). For Brains were sectioned coronally at a thickness of 50 lm to facilitate long-term culture, at this time, neuron-including coverslips were measurements. Strict morphological criteria were used in all mice to transferred into dishes containing an astrocyte monolayer, with determine the boundaries of these brain regions. Briefly, hippocampal neurons oriented facing the glia but without contacting them. outlines encompassed the CA1--3 fields of Ammon’s horn and the subiculum but not the presubiculum or fimbria hippocampus. The DNA Constructs and Transient Transfection dentate gyrus was measured separately. Starting with one of the pLVWIP-GFP was obtained by digestion of pcDNA3WIP-GFP (kindly sections, randomly selected across brains, 1 of every 6 sections was provided by Prof. N. Ramesh, Children’s Hospital, Boston, MA) and by analyzed through the extent of a hemisphere of the brain. One cloning the insert into pLV; control pLVGFP was generated in a similar hemisphere of each brain was analyzed. Using this sampling strategy, manner. Suspended WT (cortical and hippocampal) or WIP–/– (cortical) 7--10 histological sections per brain were analyzed. All volumetric neurons were nucleofected using a 6 lg DNA/100 ll suspension quantifications were performed with an Olympus BX51 with a 1.253 (Amaxa pulser; Lonza, Germany). Cells were maintained in suspension objective, a motorized XYZ axis computer-controlled stage (Prior (4 h) to permit exogenous gene expression and then plated for 24 h Scientific, Houston, TX), a digital video camera (JVC), and Stereo- before fixation for immunofluorescence analysis. Investigator, a stereology software package (version 8.03, MicroBright- Field). When calculating hippocampus volume, the boundaries were Immunofluorescence defined and the volumes determined with Stereo-Investigator software. At 3 h, 1 and 22 DIV (days in vitro) postplating, cells were fixed in 4% paraformaldehyde (PFA) in phosphate-buffered saline (PBS) (pH 7.4; 20 Intracellular Injection of Lucifer Yellow min, room temperature). Cells were then permeablilized with 0.1% Coronal sections (150 lm) were cut on a vibratome. Sections were Triton X-100 and labeled with phalloidin-tetramethylrhodamine iso- prelabeled with 4,6-diamidino-2-phenylindole, and a continuous cur- thiocyanate (Sigma) and antibodies to tyrosinated a-tubulin (1/400; rent used to inject individual cells with lucifer yellow (8% in 0.1 M Tris T9028, Sigma), MAP2 (MT-associated protein, 1/400; 514, Sanchez buffer, pH 7.4). Dye was injected into neurons in the dentate gyrus until Martin et al. 1998), PSD-95 (5 lg/ml; 75-028, NeuroMab), or GFP (1/ the individual dendrites of each cell could be traced to an abrupt end at 100, 11814460; Roche); secondary fluo-rescent antibody or biotinylated the distal tips and the dendritic spines were readily visible, indicating antibody and labeled streptavidin were then added. Cells were that the dendrites were completely filled. The sections were then excluded from analysis if they showed obvious features of toxicity, processed, first with rabbit anti-lucifer yellow (Cajal Institute, such as neurite fragmentation/blebbing or vacuoles in the cell body. 1/400 000 in stock solution of 2% Bovine serum albumin, 1% Triton X-100, 5% sucrose in PBS) and then with Alexa 488-conjugated Imaging secondary antibody (Molecular Probes, 1/1000, 4 h). Sections were Confocal images were acquired digitally on a confocal LSM510 Meta mounted on a glass slide in fresh ProLong Gold antifade reagent microscope (Zeiss) coupled to an inverted Axiovert 200 microscope (Invitrogen, Eugene, OR; 24 h, room temperature in the dark) and (Zeiss). Image stacks (logical size 1024 3 1024 pixels) consisted of 10 sealed with nail polish. image planes acquired through a 403 (numerical aperture (NA), 1.3) or a 633 oil-immersion lens (NA 1.4). Confocal Microscopy Time-lapse images of phase-contrast fields were captured on an Imaging was performed on a Leica laser scanning multispectral inverted Axiovert 200 microscope (Zeiss) equipped with a mono- confocal microscope (TCS SP5) using an argon laser. Image stacks chrome CCD camera and ultrafast filter change. Image stacks (logical (physical size 76.9 3 76.9 lm, logical size 1024 3 1024 pixels) consisted size 1024 3 1024 pixels) consisted of 6 image planes acquired through of 100--350 image planes acquired through a 633 glycerol-immersion Page 2 of 12 WIP Regulates Neuronal Maturation d Franco et al.
  • 3. lens (NA, 1.3; working distance, 280 lm; refraction index, 1.45) with murine brain and corroborate previous mRNA studies (Ramesh a calculated optimal zoom factor of 3.2 and a z-step of 0.14 lm (voxel et al. 1997; Tsuboi 2006) as well as the data included in the Allen size, 75.1 3 75.1 3 136.4 nm). These settings and optics represent the atlas (http://www.brain-map.org/). highest resolution currently possible with confocal microscopy. For each neuron (5 neurons per mouse, 55 neurons total), 1--5 randomly selected dendrites were scanned from soma to tip, and stacks were Brain Hypertrophy in WIP-Deficient Mice processed with a 3D blind deconvolution algorithm (Autodeblur; To assess general effects of WIP deficiency on the mouse brain, Autoquant, Media Cybernetics) for 10 iterations to reduce the out-of- we determined volumes of the hippocampus and of the rest of focus light. the murine forebrain (3-month-old male mice). Contours of the structures of interest were drawn on Nissl-stained serial sec- Statistical Analysis tions with the aid of Stereo-Investigator software to yield the Quantification was performed in a blind fashion by independent volume of each. There was a significant increase (30%) in researchers. For statistical analyses, we used the two-tailed Student t- forebrain volume in WIP-deficient brains (n = 5) compared test to compare means, the chi-square test to compare nominal with WT littermates (n = 6, Fig. 1B--D and Supplementary Fig. variables, and the two-way analysis of variance to compare Sholl analysis 1). The hippocampus proper (without the dentate gyrus) and or the Kolmogorov--Smirnov test to compare cumulative frequency analysis. Results are shown as mean ± standard error of the mean. the dentate gyrus itself were also hypertrophied in WIP–/– brain (Fig. 1E,F). WIP deficiency thus causes general macrocephaly that includes hippocampal hypertrophy. Results Downloaded from cercor.oxfordjournals.org at Universidad Autonoma de Madrid. Facultad de Medicina on August 2, 2011 Enhanced Early Neuronal Development in WIP –/– Neurons WIP Is Expressed in the WT Mouse Brain The enlarged brain observed in WIP–/– mice relative to their WT The expression pattern of WIP messenger RNA (mRNA) suggests littermates raised the possibility that the greater brain volume that WIP is ubiquitously expressed in all mouse tissues, including could be due to increased neuritic branching since dendrites brain (Ramesh et al. 1997; Tsuboi 2006). To date, however, the and axon collaterals account for most of the brain volume levels of endogenous WIP protein have only been analyzed in (Acebes and Ferrus 2000). To test this hypothesis at the cellular ´ fibroblasts, myoblasts, and hematopoietic-derived cells (reviewed level, we examined the effect of WIP deficiency on early in Anton et al. 2007). To study WIP expression in brain, we probed neuronal development. Primary hippocampal neurons from western blots of cell lysates from 3 regions of the adult mouse brain control and WIP–/– embryos were grown at very low density so (cortex, hippocampus, and olfactory bulb), using the WIP-specific that their neurite arbors did not overlap to avoid confusion mAb 3D10 (Koduru et al. 2007). This mAb recognized 2 protein about which neurites protruded from each cell body. Cells bands in cell lysates from each of the regions examined (Fig. 1A), were labeled with anti-tyrosinated a-tubulin and fluorescent which could represent 2 WIP isoforms and/or be the result of phalloidin (Fig. 2A,B). As a measure of neuronal development, posttranslational modifications of WIP. Neither band was present we quantified the fraction of neurons at each developmental in WIP-deficient tissue. These data demonstrate WIP expression in stage at 3 h postplating. At stage 1, the MT-containing soma is Figure 1. Brain hypertrophy in WIP-deficient mice. (A) Western blot indicates WIP expression in WT mouse brain (cortex, hippocampus, and olfactory bulb; 80 lg total protein/ lane) but not in WIPÀ/À mouse brain. Glyceraldehyde-3-phosphate dehydrogenase labeling confirmed equivalent protein loading in cortical samples. (B) Representative WT (left) and WIPÀ/À brain (right) at 3 months of age. Note: the enlargement of the WIPÀ/À brain. Scale bar, 4 mm. (C) Representative Nissl staining showing hippocampus enlargement in WIPÀ/À mice. Scale bar, 500 lm. (D--F) Average volumes of forebrain (D), hippocampus (E), and dentate gyrus (F) are increased in WIPÀ/À mice. P values were determined with Student’s t-test; *P 0.05; **P 0.01. Cerebral Cortex Page 3 of 12
  • 4. Downloaded from cercor.oxfordjournals.org at Universidad Autonoma de Madrid. Facultad de Medicina on August 2, 2011 Figure 2. WIP deficiency accelerates development of dissociated hippocampal neurons. (A) Representative confocal images of WT- and WIPÀ/À-dissociated hippocampal neurons fixed at 3 h postplating and stained for F-actin (phalloidin-tetramethylrhodamine isothiocyanate, red) and MT (Alexa488-anti-tyrosinated tubulin [TT], green). At this time point, the WIPÀ/À neuron population showed more advanced development compared with WT neurons. Scale bar, 10 lm. (B) High magnification images of the inset in A. Scale bar, 5 lm. (C) Neuron classification by developmental stage (3 h postplating) showed significantly lower frequency of WIPÀ/À at developmental stage 1 compared with WT Page 4 of 12 WIP Regulates Neuronal Maturation d Franco et al.
  • 5. surrounded by flattened actin-rich lamellipodia, while in stage (pLVGFP; Fig. 3C). At 24 h postplating, while 31% of GFP- 2, the lamellipodia are transformed into equidistant neurites expressing neurons were in stage 1, the frequency of stage 1 (Dotti et al. 1988). Stage 1+ is defined as cells with incipient WIP-GFP--expressing neurons was 80% (Fig. 3D). The de- tubulin-rich small protrusions. We found a clear difference in velopmental delay at 24 h induced by WIP overexpression was the distribution of these populations (Fig. 2C); there was confirmed in cortical embryonic neurons (Supplementary Fig. a significant decrease (35%) in the frequency of WIP–/– 2). This effect was sustained over time, since 48 h after plating, neurons classified as developmental stage 1 compared with 22.6% of neurons with increased WIP levels remained at stage 1 WT neurons (n = 4 cultures and n = 150 neurons/genotype, compared with none of the GFP-expressing neurons. Fig. 2C). Conversely, there was an increase (45%) in the We used rescue experiments to confirm the WIP contribution percentage of WIP-deficient neurons at stage 2. As another to neuron development. WIP–/– neurons were nucleofected with measure of neuronal development, we quantified the number a lentiviral-based vector coding for WIP-GFP or control GFP, and of neurites arising from each neuron at 3 h postplating and 24 h after plating, the fraction of cells in stage 1 or 2 was found a significant increase (36%) in the average number of quantified (Fig. 3E,F; WIP-GFP--expressing WIP–/– neurons). The neurites arising from WIP–/– (n = 172) compared with WT population distribution of rescued neurons was indistinguishable neurons (n = 151, Fig. 2D). This finding was confirmed in time- from that of WT neurons and differed significantly from that of lapse images made with phase-contrast microscopy for 3 h GFP-expressing WIP–/– neurons. WIP-GFP expression in WIP –/– after plating (Fig. 2E and Supplementary Videos 1 and 2). neurons also reversed the phenotype, as the mean number of These data suggest that, at 3 h after plating, WIP–/– neurons are primary neurites (Fig. 3G and Supplementary Fig. 3) and neuritic Downloaded from cercor.oxfordjournals.org at Universidad Autonoma de Madrid. Facultad de Medicina on August 2, 2011 in a more advanced developmental stage than WT neurons. bifurcations (Supplementary Fig. 3) were similar to those of GFP- This implies enhanced early neuronal development in the positive WT neurons. absence of WIP. These findings indicate that WIP acts as a negative regulator of neuronal differentiation and that neuronal development can be modulated bidirectionally by WIP levels. N-WASP Activity and Fine-Tuned WIP Levels Regulate Neuronal Development To test whether this accelerated neuronal development Enhanced Dendritic Maturation in WIP –/– Neurons continues at later developmental times, we quantified the To test whether the WIP regulatory role persists in differen- fraction of neurons that remains at developmental stage 1 at 24 h tiated neurons, we also imaged cultured hippocampal neurons postplating. Cultured hippocampal neurons were fixed and at 22 DIV (Fig. 4A). For quantitative analysis of the branching labeled with anti-tyrosinated a-tubulin antibody and fluores- pattern of the neuritic or dendritic tree at the distinct neu- cent phalloidin and imaged by confocal microscopy. As ronal developmental stages, we traced neurons (1 and 22 DIV) observed at 3 h postplating, we found a decrease of 48% in with Neurolucida software. Sholl analysis was used to measure the frequency of WIP–/– neurons at developmental stage 1 the number of neurites/dendrites crossing circles at various (Fig. 3A left and Fig. 3B), implying that WIP–/– neurons still radial distances from the soma (Sholl 1953). Both neuronal show accelerated development at this time. types showed addition of dendritic branches over time (Fig. To determine whether this phenotype depends on the 4B,D), although the number of crossings was significantly activity of N-WASP, an actin NPF with WIP-binding capacity, higher in WIP–/– neurons compared with WT neurons at both we incubated cultures (23 h) with wiskostatin, a selective time points. This finding implies that WIP–/– neurons show reversible N-WASP inhibitor (Peterson et al. 2004). Wiskosta- higher ramification of their neuritic (1 DIV) and dendritic tin is a cell-permeable N-alkylated carbazole derivative that arbor (22 DIV, Fig. 4D). Nevertheless, the difference in the selectively blocks actin filament assembly by binding to N- number of crossings between phenotypes, which was 82% at WASP, stabilizing the autoinhibited conformation, and pre- 1 DIV, decreased to 45% at 22 DIV (Supplementary Fig. 4A). venting activation of the Arp2/3 complex (Wegner et al. The increased ramification was reflected in greater total 2008). Wiskostatin produced a dose-dependent increase in neuritic or dendritic length of WIP–/– neurons at both time points the fraction of stage 1 neurons, indicating impairment in (Fig. 4C,E), a difference that was also reduced in mature neurons neuronal development after blockade of N-WASP-dependent (from 109% to 40%; Supplementary Fig. 4B). This difference was filament assembly (Fig. 3A,B) (da Silva and Dotti 2002; even smaller in neurons of adult mice (see below). Dent et al. 2007). Inhibition of N-WASP activity with 2 lM Soma size determines the number of dendrites and associated wiskostatin induced a phenotype in WIP–/– neurons resem- branches in neurons (Kernell and Zwaagstra 1989; Kollins and bling that of untreated control neurons, and with 5 lM Davenport 2005). To analyze whether WIP deficiency alters wiskostatin, WIP–/– neurons no longer showed a lower soma area, we quantified this parameter in WT and WIP–/– frequency of stage 1 neurons (Fig. 3A,B). This finding suggests neurons. Similar to our observations for the dendritic arbor, that N-WASP activity is essential in mediating the accelerated soma area was significantly larger in WIP–/– neurons (Fig. 4F,G), development of WIP–/– neurons. but this difference attenuated from 40% to 27% with time To further evaluate the role of WIP in neurite sprouting, WT (Supplementary Fig. 4C). The effect of WIP deficiency on neurons were nucleofected with lentiviral constructs for over- promoting neuritic or dendritic arborization and on soma size is expression of WIP-GFP (pLVWIP-GFP) or GFP as control thus greater in early developmental stages. neurons, implying an average more advanced developmental stage for WIPÀ/À neurons. Chi-square test, *P 5 0.0194. (D) The average number of primary neurites arising from WIPÀ/À somas was significantly larger compared with WT neurons (3 h postplating), again indicating the more advanced developmental stage of WIPÀ/À neurons at this time. Data derived from quantification of 150 neurons in each experimental group (from 4 separate experiments). Student’s t-test; ***P 0.001. (E) Selected images from a phase- contrast time-lapse series of control (WT; Supplementary Video1) and WIP-deficient (WIPÀ/À; Supplementary Video2) hippocampal embryonic neurons cultured on poly-L-lysine over a 3-h period. Cerebral Cortex Page 5 of 12
  • 6. Downloaded from cercor.oxfordjournals.org at Universidad Autonoma de Madrid. Facultad de Medicina on August 2, 2011 Figure 3. Inhibition of N-WASP prevents WIPÀ/À phenotype, WIP overexpression delays neurite protrusion, and WIP reexpression reverts the phenotype. (A) Representative images of dissociated WT and WIPÀ/À hippocampal neurons, alone or wiskostatin-treated, fixed at 24 h postplating and stained for F-actin (phalloidin-tetramethylrhodamine isothiocyanate, red) and tyrosinated MT (tyrosinated tubulin [TT], green). As at 3 h postplating (Fig. 2), a typical 24-h WIPÀ/À neuron showed more advanced development than a WT neuron. Scale bar, 10 lm. (B) Percentage of cells at developmental stage 1, alone or wiskostatin-treated. Cells were dissociated and cultured without additives (control), with DMSO, or with wiskostatin (2 or 5 lM). Wiskostatin (2 lM) caused a significant increase in the frequency of WIPÀ/À neurons at stage 1, equivalent to the frequency for untreated WT neurons. A higher wiskostatin concentration (5 lM) prevented differentiation of both WT and WIPÀ/À neurons. Student’s t-test; *P 0.05; **P 0.01. (C) Images of phalloidin-stained (red) hippocampal neurons expressing GFP (left) or GFP-WIP (right) at 24 h postplating. (D) The percentage of cells at developmental stage 1 was greater in WIP-overexpressing cells, implying that WIP overexpression delays neurite formation. Student’s t-test; *P 0.05. (E) Representative images of cortical control WT neurons nucleofected to express GFP and of WIPÀ/À neurons nucleofected to express GFP or WIP-GFP. Cortical neurons were fixed at 24 h postplating and stained for F-actin (phalloidin-tetramethylrhodamine isothiocyanate, red) and anti-GFP (green). A typical WIPÀ/À neuron--expressing WIP-GFP showed similar development to that of a WT neuron. Scale bar, 10 lm. (F) Percentage of cells at developmental stage 1 after WIP reexpression. The fraction of cells at stage 1 was quantified at 24 h postplating. Distribution of rescued WIPÀ/À neurons was similar to that of WT neurons, confirming the role of WIP in neuronal development. Student’s t-test; **P 0.01. (G) Mean number of primary neurites per GFP-expressing WT neuron is similar to that of WIP-GFP-- expressing WIPÀ/À neurons and differs significantly from that of WIPÀ/ÀGFP-expressing neurons. Student’s t-test; ***P 0.001; ns, not significant. Enhanced Neuronal Ramification in Adult WIP –/– Mice (Fig. 5A) and studied the structure of granule cells in the To test whether WIP deficiency produces a long-lasting effect dentate gyrus. Sholl analysis showed significantly more cross- on neuronal maturation in vivo, we evaluated dendritic archi- ings in the distal part of the dendrite in WIP–/– mice (n = 59 tecture in adult (3-month-old) mice after injection of lucifer neurons from 6 mice) compared with WT mice (n = 39 yellow into hippocampal neurons of fixed brain sections neurons from 5 mice, Fig. 5B). This increased number of Page 6 of 12 WIP Regulates Neuronal Maturation d Franco et al.
  • 7. Downloaded from cercor.oxfordjournals.org at Universidad Autonoma de Madrid. Facultad de Medicina on August 2, 2011 Figure 4. Increased ramification in WIPÀ/À neurons in vitro. (A) Representative images of dissociated WT and WIPÀ/À hippocampal neurons fixed at 1 and 22 DIV and stained with anti-tyrosinated-a-tubulin (1 DIV) or anti-MAP2 (22 DIV) antibodies. Note the greater ramification of WIPÀ/À neurons at both 1 and 22 DIV. (B and C) Sholl analysis of traced WT and WIPÀ/À neurons showed more ramification in WIPÀ/À neurons (B), reflected as greater total neuritic or dendritic length for these cells (C). Two-way analysis of variance; **P 0.01; ***P 0.001. (D) Total number of crossings at 1 and 22 DIV. Note the increase with time in the total number of crossings for both genotypes. (E) Total neuritic or dendritic length at 1 and 22 DIV. Note the increase over time in the total dendritic length for both genotypes. (F) Representative images of WT and WIPÀ/À somas demonstrating increased soma surface in WIPÀ/À neurons. (G) WIPÀ/À neuronal somas are larger than WT neuronal somas at 1 and 22 DIV. (D, E, and G) Student’s t-test; **P 0.01; ***P 0.001. Cerebral Cortex Page 7 of 12
  • 8. crossings reflects a larger number of ramifications at the outer neuronal development. We found that, in hippocampal primary molecular layer of the dentate gyrus. As a result of the higher neurons, loss of WIP accelerates the onset of neuritogenesis and dendritic ramification, the dendritic length in this layer is also increases neuritic and dendritic branching. Moreover, lack of increased in WIP–/– mice (Fig. 5C ), leading to greater total WIP enhances PSD-95 accumulation in hippocampal dendritic dendritic length (18%) in WIP–/– mice (Fig. 5D). Cumulative spines and increases neuronal synaptic activity. Conversely, WIP frequency analysis indicated that WIP–/– mice have a sub- overexpression blocks the initiation of neuritogenesis, suggesting population of highly ramified neurons not detected in WT that WIP is a bidirectional regulator of neuronal development. mice (Fig. 5E ). WIP deficiency thus leads to enhanced WIP Modulates Neuritic Branching During Neuronal dendritic ramification in the adult mouse. Nonetheless, the Development magnitude of change in total dendritic length of the adult Marked dendritic elaboration normally begins in neuronal WIP–/– mouse is smaller than in early developmental stages. cultures 2--3 days after axonal outgrowth (Nowakowski and Rakic 1979; Dotti et al. 1988). Our experiments showed that immature Enhanced Synaptic Maturation in WIP –/– Neurons WIP–/– neurons begin to emit neurites starting at the first day in During later stages of neuronal development, dendritic culture, much earlier than control neurons. After cell cycle exit arborization and synaptogenesis occur in parallel (Cantallops and before neuronal polarization, cortical postmitotic neurons et al. 2000; Cline 2001). The actin cytoskeleton has a pivotal make a transition through a multipolar stage, when multiple role in both processes. In the context of synaptogenesis, actin neurites emerge rapidly from the cell body (Barnes and Polleux dynamics control the morphogenesis and function of the den- 2009). This morphological transition is also controlled by WIP Downloaded from cercor.oxfordjournals.org at Universidad Autonoma de Madrid. Facultad de Medicina on August 2, 2011 dritic spines, small actin-rich protrusions from dendritic shafts during brain development, as GFP in utero electroporation shows (Ethell and Pasquale 2005). As we found that WIP modulates a notable increase in the percentage of multipolar cells in WIP–/– dendritic arborization, we examined the possibility that WIP embryos, in parallel to a reduction in the fraction of round and deficiency fosters synaptic maturation. We measured the effects unipolar neurons (not shown). In addition, neurites in immature of WIP deficiency on spontaneous miniature (action-potential WIP–/– neurons are more branched than neurites of WT neurons. independent) postsynaptic currents (mEPSC) in dissociated Dendritic branching is also increased in WIP–/– mature neurons, hippocampal neurons (22 DIV) in the presence of 1 lM although to a lesser extent than at early neuronal developmental tetrodotoxin (Fig. 6A). Both the amplitude and the frequency stages (Fig. 4 and Supplementary Fig. 4). Sholl analysis demon- of mEPSC were significantly increased in WIP–/– compared with strated that the initial morphological features acquired by WT neurons (n = 17 and 15 neurons, respectively; Fig. 6B,C ). immature WIP–/– neurons determine the pattern of dendritic mEPSC amplitude is indicative of the postsynaptic strength of branching of the more differentiated cells. We therefore suggest individual functional synapses, whereas mEPSC frequency that WIP acts as a negative modulator that controls the precise depends on synapse number and presynaptic properties timing of the correct onset of dendritic development. Our results (Turrigiano et al. 1998; Han and Stevens 2009). WIP deficiency point to the possibility that some pathologies detected in mature functionally increases both the strength and possibly the neurons might have their origin, yet unknown, in altered number of individual synapses, suggesting that at this time, processes that occur during early neuronal development. WIP –/– neurons show either a more mature phenotype or more abundant and/or enlarged dendritic spines. WIP versus Other Negative Regulators of Dendritic To examine whether WIP deficiency resulted in an increased Branching number of structural synapses, we stained dissociated hippo- Numerous molecules are involved in the positive control of campal neurons at the same developmental stage (22 DIV) for the neuritic or dendritic outgrowth and branching, many of them postsynaptic marker PSD-95, known to drive maturation of related to the control of cytoskeleton dynamics (McAllister 2000; glutamatergic synapses (El-Husseini et al. 2000). WIP–/– neurons Urbanska et al. 2008). Only a minority of these molecules, such as showed increased fluorescence intensity of individual PSD-95 PTEN (Kwon et al. 2001; Jaworski et al. 2005) and RhoA (Negishi puncta, indicating greater PSD-95 accumulation in spines in the and Katoh 2002; da Silva et al. 2003), have been identified as absence of WIP (WT, n = 117 spines from 6 neurons; WIP–/–, n = negative regulators of these processes. Similarly to our findings, 168 spines from 7 neurons, Fig. 6D,E ). Frequency analysis conditional adult Pten mutant mice show enlargement of cortex showed that in WIP–/– neurons, staining of some PSD-95--positive and hippocampus associated with dendritic hypertrophy and spines is particularly intense, whereas such a population was not increased soma size (Kwon et al. 2001, 2006). At difference from found in WT neurons (Fig. 6F ). Positive spine density was the WIP–/– phenotype observed here, PTEN is not necessary for nonetheless similar in WIP–/– and WT neurons (Fig. 6G ). As WIP–/– initiation of neuritogenesis (Lachyankar et al. 2000). The most neurons are substantially more branched than WT neurons (and striking difference between WIP- and PTEN-mediated negative their total dendritic length is therefore increased), the total regulation thus appears to depend on the temporal window of number of PSD-95--positive spines is significantly increased in neuronal development in which both proteins operate. PTEN WIP–/– neurons (Fig. 6H). These neuronal cultures were does not control early stages of neuritogenesis, and its influence maintained over WT astrocytes, excluding possible astrocyte on neuronal structure increases progressively throughout the involvement in the synaptic modulation derived from the lack of animal’s life. In contrast, WIP modulates neuronal development WIP. These results indicate that WIP deficiency increases the and synaptogenesis early in postnatal life, and its effects are number and size of structural and functional synapses. attenuated as neuronal maturation progresses. Similarly to the absence of WIP, RhoA inhibition increases both the number of primary neurites and total neuritic length Discussion in immature dissociated hippocampal neurons, whereas ex- Here, we present the first report of a role for WIP in the brain, pression of a constitutively active form of RhoA arrests cells in showing a specific role for WIP as a negative regulator of murine stage 1 (da Silva et al. 2003). In mature neurons, transfection of Page 8 of 12 WIP Regulates Neuronal Maturation d Franco et al.
  • 9. Downloaded from cercor.oxfordjournals.org at Universidad Autonoma de Madrid. Facultad de Medicina on August 2, 2011 Figure 5. Increased ramification in WIPÀ/À neurons in vivo. (A) Representative projection images of lucifer yellow--injected granule neurons of 3-month-old WT and WIPÀ/À mice. Scale bar, 25 lm. (B and C) Sholl analysis of traced WT and WIPÀ/À granule neurons showed greater ramification in the distal part of WIPÀ/À neurons (B). This is reflected as greater dendritic length for these neurons (two-way analysis of variance; *P 0.05) (C). (D) Total dendritic length is greater in neurons from WIPÀ/À mice. Student’s t-test; *P 0.05. (E) Cumulative frequency curves of total dendritic length indicating a shift toward higher values in neurons from WIPÀ/À mice. Kolmogorov--Smirnov test; *P 0.05. a dominant-negative RhoA construct does not affect dendritic Possible Mechanisms for WIP-Mediated Neurite Sprouting morphology (Nakayama et al. 2000), whereas WIP–/– neurons and Branching still show enhanced dendritic arborization. The effect of WIP Sprouting of primary neurites and interstitial branching from on neuronal maturation thus appears to be longer lasting than neuritic or dendritic shafts follows the same sequence as that of other negative modulators of neuritic and dendritic cortical cytoskeletal rearrangements (Wu et al. 1999; Dent et al. outgrowth and branching. 2007). For the extension of a single filopodium, which is Cerebral Cortex Page 9 of 12
  • 10. Downloaded from cercor.oxfordjournals.org at Universidad Autonoma de Madrid. Facultad de Medicina on August 2, 2011 Figure 6. WIP deficiency increases the strength and the number of individual synapses. (A) Representative trace (calibration: 20 pA, 500 ms) and event (calibration: 10 pA, 2 ms) of mEPSC recorded from a WT and a WIPÀ/À neuron. (B and C) Cumulative frequency representation of the amplitude (B) and frequency (C) of mEPSC, showing significant shifts to higher values in WIPÀ/À neurons. Kolmogorov--Smirnov test; ***P 0.001. (D) Representative projection confocal images of WT and WIPÀ/À dendrites, stained with anti-MAP2 (blue) and -PSD-95 antibodies (green). Note that there are intense dendritic spines in WIPÀ/À but not in WT dendrites. (E) Average intensity of PSD-95--positive spines in WT and WIPÀ/À neurons. Student’s t-test; ***P 0.001. (F) Cumulative frequency representation of the intensity of PSD-95--positive spines. Observe the subpopulation of particularly intense PSD-95--positive spines in WIPÀ/À but not in WT neurons. Kolmogorov--Smirnov test; ***P 0.001. (G) Sholl analysis of PSD-95--positive spine density in WT and WIPÀ/À neurons. (H) The number of PSD-95--positive spines was calculated as a function of the distance from soma (Sholl analysis) by multiplying spine density for each distance by total dendritic length at the same distance. Two-way analysis of variance; ***P 0.001. subsequently stabilized by MT invasion, F-actin and MT must Our data indicate that WIP modulates neuritogenesis undergo local depolymerization triggered by extracellular signals through its previously described ability to maintain N-WASP (Acebes and Ferrus 2000; da Silva and Dotti 2002; Luo 2002). ´ in its autoinhibited state (Martinez-Quiles et al. 2001). N-WASP WIP inhibits F-actin depolymerization (Martinez-Quiles et al. promotes neurite outgrowth and regulates neurite branching 2001), which could explain the neuritogenesis arrest found in in hippocampal neurons (Suetsugu, Hattori, et al. 2002; Abe WIP-overexpressing neurons. et al. 2003; Pinyol et al. 2007). At the molecular level, N-WASP Page 10 of 12 WIP Regulates Neuronal Maturation d Franco et al.
  • 11. acts as a signal integration device that can precisely target actin Daniel Gallego for electrophysiological signal processing, Jose Ramon ´ ´ polymerization to membrane sites at which PI(4,5)P2 and Valverde for statistical assistance and Catherine Mark for editorial activated Src kinases and Cdc42 are located (Prehoda et al. assistance. Conflict of Interest : None declared. 2000; Suetsugu, Miki, et al. 2002). Cdc42 stimulates the actin- polymerizing activity of N-WASP, creating free barbed ends References from which actin polymerization can take place (Miki, Suetsugu, et al. 1998), leading to filopodium formation (Miki, Abe T, Kato M, Miki H, Takenawa T, Endo T. 2003. Small GTPase Tc10 and its homologue RhoT induce N-WASP-mediated long process Sasaki, et al. 1998). In the absence of WIP, N-WASP would be formation and neurite outgrowth. J Cell Sci. 116:155--168. more easily released from inhibition and be hyperactivated to Acebes A, Ferrus A. 2000. Cellular and molecular features of axon ´ initiate premature filopodium formation. This interpretation is collaterals and dendrites. Trends Neurosci. 23:557--565. supported by pharmacological inhibition of N-WASP by wiskos- Anton IM, de la Fuente MA, Sims TN, Freeman S, Ramesh N, Hartwig JH, tatin, which blocked the accelerated neuritogenesis seen in WIP- Dustin ML, Geha RS. 2002. WIP deficiency reveals a differential role deficient neurons (Fig. 3). for WIP and the actin cytoskeleton in T and B cell activation. Immunity. 16:193--204. Anton IM, Jones GE, Wandosell F, Geha R, Ramesh N. 2007. WASP- WIP Modulates Synaptic Activity interacting protein (WIP): working in polymerisation and much We report here that mEPSC amplitude and frequency are more. Trends Cell Biol. 17:555--562. increased in WIP–/– neurons, suggesting both stronger AMPA Barnes AP, Polleux F. 2009. Establishment of axon-dendrite polarity in (alpha-amino-3-hydroxy-5-methyl-4-isoxazole propionic acid) developing neurons. Annu Rev Neurosci. 32:347--381. Downloaded from cercor.oxfordjournals.org at Universidad Autonoma de Madrid. Facultad de Medicina on August 2, 2011 receptor--mediated synaptic currents and more functional Bradke F, Dotti CG. 1999. The role of local actin instability in axon synapses in these cells (Fig. 6). In addition, we describe an formation. Science. 283:1931--1934. increase in PSD-95 accumulation at individual spines in WIP–/– Cantallops I, Haas K, Cline HT. 2000. Postsynaptic CPG15 promotes synaptic maturation and presynaptic axon arbor elaboration in vivo. neurons. These 2 events are probably linked since PSD-95 Nat Neurosci. 3:1004--1011. drives the maturation of excitatory synapses with the in- Cline HT. 2001. Dendritic arbor development and synaptogenesis. Curr corporation of AMPA receptors (El-Husseini et al. 2000). Opin Neurobiol. 11:118--126. Indeed, the contribution of AMPA receptors to synaptic Conde C, Caceres A. 2009. Microtubule assembly, organization and transmission increases gradually with neuronal maturation dynamics in axons and dendrites. Nat Rev Neurosci. 10:319--332. (Mammen et al. 1997; Petralia et al. 1999). The increase in Curcio C, Pannellini T, Lanzardo S, Forni G, Musiani P, Anton IM. 2007. PSD-95 accumulation at individual WIP–/– spines might suggest WIP null mice display a progressive immunological disorder that resembles Wiskott-Aldrich Syndrome. J Pathol. 211:67--75. that the PSD (and therefore the synapse) is enlarged in the da Silva JS, Dotti CG. 2002. Breaking the neuronal sphere: regulation of absence of WIP. Given that the synaptic area correlates the actin cytoskeleton in neuritogenesis. Nat Rev Neurosci. positively with the number of synaptic AMPA receptors (Nusser 3:694--704. et al. 1998), our electrophysiological findings are strengthened da Silva JS, Medina M, Zuliani C, Di Nardo A, Witke W, Dotti CG. 2003. by the morphological analysis of PSD-95 in spines. RhoA/ROCK regulation of neuritogenesis via profilin IIa-mediated In conclusion, this study sheds light on the molecular events control of actin stability. J Cell Biol. 162:1267--1279. that control early neuronal development by presenting WIP as de Curtis I. 2007. Intracellular mechanisms of neuritogenesis. New York: Springer. a previously undescribed regulator that prevents premature Dent EW, Kwiatkowski AV, Mebane LM, Philippar U, Barzik M, dendritic and synaptic maturation. Rubinson DA, Gupton S, Van Veen JE, Furman C, Zhang J, et al. 2007. Filopodia are required for cortical neurite initiation. Nat Cell Biol. 9:1347--1359. Supplementary Material Dong X, Patino-Lopez G, Candotti F, Shaw S. 2007. Structure-function Supplementary material can be found at: http://www.cercor. analysis of the WIP role in T cell receptor-stimulated NFAT oxfordjournals.org/. activation: evidence that WIP-WASP dissociation is not required and that the WIP NH2 terminus is inhibitory. J Biol Chem. 282:30303--30310. Funding Dotti CG, Sullivan CA, Banker GA. 1988. The establishment of polarity Grants from Consejo Superior de Investigaciones Cientı´ ficas- by hippocampal neurons in culture. J Neurosci. 8:1454--1468. Comunidad de Madrid (CCG08-CSIC/SAL-3471), CSIC (PIE2- El-Husseini AE, Schnell E, Chetkovich DM, Nicoll RA, Bredt DS. 2000. 00720I002), and the Spanish Ministry of Education and Science PSD-95 involvement in maturation of excitatory synapses. Science. 290:1364--1368. (BFU2007-64144 and BFU2010-21374) to I.M.A., from Centro de Ethell IM, Pasquale EB. 2005. Molecular mechanisms of dendritic spine Investigacion Biomedica en Red Enfermedades Neurodegener- ´ ´ development and remodeling. Prog Neurobiol. 75:161--205. ativas (Instituto de Salud Carlos III), the Plan Nacional DGCYT Ferreira A, Caceres A. 1989. The expression of acetylated microtubules (SAF2009-12249-C02-01) to F.W. and SAF2010-15676 to S.K., during axonal and dendritic growth in cerebellar macroneurons and by an institutional grant from the Fundacion Ramon Areces. ´ ´ which develop in vitro. Brain Res Dev Brain Res. 49:205--213. A.F. was a recipient of an FPU MEC fellowship (AP2005-3405), Han EB, Stevens CF. 2009. Development regulates a switch between I.B. held a contract from the Comunidad Autonoma de Madrid ´ post- and presynaptic strengthening in response to activity deprivation. Proc Natl Acad Sci U S A. 106:10817--10822. and S.K., a Ramon y Cajal contract. ´ Ho HY, Rohatgi R, Lebensohn AM, Le M, Li J, Gygi SP, Kirschner MW. 2004. Toca-1 mediates Cdc42-dependent actin nucleation by Notes activating the N-WASP-WIP complex. Cell. 118:203--216. We thank Chiara Ragazzini and Sonia Perez for their excellent technical ´ Jaworski J, Spangler S, Seeburg DP, Hoogenraad CC, Sheng M. 2005. Control assistance and Javier de Felipe for his contribution to the morphomet- of dendritic arborization by the phosphoinositide-3’-kinase-Akt- ric analysis with NeuroLucida Software. We are grateful to Raif Geha mammalian target of rapamycin pathway. J Neurosci. 25:11300--11312. and Narayanaswamy Ramesh for the 3D10 mAb and to Lola Ledesma Kaech S, Banker G. 2006. Culturing hippocampal neurons. Nat Protoc. and Carlos Dotti for helpful advice on the manuscript. We acknowledge 1:2406--2415. Cerebral Cortex Page 11 of 12
  • 12. Kakimoto T, Katoh H, Negishi M. 2004. Identification of splicing Peterson FC, Deng Q, Zettl M, Prehoda KE, Lim WA, Way M, variants of Rapostlin, a novel RND2 effector that interacts with Volkman BF. 2007. Multiple WASP-interacting protein recognition neural Wiskott-Aldrich syndrome protein and induces neurite motifs are required for a functional interaction with N-WASP. J Biol branching. J Biol Chem. 279:14104--14110. Chem. 282:8446--8453. Kernell D, Zwaagstra B. 1989. Dendrites of cat’s spinal motoneurones: Peterson JR, Bickford LC, Morgan D, Kim AS, Ouerfelli O, relationship between stem diameter and predicted input conduc- Kirschner MW, Rosen MK. 2004. Chemical inhibition of N-WASP tance. J Physiol. 413:255--269. by stabilization of a native autoinhibited conformation. Nat Struct Kettner A, Kumar L, Anton IM, Sasahara Y, de la Fuente M, Pivniouk VI, Mol Biol. 11:747--755. Falet H, Hartwig JH, Geha RS. 2004. WIP regulates signaling via the Petralia RS, Esteban JA, Wang YX, Partridge JG, Zhao HM, Wenthold RJ, high affinity receptor for immunoglobulin E in mast cells. J Exp Med. Malinow R. 1999. Selective acquisition of AMPA receptors over 199:357--368. postnatal development suggests a molecular basis for silent Koduru S, Massaad M, Wilbur C, Kumar L, Geha R, Ramesh N. 2007. A synapses. Nat Neurosci. 2:31--36. novel anti-WIP monoclonal antibody detects an isoform of WIP that Pinyol R, Haeckel A, Ritter A, Qualmann B, Kessels MM. 2007. lacks the WASP binding domain. Biochem Biophys Res Commun. Regulation of N-WASP and the Arp2/3 complex by Abp1 controls 353:875--881. Kollins KM, Davenport RW. 2005. Branching morphogenesis in neuronal morphology. PLoS One. 2:e400. vertebrate neurons. Austin (TX): Landes Bioscience. Prehoda KE, Scott JA, Mullins RD, Lim WA. 2000. Integration of multiple Kwon CH, Luikart BW, Powell CM, Zhou J, Matheny SA, Zhang W, Li Y, signals through cooperative regulation of the N-WASP-Arp2/3 Baker SJ, Parada LF. 2006. Pten regulates neuronal arborization and complex. Science. 290:801--806. social interaction in mice. Neuron. 50:377--388. Ramesh N, Anton IM, Hartwig JH, Geha RS. 1997. WIP, a protein Kwon CH, Zhu X, Zhang J, Knoop LL, Tharp R, Smeyne RJ, associated with wiskott-aldrich syndrome protein, induces actin polymerization and redistribution in lymphoid cells. Proc Natl Acad Downloaded from cercor.oxfordjournals.org at Universidad Autonoma de Madrid. Facultad de Medicina on August 2, 2011 Eberhart CG, Burger PC, Baker SJ. 2001. Pten regulates neuronal soma size: a mouse model of Lhermitte-Duclos disease. Nat Genet. Sci U S A. 94:14671--14676. 29:404--411. Sanchez Martin C, Diaz-Nido J, Avila J. 1998. Regulation of a site-specific Lachyankar MB, Sultana N, Schonhoff CM, Mitra P, Poluha W, phosphorylation of the microtubule-associated protein 2 during the Lambert S, Quesenberry PJ, Litofsky NS, Recht LD, Nabi R, et al. development of cultured neurons. Neuroscience. 87:861--870. 2000. A role for nuclear PTEN in neuronal differentiation. Sholl DA. 1953. Dendritic organization in the neurons of the visual and J Neurosci. 20:1404--1413. motor cortices of the cat. J Anat. 87:387--406. Luo L. 2002. Actin cytoskeleton regulation in neuronal morphogenesis Suetsugu S, Hattori M, Miki H, Tezuka T, Yamamoto T, Mikoshiba K, and structural plasticity. Annu Rev Cell Dev Biol. 18:601--635. Takenawa T. 2002. Sustained activation of N-WASP through Mammen AL, Huganir RL, O’Brien RJ. 1997. Redistribution and sta- phosphorylation is essential for neurite extension. Dev Cell. bilization of cell surface glutamate receptors during synapse 3:645--658. formation. J Neurosci. 17:7351--7358. Suetsugu S, Miki H, Takenawa T. 2002. Spatial and temporal regulation Martinez-Quiles N, Rohatgi R, Anton IM, Medina M, Saville SP, Miki H, of actin polymerization for cytoskeleton formation through Arp2/3 Yamaguchi H, Takenawa T, Hartwig JH, Geha RS, et al. 2001. WIP complex and WASP/WAVE proteins. Cell Motil Cytoskeleton. regulates N-WASP-mediated actin polymerization and filopodium 51:113--122. formation. Nat Cell Biol. 3:484--491. Tsuboi S. 2006. A complex of Wiskott-Aldrich syndrome protein with McAllister AK. 2000. Cellular and molecular mechanisms of dendrite mammalian verprolins plays an important role in monocyte growth. Cereb Cortex. 10:963--973. chemotaxis. J Immunol. 176:6576--6585. Miki H, Sasaki T, Takai Y, Takenawa T. 1998. Induction of filopodium Turrigiano GG, Leslie KR, Desai NS, Rutherford LC, Nelson SB. 1998. formation by a WASP-related actin-depolymerizing protein N-WASP. Activity-dependent scaling of quantal amplitude in neocortical Nature. 391:93--96. neurons. Nature. 391:892--896. Miki H, Suetsugu S, Takenawa T. 1998. WAVE, a novel WASP-family Urbanska M, Blazejczyk M, Jaworski J. 2008. Molecular basis of dendritic protein involved in actin reorganization induced by Rac. EMBO J. 17:6932--6941. arborization. Acta Neurobiol Exp (Wars). 68:264--288. Nakayama AY, Harms MB, Luo L. 2000. Small GTPases Rac and Rho in Volkman BF, Prehoda KE, Scott JA, Peterson FC, Lim WA. 2002. Structure the maintenance of dendritic spines and branches in hippocampal of the N-WASP EVH1 domain-WIP complex: insight into the pyramidal neurons. J Neurosci. 20:5329--5338. molecular basis of Wiskott-Aldrich Syndrome. Cell. 111:565--576. Negishi M, Katoh H. 2002. Rho family GTPases as key regulators for Wegner AM, Nebhan CA, Hu L, Majumdar D, Meier KM, Weaver AM, neuronal network formation. J Biochem. 132:157--166. Webb DJ. 2008. N-wasp and the arp2/3 complex are critical Nowakowski RS, Rakic P. 1979. The mode of migration of neurons to regulators of actin in the development of dendritic spines and the hippocampus: a Golgi and electron microscopic analysis in synapses. J Biol Chem. 283:15912--15920. foetal rhesus monkey. J Neurocytol. 8:697--718. Wiggin GR, Fawcett JP, Pawson T. 2005. Polarity proteins in axon Nusser Z, Lujan R, Laube G, Roberts JD, Molnar E, Somogyi P. 1998. Cell specification and synaptogenesis. Dev Cell. 8:803--816. type and pathway dependence of synaptic AMPA receptor number Wu GY, Zou DJ, Rajan I, Cline H. 1999. Dendritic dynamics in vivo and variability in the hippocampus. Neuron. 21:545--559. change during neuronal maturation. J Neurosci. 19:4472--4483. Page 12 of 12 WIP Regulates Neuronal Maturation d Franco et al.