You are on page 1of 15

Neurobiology of Learning and Memory xxx (2016) xxx–xxx

Contents lists available at ScienceDirect

Neurobiology of Learning and Memory


journal homepage: www.elsevier.com/locate/ynlme

The memory gene KIBRA is a bidirectional regulator of synaptic


and structural plasticity in the adult brain
Fabrice D. Heitz a, Mélissa Farinelli a, Safa Mohanna a, Martin Kahn a, Kerstin Duning b, Marco C. Frey c,
Hermann Pavenstädt d, Isabelle M. Mansuy a,⇑
a
Laboratory of Neuroepigenetics, Medical Faculty of the University of Zürich and Department of Health Science and Technology of the Swiss Federal Institute of Technology
Zürich, Brain Research Institute, Winterthurerstrasse 190, CH-8057 Zürich, Switzerland
b
Internal Medicine D, Molecular Nephrology, University Hospital Münster, Domagkstrasse 3a, D-48149 Münster, Germany
c
Division of Psychiatry Research, Medical Faculty of the University of Zürich, August Forel Strasse 1, CH-8008 Zürich, Switzerland
d
Medizinische Klinik und Poliklinik D, Molecular Nephrology, University of Münster, Domagkstrasse 3a, D-48149 Münster, Germany

a r t i c l e i n f o a b s t r a c t

Article history: Memory formation is associated with activity-dependent changes in synaptic plasticity. The mechanisms
Received 29 May 2016 underlying these processes are complex and involve multiple components. Recent work has implicated
Revised 23 July 2016 the protein KIBRA in human memory, but its molecular functions in memory processes remain not fully
Accepted 28 July 2016
understood. Here, we show that a selective overexpression of KIBRA in neurons increases hippocampal
Available online xxxx
long-term potentiation (LTP) but prevents the induction of long-term depression (LTD), and impairs spa-
tial long-term memory in adult mice. KIBRA overexpression increases the constitutive recycling of AMPA
Keywords:
receptors containing GluA1 (GluA1-AMPARs), and favors their activity-dependent surface expression. It
Neurons
LTP
also results in dramatic dendritic rearrangements in pyramidal neurons both in vitro and in vivo.
GluA1 KIBRA knockdown in contrast, abolishes LTP, decreases GluA1-AMPARs recycling and reduces dendritic
arborization. These results establish KIBRA as a novel bidirectional regulator of synaptic and structural
plasticity in hippocampal neurons, and of long-term memory, highly relevant to cognitive processes
and their pathologies.
Ó 2016 Elsevier Inc. All rights reserved.

1. Introduction as WWC1) has been newly associated with episodic memory in


human. A genetic variant of KIBRA with a single nucleotide poly-
Episodic memory in mammals is a complex form of information morphism (SNP) leading to T-C exchange (rs17070145) in the
storage primarily processed in the hippocampus. It recruits multi- ninth intron of the KIBRA gene was found to correlate with a vari-
ple molecular cascades and components which to date, remain ability in memory performance in healthy subjects (Almeida et al.,
only partially understood. Recently, the gene KIBRA (also known 2008; Bates et al., 2009; Burgess et al., 2010; Papassotiropoulos
et al., 2006; Preuschhof et al., 2009; Schaper, Kolsch, Popp,
Wagner, & Jessen, 2008; Schwab, Luo, Clarke, & Nathan, 2014).
Abbreviations: AMPAR, a-amino-3-hydroxyl-5-methyl-4-isoxazole-proprionate KIBRA is a large protein that is highly expressed in the brain, in
receptor; BiTetO, rtTA2-responsive Tet-On bidirectional promoter; CaMKIIa,
particular in structures important for memory including the hip-
calcium/calmodulin-dependent protein kinase alpha; CNQX, 6-cyano-7-nitroqui
noxaline-2,3-dione; C-term, cytoplasmic C-terminus domain; dox, doxycycline; pocampus and cortex (Johannsen, Duning, Pavenstadt,
EGFP, enhanced green fluorescent protein; EPSC, excitatory postsynaptic current; Kremerskothen, & Boeckers, 2008; Kremerskothen et al., 2003;
f-EPSP, field excitatory postsynaptic potential; acGFP, acetylated green fluorescent Papassotiropoulos et al., 2006). KIBRA has a somatodendritic distri-
protein; HFS, high frequency stimulation; LFS, low frequency stimulation; LTD, bution in neuronal cells and is enriched in the postsynaptic density
long-term depression; LTP, long-term potentiation; cLTP, chemical long-term
(PSD). Recent studies have shown that it interacts with the
potentiation; MAP2, microtubule-associated protein 2; NMDAR, N-methyl-D-
Aspartate receptor; PKMf, protein kinase M zeta; PSD, postsynaptic density; cytoskeleton-associated proteins dendrin and synaptopodin, which
PSD-95, postsynaptic density protein 95; shRNA, small hairpin RNA; SNP, single are both involved in synaptic plasticity and higher-order brain
nucleotide polymorphism; SNX4, sorting nexin-4; rtTA2, reverse tetracycline- functions (Buther, Plaas, Barnekow, & Kremerskothen, 2004;
controlled transactivator.
⇑ Corresponding author.
Duning et al., 2008; Johannsen et al., 2008; Kremerskothen et al.,
2003). Importantly, KIBRA also binds to PKMf, a protein kinase
E-mail address: mansuy@hifo.uzh.ch (I.M. Mansuy).

http://dx.doi.org/10.1016/j.nlm.2016.07.028
1074-7427/Ó 2016 Elsevier Inc. All rights reserved.

Please cite this article in press as: Heitz, F. D., et al. The memory gene KIBRA is a bidirectional regulator of synaptic and structural plasticity in the adult
brain. Neurobiology of Learning and Memory (2016), http://dx.doi.org/10.1016/j.nlm.2016.07.028
2 F.D. Heitz et al. / Neurobiology of Learning and Memory xxx (2016) xxx–xxx

involved in AMPARs trafficking (Yao et al., 2008) that is critical for generations) and heterozygous for either one (control mice) or
the maintenance of hippocampal LTP and for memory persistence both transgenes (double positive mice, ‘‘KIBRA overexpressing”
(Pastalkova et al., 2006; Shema, Sacktor, & Dudai, 2007; mice, OE) were used. Mice were housed under constant tempera-
Yoshihama, Hirai, Ohtsuka, & Chida, 2009). Owing to its cellular ture and humidity in a reversed 12 h light-dark cycle (dark
distribution and its link with synaptic proteins, KIBRA appears as 8 am–8 pm). Food and water were available at libitum. Genotyping.
a potentially important regulator of synaptic plasticity. Transgenic mouse identification was performed by polymerase
In the hippocampus, the forms of activity-dependent plasticity chain reaction (PCR) on isolated genomic DNA for acGFP and rtTA2
thought to be implicated in memory formation involve the transgenes. Pups were ear clipped at weaning. Genomic DNA was
strengthening or weakening of synaptic connections between neu- extracted from ear biopsies by alkaline lysis (HotSHOT method)
rons (Bliss & Collingridge, 1993). Such adaptive changes occur in prior to PCR. The PCR reaction was performed under standard con-
part, through the activation of a-amino-3-hydroxyl-5-methyl-4-i ditions, using the following forward and reverse primers for acGFP
soxazole-proprionate (AMPARs) and N-methyl-D-Aspartate recep- and rtTA2 transgenes detection: acGFP for.: 50 -GTAGATCATGT
tors (NMDARs) in response to glutamate release, and through the GATCGCGC-30 , acGFP rev.: 50 -GTGAATCGCATCGAGCTGAC-30 ,
recruitment of downstream intracellular signaling pathways. F1: 50 -TGCCTTTCTCTCCACAGGTGTCC-30 , rtTA2-278-R: 50 -GAGAGCA
AMPARs are tetrameric receptors composed of GluA1-4 subunits CAGCGGAATGAC-30 . PCR products were separated by electrophoresis
that mediate fast synaptic transmission at most excitatory on a 2% agarose gel and visualized with GelRed staining
synapses. The combination of these subunits determines the func- (5 ll GelRed/100 ml) under UV light. Doxycycline (dox) administration.
tional properties of the receptor subtypes (Dingledine, Borges, Double positive mice and control littermates were fed with dox-
Bowie, & Traynelis, 1999). In the hippocampus, AMPARs are mostly containing food for 10 days. Briefly, food pellets (Alleinfuttermittel
composed of tetramers of GluA1A2 and GluA2A3 (Wenthold, für Versuchstiere, KLIBA NAFAG) were mixed with water at a ratio
Petralia, Blahos, & Niedzielski, 1996) with GluA1A2 heteromers at 1:1 for a few hours to overnight. 6 mg dox (Pelodis-doxycyline
CA1 pyramidal neuron synapses (Lu et al., 2009). They are mini- hyclate capsules; Apical Pharmaceutical Corp.) were added per gram
mally present at synapses in basal conditions, and are recruited of food. Medicated food was prepared freshly daily.
by constitutive vesicle transport mechanisms for synaptic trans-
mission and plasticity. Upon neuronal activity, new AMPARs are 2.2.2. RT-PCR
inserted to the plasma membrane by exocytosis and lateral diffu- Prefrontal cortex, insular cortex, and hippocampus were dis-
sion. They are then endocytosed and recycled back to the mem- sected and total RNA was extracted using the NucleoSpin Kit II
brane (Hanley, 2008; Kessels & Malinow, 2009). AMPAR (Macherey-Nagel), purified with RQ1 DNase (Promega) and
trafficking and recycling are critical for synaptic plasticity, in par- reverse-transcribed using the SuperScript First-Strand Synthesis
ticular for long-term potentiation (LTP) and long-term depression System for RT-PCR II (Invitrogen). PCR was performed on 2 ll
(LTD) (Park, Penick, Edwards, Kauer, & Ehlers, 2004; Shepherd & cDNA, using the primers acGFP for. (50 -GTAGATCATGT
Huganir, 2007). Here, we investigated the potential contribution GATCGCGC-30 ) and acGFP rev. (50 -GTGAATCGCATCGAGCTGAC-30 ).
of KIBRA to synaptic plasticity and to memory processes using PCR products were resolved on a 2% agarose gel. b-actin was used
in vitro and in vivo models. as an internal control for normalization.

2.3. Generation of a monoclonal anti-KIBRA antibody


2. Methods
Mouse monoclonal KIBRA antibodies were raised against a puri-
2.1. Ethical approval
fied recombinant human KIBRA fragment (aa 923-1113) fused to
GST. Antibodies from mouse hybridoma cell culture supernatants
Animal experiments were approved by Zürich cantonal author-
were affinity-purified using a GST-KIBRA 923-1113 affinity column
ities (License No. 205/2007) and performed as recommended by
and were tested by Western blot analysis with cell lysates from
the Federal Veterinary Office (FVO).
KIBRA overexpressing cells.

2.2. Transgenic mice 2.4. Western blot analysis

2.2.1. Generation The mouse hippocampus was dissected out and homogenized
Double transgenic mice for inducible and forebrain-specific using a 26G syringe in 10 mM HEPES, 1 mM MgCl2, 5 mM EDTA,
KIBRA expression were generated by crossing CaMKIIa promoter- 0.2% (v/v) Triton X-100 (Sigma), 10% (v/v) glycerol (Sigma), pro-
rtTA2 mice (Michalon, Koshibu, Baumgartel, Spirig, & Mansuy, tease inhibitor cocktail (Sigma), 250 lM PMSF (Sigma), and
2005) with pTRE-Tight-BI-AcGFP1-KIBRA mice carrying mouse 15 mM b-mercaptoethanol (Sigma). About 20 lg total protein
KIBRA cDNA (GenBank acc. No. NM_170779) and an acGFP reporter was resolved on 10% SDS-PAGE and transferred onto a nitrocellu-
driven by a rtTA2-responsive Tet-On bidirectional promoter. pTRE- lose membrane (BioRad). Membranes were blocked (Rockland IR
Tight-BI-AcGFP1-KIBRA transgenic mice were generated as fol- blocking buffer, Rockland), and incubated in monoclonal anti-
lows: KIBRA cDNA was excised from the vector pBSK(-)-KIBRA by mouse KIBRA (1:1000) and mouse anti-mouse b-actin (1:2000;
EcoRI/XhoI digestion, blunt-ended with T4 polymerase (New Eng- AC-15, Sigma) primary antibodies. Membranes were then incu-
land Biolabs), and reinserted into the EcoRV site of pNN265. A NotI bated in goat anti-rabbit IRDye 800 and goat anti-mouse IRDye
fragment was excised (5.2 kb) and introduced in the NotI site of 680 secondary antibodies (1:5000, Li-Cor Biosciences). Band inten-
pTRE-Tight-BI-AcGFP1 (Clontech). XmnI/EcoRV digest resulted in sity was determined and quantified using an Odyssey IR scanner
a 7 kb fragment that was microinjected into fertilized oocytes with (Li-Cor Biosciences). The protein signal was normalized to b-actin.
a mixed C57Bl/6J and DBA2 background. Integration of the KIBRA
construct occurred in 4 founders that transmitted the transgene 2.5. Immunohistochemistry
to the F1 generation, giving rise to 4 independent lines. The 4 lines
were backcrossed to CaMKIIa promoter-rtTA2 mice with a Mice were sacrificed and transcardially perfused with Ringer
C57Bl/6J background. For the experiments, 3–4 months old trans- solution followed by 4% paraformaldehyde (Sigma) in phosphate
genic males from the line 2 F4 generation (backcrossed for 3 buffer (0.1 M, pH7.4; flow: 20 ml/min). Brains were quickly

Please cite this article in press as: Heitz, F. D., et al. The memory gene KIBRA is a bidirectional regulator of synaptic and structural plasticity in the adult
brain. Neurobiology of Learning and Memory (2016), http://dx.doi.org/10.1016/j.nlm.2016.07.028
F.D. Heitz et al. / Neurobiology of Learning and Memory xxx (2016) xxx–xxx 3

removed, post-fixed in the same solution and transferred into a an interface chamber continuously flowed with aCSF at 1–2 ml/
30% sucrose solution. Coronal sections (40 lm thick) were cut with min. A monopolar electrode was placed in the Schaffer collateral
a cryostat. Free-floating brain sections were washed in 0.1 M PB, fibers, and stimulation pulses applied at 0.033 Hz with stimulus
blocked and permeabilized in 0.1 M PB, 0.4% Triton X-100 (Sigma) intensity ranging from 20 to 80 lA, yielding evoked field EPSPs
and 10% heat-inactivated horse serum (HS; Sigma) for 12 h at 4 °C. (f-EPSPs) of 0.2–0.5 V. f-EPSPs were recorded in the stratum radia-
Slices were then incubated with a primary rabbit polyclonal anti- tum using a borosilicate micropipette filled with aCSF. The signal
KIBRA antibody (V-13, Santa Cruz Biotechnology) with or without was amplified with an Axopatch 200A amplifier (Molecular
KIBRA blocking peptide (sc-133374 P, Santa Cruz Biotechnology) Devices, Union City, CA), digitized by a Digidata 1200 interface
and a primary mouse monoclonal anti-NeuN antibody (A60, Milli- (Molecular Devices) and sampled at 10 kHz with Clampex 8.2
pore) for 12 h at 4 °C in 0.1 M PB, 0.4% Triton X-100 and 5% HS. (Molecular Devices). Baseline was recorded for a minimum of
Slices were washed in 0.1 M PB, 0.4% Triton X-100 and incubated 20 min or until stable. Plasticity was then induced by stimulation
overnight at 4 °C with a goat anti-rabbit FITC and a donkey anti- with either 1 Hz for 15 min (900 pulses), 100 Hz for one train of
mouse TRITC fluorescence-conjugated secondary antibody (Jack- 1 s tetanus, or 100 Hz for three trains of 1 s tetanus separated by
son ImmunoResearch). DAPI (Invitrogen) was used for the staining 20 s. Data were analyzed by measuring the slope of individual
of neuronal nuclei. After washing in 0.1 M PB, slices were mounted f-EPSPs at 1–1.5 ms after the stimulus pulse by linear fitting using
using Moviol (Molecular Probes) and stored in the dark at 4 °C. Low Clampfit (Molecular Devices).
magnification fluorescence images were acquired at 20 with a
CoolSNAPK4 digital camera (Roper Scientific) mounted on an Axio- 2.7.2. Whole-cell recording
phot microscope (Zeiss) and analyzed using MCID Elite 7.0 soft- Transversal slices (250 lm thickness) were cut using a tissue
ware (MCID). High magnification images were acquired at 80 vibratome (VT 1000S; Leica Microsystems, Bannockburn, IL). Slices
and 160 with a Zeiss LSM 410 confocal laser-scanning micro- were kept in aCSF at room temperature before recording. Then,
scope equipped with a 40 oil-immersion objective (N.A. = 1.25) slices were transferred to a submerged recording chamber and per-
and laser excitation at 460 nm (DAPI), 488 nm (FITC) and 570 nm fused with aCSF containing 2.5 lM bicuculline and bubbled with
(TRITC). Each image was a z series projection of 10 stacks taken 95% O2/5% CO2. All experiments were performed at room temper-
at 1 lm depth intervals. ature. Patch pipettes (2–5 MX) for whole-cell voltage recordings
were filled with intracellular solution containing (in mM) 120
2.6. Behavioral testing Cs-gluconate, 10 CsCl, 10 HEPES, 8 NaCl, 0.2 EGTA, 2 MgATP, 0.3
Na3GTP, and 10 phosphocreatine (pH 7.3 with CsOH, osmolarity
Morris water maze testing was carried out using a white round 290–300 mOsm). In some experiments 100 lM spermine was
pool (150 cm in diameter and 50 cm high) filled with opaque water added to the intracellular solution. Liquid junction potentials were
maintained at 21–22 °C. The pool was in a dimly lit room with var- not corrected. Pyramidal cells in the CA1 region of the hippocam-
ious distal cues on white walls. Each animal was tracked using a pus were identified by infrared differential interference contrast
video tracking system (EthoVision, Noldus). Visible platform train- video microscopy with an Olympus BX51 microscope. Whole-cell
ing (non-spatial or cued learning): Mice were trained to reach a ran- voltage clamp recordings were made with a MultiClamp 700A
domly located visible (flagged) platform with four trials per day (Molecular Devices) amplifier. Signals were filtered at 2 kHz and
(max 90 s) for two days with 15 min inter-trial intervals (ITIs). digitized at 10 kHz by an analog-to-digital converter (Digidata
Mice were left on the platform for 20 s after each trial. This task 1322A, Molecular Devices). To evoke synaptic potentials and cur-
was conducted before the spatial version as a control condition rents from two independent pathways, two low-resistance
to test the ability to learn to swim towards a cued goal. Hidden plat- (2 MX) glass electrodes filled with standard ACSF were placed at
form training (spatial learning): Mice were trained to find an hidden a distance of 100 lm from the soma. Excitatory synaptic potentials
platform with four trials per day (max 90 s), 15 min ITIs for six were evoked by extracellular field stimulation (100 ls) at 0.125 Hz
days. Mice were split into subgroups, each trained with a different with a delay of 4 s between control and paired pathway. Stimulus
platform location to counterbalance potential quadrant effects. intensity was adjusted to produce a single excitatory postsynaptic
Mice were left on the platform for 15 s after each trial. Memory test current (EPSC) with an average amplitude of 50–150 pA. For anal-
(probe trial): 24 h and 28 days after the last training trial, mice ysis of synaptic AMPARs, the AMPA component of the mixed
were tested for their memory for the platform location after the AMPAR and NMDAR-mediated EPSC at a holding potential of
platform was removed from the water (60 s). Percent time spent +40 mV was measured at the beginning of the EPSC peak between
in each quadrant and number of platform crossings were 0 and 70 ms. Series and input resistance was continuously moni-
measured. tored during the recording in response to a small, hyperpolarizing
current or voltage step (10 mV; 250 ms). Cells were clamped at
2.7. Electrophysiological recordings 70 mV during EPSC recording and series resistance was compen-
sated 80–90%. LTP experiments were performed in a modified aCSF
Mice were anesthetized with isoflurane then decapitated. The containing 4 mM MgCl2, 4 mM CaCl2 and 10 lM Glycine. Extracel-
brain was quickly removed and immediately immersed in ice- lular stimulation was adjusted until a stable baseline of EPSCs
cold freshly prepared artificial CSF (aCSF; field recording: could be recorded for 10 min (75 sweeps). The peak EPSC ampli-
119 mM NaCl, 26 mM NaHCO3, 2.5 mM KCl, 1.3 mM NaH2PO4, tude of the synaptic response was calculated as the difference
1.3 mM MgCl2, 11 mM glucose, 2.5 mM CaCl2, gassed with 95% between maximum current in a time window of 25 ms after extra-
O2/5% CO2; whole-cell recording: 125 mM NaCl, 25 mM NaHCO3, cellular stimulation and the average current during the 50 ms pre-
2.5 mM KCl, 1.25 mM NaH2PO4, 1 mM MgCl2, 25 mM glucose, ceding extracellular stimulation. EPSC amplitudes were
2 mM CaCl2, gassed with 95% O2/5% CO2. normalized to the average amplitude before LTP induction. EPSC
decays were fitted with a single exponential from 90% to 10% of
2.7.1. Field recording the EPSC peak. A postsynaptic depolarization pairing protocol
Acute slices (400 lm thick) were prepared with a vibratome (VT was used for LTP induction in voltage clamp experiments. For
1000S; Leica Microsystems, Bannockburn, IL) in ice-cold gassed depolarization pairing 180 paired pathway EPSCs were evoked at
aCSF. Sections were incubated in aCSF at 34 °C for 20 min then kept 1 Hz and were paired with continuous postsynaptic depolarization
at room temperature for at least 1 h. Recording was performed in at 0 mV. After LTP induction, EPSC sampling was continued for up

Please cite this article in press as: Heitz, F. D., et al. The memory gene KIBRA is a bidirectional regulator of synaptic and structural plasticity in the adult
brain. Neurobiology of Learning and Memory (2016), http://dx.doi.org/10.1016/j.nlm.2016.07.028
4 F.D. Heitz et al. / Neurobiology of Learning and Memory xxx (2016) xxx–xxx

to 30 min. Data analysis was performed using custom written soft- Neurobasal growth medium supplemented with 2% B27, 2 mM
ware in IGOR (Wavemetrics, Lake Oswego, OR). Bath application of Glutamax and 5% FBS.
AMPA was performed as previously described {Schnell, 2002 Mouse neurons in culture were stained using mouse mono-
#142}. Values are expressed as mean ± SEM. Two-tailed Student’s clonal anti-KIBRA, rabbit polyclonal anti-GluA1 (ABN241, Milli-
t-tests were used for the calculation of statistical significance. pore) and mouse monoclonal anti-PSD-95 (7E3-1B8, Sigma)
primary antibodies. Images were acquired with a Zeiss LSM 410
2.8. Hippocampal slice culture and viral infection confocal laser-scanning microscope equipped with a 63 oil-
immersion objective (N.A. = 1.32) and laser excitation at 488 nm
Organotypic hippocampal slices from 5 to 7 day old postnatal (FITC) and 570 nm (TRITC). Each image was a z series projection
wild-type C57/BL6 mice were prepared and cultured for 2 weeks of 16–17 stacks taken at 0.27 lm depth intervals.
using the roller-tube technique (Gahwiler, Capogna, Debanne,
McKinney, & Thompson, 1997). Slices were then transferred to an 2.12. Colocalization and co-immunoprecipitation assays
electrophysiological recording chamber at room temperature and
the CA1 pyramidal cell layer was injected with 2 ll of lentiviral Green monkey CV1 and HEK293T cells were cultured as
particles (5  106 infectious units/ml) containing several expression described previously (Duning et al., 2008; Kremerskothen et al.,
constructs, each encoding a 19–25 nucleotide shRNA designed to 2003). CV1 and HEK293T cells were transfected with native
specifically knockdown kibra expression (sc-146464-V, Santa Cruz pQCXIP-mCherry, pQCXIP-mCherry-KIBRA and pcDNA6-Flag-
Biotechnology). Lentiviral particles containing constructs encoding KIBRA. Modified pEGFP-C1 and V37/V180-3-Flag plasmids were
a scrambled shRNA sequence were used as control (sc-108080, used to express the full-length human KIBRA sequence (Duning
Santa Cruz Biotechnology). Slices were returned to roller tubes et al., 2008; Kremerskothen et al., 2003) and the rat GluA1
after injection, and cultured for 1 week in medium supplemented C-Terminus (aa 821-907) fused to EGFP (pEGFP-C1-KIBRA and
with penicillin/streptomycin (1:500; Sigma) until recorded. pEGFP-C1-GluA1 C-term) and to a Flag tag (V37/V180-3-Flag-
KIBRA and V37/V180-3-Flag-GluA1 C-term). Rat hippocampal
2.9. Quantitative real-time RT-PCR neurons were co-transfected with pEGFP-C1-GluA1 C-term or pSIN
REP5-GFP-GluA2 and pQCXIP-mCherry or pQCXIP-mCherry-KIBRA
Quantitative PCR was performed on 0.5 ll cDNA using specific at DIV 14-15. Live-imaging assays were performed at 488 nm and
Taqman probes for the mouse KIBRA full-length mRNA 561 nm excitation to detect GFP and mCherry 48 h post-
(Mm01325387_m1; Applied Biosystems) and an Applied Biosys- transfection. Transient transfection was performed using Lipofec-
tems 7500 Thermal Cycler. Each sample was analyzed in triplicate tamine 2000 (Invitrogen, Germany) according to the manufac-
and an equal amount of cDNA was plated for each sample. Values turer’s instructions. For immunocytochemical staining, CV1 cells
were chosen in the linear range of amplification and the compara- grown on coverslips were fixed 24 h post-transfection in 4%
tive Ct method was used to assess differences in gene expression paraformaldehyde (PFA) in PBS at room temperature for 20 min.
between samples. b-actin was used as an internal control for Samples were washed with PBS and incubated for 10 min in
normalization. 50 mM NH4Cl in PBS to quench reactive amino groups. Staining
was performed using mouse monoclonal JL8 antibody directed
2.10. Subcellular fractionation against EGFP (Invitrogen). After washing in PBS, coverslips were
rinsed in water and cells were mounted in Moviol. Images were
Subcellular fractionation of adult mouse brain was conducted as obtained using a Leitz Observer Z1 fluorescence microscope. For
follows. The hippocampus was dissected and homogenized to 10% co-IP assays, lysates from co-transfected HEK293T cells were pre-
(wt/vol) in ice cold 0.32 M sucrose buffer (0.32 M sucrose, 20 mM pared by scraping cells into IP buffer (1% Triton-X 100, 20 mM
HEPES (pH 7.4), 5 mM EDTA, and protease inhibitor cocktail Tris-HCl (pH 7.5), 25 mM NaCl, 50 mM NaF, 15 mM Na4P2O7, and
(Sigma) using a tissue lyser (Qiagen). The homogenate was pro- 1.5 mM EDTA) containing protease inhibitor (Complete; Roche,
cessed by sequential centrifugation to collect different fractions Mannheim, Germany). Lysates were centrifuged at 10,000g for
(P2, LS1, LP1, LS2, LP2, SM) as described previously (Suh et al., 30 min at 4 °C. For precipitation of EGFP-tagged KIBRA and GluA1
2010). For each fraction, 20 lg of total protein were used for Wes- C-term, GFP-Trap-A (Chromotec) was used according to the manu-
tern blotting and the following primary antibodies: mouse mono- facturer’s instructions. For precipitation of Flag-tagged KIBRA and
clonal anti-KIBRA, mouse monoclonal anti-PSD-95 (7E3-1B8, GluA1 C-term, supernatants were incubated with anti-FLAG affin-
Sigma), and mouse monoclonal anti-mouse GluN1 (54.1, ity beads (Sigma-Aldrich) overnight at 4 °C on a rocking platform.
Millipore). For endogenous IP, rat brains were washed and homogenized in
buffer (4 mM HEPES/KOH, pH 7.4; 320 mM sucrose) and cen-
2.11. Neuronal cultures trifuged at 800g for 10 min at 4 °C. Supernatant was centrifuged
at 9,000g for 15 min at 4 °C and resulted pellet was resuspended
Primary hippocampal neurons were isolated from embryonic in homogenization buffer and centrifuged at 11,000g for 15 min
day 18 (E18) control or transgenic mice and rat pups hippocampi. at 4 °C. The resulting pellet (P2) was solubilized in 0.5% Tri-
Mouse hippocampal neurons were cultured on 12-well dishes as ton/50 mM Tris/HCl, pH 9.0 and centrifuged 10 min at 100,000g.
previously described (Kaech & Banker, 2006). KIBRA transgene 800 lg of each P2 supernatant was used for each immunoprecipi-
expression was induced by adding dox (1 lM) to the culture med- tation. Extracts were pre-cleared for 2 h with 40 ll of protein A/G
ium for 48 h. To knockdown KIBRA in neurons, we used commer- sepharose beads (Pierce) at 4 °C. Rabbit KIBRA antibodies or rabbit
cially available lentiviral particles containing several expression IgG control were coupled 90 min at 4 °C to 40 ll protein A/G
constructs each harboring a target-specific shRNA (sc-146464-V, sepharose beads. Pre-cleared P2 fraction was added to protein
Santa Cruz Biotechnology). Lentiviral particles (5  106 infectious A/G sepharose beads and incubated overnight at 4 °C on an over-
units/ml) containing a construct encoding a scrambled shRNA head rotator. For elution of bound proteins, washed beads were
sequence were used as control (sc-108080, Santa Cruz Biotechnol- resuspended in sample buffer and boiled for 5 min at 95 °C.
ogy). Neuronal infections were performed at DIV12-18 by using Samples were subsequently subjected to SDS-PAGE and Western
2 ll of lentivirus per well on a 12-well dish. Rat hippocampal blot analysis as described earlier (Duning et al., 2008). 3Flag
neurons were cultured onto poly-L-lysine coated coverslips in was used as negative control samples.

Please cite this article in press as: Heitz, F. D., et al. The memory gene KIBRA is a bidirectional regulator of synaptic and structural plasticity in the adult
brain. Neurobiology of Learning and Memory (2016), http://dx.doi.org/10.1016/j.nlm.2016.07.028
F.D. Heitz et al. / Neurobiology of Learning and Memory xxx (2016) xxx–xxx 5

2.13. Recycling and internalization assays imaged through a 63 oil objective (N.A. = 1.10–1.30) and
collected at a rate of 1 image per min. Images were analyzed using
AMPAR recycling was measured in primary hippocampal neu- ImageJ software (NIH) by calculating the normalized change in
rons as previously described (Suh et al., 2010), with some modifi- average pHluorin over mCherry fluorescence intensities from a
cations. Briefly, dissociated neurons in culture (DIV17-23) were somatic and dendritic shaft area defined manually to compensate
incubated on coverslips in conditioned medium for 15 min at for x-y movements of the recorded neurons. The fluorescence
20–25 °C with rabbit polyclonal anti-GluA1 primary antibody intensity change is expressed as DF/Fo and the maximal amplitude
(ABN241, Millipore) directed to the N-terminus to label surface of fluorescence change (DFmax/Fo) represents the extent of GluA1
AMPARs. Neurons were then incubated in conditioned medium exocytosis.
for 30 min at 37 °C to allow internalization of the receptors. Non-
internalized surface bound antibody was removed by stripping in
0.5 M NaCl and 0.2 M acetic acid for 2 min at 4 °C. Stripping was 2.15. Morphological analysis
performed directly after primary antibody incubation as control.
Neurons were then incubated in conditioned medium for 1 h at Immunostaining with a mouse monoclonal anti-MAP2 primary
37 °C to allow the recycling of receptors back to the plasma mem- antibody (AP20, Novus Biologicals) was used on mouse primary
brane. Neurons were fixed in 4% PFA in 0.1 M PBS for 10 min at neurons. Golgi staining was performed on mouse brain slices using
4 °C, washed (in PBS 0.1 M, 3% BSA), and incubated for 45 min at FD Rapid GolgiStain Kit (FD NeuroTechnologies) and according to
room temperature (RT) with an excess of Cy2-conjugated sec- the manufacturer’s instructions. Reconstruction and morphological
ondary antibody (in PBS 0.1 M, 3% BSA) to label surface-recycled analyses were performed with Imaris 7 software (Bitplane).
receptors. Internalized receptors that failed to recycle back to the Fluorescent images were taken with a Zeiss LSM 410 confocal
plasma membrane were subsequently labeled by permeabilization laser-scanning microscope equipped with a 63 oil objective
with 0.1% Triton X-100 (Sigma) in PBS 0.1 M for 30 min at RT and (N.A. = 1.32) and laser excitation at 488 nm. Each image was a z
by incubation with Cy5-conjugated rabbit secondary antibody (in series projection of 16–17 stacks taken at 0.27 lm depth intervals.
PBS 0.1 M, 3% BSA) for 45 min at RT. To control for the specificity Golgi images were acquired on an Axiophot microscope (Zeiss). We
of internalized receptors staining, some neurons did not undergo also used unstained brain slices of Thy1-YFP (Yellow Fluorescent
permeabilization. Endocytosis of GluA1-AMPARs was evaluated Protein) mice crossed with KIBRA transgenic mice (F4) to analyze
by simultaneous staining surface and internalized receptors. The the morphology of YFP expressing neurons in the CA1 area of the
assay was performed as described for the recycling of GluA1- hippocampus. Transgenic males heterozygous for YFP (control
AMPARs, except that neurons were incubated for 15 min at 37 °C mice) or for both YFP and KIBRA (OE mice) were fed with
to allow AMPARs internalization and immediately fixed before doxycycline-containing food for 10 days prior sacrifice and histo-
incubation with the secondary antibody. Imaging was performed logical analysis.
with a Zeiss LSM 410 confocal laser-scanning microscope equipped
with a 40 oil-immersion objective (N.A. = 1.25) and laser excita- 2.16. Statistical analysis
tion at 488 nm (Cy2) and 633 nm (Cy5). Each image was a z series
projection of 10 stacks taken at 1 lm depth intervals and using Data are presented as mean normalized to baseline or con-
identical acquisition parameters. The amount of recycling and trol ± SEM. Student’s t-test and analysis of variance (ANOVA) were
endocytosis was quantified using Imaris 7 software (Bitplane). used to compare non-normalized data. Statistical significance was
Data for recycled receptors are presented as a recycling index, set at ⁄p 6 0.05, ⁄⁄p 6 0.01 and ⁄⁄⁄p 6 0.001.
which is the ratio of the surface (recycled) fraction and the total
(internalized and recycled) fraction. Data for internalized receptors
are presented as an internalization index, which is the ratio of the 3. Results
internalized fraction and the total (surface and internalized)
fraction. 3.1. KIBRA overexpression in hippocampal neurons in vivo

2.14. Live-cell imaging We studied the role of KIBRA in memory formation and hip-
pocampal plasticity by selectively modulating its level of expres-
Hippocampal neurons from E18 rat pups were plated onto sion in mouse forebrain neurons in vitro and in vivo. KIBRA was
poly-L-lysine coated coverslips in Neurobasal growth medium sup- conditionally overexpressed selectively in forebrain neurons by
plemented with 2% B27, 2 mM Glutamax, 50 U/mL penicillin, generating transgenic mice carrying a full-length KIBRA gene
50 lg/mL streptomycin and 5% FBS. Neurons were transfected at under the control of a tetracycline-responsive bidirectional pro-
DIV 14-17 using lipofectamine 2000 (Invitrogen) and SEP-GluA1 moter (BiTetO) fused to a green fluorescent protein (acGFP) repor-
live-imaging assays were performed 48 h post-transfection as ter. These animals were crossed with mice expressing rtTA2 under
described previously (Ashby et al., 2004). Briefly, coverslips con- the control of a CaMKIIa promoter (CaMKIIa-rtTA2) (Michalon
taining neurons were assembled onto a closed perfusion chamber et al., 2005) (Fig. 1A). In double transgenic mice (heterozygous
and continuously perfused with recording buffer (125 mM NaCl, for both transgenes), treatment with doxycycline (dox) induced
25 mM NaHCO3, 2.5 mM KCl, 1.25 mM NaH2PO4, 1 mM MgCl2, KIBRA mRNA overexpression in neurons in forebrain structures
25 mM glucose, 2 mM CaCl2, 1 lM TTX, pH 7.4), After 3 min of including the prefrontal and somatosensory cortex, and the hip-
baseline recording (F0), neurons were perfused with recording pocampus (Fig. 1B). Quantitative real-time PCR and Western blot
buffer supplemented with 200 lM glycine for 3 min before the analysis using a selective antibody against KIBRA (Supplementary
perfusion was switched back to recording buffer for the remainder Fig. 1A) showed that KIBRA (Kremerskothen et al., 2003;
of the recording session. All imaging experiments were performed Papassotiropoulos et al., 2006) (Fig. 1C) was expressed in the trans-
at 37 °C using a Leica SP5 confocal microscope. The pHluorin genic mouse hippocampus (Supplementary Fig. 1B), with a 2-fold
fluorescence was imaged at 488 nm excitation and collected increase in KIBRA protein (Fig. 1D). The increase in KIBRA protein
through a 505–550 nm filter, while the mCherry signal was imaged was detected in neurons, particularly in CA1 pyramidal neurons
at 561 nm excitation and 575–615 nm emission. Neurons were as shown by immunostaining (Fig. 1E and F).

Please cite this article in press as: Heitz, F. D., et al. The memory gene KIBRA is a bidirectional regulator of synaptic and structural plasticity in the adult
brain. Neurobiology of Learning and Memory (2016), http://dx.doi.org/10.1016/j.nlm.2016.07.028
6 F.D. Heitz et al. / Neurobiology of Learning and Memory xxx (2016) xxx–xxx

Fig. 1. Conditional KIBRA overexpression (OE) in the adult mouse forebrain. (A) Schematic representation of the rtTA2 system used for inducible and neuron-specific
expression of KIBRA in the mouse brain. Doxycycline (Dox) binds to rtTA2, which is expressed in forebrain neurons due to the CaMKIIa promoter, and leads to rtTA2-mediated
activation of the tetracycline-responsive BiTetO promoter, and KIBRA and acGFP expression in forebrain neurons. (B) RT-PCR analyses of acGFP expression reflecting KIBRA
expression in prefrontal cortex (PfCx), cortex (Cx), and hippocampus (Hi) in transgenic (OE + dox) and control (Control) littermates treated with dox, and in transgenic mice
not treated with dox (OE-dox). No transgene expression was detected in the absence of dox. -RT = no reverse transcription, H2O = water control. (C) Schematic representation
of full-length KIBRA protein including from the N- (N-term) to C-terminal (C-term) region two WW domains for binding to proline-rich sequences, a putative nuclear
localization signal (NLS), a calcium-dependent binding domain (C2), a glutamic acid-rich (Glu-rich) domain, two phosphorylation sites on Ser residues (p-Ser sites), and a PDZ
binding motif. (D) Immunoblot analyses of KIBRA protein in two independent samples from transgenic (OE) and control mice treated with dox (left panel). b-actin was used as
loading control. Quantification relative to control (right panel; KIBRA protein level: control = 1 ± 0.1 (n = 4) versus OE = 2.6 ± 0.7 (n = 3), t(5) = 2.67, ⁄p < 0.05. Data are
expressed as fold change (mean ± SEM). (E) Immunohistochemical analyses of KIBRA expression (green) in the hippocampus in transgenic (OE, upper panel) and control
(control, middle panel) mice. No fluorescence was observed in the presence of a KIBRA antibody blocking peptide (bottom panel), indicating no unspecific staining. Scale bar:
200 lm. (F) KIBRA (green) co-immunostaining with the neuronal marker NeuN (red), and DAPI (blue) in hippocampus area CA1 in transgenic (OE, left) and control (Control,
right) mice. Scale bar: 50 lm and 10 lm. (For interpretation of the references to colour in this figure legend, the reader is referred to the web version of this article.)

3.2. KIBRA overexpression impairs spatial memory No difference in performance between control and KIBRA
transgenic mice was however observed on the open field
Since KIBRA has been implicated in human memory, we exam- (Supplementary Fig. 2B), or in contextual or tone fear memory
ined the impact of its overexpression on memory in the transgenic (Supplementary Fig. 2C).
animals using a water maze (Morris, 1984; Vorhees & Williams,
2006). On a non-spatial version of the task, both control and trans- 3.3. KIBRA acts as a positive regulator of synaptic potentiation in CA1
genic animals learned the platform position similarly well, and had neurons
comparable swimming distance and latency to reach the platform
(Supplementary Fig. 2A). In contrast, on a spatial version of the We next examined the effect of KIBRA overexpression on synap-
task, while learning was normal in both groups (Fig. 2A), spatial tic plasticity in the hippocampus after one- or three-train of high
memory 24 h after the end of training was impaired in the trans- frequency stimulation (HFS) of Schaffer collaterals. LTP was signif-
genic mice. Transgenic mice spent significantly less time in the tar- icantly increased in hippocampal area CA1 after one-train HFS in
get quadrant (Fig. 2B, left) and had fewer platform crossings slices from the transgenic mice compared to controls, but it was
(Fig. 2B, right), suggesting impaired spatial memory. The memory lower after three-train HFS (Fig. 3A and B). In the slices from trans-
impairment was persistent and could still be detected based on genic mice, the overall level of potentiation after one or three-train
time spent in the target quadrant after 28 days (Fig. 2C, left). Both HFS was similar, suggesting that LTP is at its maximum level in
control and transgenic mice showed few platform crossings transgenic slices already after only one tetanus. Indeed, when a
(Fig. 2C, right), indicative of a gradual decline in memory strength. second tetanus was delivered 15 min after the first tetanus, LTP

Please cite this article in press as: Heitz, F. D., et al. The memory gene KIBRA is a bidirectional regulator of synaptic and structural plasticity in the adult
brain. Neurobiology of Learning and Memory (2016), http://dx.doi.org/10.1016/j.nlm.2016.07.028
F.D. Heitz et al. / Neurobiology of Learning and Memory xxx (2016) xxx–xxx 7

Fig. 2. KIBRA overexpression in forebrain neurons impairs spatial memory. (A) Path length (left panel) and escape latency (right panel) to find a hidden platform in a water
maze were similar in control (n = 9) and transgenic mice (OE, n = 8). Comparable decrease in path length (control: F(5,20) = 5.3, ⁄⁄p < 0.01; OE: F(5,40) = 3.0, ⁄p < 0.05; left
panel) and escape latency (control: F(5,20) = 5.0, ⁄⁄p < 0.01; OE: F(5,40) = 3.3, ⁄p < 0.05; right panel) over the 6 days of acquisition (4 trials per day) in control and transgenic
mice. (B) Performance on a spatial memory test 24 h after the last acquisition trial. Percentage of total time spent in each quadrant (left panel); chance is at 25%. Time spent in
the target quadrant was significantly above chance in control and transgenic mice (control: t9 = 3.4, ⁄p < 0.05; OE: t8 = 4.4, ⁄⁄p < 0.01) but, transgenic mice spent significantly
less time in the target quadrant (control: 57.8 ± 5.4%; OE: 34.1 ± 2.1%; F(1,11) = 8.5, ⁄⁄p < 0.01), and had fewer crossings of the platform location (control: 4.2 ± 0.4; OE:
2.1 ± 0.5; F(1,12) = 6.5, ⁄⁄p < 0.01; right panel) than control mice. (C) Performance on a spatial memory test 28 days after the last acquisition trial. Transgenic mice spent
significantly less time in the target quadrant than control mice (control: 60.4 ± 5.3%; OE: 35.2 ± 2.2%; F(1,11) = 12.9, ⁄⁄p < 0.01; right panel), with no differences in crossings of
the platform location (control: 1.7 ± 0.8; OE: 1.9 ± 0.3; right panel). Chance is at 25%. Target, left, opposite, right indicate the quadrant position relative to the platform’s last
position. Data are mean ± SEM.

was not further increased in the transgenic slices unlike in controls overexpression and knockdown have an opposite effect on LTP
(Supplementary Fig. 3A and B). The effect on LTP observed in trans- and confirm the involvement of KIBRA in this form of synaptic
genic slices resulted from KIBRA overexpression and not from any plasticity. Next, we examined whether KIBRA is also involved in
effect of transgene integration since LTP was unaffected in double LTD. Prolonged low frequency stimulation (LFS: 15 min at 1 Hz)
transgenic mice not treated with dox (Supplementary Fig. 3C). Fur- at Schaffer collaterals induced LTD in control slices, but not in
ther, LTP change upon KIBRA overexpression was not due to any transgenic slices in which it rather led to a robust LTP in the CA1
gross alteration in basal synaptic transmission since field excita- area (Fig. 3E). This LTP slowly increased and reached an amplitude
tory postsynaptic potentials (f-EPSPs) in response to increasing comparable to that induced by one-train HFS about 35 min after
stimulation intensity were similar in transgenic and control slices stimulation. Together, the data show that KIBRA overexpression
(Fig. 3B, right). To confirm the involvement of KIBRA in LTP, we shifts hippocampal plasticity in favor of LTP and prevents LTD
next tested the effect of KIBRA knockdown. For this, a lentivirus (Fig. 3F), pointing to a function of KIBRA as positive regulator of
expressing a KIBRA shRNA was injected in the CA1 pyramidal cell synaptic potentiation. The saturating effect of KIBRA overexpres-
layer in organotypic hippocampal slices (Koshibu et al., 2009), sion on LTP and the impairing effect on LTD may explain the mem-
and fEPSPs were recorded five days later. KIBRA shRNA signifi- ory impairment, as inappropriate synaptic strengthening in the
cantly decreased the expression of full-length KIBRA mRNA hippocampus is likely detrimental to memory processes. It is also
(Fig. 3C). This led to an impairment of one-train LTP in CA1 neurons consistent with the hypothesis that hippocampal LTD is needed
(Fig. 3D), an effect that was not seen in neurons infected with a for memory formation (Kemp & Manahan-Vaughan, 2007, 2008;
scrambled control shRNA. These data reveal that KIBRA Manahan-Vaughan & Braunewell, 1999; Migaud et al., 1998;

Please cite this article in press as: Heitz, F. D., et al. The memory gene KIBRA is a bidirectional regulator of synaptic and structural plasticity in the adult
brain. Neurobiology of Learning and Memory (2016), http://dx.doi.org/10.1016/j.nlm.2016.07.028
8 F.D. Heitz et al. / Neurobiology of Learning and Memory xxx (2016) xxx–xxx

Fig. 3. KIBRA overexpression increases hippocampal CA1 LTP while KIBRA knockdown blocks LTP. (A) f-EPSP slope is increased in transgenic slices (OE) compared to controls
following one-train of 100 Hz stimulation (1  100 Hz HFS) (f-EPSP slope averaged from min 20 to 45: control = 119.1 ± 6.9% (n = 6) versus OE = 152.1 ± 11.2% (n = 7), t(11)
=2.40, ⁄p < 0.05). (B) LTP induced by three-train HFS (3  100 Hz HFS) is higher in control slices than in transgenic slices (f-EPSP slope averaged from min 20 to 45:
control = 206.3 ± 14.5% (n = 9) versus OE = 162.1 ± 12.3% (n = 7), t(14)=2.24, ⁄p < 0.05). Stimulation was delivered at time zero. Inset: input-output curve with f-EPSPs over
increasing stimulation intensity showing comparable basal synaptic transmission in CA1 neurons in transgenic slices overexpressing KIBRA (OE, n = 5) and control slices
(n = 5). Data are expressed as mean f-EPSP slope. (C) Quantitative real-time PCR analyses of KIBRA full-length mRNA expression in hippocampal slice cultures injected in area
CA1 with a lentivirus expressing a KIBRA shRNA for knockdown (KD) or a lentivirus expressing a scrambled shRNA (control). Five days after infection, full-length mRNA was
significantly decreased in slices injected with the KIBRA shRNA lentivirus (control = 1.00 ± 0.08 (n = 3) versus KD = 0.71 ± 0.05 (n = 4), t(5) = 3.25, ⁄p < 0.05). b-actin was used
as internal control. Data are expressed as fold change (mean ± SEM). (D) No LTP was observed in slices with KIBRA knockdown while control slices have normal LTP following
one-train of 100 Hz stimulation (1  100 Hz HFS) delivered at time zero (f-EPSP slope averaged from min 20 to 30: control = 146.3 ± 8.6% (n = 3) versus KD = 90.2 ± 6.6%
(n = 4), t(5) = 5.28, ⁄⁄p < 0.01). Horizontal bars indicate the time window for calculation of mean f-EPSPs in control and KIBRA knockdown slices before (1) and after (2) LTP
induction, as shown in representative traces (top insets). (E) Low frequency stimulation (15 min at 1 Hz, delivered at time zero) results in LTD in control slices, but induces LTP
in transgenic slices (f-EPSP slope averaged from min 20 to 45: control = 76.9 ± 4.3% (n = 4) versus OE = 151.2 ± 13.1% (n = 4), t(6) = 5.389, ⁄⁄p < 0.01). (F) Frequency response
curve in control and transgenic slices. 0.033 Hz corresponds to the stimulation frequency used to record basal synaptic transmission. (G) Whole-cell two-pathway recording
of CA1 pyramidal neurons in acute slices from control and KIBRA OE mice. EPSCs were evoked at membrane potential by presynaptic stimulation and LTP was induced by
postsynaptic depolarization (delivered at time zero). LTP is increased in the stimulated pathway in transgenic neurons (OE LTP-path, n = 9) compared to controls (control
LTP-path, n = 6). No LTP was induced in the non-stimulated pathway (control and OE Ctrl-path). (H) Analysis of mean EPSCs amplitude in control and KIBRA OE CA1 neurons
at baseline (averaged from min 10 to 0), 10 min after LTP induction (averaged from min 0 to 10; stimulated pathway (LTP-path): control = 147 ± 16% (n = 6) versus
OE = 229 ± 14% (n = 9), t(13) = 3.80, ⁄⁄p < 0.01; non-stimulated pathway (Ctrl-path): control = 99 ± 8% (n = 6) versus OE = 90 ± 6% (n = 9), t(13) = 0.92, n.s.), and over the last
20 min (averaged from min 20 to 40 after pairing; stimulated pathway: control = 123 ± 12% (n = 6) versus OE = 183 ± 13% (n = 9), t(13) = 3.20, ⁄⁄p < 0.01; non-stimulated
pathway: control = 98 ± 9% (n = 6) versus OE = 72 ± 4% (n = 9), t(13) = 2.76, n.s.). Baseline (100%) is represented by a dashed line. Data are expressed as mean f-EPSP slope and
mean EPSC amplitude of five-binned consecutive sweeps normalized to baseline (100%) ± SEM.

Moser, Krobert, Moser, & Morris, 1998; Nakao, Ikegaya, Yamada, Since KIBRA is present in synaptic terminals (Schneider et al.,
Nishiyama, & Matsuki, 2002; Willshaw & Buckingham, 1990). 2010), we tested whether it can modulate a form of
GluA1-dependent LTP in CA1 pyramidal neurons using whole-cell
3.4. KIBRA is specifically involved in activity- and GluA1-dependent patch-clamp recording. AMPAR-dependent excitatory post-
synaptic potentiation in CA1 neurons synaptic currents (EPSCs) were measured following a paired stim-
ulation using a protocol known to induce GluA1-dependent LTP
The expression and maintenance of LTP in the hippocampus lar- (Frey, Sprengel, & Nevian, 2009). This stimulation induced a robust
gely depend on postsynaptic AMPARs, and require the insertion of LTP that was specific to the stimulated pathway in control neurons.
new AMPARs in the postsynaptic membrane (Hayashi et al., 2000; In transgenic neurons, this form of LTP was significantly increased
Makino & Malinow, 2009; Takahashi, Svoboda, & Malinow, 2003). (Fig. 3G and H). Again, this effect was not due to a change in basal

Please cite this article in press as: Heitz, F. D., et al. The memory gene KIBRA is a bidirectional regulator of synaptic and structural plasticity in the adult
brain. Neurobiology of Learning and Memory (2016), http://dx.doi.org/10.1016/j.nlm.2016.07.028
F.D. Heitz et al. / Neurobiology of Learning and Memory xxx (2016) xxx–xxx 9

synaptic transmission since current-voltage relationship of 3.5. KIBRA colocalizes and interacts with GluA1-AMPARs in
AMPAR-mediated EPSCs at different holding potential was compa- hippocampal neurons
rable in control and transgenic neurons (Fig. 4A and B). A rectifica-
tion index calculated from the ratio of EPSCs amplitude at 60 and To further explore the relationship between KIBRA and GluA1-
+40 mV holding potential was also similar in control and trans- containing AMPARs (GluA1-AMPARs), we next examined the dis-
genic slices (data not shown). Further, the effect was not linked tribution of KIBRA in hippocampal neurons and its localization
to a change in NMDAR currents since the AMPAR/NMDAR ratio with respect to AMPARs. Various subcellular fractions were pre-
was comparable in control and transgenic cells (Fig. 4C). AMPAR- pared from adult mouse hippocampus, and assayed for KIBRA by
mediated spontaneous activity was also unaffected in transgenic Western blotting. KIBRA was enriched in synaptic fractions, specif-
neurons as seen by comparable spontaneous AMPARs EPSC (sEPSC) ically in synaptic membranes that also contained the postsynaptic
amplitude, sEPSC frequency, and sEPSC decay time course in CA1 components PSD-95 and GluN1 (Fig. 5A), consistent with the pres-
neurons in transgenic and control slices (Fig. 4D and E). The mea- ence of KIBRA in PSDs (Johannsen et al., 2008). At a subcellular
surements were specific to AMPAR EPSCs, since the AMPAR chan- level, immunostaining showed that KIBRA colocalizes with GluA1
nel blocker CNQX abolished spontaneous responses (Fig. 4E). clusters on dendrites, but is also present in somato-dendritic com-
Moreover, bath application of AMPA evoked a comparable partments in cultured neurons (Supplementary Fig. 4A and B). It
response in control and transgenic slices (Fig. 4F). These results also colocalizes with PSD-95 in these structures, confirming its
demonstrate that KIBRA is involved in GluA1-dependent synaptic presence in the PSD (Supplementary Fig. 4C and D). KIBRA is also
plasticity, but does not alter AMPAR subunit composition at distributed in the nucleus of hippocampal neurons, consistent with
synapses or the overall AMPAR content at the cell surface in resting its putative nuclear localization sequence and nuclear function
conditions (Meng, Zhang, & Jia, 2003). (Rayala et al., 2006). Next, the link between KIBRA and GluA1

Fig. 4. KIBRA overexpression does not affect AMPAR spontaneous activity in CA1 neurons. (A) Mean amplitude of AMPAR EPSCs recorded at different membrane holding
potential (70 mV to +40 mV) in patched CA1 pyramidal neurons in acute hippocampal slices from control (n = 7) and transgenic mice (OE, n = 4) using the same stimulation
position and intensity as during LTP recording. The amplitude of the evoked AMPAR EPSCs component was calculated over the first 20 ms and averaged for each potential. (B)
Representative traces in control and transgenic slices recorded at a holding potential of +40 and 70 mV. (C) AMPAR/NMDAR ratio calculated as the mean AMPAR EPSC
amplitude at 70 mV compared to the mean NMDAR EPSC amplitude at +40 mV (AMPAR/NMDAR ratio: control = 2.1 ± 0.3 (n = 7) versus OE = 2.3 ± 0.6 (n = 4), t(9) = 0.34, n.
s.). (D) Quantitative histograms showing AMPAR sEPSC amplitude (control = 21.2 ± 2.1 pA (n = 6) versus OE = 19.4 ± 1.4 pA (n = 8), t(12) = 0.74, n.s.), frequency
(control = 0.5 ± 0.2 Hz (n = 6) versus OE = 0.4 ± 0.1 Hz (n = 8), t(12) = 0.48, n.s.), and decay time (control = 6.8 ± 0.9 ms (n = 6) versus OE = 5.5 ± 1.6 ms (n = 8), t(12) = 0.64,
n.s.) over 15 min starting 10 min after cell clamping. (E) Representative traces of AMPAR sEPSCs at 70 mV holding potential in CA1 neurons from control and transgenic (OE)
slices. AMPAR sEPSCs were blocked by bath perfusion of the AMPAR blocker 6-cyano-7-nitroquinoxalene-2,3-dione (CNQX, 3 lM). (F) KIBRA OE (n = 10) does not change the
response to bath application of 500 nM AMPA when compared to control (n = 8). Data are expressed as mean ± SEM.

Please cite this article in press as: Heitz, F. D., et al. The memory gene KIBRA is a bidirectional regulator of synaptic and structural plasticity in the adult
brain. Neurobiology of Learning and Memory (2016), http://dx.doi.org/10.1016/j.nlm.2016.07.028
10 F.D. Heitz et al. / Neurobiology of Learning and Memory xxx (2016) xxx–xxx

Fig. 5. KIBRA is enriched at synapses and interacts with GluA1. (A) Subcellular fractionation of mouse hippocampal extracts showing that KIBRA is present in the synaptic
membrane (SM) fraction but not in other fractions except in low amount in crude synaptosomes (P2). The postsynaptic components PSD-95 and GluN1 are abundant in SM.
Input, crude hippocampal fraction; P2, crude synaptosomal fraction; LS1, synaptic vesicle and cytosolic supernatant; LP1, synaptosomal membrane fraction; LS2,
synaptosomal-cytosolic fraction; LP2, crude synaptic vesicle-enriched fraction; SM, synaptic membrane fraction. (B) and (C) CV1 cells and hippocampal neurons
co-transfected with constructs encoding the intracellular GluA1 C-terminal domain fused to EGFP (EGFP-GluA1 C-term) and mCherry as control (upper panels) or mCherry-
KIBRA (lower panels), respectively. EGFP-GluA1 C-term and mCherry are diffusely distributed in transfected cells, while expression of mCherry-KIBRA leads to the formation
of cytoplasmic aggregates containing EGFP-GluA1 C-term in CV1 cells and to somato-dendritic EGFP-GluA1 C-term redistribution with mCherry-KIBRA in hippocampal
neurons. Scale bar: 20 lm. (D) Colocalization of EGFP-GluA1 C-term and mCherry-KIBRA on dendrites of hippocampal neurons in culture (DIV17-23). Images were generated
by confocal immunofluorescence microscopy with a 63 oil-immersion objective. Scale bar: 5 lm. (E) Immunoprecipitation (IP) assays with KIBRA and GluA1 C-term.
HEK293 cells were co-transfected with constructs encoding EGFP-KIBRA and Flag-GluA1 C-term (left panel) or with Flag-tagged KIBRA and EGFP-GluA1 (right panel). Flag
only was used as negative control. Lysates from transfected cells were incubated with immobilized anti-GFP and anti-Flag antibody and immunoprecipitates were isolated by
centrifugation. Western blot (WB) analysis with anti-GFP and anti-Flag antibodies. (F) Immunoprecipitation (IP) assay on rat P2 brain fractions with rabbit anti-KIBRA
antibodies and normal rabbit IgG. Western blot analysis with anti-KIBRA and anti-GluA1 antibodies.

was examined by co-expression assays. A KIBRA variant fused to incorporate into synapses (Hanley, 2008; Makino & Malinow,
the mCherry reporter (mCherry-KIBRA) was co-expressed with 2009). Trafficking is controlled in part by the interaction of the
the GluA1 cytoplasmic C-terminus domain (aa 821–907) fused to C-terminus cytoplasmic tail of GluA1 with postsynaptic scaffolding
EGFP (EGFP-GluA1 C-term) in CV1 cells and hippocampal neurons. proteins (Bredt & Nicoll, 2003; Collingridge, Isaac, & Wang, 2004;
24 h after transfection, KIBRA and GluA1 colocalized in these two Groc & Choquet, 2006; Hayashi et al., 2000; Malinow & Malenka,
cell types (Fig. 5B and C). In CV1 cells, KIBRA and GluA1 formed 2002). Since KIBRA is associated with GluA1, we examined
large clusters, while in the absence of KIBRA, GluA1 was diffusely whether it can modulate the constitutive recycling of GluA1-
distributed in the cytoplasm (Fig. 5B). When alone, KIBRA itself containing AMPARs in basal conditions. We monitored GluA1 traf-
remained concentrated in punctates, probably due to its oligomer- ficking in primary hippocampal neurons in which KIBRA was either
ization activity after prolonged expression (Johannsen et al., 2008). knockdown or overexpressed. For knockdown, cultured neurons
Likewise in hippocampal neurons, KIBRA colocalized with GluA1 were infected with a lentivirus expressing a KIBRA shRNA, and
clusters, and both proteins were found in soma and dendrites for overexpression, neurons were prepared from KIBRA transgenic
including in dendritic spines (Fig. 5C and D). Moreover, co- mice and cultured. Quantitative immunostaining confirmed that
immunoprecipitation assays using recombinant EGFP- and Flag- KIBRA expression was significantly decreased by the shRNA and
tagged KIBRA and EGFP-GluA1 C-term showed that KIBRA and increased in transgenic neurons under these experimental condi-
GluA1 are present in the same protein complex (Fig. 5E). These tions (Supplementary Fig. 5). We then measured the constitutive
results were confirmed in vivo on brain fractions (Fig. 5F). Overall, recycling of GluA1-AMPARs in the soma and proximal dendrites
these results strongly suggest that KIBRA preferentially colocalizes of the cultured neurons using a GluA1 (N-terminus)-specific anti-
with GluA1 in hippocampal neurons. body for detection of both internalized and surface receptors
(Bhattacharyya, Biou, Xu, Schluter, & Malenka, 2009; Suh et al.,
3.6. KIBRA regulates constitutive and activity-dependent GluA1 2010). The signal for internalized GluA1 was significantly increased
recycling in hippocampal neurons by KIBRA knockdown in the soma and dendrites of infected neu-
rons, suggesting an accumulation of intracellular GluA1
In resting conditions in neurons, GluA1-containing AMPARs (Fig. 6A and B). This effect was likely due to an impaired GluA1
constitutively traffic to the membrane, while during LTP, new recycling, since GluA1 recycling index was decreased by KIBRA
AMPARs are recruited by exocytosis and lateral diffusion, and knockdown (Fig. 6B). In contrast, KIBRA overexpression increased

Please cite this article in press as: Heitz, F. D., et al. The memory gene KIBRA is a bidirectional regulator of synaptic and structural plasticity in the adult
brain. Neurobiology of Learning and Memory (2016), http://dx.doi.org/10.1016/j.nlm.2016.07.028
F.D. Heitz et al. / Neurobiology of Learning and Memory xxx (2016) xxx–xxx 11

Fig. 6. KIBRA regulates the constitutive and activity-dependent recycling of GluA1-AMPARs. (A) Differential staining of surface (recycled, top panels) and internalized
(bottom panels) GluA1-AMPARs in mouse primary hippocampal neurons infected with a scrambled shRNA lentivirus (Control) or a KIBRA shRNA lentivirus for knockdown
(KD), and in transgenic neurons overexpressing KIBRA (OE, 48 h of dox treatment). Images of representative neurons show that internalized GluA1 staining is increased by
KIBRA KD while surface GluA1 staining is increased by KIBRA OE in soma and dendrites. Neurons stripped immediately after incubation with GluA1 primary antibody
(Stripping 00 ) and neurons not permeabilized prior to secondary antibody incubation (Non perm.) were used as controls. Images were generated by confocal
immunofluorescence microscopy with a 40 objective. Scale bar: 60 lm. (B) Quantitative analyses of GluA1 recycling. A GluA1 recycling index was calculated as the
percentage of recycled receptors compared to total number of receptors (recycled and internalized) for the soma (Control = 1.00 ± 0.02 (n = 13); KD = 0.90 ± 0.03 (n = 13), t
(24) = 2.77, ⁄p < 0.05; OE = 1.12 ± 0.01 (n = 10), t(21) = 4.89, ⁄⁄⁄p < 0.001) and dendrites (Control = 1.00 ± 0.02 (n = 13); KD = 0.86 ± 0.03 (n = 10), t(21) = 4.03, ⁄⁄p < 0.01;
OE = 1.11 ± 0.01 (n = 11), t(22) = 4.65, ⁄⁄⁄p < 0.001). Quantitative histograms are mean ± SEM. (C)–(E) Cultured hippocampal neurons (DIV14-17) were co-transfected with
SEP-GluA1 and mCherry (control, n = 8) or SEP-GluA1 and mCherry-KIBRA (OE, n = 8)) constructs to analyze activity-dependent increase in GluA1 surface expression by
KIBRA. Surface SEP-GluA1 fluorescence intensity was measured 48 h post-transfection by live-cell confocal microscopy upon neuronal stimulation with 200 lM glycine for
3 min. C, Representative images of control and KIBRA overexpressing (OE) neurons with SEP and mCherry fluorescence visualization at low (top panels) and high (bottom
panels) magnification to observe individual dendrites. Images were generated by live confocal fluorescence microscopy with a 63 oil-immersion objective and converted to
a monochrome signal. Scale bar: 20 lm. (D) Time course of SEP-GluA1 fluorescence change (DF/F0) in dendrites and soma (left and right panels). Horizontal bars indicate the
time window of glycine perfusion delivered at 3 min. (E) Quantitative histogram showing the averaged maximum amplitude of SEP-GluA1 fluorescence change (DFmax/F0) in
response to glycine in dendrites (control = 1.11 ± 0.02 (n = 8) versus OE = 1.22 ± 0.04 (n = 8), t(14)=2.46, ⁄p < 0.05) and soma (control = 1.11 ± 0.09 (n = 3) versus
OE = 1.33 ± 0.12 (n = 8), t(9) = 1.002, n.s.). Data are mean ± SEM.

GluA1-AMPARs exocytosis and return to the membrane in soma confocal imaging using an adapted pH-sensitive GFP (super ecliptic
and dendrites (Fig. 6A and B). No fluorescence was detected when pHluorin) fused to GluA1 (SEP-GluA1) (Ashby et al., 2004; Lin &
surface GluA1 antibody was stripped before recycling, or only sur- Huganir, 2007) (Fig. 6C–E). Surface expression of SEP-GluA1 was
face signal in non-permeabilized neurons indicating no unspecific rapidly increased on soma and dendrites in control neurons upon
staining. Further, there was no change in GluA1-AMPARs recycling glycine perfusion, before reaching a plateau. However,
with a scrambled shRNA, suggesting no unspecific effect. somato-dendritic SEP-GluA1 recycling rate and surface number
We next examined the effect of KIBRA overexpression on were further increased in response to glycine in neurons
activity-dependent GluA1 recycling in cultured hippocampal overexpressing KIBRA when compared to control neurons
neurons. Chemical LTP (cLTP) was induced by perfusion of glycine (Fig. 6D and E). These results demonstrate that KIBRA is involved
for 3 min and GluA1 surface expression was monitored by live in activity-dependent GluA1-AMPAR surface expression, and are

Please cite this article in press as: Heitz, F. D., et al. The memory gene KIBRA is a bidirectional regulator of synaptic and structural plasticity in the adult
brain. Neurobiology of Learning and Memory (2016), http://dx.doi.org/10.1016/j.nlm.2016.07.028
12 F.D. Heitz et al. / Neurobiology of Learning and Memory xxx (2016) xxx–xxx

consistent with the effect of KIBRA overexpression and knockdown pression significantly increased the dendritic volume, the number
observed on LTP. of branch points, and the number of tertiary dendrites, although
it had no effect on primary dendritic length. These effects were
not only observed in cultured neurons in vitro, but were also pre-
3.7. KIBRA overexpression promotes neuronal growth in vitro and in sent in the brain of adult transgenic mice overexpressing KIBRA.
the adult mouse hippocampus in vivo In these mice, dendritic volume, mean number of branch points,
and mean number of secondary dendrites were significantly
GluA1-AMPAR recycling has been implicated in neuronal increased in the hippocampus only a few days after induction of
growth and dendritic branching complexity (Haas, Li, & Cline, KIBRA transgene by doxycycline (Fig. 7B and C). These results
2006; Prithviraj et al., 2008; Zhou et al., 2008). Therefore, we strongly suggest a critical role for KIBRA in dendritic organization
examined whether KIBRA influences these processes. Primary hip- in the adult hippocampus.
pocampal neurons in which KIBRA was knocked-down or overex-
pressed were stained with an antibody directed against the
dendritic marker protein MAP2 and the morphology of dendrites 4. Discussion
was analyzed. KIBRA knockdown led to a significant decrease in
the total dendritic volume, in the mean number of branch points, Here, we provide novel evidence that KIBRA, a protein associ-
and in the mean number of tertiary dendrites, as well as in the ated with memory performance in human (Papassotiropoulos
length of primary dendrites (Fig. 7A). In contrast, KIBRA overex- et al., 2006; Schneider et al., 2010; Wersching et al., 2011) is a crit-

Fig. 7. Dendritic growth and arborization are regulated by KIBRA. (A) Representative images with reconstructions of the dendritic arborization and morphometric analyses of
cultured hippocampal neurons infected with a scrambled shRNA lentivirus (Control) or with a KIBRA shRNA lentivirus (KD), and of neurons from transgenic mice
overexpressing KIBRA (OE). To reveal neuronal morphology, neurons were fixed and stained with an anti-MAP2 primary antibody and a Cy2-conjugated secondary antibody
5 days after lentiviral infection, and 48 h after the induction of KIBRA expression with dox in OE neurons. The following parameters were analyzed: total dendritic volume
(R dendrite volume), number of branch points, number of primary (1°), secondary (2°) and tertiary (3°) dendrites, primary dendrite length (1° dendrite length). While control
neurons have a complex dendritic arborization, neurons with KIBRA KD show a decrease in dendritic volume (KD = 1996.7 ± 55.5 lm3 (n = 9) versus con-
trol = 2901.5 ± 239.1 lm3 (n = 15), t(22) = 2.88, ⁄⁄p < 0.01), in the mean number of branch points (KD = 22.1 ± 1.8 (n = 10) versus control = 39.7 ± 2.6 (n = 23), t(31) = 4.14,
⁄⁄⁄
p < 0.001), in the mean number of tertiary dendrites (KD = 1.2 ± 0.3 (n = 9) versus control = 5.5 ± 0.8 (n = 15), t(22) = 3.57, ⁄⁄p < 0.01), and in primary dendrite length
(KD = 131.4 ± 2.4 lm (n = 6) versus control = 175.3 ± 4.9 lm (n = 15), t(19) = 5.44, ⁄⁄⁄p < 0.001). In contrast, neurons OE KIBRA have a more complex arborization with an
increased dendritic volume (OE = 3677.1 ± 242.2 lm3 (n = 9) versus control = 2901.5 ± 239.1 lm3 (n = 15), t(22) = 2.14, ⁄p < 0.05), more branch points (OE = 50.4 ± 1.6 (n = 11)
versus control = 39.7 ± 2.6 (n = 23), t(32) = 2.65, ⁄p < 0.05), and more tertiary dendrites (OE = 9.1 ± 1.1 (n = 9) versus control = 5.5 ± 0.8 (n = 15), t(22) = 2.51, ⁄p < 0.05). KIBRA
KD or OE did not significantly affect the mean number of primary (KD = 4.4 ± 0.2 (n = 9) or OE = 4.6 ± 0.5 (n = 9) versus control = 3.1 ± 0.5 (n = 15), t(22) = 1.93 or t(22) = 1.90,
n.s.) or secondary dendrites (KD = 9.6 ± 0.9 (n = 9) or OE = 16.8 ± 2.8 (n = 9) versus control = 11.6 ± 2.2 (n = 15), t(22) = 0.68 or t(22) = 1.44, n.s.). Scale bar: 60 lm. (B) Golgi
staining and morphometric analyses of CA1 hippocampal neurons in adult control and KIBRA OE mice treated with dox. Top images show whole neurons in control and KIBRA
OE mice. Neurons overexpressing KIBRA have increased dendrite volume (OE = 280.3 ± 59.0% (n = 4) versus control = 100.0 ± 3.8% (n = 3), t(5) = 2.58, ⁄p < 0.05), more branch
points (OE = 48.8 ± 2.5 (n = 4) versus control = 35.7 ± 4.9 (n = 3), t(5) = 2.59, ⁄p < 0.05), and more secondary dendrites (OE = 30.5 ± 3.0 (n = 4) versus control = 18.7 ± 2.7 (n = 3),
t(5) = 2.81, ⁄p < 0.05). KIBRA OE did not significantly affect the number of primary dendrites (OE = 4.3 ± 0.3 (n = 4) versus control = 4.2 ± 0.6 (n = 3), t(5) = 0.10, n.s.). Scale bar:
30 lm. Morphometric analyses were performed with Imaris software. Data are mean ± SEM. (C) Images of yellow fluorescent protein (YFP)-containing neurons (green) from
hippocampus CA1 area in Thy1-YFP and KIBRA double transgenic mice (OE) compared to Thy1-YFP transgenic mice (control). Scale bar: 30 lm. (For interpretation of the
references to colour in this figure legend, the reader is referred to the web version of this article.)

Please cite this article in press as: Heitz, F. D., et al. The memory gene KIBRA is a bidirectional regulator of synaptic and structural plasticity in the adult
brain. Neurobiology of Learning and Memory (2016), http://dx.doi.org/10.1016/j.nlm.2016.07.028
F.D. Heitz et al. / Neurobiology of Learning and Memory xxx (2016) xxx–xxx 13

ical regulator of synaptic plasticity and memory that controls GluA1 knockout mice (Zamanillo et al., 1999; Reisel et al., 2002)
AMPARs. We show that the selective overexpression of KIBRA in that may be caused by compensatory homeostatic mechanisms.
forebrain neurons in adult mice does not affect learning but Since LTD impairment is associated with spatial memory deficit
impairs spatial long-term memory. We show further that KIBRA in both GluA1 phosphomutant and KIBRA overexpressing mice,
overexpression increases hippocampal LTP, in particular a GluA1- these data also confirm the essential role of hippocampal CA1
dependent form of LTP, but prevents LTD in area CA1, and results LTD in long-term spatial memory (Ge PNAS 2010).
in a shift of synaptic plasticity in favor of potentiation. KIBRA The exact mechanisms by which KIBRA influences AMPAR
knockdown in contrast, abolishes LTP. These effects are associated trafficking are not known but likely involve some of its known
with a modulation of the constitutive recycling of GluA1-AMPARs neuronal binding partners. This may include partners implicated
that is increased by KIBRA overexpression and decreased by KIBRA in AMPAR regulation such as dynein light chain 1, a cytoplasmic
knockdown, and with an activity-dependent recruitment of GluA1 protein required for the transport of AMPARs to dendrites
at the surface. They also correlate with an increase and decrease in (Kapitein et al., 2010; Rayala et al., 2006), the exocyst complex
dendritic growth and arborization respectively, both in vitro and proteins Sec3/5/8 involved in synaptic insertion of AMPARs
in vivo. Overall, the present data establish KIBRA as a novel regula- (Gerges, Backos, Rupasinghe, Spaller, & Esteban, 2006; Rosse
tor of GluA1 essential for bidirectional synaptic plasticity in the et al., 2009), or the early endosome-to-endocytic recycling
hippocampus and for long-term memory. compartment protein sorting nexin 4 (SNX4) involved in protein
We show that KIBRA co-localizes with AMPARs in hippocampal recycling (Traer et al., 2007). It should be noted that, since KIBRA
neurons both in vitro and in vivo, and that both proteins co- also co-localizes with GluA2 to some extent, and a large number
immunoprecipitate. In dendrites, KIBRA preferentially colocalizes of AMPARs are heteromeric and contain GluA1 and GluA2 (Shi,
with GluA1 and favors the constitutive and activity-dependent Hayashi, Esteban, & Malinow, 2001), KIBRA can also regulate GluA2
recycling of GluA1-containing AMPARs in hippocampal neurons. as reported in a recent study (Makuch et al., 2011).
In resting conditions, this does not affect basal synaptic transmis- Further to synaptic plasticity, KIBRA overexpression and knock-
sion or the activity of AMPARs, since more GluA1 is not expected to down are also shown to alter the morphology and dendritic orga-
be recruited and stabilized at synapses. However upon neuronal nization of hippocampal neurons. They oppositely modulate the
activity, it leads to an increase in GluA1 surface receptors that length, number and volume of dendrites, and the number of branch
likely leads to the enhanced LTP and impaired LTD observed after points. Such bidirectional effect of KIBRA is striking and points to a
KIBRA overexpression. This effect may be mediated by an increase remarkable property of KIBRA to actively contribute to dendritic
in recycling and receptor delivery at perisynaptic sites followed by remodeling, in particular in the adult brain. This suggests that
lateral diffusion to synapses. More surface GluA1 due to KIBRA KIBRA is likely an important player in the structural dynamics of
would lead to synaptic strengthening and favor synaptic potentia- neuronal networks in adulthood, and may be implicated in
tion over depression, which likely induces LTP saturation and pre- structural changes induced by activity-dependent experiences or
vent the induction of LTD. This is consistent with the demonstrated environmental enrichment, but also after stress and neurodegener-
role of AMPAR recycling and activity-dependent surface expression ation (Holtmaat & Svoboda, 2009; Tavosanis, 2012). Notably, the
in LTP in CA1 pyramidal neurons (Park et al., 2004; Petrini et al., extent of the effect of KIBRA overexpression and knockdown on
2009; Yang, Wang, Frerking, & Zhou, 2008), that specifically impli- different parameters of neuronal morphology varies. For instance,
cates calcium-permeable GluA1-containing AMPARs (Makino & the number of tertiary dendrites is more affected than the number
Malinow, 2009; Plant et al., 2006). It is also consistent with the of primary dendrites maybe because tertiary dendrites have a dif-
impairing effect of GluA1 knockout on CA1 hippocampal LTP ferent requirement for KIBRA and their growth and establishment
(Mack et al., 2001; Zamanillo et al., 1999), but the lack of effect may be easier to modulate than primary dendrites. Finally, the
of GluA2 or GluA3 knockout (Meng et al., 2003). In addition, LTP effect of KIBRA on neuronal morphology is likely linked to GluA1,
saturation identified after 3-train HFS and repeated 1-train HFS since GluA1 has been demonstrated to be implicated in dendrite
in transgenic mice could be indicative of deficits in synaptic scaling growth (Haas et al., 2006; Prithviraj et al., 2008; Zhou et al., 2008).
reflecting a lack of homeostatic compensation (Vitureira & Goda, Finally, the present data are clinically relevant since KIBRA has
2013). In this view, KIBRA overexpression may increase synaptic been associated with memory performance in human, and a SNP
density of GluA1-containing AMPA receptors at activated synapses (rs17070145) has been linked to an increased risk for late-
upon LTP induction but may also lead to recruitment and stabiliza- onset Alzheimer’s disease (Rodriguez-Rodriguez et al., 2009;
tion of GluA1-containing AMPA receptors at a larger number of Corneveaux et al., 2010). This link may also be related to the obser-
synapses including synapses whose strengths are normally weak- vation that both AMPAR trafficking and LTP are impaired in Alzhei-
ened by homeostatic mechanisms. mer’s patients (Hsieh et al., 2006; Rui, Gu, Yu, Hartzell, & Zheng,
We show that KIBRA overexpression in transgenic mice impairs 2010). It is also supported by our observation that KIBRA plays a
spatial long-term memory on the hidden platform Morris water critical role in dendritic arborization and neuronal morphology,
maze task, but does not affect spatial learning. In KIBRA transgenic and may therefore contribute to altered neuronal structures and
mice, spatial memory deficits are associated with enhanced integrity in neurodegeneration, Alzheimer’s disease and other cog-
hippocampal CA1 LTP and impaired LTD, together with increased nitive pathologies. The present data may ultimately provide a
surface GluA1 receptors in CA1 pyramidal neurons. This is novel therapeutic target for the development of potential treat-
consistent with spatial memory deficits in mice with mutations ment of disorders associated with impaired plasticity and cognitive
in GluA1 phosphorylation sites critical for LTP and LTD expression defects.
(Lee et al., 2003). In particular, GluA1 phosphomutant mice essen-
tially show a lack of LTD and a reduced LTP in the CA1 region of the
hippocampus associated with a deficit in spatial memory but not Author contributions
spatial learning on the Morris water maze task. These findings
together with our results establish that activity-dependent regula- F.D.H. and I.M.M. conceived the project. F.D.H. designed and
tion of GluA1-containing AMPA receptor function at synapses is conducted the experiments, and prepared the manuscript with I.
critical for spatial memory. However, these findings contrast with M.M. All authors technically contributed to the results. All the
normal performance on the spatial memory water maze task in authors approved the final version of the manuscript.

Please cite this article in press as: Heitz, F. D., et al. The memory gene KIBRA is a bidirectional regulator of synaptic and structural plasticity in the adult
brain. Neurobiology of Learning and Memory (2016), http://dx.doi.org/10.1016/j.nlm.2016.07.028
14 F.D. Heitz et al. / Neurobiology of Learning and Memory xxx (2016) xxx–xxx

Acknowledgements Hayashi, Y., Shi, S. H., Esteban, J. A., Piccini, A., Poncer, J. C., & Malinow, R. (2000).
Driving AMPA receptors into synapses by LTP and CaMKII: Requirement for
GluR1 and PDZ domain interaction. Science, 287, 2262–2267.
We are grateful to Joachim Kremerskothen for generously pro- Holtmaat, A., & Svoboda, K. (2009). Experience-dependent structural synaptic
viding KIBRA vectors and antibody, for support with the IPs and for plasticity in the mammalian brain. Nature Reviews Neuroscience, 10, 647–658.
Hsieh, H., Boehm, J., Sato, C., Iwatsubo, T., Tomita, T., Sisodia, S., & Malinow, R.
reading an initial version of the manuscript, Aniña Pescatore,
(2006). AMPAR removal underlies Abeta-induced synaptic depression and
Osvaldo Mirante, Rreze Gecaj-Mehmeti and Nina Meyer for techni- dendritic spine loss. Neuron, 52, 831–843.
cal help, Dr. Holger Russig for advice on behavioral testing, Dr. Cris- Johannsen, S., Duning, K., Pavenstadt, H., Kremerskothen, J., & Boeckers, T. M.
(2008). Temporal-spatial expression and novel biochemical properties of the
tina Fernandez for conceptual contribution, Philipp Schätzle, Stella
memory-related protein KIBRA. Neuroscience, 155, 1165–1173.
Kramer and Giovanna Bosshard for providing mouse and rat neu- Kaech, S., & Banker, G. (2006). Culturing hippocampal neurons. Nature Protocols, 1,
ronal cultures We acknowledge staff members of the Center for 2406–2415.
Microscopy and Image Analysis of the University of Zürich (ZMB). Kapitein, L. C., Schlager, M. A., Kuijpers, M., Wulf, P. S., van Spronsen, M.,
MacKintosh, F. C., & Hoogenraad, C. C. (2010). Mixed microtubules steer
This work was supported by the University of Zürich, the Swiss dynein-driven cargo transport into dendrites. Current Biology, 20, 290–299.
Federal Institute of Technology, the Human Frontier Science Pro- Kemp, A., & Manahan-Vaughan, D. (2007). Hippocampal long-term depression:
gram, the Swiss National Foundation, The National Center for Com- Master or minion in declarative memory processes? Trends in Neurosciences, 30,
111–118.
petence in Research ‘‘Neural Plasticity and Repair”, the Slack-Gyr Kemp, A., & Manahan-Vaughan, D. (2008). The hippocampal CA1 region and dentate
and Bitterlin foundations, and a DFG grant PA 483/14-2 to H.P. gyrus differentiate between environmental and spatial feature encoding
The authors declare no conflict of interest. through long-term depression. Cerebral Cortex, 18, 968–977.
Kessels, H. W., & Malinow, R. (2009). Synaptic AMPA receptor plasticity and
behavior. Neuron, 61, 340–350.
Koshibu, K., Graff, J., Beullens, M., Heitz, F. D., Berchtold, D., Russig, H., ... Mansuy, I.
Appendix A. Supplementary material M. (2009). Protein phosphatase 1 regulates the histone code for long-term
memory. Journal of Neuroscience, 29, 13079–13089.
Kremerskothen, J., Plaas, C., Buther, K., Finger, I., Veltel, S., Matanis, T., ... Barnekow,
Supplementary data associated with this article can be found, in A. (2003). Characterization of KIBRA, a novel WW domain-containing protein.
the online version, at http://dx.doi.org/10.1016/j.nlm.2016.07.028. Biochemical and Biophysical Research Communications, 300, 862–867.
Lee, H. K., Takamiya, K., Han, J. S., Man, H., Kim, C. H., Rumbaugh, G., ... Huganir, R. L.
(2003). Phosphorylation of the AMPA receptor GluR1 subunit is required for
synaptic plasticity and retention of spatial memory. Cell, 112, 631–643.
References Lin, D. T., & Huganir, R. L. (2007). PICK1 and phosphorylation of the glutamate
receptor 2 (GluR2) AMPA receptor subunit regulates GluR2 recycling after NMDA
receptor-induced internalization. Journal of Neuroscience, 27, 13903–13908.
Almeida, O. P., Schwab, S. G., Lautenschlager, N. T., Morar, B., Greenop, K. R., Flicker,
Lu, W., Shi, Y., Jackson, A. C., Bjorgan, K., During, M. J., Sprengel, R., ... Nicoll, R. A.
L., & Wildenauer, D. (2008). KIBRA genetic polymorphism influences episodic
(2009). Subunit composition of synaptic AMPA receptors revealed by a single-
memory in later life, but does not increase the risk of mild cognitive
cell genetic approach. Neuron, 62, 254–268.
impairment. Journal of Cellular and Molecular Medicine, 12, 1672–1676.
Mack, V., Burnashev, N., Kaiser, K. M., Rozov, A., Jensen, V., Hvalby, O., ... Sprengel, R.
Ashby, M. C., De La Rue, S. A., Ralph, G. S., Uney, J., Collingridge, G. L., & Henley, J. M.
(2001). Conditional restoration of hippocampal synaptic potentiation in Glur-A-
(2004). Removal of AMPA receptors (AMPARs) from synapses is preceded by
deficient mice. Science, 292, 2501–2504.
transient endocytosis of extrasynaptic AMPARs. Journal of Neuroscience, 24,
Makino, H., & Malinow, R. (2009). AMPA receptor incorporation into synapses
5172–5176.
during LTP: The role of lateral movement and exocytosis. Neuron, 64, 381–390.
Bates, T. C., Price, J. F., Harris, S. E., Marioni, R. E., Fowkes, F. G., Stewart, M. C., ...
Makuch, L., Volk, L., Anggono, V., Johnson, R. C., Yu, Y., Duning, K., ... Huganir, R. L.
Deary, I. J. (2009). Association of KIBRA and memory. Neuroscience Letters, 458,
(2011). Regulation of AMPA receptor function by the human memory-
140–143.
associated gene KIBRA. Neuron, 71, 1022–1029.
Bhattacharyya, S., Biou, V., Xu, W., Schluter, O., & Malenka, R. C. (2009). A critical
Malinow, R., & Malenka, R. C. (2002). AMPA receptor trafficking and synaptic
role for PSD-95/AKAP interactions in endocytosis of synaptic AMPA receptors.
plasticity. Annual Review of Neuroscience, 25, 103–126.
Nature Neuroscience, 12, 172–181.
Manahan-Vaughan, D., & Braunewell, K. H. (1999). Novelty acquisition is associated
Bliss, T. V., & Collingridge, G. L. (1993). A synaptic model of memory: Long-term
with induction of hippocampal long-term depression. Proceedings of the
potentiation in the hippocampus. Nature, 361, 31–39.
National Academy of Sciences USA, 96, 8739–8744.
Bredt, D. S., & Nicoll, R. A. (2003). AMPA receptor trafficking at excitatory synapses.
Meng, Y., Zhang, Y., & Jia, Z. (2003). Synaptic transmission and plasticity in the
Neuron, 40, 361–379.
absence of AMPA glutamate receptor GluR2 and GluR3. Neuron, 39, 163–176.
Burgess, J. D., Pedraza, O., Graff-Radford, N. R., Hirpa, M., Zou, F., Miles, R., ... Ertekin-
Michalon, A., Koshibu, K., Baumgartel, K., Spirig, D. H., & Mansuy, I. M. (2005).
Taner, N. (2010). Association of common KIBRA variants with episodic memory
Inducible and neuron-specific gene expression in the adult mouse brain with
and AD risk. Neurobiology of Aging, 32(557), e551–e559.
the rtTA2S-M2 system. Genesis, 43, 205–212.
Buther, K., Plaas, C., Barnekow, A., & Kremerskothen, J. (2004). KIBRA is a novel
Migaud, M., Charlesworth, P., Dempster, M., Webster, L. C., Watabe, A. M.,
substrate for protein kinase Czeta. Biochemical and Biophysical Research
Makhinson, M., ... Grant, S. G. (1998). Enhanced long-term potentiation and
Communications, 317, 703–707.
impaired learning in mice with mutant postsynaptic density-95 protein. Nature,
Collingridge, G. L., Isaac, J. T., & Wang, Y. T. (2004). Receptor trafficking and synaptic
396, 433–439.
plasticity. Nature Reviews Neuroscience, 5, 952–962.
Morris, R. (1984). Developments of a water-maze procedure for studying spatial
Corneveaux, J. J., Liang, W. S., Reiman, E. M., Webster, J. A., Myers, A. J., Zismann, V.
learning in the rat. Journal of Neuroscience Methods, 11, 47–60.
L., ... Huentelman, M. J. (2010). Evidence for an association between KIBRA and
Moser, E. I., Krobert, K. A., Moser, M. B., & Morris, R. G. (1998). Impaired spatial
late-onset Alzheimer’s disease. Neurobiology of Aging, 31, 901–909.
learning after saturation of long-term potentiation. Science, 281, 2038–2042.
Dingledine, R., Borges, K., Bowie, D., & Traynelis, S. F. (1999). The glutamate receptor
Nakao, K., Ikegaya, Y., Yamada, M. K., Nishiyama, N., & Matsuki, N. (2002).
ion channels. Pharmacological Reviews, 51, 7–61.
Hippocampal long-term depression as an index of spatial working memory.
Duning, K., Schurek, E. M., Schluter, M., Bayer, M., Reinhardt, H. C., Schwab, A., ...
European Journal of Neuroscience, 16, 970–974.
Pavenstadt, H. (2008). KIBRA modulates directional migration of podocytes.
Papassotiropoulos, A., Stephan, D. A., Huentelman, M. J., Hoerndli, F. J., Craig, D. W.,
Journal of the American Society of Nephrology, 19, 1891–1903.
Pearson, J. V., ... de Quervain, D. J. (2006). Common Kibra alleles are associated
Frey, M. C., Sprengel, R., & Nevian, T. (2009). Activity pattern-dependent long-term
with human memory performance. Science, 314, 475–478.
potentiation in neocortex and hippocampus of GluA1 (GluR-A) subunit-
Park, M., Penick, E. C., Edwards, J. G., Kauer, J. A., & Ehlers, M. D. (2004). Recycling
deficient mice. Journal of Neuroscience, 29, 5587–5596.
endosomes supply AMPA receptors for LTP. Science, 305, 1972–1975.
Gahwiler, B. H., Capogna, M., Debanne, D., McKinney, R. A., & Thompson, S. M.
Pastalkova, E., Serrano, P., Pinkhasova, D., Wallace, E., Fenton, A. A., & Sacktor, T. C.
(1997). Organotypic slice cultures: A technique has come of age. Trends in
(2006). Storage of spatial information by the maintenance mechanism of LTP.
Neurosciences, 20, 471–477.
Science, 313, 1141–1144.
Gerges, N. Z., Backos, D. S., Rupasinghe, C. N., Spaller, M. R., & Esteban, J. A. (2006).
Petrini, E. M., Lu, J., Cognet, L., Lounis, B., Ehlers, M. D., & Choquet, D. (2009).
Dual role of the exocyst in AMPA receptor targeting and insertion into the
Endocytic trafficking and recycling maintain a pool of mobile surface AMPA
postsynaptic membrane. EMBO Journal, 25, 1623–1634.
receptors required for synaptic potentiation. Neuron, 63, 92–105.
Groc, L., & Choquet, D. (2006). AMPA and NMDA glutamate receptor trafficking:
Plant, K., Pelkey, K. A., Bortolotto, Z. A., Morita, D., Terashima, A., McBain, C. J., ...
Multiple roads for reaching and leaving the synapse. Cell and Tissue Research,
Isaac, J. T. (2006). Transient incorporation of native GluR2-lacking AMPA
326, 423–438.
receptors during hippocampal long-term potentiation. Nature Neuroscience, 9,
Haas, K., Li, J., & Cline, H. T. (2006). AMPA receptors regulate experience-dependent
602–604.
dendritic arbor growth in vivo. Proceedings of the National Academy of Sciences
Preuschhof, C., Heekeren, H. R., Li, S. C., Sander, T., Lindenberger, U., & Backman, L.
USA, 103, 12127–12131.
(2009). KIBRA and CLSTN2 polymorphisms exert interactive effects on human
Hanley, J. G. (2008). AMPA receptor trafficking pathways and links to dendritic
episodic memory. Neuropsychologia, 48, 402–408.
spine morphogenesis. Cell Adhesion & Migration, 2, 276–282.

Please cite this article in press as: Heitz, F. D., et al. The memory gene KIBRA is a bidirectional regulator of synaptic and structural plasticity in the adult
brain. Neurobiology of Learning and Memory (2016), http://dx.doi.org/10.1016/j.nlm.2016.07.028
F.D. Heitz et al. / Neurobiology of Learning and Memory xxx (2016) xxx–xxx 15

Prithviraj, R., Kelly, K. M., Espinoza-Lewis, R., Hexom, T., Clark, A. B., & Inglis, F. M. Takahashi, T., Svoboda, K., & Malinow, R. (2003). Experience strengthening
(2008). Differential regulation of dendrite complexity by AMPA receptor transmission by driving AMPA receptors into synapses. Science, 299,
subunits GluR1 and GluR2 in motor neurons. Developmental Neurobiology, 68, 1585–1588.
247–264. Tavosanis, G. (2012). Dendritic structural plasticity. Developmental Neurobiology, 72,
Rayala, S. K., den Hollander, P., Manavathi, B., Talukder, A. H., Song, C., Peng, S., ... 73–86.
Kumar, R. (2006). Essential role of KIBRA in co-activator function of dynein light Traer, C. J., Rutherford, A. C., Palmer, K. J., Wassmer, T., Oakley, J., Attar, N., ... Cullen,
chain 1 in mammalian cells. Journal of Biological Chemistry, 281, 19092–19099. P. J. (2007). SNX4 coordinates endosomal sorting of TfnR with dynein-mediated
Reisel, D., Bannerman, D. M., Schmitt, W. B., Deacon, R. M., Flint, J., Borchardt, T., ... transport into the endocytic recycling compartment. Nature Cell Biology, 9,
Rawlins, J. N. (2002). Spatial memory dissociations in mice lacking GluR1. 1370–1380.
Nature Neuroscience, 5, 868–873. Vitureira, N., & Goda, Y. (2013). Cell biology in neuroscience: The interplay between
Rodriguez-Rodriguez, E., Infante, J., Llorca, J., Mateo, I., Sanchez-Quintana, C., Garcia- Hebbian and homeostatic synaptic plasticity. Journal of Cell Biology, 203,
Gorostiaga, I., ... Combarros, O. (2009). Age-dependent association of KIBRA 175–186.
genetic variation and Alzheimer’s disease risk. Neurobiology of Aging, 30, Vorhees, C. V., & Williams, M. T. (2006). Morris water maze: Procedures for
322–324. assessing spatial and related forms of learning and memory. Nature Protocols, 1,
Rosse, C., Formstecher, E., Boeckeler, K., Zhao, Y., Kremerskothen, J., White, M. D., ... 848–858.
Parker, P. J. (2009). An aPKC-exocyst complex controls paxillin phosphorylation Wenthold, R. J., Petralia, R. S., Blahos, J., II, & Niedzielski, A. S. (1996). Evidence for
and migration through localised JNK1 activation. PLoS Biology, 7, e1000235. multiple AMPA receptor complexes in hippocampal CA1/CA2 neurons. Journal of
Rui, Y., Gu, J., Yu, K., Hartzell, H. C., & Zheng, J. Q. (2010). Inhibition of AMPA receptor Neuroscience, 16, 1982–1989.
trafficking at hippocampal synapses by beta-amyloid oligomers: The Wersching, H., Guske, K., Hasenkamp, S., Hagedorn, C., Schiwek, S., Jansen, S., ...
mitochondrial contribution. Molecular Brain, 3, 10. Floel, A. (2011). Impact of common KIBRA allele on human cognitive functions.
Schaper, K., Kolsch, H., Popp, J., Wagner, M., & Jessen, F. (2008). KIBRA gene variants Neuropsychopharmacology, 36, 1296–1304.
are associated with episodic memory in healthy elderly. Neurobiology of Aging, Willshaw, D. J., & Buckingham, J. T. (1990). An assessment of Marr’s theory of the
29, 1123–1125. hippocampus as a temporary memory store. Philosophical Transactions of the
Schneider, A., Huentelman, M. J., Kremerskothen, J., Duning, K., Spoelgen, R., & Royal Society of London. Series B, Biological sciences, 329, 205–215.
Nikolich, K. (2010). KIBRA: A new gateway to learning and memory? Frontiers in Yang, Y., Wang, X. B., Frerking, M., & Zhou, Q. (2008). Delivery of AMPA receptors to
Aging Neuroscience, 2, 4. perisynaptic sites precedes the full expression of long-term potentiation.
Schwab, L. C., Luo, V., Clarke, C. L., & Nathan, P. J. (2014). Effects of the KIBRA single Proceedings of the National Academy of Sciences USA, 105, 11388–11393.
nucleotude polymorphism on synaptic plasticity and memory: A review of the Yao, Y., Kelly, M. T., Sajikumar, S., Serrano, P., Tian, D., Bergold, P. J., ... Sacktor, T. C.
literature. Current Neuropharmacology, 12, 281–288. (2008). PKM zeta maintains late long-term potentiation by N-ethylmaleimide-
Shema, R., Sacktor, T. C., & Dudai, Y. (2007). Rapid erasure of long-term memory sensitive factor/GluR2-dependent trafficking of postsynaptic AMPA receptors.
associations in the cortex by an inhibitor of PKM zeta. Science, 317, 951–953. Journal of Neuroscience, 28, 7820–7827.
Shepherd, J. D., & Huganir, R. L. (2007). The cell biology of synaptic plasticity: AMPA Yoshihama, Y., Hirai, T., Ohtsuka, T., & Chida, K. (2009). KIBRA Co-localizes with
receptor trafficking. Annual Review of Cell and Developmental Biology, 23, protein kinase Mzeta (PKMzeta) in the mouse hippocampus. Bioscience,
613–643. Biotechnology, and Biochemistry, 73, 147–151.
Shi, S., Hayashi, Y., Esteban, J. A., & Malinow, R. (2001). Subunit-specific rules Zamanillo, D., Sprengel, R., Hvalby, O., Jensen, V., Burnashev, N., Rozov, A., ...
governing AMPA receptor trafficking to synapses in hippocampal pyramidal Sakmann, B. (1999). Importance of AMPA receptors for hippocampal synaptic
neurons. Cell, 105, 331–343. plasticity but not for spatial learning. Science, 284, 1805–1811.
Suh, Y. H., Terashima, A., Petralia, R. S., Wenthold, R. J., Isaac, J. T., Roche, K. W., & Zhou, W., Zhang, L., Guoxiang, X., Mojsilovic-Petrovic, J., Takamaya, K., Sattler, R., ...
Roche, P. A. (2010). A neuronal role for SNAP-23 in postsynaptic glutamate Kalb, R. (2008). GluR1 controls dendrite growth through its binding partner,
receptor trafficking. Nature Neuroscience, 13, 338–343. SAP97. Journal of Neuroscience, 28, 10220–10233.

Please cite this article in press as: Heitz, F. D., et al. The memory gene KIBRA is a bidirectional regulator of synaptic and structural plasticity in the adult
brain. Neurobiology of Learning and Memory (2016), http://dx.doi.org/10.1016/j.nlm.2016.07.028

You might also like