Sunteți pe pagina 1din 16

doi:10.

1093/brain/awt303 Brain 2014: 137; 137–152 | 137

BRAIN
A JOURNAL OF NEUROLOGY

Zinc deficiency dysregulates the synaptic


ProSAP/Shank scaffold and might contribute
to autism spectrum disorders
Stefanie Grabrucker,1,2,* Linda Jannetti,1,* Matti Eckert,1 Simone Gaub,2 Resham Chhabra,1
Stefanie Pfaender,3 Katharina Mangus,1 Parameshwar Pasham Reddy,4 Vladan Rankovic,4
Michael J. Schmeisser,3 Michael R. Kreutz,4 Günter Ehret,2 Tobias M. Boeckers3 and
Andreas M. Grabrucker1,3

1 WG Molecular Analysis of Synaptopathies, Neurology Department, Neurocentre of Ulm University, Ulm, Germany
2 Institute of Neurobiology, Ulm University, Ulm, Germany
3 Institute for Anatomy and Cell Biology, Ulm University, Ulm Germany
4 RG Neuroplasticity, Leibniz Institute for Neurobiology, Magdeburg, Germany

*These authors contributed equally to this work.

Correspondence to: Prof. Dr. Andreas M. Grabrucker,


WG Molecular Analysis of Synaptopathies,
Neurology Dept. of Ulm University,
Albert Einstein Allee 11,
89081 Ulm Germany
E-mail: andreas.grabrucker@uni-ulm.de

Proteins of the ProSAP/Shank family act as major organizing scaffolding elements within the postsynaptic density of excitatory
synapses. Deletions, mutations or the downregulation of these molecules has been linked to autism spectrum disorders, the
related Phelan McDermid Syndrome or Alzheimer’s disease. ProSAP/Shank proteins are targeted to synapses depending on
binding to zinc, which is a prerequisite for the assembly of the ProSAP/Shank scaffold. To gain insight into whether the
previously reported assembly of ProSAP/Shank through zinc ions provides a crossing point between genetic forms of autism
spectrum disorder and zinc deficiency as an environmental risk factor for autism spectrum disorder, we examined the interplay
between zinc and ProSAP/Shank in vitro and in vivo using neurobiological approaches. Our data show that low postsynaptic
zinc availability affects the activity dependent increase in ProSAP1/Shank2 and ProSAP2/Shank3 levels at the synapse in vitro
and that a loss of synaptic ProSAP1/Shank2 and ProSAP2/Shank3 occurs in a mouse model for acute and prenatal zinc
deficiency. Zinc-deficient animals displayed abnormalities in behaviour such as over-responsivity and hyperactivity-like behav-
iour (acute zinc deficiency) and autism spectrum disorder-related behaviour such as impairments in vocalization and social
behaviour (prenatal zinc deficiency). Most importantly, a low zinc status seems to be associated with an increased incidence rate
of seizures, hypotonia, and attention and hyperactivity issues in patients with Phelan-McDermid syndrome, which is caused by
haploinsufficiency of ProSAP2/Shank3. We suggest that the molecular underpinning of prenatal zinc deficiency as a risk factor
for autism spectrum disorder may unfold through the deregulation of zinc-binding ProSAP/Shank family members.

Keywords: PSD; ASD; Shank3; synapse; Zn2 +


Abbreviation: MT = metallothionein

Received May 24, 2013. Revised September 2, 2013. Accepted September 8, 2013. Advance Access publication November 25, 2013
ß The Author (2013). Published by Oxford University Press on behalf of the Guarantors of Brain. All rights reserved.
For Permissions, please email: journals.permissions@oup.com
138 | Brain 2014: 137; 137–152 S. Grabrucker et al.

Introduction Materials and methods


The three members of the ProSAP/Shank family of scaffolding
proteins are key molecules for the spatial organization of the post- Materials
synaptic density of excitatory synapses (Boeckers et al., 2002). ZnCl2, CaEDTA and TPEN [N,N,N0 ,N0 -tetrakis(2-pyridylmethyl) ethyl-
Recent studies report mutations in ProSAP1/Shank2, ProSAP2/ enediamine], Zinquin ethyl-ester and Zinpyr1 were purchased from
Shank3 and Shank1 associated with autism (Durand et al., Sigma-Aldrich as were all other chemicals unless indicated otherwise.
2007; Moessner et al., 2007; Gauthier et al., 2009; Berkel Primary antibodies were purchased from Synaptic Systems (Gephyrin,
et al., 2010; Pinto et al., 2010; Leblond et al., 2012; Sato Homer1, Homer1b/c, GluA1, GluA2, GluA3), Stressgen (bassoon),
Abcam (PSD95) Novus Biological [GKAP/SAPAP, Shank1 (immuno-
et al., 2012). Moreover, the loss of one copy of ProSAP2/
fluorescence, immunohistochemistry), mGluR5], Sigma [b-actin,
Shank3 in humans is correlated with Phelan-McDermid syndrome
GluN1, Shank1 (western blotting)], Santa Cruz (MT-3), Chemicon
(22q13 deletion syndrome) (Bonaglia et al., 2001; Phelan et al., (MAP2, mGluR5) and Millipore (GluN2A, GluN2B). ProSAP1/Shank2
2001; Wilson et al., 2003; Manning et al., 2004). This syndrome is and ProSAP2/Shank3 antibodies have been described previously
characterized by moderate to profound mental retardation, neo- (Schmeisser et al., 2012). Secondary Alexa FluorÕ conjugated antibo-
natal hypotonia, global developmental delay, absent or severely dies were purchased from Invitrogen.
impaired speech (Manning et al., 2004), occurrence of seizures
and ‘autistic-like’ behaviour. Besides genetic factors, it has been
suggested previously that a dysregulation of metal-ion homeosta-
Expression constructs and transfection
sis might contribute to the pathogenesis of autism spectrum The pEGFP (C1-3) vector system (Clontech) was used for MT-3 ex-
disorders (Curtis and Patel, 2008). Intriguingly, the incidence pression constructs and the pLVX vector system (Clontech) for MT-3
rate of Zn2 + -deficiency is as high as 50% for autistic patients knockdown. Short hairpin RNA oligonucleotides were purchased from
MWG-Eurofins. Two sequences were used for MT-3 knockdown:
between 0 and 3 years of age compared with 51% in a healthy
AAGGGCTGCAAATGCACGA and CCTGCCCCTGTCCTACTGG.
control group (Yasuda et al., 2011).
Hippocampal cells were transfected at Day 10 and fixed on Day 14
Recent reports demonstrate a tight interplay between in vitro using OptiFectTM (Invitrogen).
Zn2 + -binding of ProSAP1/Shank2 and ProSAP2/Shank3, but not
Shank1 (Baron et al., 2006; Gundelfinger et al., 2006; Grabrucker
et al., 2011a) and their synaptic association and function Immunohistochemistry
(Grabrucker et al., 2011a). Zn2 + is enriched in cortical, striatal Cryosections were thawed for 20 min and fixed in paraformaldehyde
and hippocampal regions of mammals and selectively stored in, for 20–30 min. After fixation, sections were washed 3  10 min with
and co-released with glutamate from presynaptic vesicles (Lin PBS. Sections were permeabilized with 0.2% Triton in PBS for 2 h and
et al., 2001; Wei et al., 2004). It has been argued that Zn2 + washed again for 3  10 min with PBS containing 0.05% Triton.
released from presynaptic vesicles might influx into the postsynap- Sections were then placed in 10% foetal calf serum in PBS for 2 h.
tic compartment through NMDA and AMPA receptors, and After blocking, sections were incubated with the primary antibody at
37 C for 2 h before 3  10 min washing with PBS containing 0.05%
Ca2 + -channels (Frederickson et al., 2005) and thereby provide a
Triton. Sections were incubated with the secondary antibody at 37 C
pool of free chelatable Zn2 + . The postsynaptic density has an
for 1.5 h. After washing 3  15 min with PBS containing 0.05% Triton,
unusually high Zn2 + -content (Jan et al., 2002) and postsynaptic sections were washed for 2  5 min with PBS containing DAPI, rinsed
Zn2 + is probably bound to metallothioneins (MTs) as well as to with distilled H2O and mounted with VectaMountTM (Vector
structural proteins such as ProSAP1/Shank2 and ProSAP2/Shank3 Laboratories).
(Lee et al., 2003; Grabrucker et al., 2011a). Although indirect Timm’s staining was performed as described by Jaarsma and Korf
evidence for an activity-dependent increase of postsynaptic (1990) (Supplementary material). Zinc staining was performed using
Zn2 + -levels was reported (Bitanihirwe and Cunningham, 2009), 10 mM Zinpyr1 for 1 h at room temperature.
it is currently unknown whether the postsynaptic Zn2 + -pool is
dynamic and whether free Zn2 + can contribute to the folding Hippocampal culture, stainings and
and synaptic association of ProSAP1/Shank2 and ProSAP2/
Shank3. We therefore studied whether the postsynaptic density
treatments
scaffold at the synapse is regulated by Zn2 + -levels, and whether Hippocampal cultures were prepared from rat (embryonic Day 18) as
Zn2 + -deficiency is accompanied by a dysregulation of the ProSAP/ described previously (Grabrucker et al., 2009). For fluorescent
Shank postsynaptic density scaffold in vitro and in vivo. Zn2 + -staining, growth medium was discarded and cells were washed
Moreover, we addressed the possibility that this mechanism 3  with Hank’s Balanced Salt Solution. Coverslips were incubated
with a solution of 25 mM Zinquin ethyl ester or 5 mM Zinpyr1 in
might constitute a plausible interplay between an environmental
Hank’s Balanced Salt Solution for 40 min at 37 C (27). Immunofluor-
risk factor and a known molecular pathway associated with autism
escence was performed as described previously with minor modifi-
spectrum disorder. To that end, we used acute and prenatal
cations (Grabrucker et al., 2011a) (Supplementary material).
Zn2 + -deficient animals and performed behavioural analyses testing Growth media were supplemented with Zn2 + -chelators (TPEN or
for autism-related phenotypes. Finally, we investigated, whether CaEDTA) or ZnCl2 (10 mM). CNQX (10 mM) and AP5 (50 mM) were
Zn2 + -deficiency augments specific symptoms observed in patients used to block synaptic activity. For stimulations, 10 mM ZnCl2, 50 mM
with Phelan-McDermid syndrome. KCl (HiK + ) or 30 mM glutamate was used. Generation of nitric oxide
Zn deficiency dysregulates ProSAP/Shank Brain 2014: 137; 137–152 | 139

(NO) was manipulated using the NO agonist spermine nonoate (1 mM) plastic cages were rinsed with HCl and deionized water. After 5 weeks,
and antagonist N-nitro-L-arginine (100 mM). females of the control and Zn2 + -deficient group were mated. For
behavioural tests, acute Zn2 + -deficient animals were tested as well
as their offspring (prenatal Zn2 + -deficient) at the same age and
Quantitative real-time polymerase compared with control animals. A detailed description of the behav-
chain reaction ioural tests performed can be found in the Supplementary material.
All animal experiments were performed in compliance with the
Isolation of total RNA from three control and three Zn2 + -deficient
guidelines for the welfare of experimental animals issued by the
mice as well as 10 pups from control and Zn2 + -deficient mice each
Federal Government of Germany and the local ethics committee
was performed using the RNeasyÕ kit (Qiagen). First strand synthesis
(Ulm University) ID Number: 0.103.
and quantitative real-time-PCR amplification were carried out in a
one-step, single-tube format using the QuantiFastTM SYBRÕ Green
RT-PCR kit (Qiagen). Thermal cycling and fluorescent detection were Human subjects
performed using the Rotor-GeneÕ Q real-time PCR machine (model 2-
Blood samples from 37 participants diagnosed with Phelan-McDermid
Plex HRM) (Qiagen). The SYBRÕ Green I reporter dye signal was
syndrome were obtained from Nelson Biological Laboratories at
measured against the internal passive reference dye (ROX) to normal-
Rutgers University and atomic absorption spectroscopy performed at
ize non-PCR-related fluctuations (Supplementary material). Resulting
the ‘ZE klinische Chemie’ of Ulm University Hospital to quantify Cu2 +
data were analysed using the hydroxymethylbilane synthase gene as
and Zn2 + levels. Blood samples were mostly from fasting blood dona-
an internal standard to normalize transcript levels. Cycle threshold (ct)
tions and participants were evaluated regarding their status on dietary
values were calculated by the Rotor-GeneÕ Q Software (version
zinc or vitamin supplementations. All experiments were performed in
2.0.2). All quantitative real-time PCR reactions were run in technical
compliance with the local ethics committee (Ulm University, ID: 282/
triplicates and mean ct-values for each reaction were taken into
12) and informed consent was obtained from all subjects. Clinical and
account for calculations.
developmental data of the participants were obtained through the
Phelan-McDermid syndrome registry. A total of 26 parameters were
Protein biochemistry evaluated based on the categorization of participants into three groups
(no, mild and severe Zn2 + -deficiency), including age, gender, diagno-
Subcellular fractions from mouse brain were isolated as described
sis, birth weight, number of ear infections, possible signs of primary
previously with minor modifications (Schmeisser et al., 2012)
immunodeficiency, occurrence of: hypotonia, seizures, nose and throat
(Supplementary material). Proteins were separated by SDS-PAGE and
problems, scoliosis, dysplastic finger/toenails, eczema, gastrointestinal
blotted onto nitrocellulose membranes. Immunoreactivity was visua-
problems, allergy-related problems, endocrine conditions, abnormal
lized using horseradish peroxidase-conjugated secondary antibodies
involuntary movements, sensitivity problems, other neurological con-
and the SuperSignalÕ detection system (Pierce).
ditions, abnormal MRI scans, abnormal CAT scans, feeding disorder,
anxiety disorder, diagnosed language disorder, and attention deficit
Intrinsic fluorescence spectroscopy disorder/attention deficit hyperactivity disorder or other attention or
hyperactivity issues. The overall occurrence of a parameter was calcu-
Tryptophan fluorescence emission spectra were recorded for the lated and the fraction of each Zn2 + status-group showing the param-
ProSAP2/Shank3-SAM domain (rat ProSAP2/Shank3 aa1464–1815, eter identified. Zn2 + and Cu2 + levels were compared with (whole
Genscript) on a Hitachi F7000 spectrofluorimeter. The quartz cuvette blood) reference values for age matched healthy controls (age 0–1:
was cleaned with chromic acid and double-distilled H2O. EDTA 41–58 mmol/l; age 1–4: 40–50 mmol/l; age 4–6: 49–52 mmol/l; age
(100 mM) solution was used to rinse the cuvette followed by double- 6–14: 51–60 mmol/l; age 14–18; 50–56 mmol/l; age 18 and up:
distilled H2O. One millilitre of protein sample (0.1–0.3 mg/ml) in 66–117 mmol/l). Adult Zn2 + concentrations show no significant correl-
50 mM Tris-HCl pH 7.4, 100 mM KCl was used for measurements. ation with age, although some studies report a decrease in very old
Excitation wavelength of 295 nm was used and spectra were recorded age (Rükgauer et al., 1997).
in the range of 300–450 nm by subtracting time-dependent reduction
of fluorescence emission. All spectra were recorded at room tempera-
ture in corrected spectra mode using an excitation and emission band Statistics
pass of 10 nm and 10 nm, respectively. The response-time was set to
2 s with a scan speed of 240 nm/min. The effects of Mg and Zn Cell culture experiments
standard solutions (Fluka) were studied by titrating them into protein For cell culture experiments, 10 cells of each condition were imaged.
solution after 1–2 min incubation periods. For brain sections, three animals of each group (control,
Zn2 + -deficient) were used and each of the four brain regions (cere-
bellum, cortex, hippocampus, striatum) imaged by at least three optic
Animals fields. Fluorescence images were obtained with an upright Axioscope
Ten-week-old mice were purchased from Janvier and housed in plastic microscope equipped with a Zeiss CCD camera (16 bits; 1280  1024
cages under the standard laboratory conditions (average temperature ppi) using Axiovision software (Zeiss). The signal intensity of all signals
of 22 C, food and water available ad libitum). Lights were automat- within the optic fields was quantified using ImageJ 1.46p. Statistical
ically turned on/off in a 12 h rhythm (lights on at 7 am). After one analysis was performed using Microsoft Excel and tested for signifi-
week of acclimation, mice were divided into two groups, one group cance using t-tests followed by ANOVA.
(12 females) was fed a Zn2 + -deficient diet (4 ppm zinc, TestDiet) with
distilled, demineralized drinking water, whereas the control group (12 Western blot data
females) was fed with standard laboratory food (35 ppm zinc) and tap Evaluation of western blot bands was performed using ImageJ 1.46p.
water. To prevent zinc contamination, feeding jars, water bottles and Three independent experiments were performed and the integrated
140 | Brain 2014: 137; 137–152 S. Grabrucker et al.

density of bands was measured. All bands were normalized to b-actin activity-dependent upregulation of ProSAP1/Shank2 and ProSAP2/
and the ratios averaged and tested for significance. The level of Shank3 to a lesser extent (Supplementary Fig. 1H).
significance was set at 0.05 (*P 5 0.05; **P 5 0.01; ***P 5 0.001). An activity-dependent modification of the postsynaptic density
by a Zn2 + -dependent assembly of ProSAP/Shank proteins requires
Behavioural data a modest Zn2 + binding affinity in a range that does not allow
Normal distribution of data was determined by Shapiro-Wilk test. Two multimerization with cytoplasmic free Zn2 + levels, which are in
sets of data were compared with t-test, if normally distributed, or the low picomolar range (Frederickson, 1989; Outten and
Wilcoxon-Mann-Whitney U-test if not normally distributed. Several O’Halloran, 2001; Krezel and Maret, 2006). Thus, to determine
data sets were compared with one-way ANOVA or ANOVA on the dynamic range in which Zn2 + -dependent conformational
ranks and Chi-square test. Although not all data are normally distrib- changes and oligomerization are apparent, we performed intrinsic
uted, data are sometimes shown in figures as means  SEM to facili- fluorescence spectroscopy and Dynamic light scattering experi-
tate comparisons. Statistical tests were performed using absolute ments with the ProSAP2/Shank3 SAM domain (Fig. 1E and F
values, despite some diagrams displaying percentages for better and Supplementary Fig. 1I–L). Taken together, we found that
comparison. All tests were run by SPSS (version 19) and SigmaPlot
the Zn2 + -dependent multimerization of ProSAP2/Shank3
software (version 2.0). Statistical tests were two-tailed with a signifi-
occurs at nanomolar Zn2 + -concentrations and it is therefore
cance level of 4 0.05 (*P 5 0.05; **P 5 0.01; ***P 5 0.001).
plausible that a cytoplasmic pool of ProSAP2/Shank3 exists that
is not Zn2 + bound.
A regression correlation analysis revealed that
Results Zn2 + -supplementation or depletion had significantly stronger
effects on the synaptic localization of ProSAP2/Shank3 than
ProSAP1/Shank2 (Supplementary Fig. 2A). Given that dissociated
The local zinc concentration regulates hippocampal cell cultures are low in presynaptic vesicular Zn2 +
ProSAP/Shank protein levels at the and that application of Zn2 + -chelators during 1 min HiK + stimu-
postsynaptic density lation and subsequent re-supplementation with conditioned
medium still results in an increase of synaptic ProSAP1/Shank2
We first investigated whether alterations of Zn2 + -concentrations
and ProSAP2/Shank3 levels (Supplementary Fig. 2B), we also
regulate ProSAP1/Shank2 and ProSAP2/Shank3 levels at the syn-
investigated whether releasable postsynaptic Zn2 + is a potential
apse. Therefore, we supplemented hippocampal primary neurons for
source for binding to ProSAP/Shank. Important intracellular Zn2 +
1 h with 10 mM ZnCl2, which leads to an increase of postsynaptic
buffers are MTs, in particular MT-3, which is prominently
Zn2 + concentration as evidenced by elevated Zinquin ethyl-ester expressed in neurons (Lee et al., 2003). It has been reported
fluorescence (Coyle et al., 1994) (Supplementary Fig. 1A). that Ca2 + transients can rapidly and persistently release Zn2 +
Supplementation of neurons with Zn2 + leads to a significant from MT through endogenously generated NO (Wang et al.,
increase in synaptic ProSAP1/Shank2 and ProSAP2/Shank3 2008). As such a mechanism can conceivably also occur in syn-
immunofluorescence levels (Supplementary Fig. 1B and Fig. 1A apses we therefore overexpressed MT-3 and applied a NO antag-
and C), which was blocked in the presence of AMPA and NMDA onist (N-nitro-L-arginine) as well as an agonist (spermine nonoate)
receptor antagonists (Supplementary Fig. 1C). Increasing neuronal (Fig. 1G and H). Stimulation of neurons in the presence of the NO
activity with 2 min bath application of HiK + (Fig. 1A–C) or 30 mM antagonist inhibited an increase in synaptic ProSAP1/Shank2 and
glutamate (Supplementary Fig. 1D) induces similar changes in syn- ProSAP2/Shank3 levels, whereas application of the NO agonist
aptic ProSAP/Shank immunofluorescence, and protein levels in a slightly increased ProSAP2/Shank3 platform formation (Fig. 1G).
synapse-enriched P2 fraction after 30 min without increase in overall Moreover, analysis of MT-3 overexpressing neurons compared
protein levels (Supplementary Fig. 1F). Application of HiK + increased with control cells showed an increase in ProSAP2/Shank3 levels
synaptic Zn2 + measured by Zinquin (Supplementary Fig. 1E). HiK + in transfected cells (Fig. 1H), indicating that releasable postsynap-
stimulation in presence of Zn2 + leads to a similar increase and does tic Zn2 + can also contribute to the assembly of the postsynaptic
not act in an additive manner hinting towards a mechanism triggered ProSAP/Shank scaffold. Stimulation with HiK + further increased
by stimulation similar to those observed with increasing Zn2 + levels synaptic ProSAP1/Shank2 and ProSAP2/Shank3 levels in MT-3
(Fig. 1C). To exclude toxic effects of HiK + stimulation in the pres- overexpressing cells, whereas knockdown of MT-3
ence of Zn2 + , we quantified the number of cells per optic field. Our (Supplementary Fig. 2C) blocked activity-dependent increases
data show no significant increase in cell death (Supplementary Fig. (Fig. 1H).
1G), nor could we detect significantly more swelling or pinching off Finally, the synaptic localization of Shank1 (which does not
of dendrites. Most important, bath application of HiK + in the pres- bind Zn2 + as well as Homer1b/c) PSD95, mGluR5 and gephyrin
ence of a Zn2 + -chelator (CaEDTA or TPEN) was unable to elicit (a scaffold protein of inhibitory synapses that does not contain
activity dependent increases in synaptic ProSAP1/Shank2 and ProSAP/Shank proteins) were analysed. The results show that
ProSAP2/Shank3 levels and the application of CaEDTA or TPEN there is no general increase in postsynaptic density protein levels
even led to a decrease in ProSAP1/Shank2 and ProSAP2/Shank3 after Zn2 + -supplementation. Shank1 concentrations at the postsy-
levels (Fig. 1C). Although CaEDTA is cell impermeable, at the con- naptic density are not affected by either Zn2 + supplementation
centration used it will also deplete intracellular Zn2 + (Frederickson or HiK + stimulation (Fig. 1D), whereas Homer1b/c seems to be
et al., 2002). Lower CaEDTA concentrations in contrast inhibited the negatively regulated by activity and Zn2 + supplementation, but
Zn deficiency dysregulates ProSAP/Shank Brain 2014: 137; 137–152 | 141

Figure 1 Synaptic levels of zinc-binding ProSAP/Shank family members change under Zn2 + -supplementation and depletion in a MT-3
and NO-dependent manner. Hippocampal neurons (14 days in vitro) were treated with HiK + and/or Zn2 + -chelators (TPEN, CaEDTA)
and/or were supplemented with Zn2 + . (A, C, D, G and H) A puncta by puncta analysis of the synaptic signal intensity was performed from
10 cells (14 days in vitro, n = 10) of each treatment group. Signal intensities were normalized against untreated cells (Control) or in case of
transfected cells (H) against an untransfected cell in the same optic field of view. (A) Example images are shown; merged images
additionally show DAPI staining of the nucleus (scale bar = 20 mm). (B) Protein biochemistry confirms an increase of ProSAP2/Shank3

(continued)
142 | Brain 2014: 137; 137–152 S. Grabrucker et al.

unaffected from Zn2 + depletion. In contrast, synaptic immuno- cerebellum (Frederickson and Moncrieff, 1994), we failed to
fluorescence for PSD95 and mGluR5 receptors, was also elevated detect a significant reduction (Supplementary Fig. 4A–D).
following Zn2 + supplementation or HiK + stimulation (Fig. 1D), The general brain morphology and cell density assessed by 4’,6-
although only the increase in mGluR5, but not PSD95 was blocked diamidino-2-phenylindole (DAPI) and Nissl staining was not
by stimulation in the presence of CaEDTA. altered in Zn2 + -deficient animals (Supplementary Fig. 4E–H).
Labelling of the pre- and postsynaptic marker proteins bassoon,
Zinc deficiency leads to synaptic deficits homer1 and gephyrin in brain sections of Zn2 + -deficient animals
shows that compared with control animals, a general decrease in
associated with the loss of synaptic the number of homer1- and bassoon-positive signals (excitatory
ProSAP2/Shank3 in vivo synapses), visible as a trend (P = 0.09) in the hippocampus but
Autism spectrum disorders are neurodevelopmental disorders statistically significant in the cortex and striatum (Fig. 2A), can
and we therefore asked whether acute or chronic be detected. The number of gephyrin- and bassoon-positive
Zn2 + -supplementation or depletion influence synapse maturation signals (inhibitory synapses) was unchanged. Next, we performed
during development. Cultivation of hippocampal neurons under immunohistochemistry to label ProSAP/Shank proteins. The
Zn2 + -deficient conditions leads to a decrease in synapse density, immunofluorescence intensity correlating to protein levels of
whereas the number of inhibitory synapses was not altered ProSAP2/Shank3 was significantly reduced in all four brain
(Supplementary Fig. 3). In contrast, we observed a significant regions: striatum, hippocampus, cortex and cerebellum (Fig. 2B
upregulation of ProSAP1/Shank2 signal density in cells supple- and C). Moreover, we found a significant reduction of ProSAP1/
mented with ZnCl2 (Supplementary Fig. 3). Thus, chronic Shank2 immunoreactivity in cortex (Fig. 2C). The density and in-
Zn2 + -deficiency might contribute to defects that are observed tensity of gephyrin immunofluorescence puncta used as a marker
in ProSAP/Shank associated brain disorders such as autism of inhibitory synapses was not affected in mice fed with a Zn2 +
spectrum disorder. We therefore analysed the impact of deficient diet (Fig. 2A and C).
Zn2 + -deficiency on ProSAP/Shank scaffold formation in vivo by Analysis of P2 fractions from striatum, hippocampus, cortex and
inducing Zn2 + -deficiency in mice. cerebellum also revealed a significant reduction of protein levels of
To this end, mice were fed a Zn2 + -deficient diet for 8 weeks. ProSAP1/Shank2 in cortex and striatum, ProSAP2/Shank3 in all
Timm’s staining to visualize free Zn2 + ions revealed a significant four brain regions, and of Shank1 in hippocampus (Fig. 2D) in
reduction of brain Zn2 + levels in cortical, striatal and hippocampal Zn2 + -deficient mice. Along with this reduction we observed a
regions (Supplementary Fig. 4A and B). The significant reduction shift from the insoluble P2 to the soluble S2 fraction for several
in Zn2 + levels was further confirmed in brain sections using synaptic proteins including ProSAP/Shanks (Supplementary Fig. 5A
Zinpyr1, a Zn2 + -staining fluorophore (Supplementary Fig. 4C and B). Importantly, the loss of ProSAP1/Shank2 in the synapse
and D). Given that levels of vesicular Zn2 + are generally low in enriched P2 fraction translates into a decrease of excitatory

Figure 1 Continued
normalized against actin after HiK + stimulation and Zn2 + -supplementation in the P2 fraction of hippocampal neurons (14 days in vitro,
n = 3). (C and D) Quantification of the signal intensities obtained by immunofluorescence. A Zn2 + -concentration dependent regulation of
ProSAP1/Shank2 and ProSAP2/Shank3 can be observed. HiK + stimulation and Zn2 + -supplementation do not show additive effects. Zn2 +
chelation prevents changes induced by HiK + [no significant differences were detected between HiK + stimulation, Zn2 + -supplementation
and HiK + stimulation plus Zn2 + -supplementation, whereas treatment with CaEDTA or TPEN with or without HiK + stimulation leads to a
highly significant (P 5 0.001) decrease compared to the HiK + , Zn2 + , and HiK + + Zn2 + conditions]. (D) Along with the regulation of
ProSAP1/Shank2 and ProSAP2/Shank3, the level of PSD95 and mGluR5 receptors at the synapse changes. However, Shank1 and
Gephyrin signals do not increase with Zn2 + -supplementation. Homer1b/c levels seem to decrease with synaptic stimulation and
Zn2 + -supplementation, but are unaffected by Zn2 + -depletion. (E and F) Intrinsic fluorescence spectroscopy experiments show Zn2 + and
Mg2 + titration for the ProSAP2/Shank3 SAM domain. Mg2 + and Zn2 + titration of the ProSAP2/Shank3 SAM domain led to a decrease in
Trp fluorescence emission. The apo-SAM domain exhibited a decrease in Trp fluorescence in the range of 3–4 nM. Higher
Zn2 + -concentrations led to precipitation of the protein (data not shown). As Zn2 + -binding of the protein will always occur in the presence
of Mg2 + titration we also performed Zn2 + titration of the Mg2 + bound-SAM domain and found here a decrease in Trp fluorescence in the
lower nanomolar but not picomolar range. (E) Zn2 + titration of the apo-SAM domain with low nM Zn2 + decreases fluorescence intensity.
(F) Mg2 + titration of the apo-SAM domain decreases fluorescence intensity and Zn2 + titration in the low nanomolar range to Mg2 +
bound protein further decreases the fluorescence intensity. (G) HiK + stimulation of cells significantly increases ProSAP1/Shank2 and
ProSAP2/Shank3 levels compared with control cells treated with N-nitro-L-arginine, and cells treated with spermine. Stimulation of cells in
the presence of a NO agonist or antagonist shows that HiK + together with NO antagonist (N-nitro-L-arginine) blocks the increase in
ProSAP1/Shank2 and ProSAP2/Shank3. (H) Overexpression of MT-3 (10–14 days in vitro) increases synaptic ProSAP1/Shank2 and
ProSAP2/Shank3 levels. Stimulation of neurons increases ProSAP1/Shank2 and ProSAP2/Shank3 levels in control (GFP transfected) cells
and even further increases the levels in MT-3 overexpressing cells. In contrast, stimulation-induced changes are prevented by knockdown
of MT-3 (10–14 days in vitro). No significant differences were detected between HiK + stimulated and unstimulated GFP-MT-3 shRNA
transfected cells. The ratio between transfected and untransfected neurons within the same optic field are shown. All values were normally
distributed and tested for significance using t-test followed by ANOVA. *P 5 0.05; **P 5 0.01; ***P 5 0.001.
Zn deficiency dysregulates ProSAP/Shank Brain 2014: 137; 137–152 | 143

Figure 2 Dietary Zn2 + deficiency reduces the amount of ProSAP/Shank and postsynaptic density (PSD) proteins. (A) Sections of
three control and three Zn2 + -deficient mice (18 weeks of age) were stained for bassoon, homer1 and gephyrin and the number of
immune-reactive signals counted in at least three optic fields of view per region. Bassoon- and gephyrin-positive signals correspond to
inhibitory synapses, whereas bassoon- and homer1-positive signals were considered as excitatory synapses. A significant decrease in
excitatory synapse density in cortex and striatum can be seen in Zn2 + -deficient animals. A trend towards an increase in inhibitory synapse
numbers can be seen, although not significant in Zn2 + -deficient animals. (B and C) Sections of three control and three Zn2 + -deficient mice

(continued)
144 | Brain 2014: 137; 137–152 S. Grabrucker et al.

receptors in this fraction, including GluA1 (striatum and hippocam- ProSAP/Shank puncta in different brain regions of Zn2 + -deficient
pus), GluA2 (cerebellum and striatum), GluA3 (striatum), GluN1 pups nursed by control mothers was not significantly different for
(striatum and hippocampus) and GluN2B (cortex) (Fig. 2D). ProSAP1/Shank2 and ProSAP2/Shank3. In contrast, Shank1 levels
Interestingly, mGluR5 levels were significantly decreased in hippo- were significantly decreased averaging all brain regions. Total
campus but significantly increased in striatum (Fig. 2D). gephyrin levels were again not altered. The number of signals
We also prepared synaptic junction fractions of whole brain per optic field was not significantly different for all ProSAP/
(without cerebellum) of Zn2 + -deficient animals and compared pro- Shank family members (Fig. 3H), whereas a slight decrease of
tein levels to those of control mice (Fig. 2E). We detected a sig- gephyrin positive puncta in the cerebellum was measured
nificant loss of ProSAP2/Shank3 proteins and GluA1 receptors in (Fig. 3H).
Zn2 + -deficient animals. However, we could not see the decrease
in ProSAP1/Shank2 and Shank1 (Fig. 2E). Quantitative real-time
PCR confirmed that the observed changes occurred on protein Mouse models for acute and prenatal
rather than on messenger RNA level, since the transcriptional
levels of ProSAP1/Shank2 and ProSAP2/Shank3 were not altered
zinc deficiency are hyper-responsive,
between control and Zn2 + -deficient mice (Fig. 2F). hyperactive and show impairments in
Analysing the pups of control and Zn2 + -deficient animals on social behaviour and ultrasonic
post-natal Day 3, we also detected a significant reduction in
brain Zn2 + -levels in pups from Zn2 + -deficient mothers (Fig. 3A
vocalization
and B). However, all pups from and nursed by Zn2 + -deficient mice To investigate the influence of Zn2 + -deficiency on behaviour, we
died on post-natal Day 20, whereas pups from Zn2 + -deficient generated acute and prenatal Zn2 + -deficient mice. In a first set of
mice but nursed by control mice survived. The brain Zn2 + levels experiments, we analysed the influence of acute Zn2 + -deficiency
of Zn2 + -deficient pups nursed by control mice were no longer in a test for maternal behaviour (response to wriggling calls)
significantly different from those of control pups (Fig. 3C). (Fig. 4A–E and Supplementary Fig. 6). Zn2 + -deficient mice
Zn2 + -deficient pups did not show significantly fewer synapses demonstrated increased activity and reactivity in the situation of
(Fig. 3D), but displayed a trend towards a decrease, and the tran- maternal care compared to normal mothers (Fig. 4A–E and
scriptional levels of ProSAP1/Shank2 and ProSAP2/Shank3 were Supplementary Fig. 6D–I).
not altered in Zn2 + -deficient pups (Fig. 3E). However, protein In a second set of experiments, we performed the same test for
biochemistry using P2 fractions from post-natal Day 3 pups maternal behaviour, this time using mice that were exposed to a
shows that a significant loss of all ProSAP/Shank family members prenatal Zn2 + -deficiency (Supplementary Fig. 7A–D). In contrast
occurred in pups from Zn2 + -deficient mothers (Fig. 3F). Along to acute Zn2 + -deficient mothers, mothers that had experienced a
with this, we again detected a significant decrease in GluA1, prenatal Zn2 + -deficiency during development showed no hyper-
GluN1 and GluN2B levels. In contrast, pups from Zn2 + -deficient responsivity (Supplementary Fig. 7E–H). However, prenatal
mothers that have been nursed by control animals (fed a normal Zn2 + -deficient mothers showed less maternal behaviour in
diet) after birth show a significant rescue of ProSAP/Shank and response to calls (Fig. 4F).
GluA1, GluN1 and GluN2B protein levels. However, Shank1, This decrease of maternal behaviour prompted us to analyse
GluA1 and GluN1 still were significantly reduced compared with prenatal Zn2 + -deficient mice, performing behavioural tests (pup
control animals (Fig. 3F). Although the amount of mGluR5 was retrieval, analysis of ultrasonic vocalization and maternal resident
significantly decreased in Zn2 + -deficient pups, a significant intruder test) indicative for autism spectrum disorder-related be-
increase was detected in pups from Zn2 + -deficient mothers that haviour. The results show that although we could not detect sig-
have been nursed by control animals (Fig. 3F). Using immunohis- nificant changes in the pup retrieval test between prenatal
tochemistry, similar results were obtained for ProSAP/Shank pro- Zn2 + -deficient and control mice regarding the latency to respond
teins (Fig. 3G). Although all synaptic concentrations of ProSAP/ to the stimulus, the number of control walks performed and the
Shank family members are decreased in Zn2 + -deficient pups number of pup retrievals (Supplementary Fig. 7I–K), prenatal
(nursed by Zn2 + -deficient mothers), the signal intensity of labelled Zn2 + -deficient mice showed an impairment in auditory

Figure 2 Continued
were stained for ProSAP1/Shank2, ProSAP2/Shank3, Shank1 and gephyrin and the signal intensity of synaptic puncta evaluated [scale
bar = 500 mm (overview and 5); scale bar = 20 mm ( 40 and enlargements)]. A significant decrease of ProSAP2/Shank3 (all brain
regions) and ProSAP1/Shank2 (cortex) could be detected. The levels of Shank1 and gephyrin were not altered. (D) Protein biochemistry
using P2 fractions of four brain regions of Zn2 + -deficient and control mice (n = 3). Protein concentrations were normalized to actin and are
shown relative to the expression in control mice. A decrease of ProSAP2/Shank3 in all brain regions can be confirmed as well as a loss of
GluA1 and GluN1 receptors most prominent in hippocampus and striatum. (E) Protein biochemistry using synaptic junction fractions of
three Zn2 + -deficient and three control mice normalized against actin and relative to the expression levels of control mice. A significant
decrease in ProSAP2/Shank3 and GluA1 levels can be seen. (F) Quantitative real-time PCR shows no differences in ProSAP1/Shank2 and
ProSAP2/Shank3 messenger RNA expression levels between Zn2 + -deficient and control mice (n = 3). All values were normally distributed
and tested for significance using t-test followed by ANOVA. *P 5 0.05; **P 5 0.01; ***P 5 0.001.
Zn deficiency dysregulates ProSAP/Shank Brain 2014: 137; 137–152 | 145

Figure 3 Maternal Zn2 + deficiency reduces the amount of ProSAP/Shank and postsynaptic density proteins in pups. (A) Using the Zn2 +
fluorophore Zinpyr1 to evaluate Zn2 + -levels reveals a significant reduction in signal intensity in brains from pups (post-natal Day 3, n = 3)
from Zn2 + -deficient mothers and nursed by Zn2 + -deficient mothers. Sections of control and Zn2 + -deficient mice were imaged and
quantified by measuring at least three optic fields of view (scale bar = 300 mm) and controls were set to 100%. (B) Timm’s staining
similarly shows a significant reduction in signal intensity in pups from Zn2 + -deficient mice (post-natal Day 3, n = 3, controls set to 100%)
(scale bar = 150 mm). (C) Pups (post-natal Day 3) nursed by control mothers show a rescue in Zn2 + -levels almost up to brain

(continued)
146 | Brain 2014: 137; 137–152 S. Grabrucker et al.

discrimination between a meaningful (50 kHz) and neutral samples of 38 participants diagnosed with Phelan-McDermid syn-
(20 kHz) sound stimulus (Fig. 4G). drome. Similar to former studies with autistic patients (Faber et al.,
The most prominent impairments were observed evaluating 2009; Lakshmi Priya and Geetha, 2011; Yasuda et al., 2011), we
parameters of ultrasonic vocalizations of adult males to female detected an increased incidence rate for Zn2 + -deficiency in
urine (Fig. 4H–L). Besides several parameters that were not patients with Phelan-McDermid syndrome primarily in the age
altered in prenatal Zn2 + -deficient mice compared with control group 512 years (Supplementary Fig. 9A) as previously reported
mice (Supplementary Fig. 8A–H), we found that prenatal for autistic children (Yasuda et al., 2011), with the exception of
Zn2 + -deficient mice vocalized significantly less calls per minute two cases at 19 and 46 years of age. From 37 participants,
observation time (Fig. 4H), had a significantly reduced loudness 30 were found to have normal blood Zn2 + -levels, four showed
of calls (Fig. 4I), a significantly increased latency to start calling a mild Zn2 + -deficiency (1–20% less than the lowest concentration
(Fig. 4J) and a trend to a reduced average call duration compared of the age matched reference values for healthy controls)
to control animals (Fig. 4K). Moreover, the number of overtones and three a severe Zn2 + -deficiency (420% less than the lowest
and harmonics was significantly reduced in the calls (Fig. 4L). concentration of the age matched reference values for healthy
Finally, we conducted a maternal resident intruder test. Besides controls) (Fig. 5A and B). Zn2 + -deficiency was not correlated
parameters that were not altered (Fig. 4M and Supplementary Fig. with gender (Supplementary Fig. 9B) and the participants
8I–L), the number of attacks performed by the mother was had no dietary problems (no underweight, no physical disability
significantly increased in prenatal Zn2 + -deficient mice (Fig. 4N). hampering food consumption, normal dietary habits), that might
explain the deficiency.
Zinc deficiency in Phelan-McDermid Participants were evaluated based on 26 categories (see
‘Materials and methods’ section.). We found that 61% of partici-
syndrome is associated with a higher pants suffering from seizures showed a Zn2 + -deficiency (mild
incidence rate of seizures, hypotonia and severe combined) compared with only 12% of participants
and the occurrence of attention without seizures (Fig. 5C). Additionally, 60% of participants
with attention deficit disorder/attention deficit hyperactivity
deficits and hyperactivity disorder or other attention or hyperactivity issues were
Given that patients with Phelan-McDermid syndrome suffer from severely Zn2 + -deficient and 27% of participants with hypotonia
a haplo-insufficiency of ProSAP2/Shank3 we investigated whether compared with 15% and 0%, respectively, of participants with-
Zn2 + -deficiency in patients with Phelan-McDermid syndrome out the symptom (Fig. 5C). Additionally, the occurrence
leads to a more pronounced phenotype. Using atomic absorption of Zn2 + -deficiency was positively correlated with signs of
spectroscopy, we evaluated Cu2 + and Zn2 + -levels in blood immunodeficiency (Fig. 5C).

Figure 3 Continued
Zn2 + -concentration of control pups. Again Timm’s staining (left) and Zinpyr1 labelling (right) was performed as shown in A and B.
(D) Sections of three control and three Zn2 + -deficient mice were stained for bassoon and homer1 and the number of positive signals
counted in multiple fields of view. Bassoon- and homer1-positive signals were considered as excitatory synapses. A trend towards a
decrease in excitatory synapse density, although not significant, can be seen in Zn2 + -deficient animals. (E) Quantitative real-time PCR
shows no differences in ProSAP1/Shank2 and ProSAP2/Shank3 messenger RNA expression levels normalized to HMBS expression
between pups (post-natal Day 3) from Zn2 + -deficient and control mice (n = 3). (F) P2 fractions from post-natal Day 3 pups show a
significant reduction of all ProSAP/Shank family members in pups from Zn2 + -deficient mothers. Along with this, a significant decrease in
GluA1, GluN1 and GluN2B can be observed. Pups from Zn2 + -deficient mothers that have been nursed by control animals show a
significant rescue of ProSAP/Shank and GluA1, GluN1 and GluN2B levels, although Shank1, GluA1 and GluN1 stay significantly reduced
compared with control animals. Expression levels were normalized against actin and are shown relative to the expression of control
mice (n = 3). (G) Sections from three control, Zn2 + -deficient pups (post-natal Day 3) raised by control mothers, and Zn2 + -deficient pups
(post-natal Day 3) raised by Zn2 + -deficient mothers were stained for ProSAP1/Shank2, ProSAP2/Shank3, Shank1 and gephyrin and the
signal intensity of synaptic puncta evaluated. Similar to the western blot results shown in (F), a significant decrease of ProSAP1/Shank2,
ProSAP2/Shank3 and Shank1 (total of all brain regions) could be detected in Zn2 + -deficient pups. The levels of ProSAP1/Shank2 and
ProSAP2/Shank3 in rescued Zn2 + -deficient pups (raised by control mothers) were not different from control animals. The levels of Shank1
however, were still significantly reduced considering the average values of all brain regions. The synaptic concentration of gephyrin for all
brain regions is not altered, although a decrease in cerebellum and an increase in hippocampus is seen in rescued Zn2 + -deficient pups. The
data are shown normalized to signal intensity values of controls. (H) Sections of three control and three Zn2 + -deficient pups (post-natal
Day 3) raised by control mothers were stained for ProSAP1/Shank2, ProSAP2/Shank3, Shank1 and gephyrin and the mean number of
positive signals counted per optic field from five images. The data are shown normalized to signal density values of controls. A significant
decrease in gephyrin-positive signals can be seen in Zn2 + -deficient pups raised by control mothers. The signal density of ProSAP/Shank
immunoreactive puncta was not significantly different from control pups [images show exemplary stainings of ProSAP1/Shank2 (green),
ProSAP2/Shank3 (red) and DAPI (blue) of the analysed brain regions] (scale bar = 10 mm). Although ratios are shown, statistical tests were
performed using the actual signal density of the Zn2 + -deficient (rescued) animals compared to the actual signal density of control animals.
(A–H) All values were normally distributed and tested for significance using t-test followed by ANOVA. *P 5 0.05; **P 5 0.01;
***P 5 0.001.
Zn deficiency dysregulates ProSAP/Shank Brain 2014: 137; 137–152 | 147

Figure 4 Behavioural deficits in a mouse model for acute and prenatal Zn2 + -deficiency. (A–E) Acute Zn2 + -deficient mice demonstrated
increased activity and reactivity in the situation of maternal care compared to normal mothers. (A) In a test for maternal behaviour, adult
Zn2 + -deficient mice (n = 20, A–E) demonstrate a significantly higher number of interruptions of nursing/warming position compared to
controls (n = 18, A–E, t-test). Additionally, Zn2 + -deficient mice showed significantly more spontaneous attention behaviour (lifting or
turning the head, U-test) (B) and spontaneous maternal behaviour (C) in the absence of calls independent of the Zn2 + status of pups
(U-test) (Supplementary Fig. 6D–F). (D) The attention response to natural wriggling calls and the synthesized call model was significantly

(continued)
148 | Brain 2014: 137; 137–152 S. Grabrucker et al.

Zn2 + -deficiency in adult mice only significantly changes Shank1


Discussion levels in hippocampus, a much more pronounced reduction
Recent first in vitro evidence indicated Zn2 + incorporation in large of Shank1 is seen in pups from Zn2 + -deficient mothers.
macromolecular platforms that are built by the SAM domains of One might speculate that in adult mice, synapses have already
the autism-associated postsynaptic density scaffolding molecules been stabilized by Shank1 and that loss of Shank1 because
ProSAP2/Shank3 and ProSAP1/Shank2. Here, we propose a of impaired synapse maturation needs longer periods of
model where the synaptic assembly of ProSAP1/Shank2 and Zn2 + -deficiency in less plastic brain regions than the hippocampus.
ProSAP2/Shank3 proteins is influenced by Zn2 + -deficiency. In contrast, in Zn2 + -deficient pups upon initial synapse formation,
Zn2 + -depletion from neuronal cultures or Zn2 + -sequestration by postsynaptic densities might not be targeted by Shank1 from
amyloid-b decreased the synaptic levels of Zn2 + -binding ProSAP/ the beginning because of their weakened ProSAP1/Shank2 and
Shank family members as shown here and in former studies ProSAP2/Shank3 scaffold.
(Grabrucker et al., 2011a, b) and results in a reduced synapse Intriguingly, we observed most alterations in hippocampus and
density. Analysing Zn2 + -deficient mice, here, we also detected a especially striatum, being the brain regions with the highest
reduction in synapse number in the cortex and striatum. Along expression levels of ProSAP2/Shank3 compared with other
with this, we could show a significant decrease of ProSAP1/ ProSAP/Shank family members (Peça et al., 2011). Similar to a
Shank2 (cortex) and ProSAP2/Shank3 (cortex, hippocampus, stri- ProSAP2/Shank3 knockout mouse model (Peça et al., 2011), we
atum, cerebellum) through immunofluorescence and a significant detected a reduction in spine density in the striatum. Given that
decrease of ProSAP1/Shank2 (cortex, striatum), ProSAP2/Shank3 the striatum contains a low amount of presynaptic Zn2 + but has a
(cortex, hippocampus, striatum, cerebellum) and Shank1 (hippo- high expression of ProSAP2/Shank3 scaffolds that can only be
campus) through protein biochemistry in Zn2 + -deficient mice. This built in presence of Zn2 + , indeed Zn2 + may be bound within
loss of postsynaptic density scaffold proteins was accompanied by the postsynaptic density in all brain regions regulated by postsy-
a downregulation of receptors, especially GluA1 and GluN1 naptic Zn2 + -stores. Thus, if postsynaptic stores are affected and
(hippocampus and striatum) reminiscent of a similar downregula- ProSAP2/Shank3 has a predominant role in the striatum, it is likely
tion of AMPA receptors reported in heterozygous and homozy- that synapse density in this brain region is most affected by
gous ProSAP2/Shank3 b knockout mice (Bozdagi et al., 2010; Zn2 + -deficiency, independent from the presynaptic pool of Zn2 +
Wang et al., 2011). ProSAP2/Shank3 b knockout and ProSAP2/ ions. This might also explain the reduction of ProSAP2/Shank3
Shank3C mice were also reported to have decreased GluN1 and levels in cerebellum.
GKAP levels (Bangash et al., 2011; Wang et al., 2011). As it was also shown before that there is a rapid turnover of
Furthermore, ProSAP1/Shank2 knockout mice were reported to ProSAP/Shank proteins at the synapse (Tsuriel et al., 2006) and
display a decrease in NMDA receptors (Won et al., 2012). that ProSAP1/Shank2 and ProSAP2/Shank3 can be detected in
Previous studies have shown that especially nascent synapses the soluble (S2) as well as postsynaptic density-bound (P2) frac-
are sensitive to Zn2 + -depletion (Grabrucker et al., 2011a) and a tion of brain lysates, it is plausible that both proteins undergo a
model was proposed where ProSAP1/Shank2 is the first ProSAP/ shift from P2 to S2 upon Zn2 + -depletion (Grabrucker et al.,
Shank family member found at forming synapses followed 2011a). One can therefore assume that the soluble and postsy-
by ProSAP2/Shank3. Only if a sufficient stabilized synapse naptic density-bound pool exist in equilibrium. Synaptic activity
with preformed scaffold is established, Shank1 is targeted to post- and Zn2 + -release may rapidly shift this equilibrium to a more
synaptic densities, which leads to less Zn2 + -sensitive mature postsynaptic density-bound pool of ProSAP/Shank (Grabrucker,
synapses. The results of the present study show that although 2013).

Figure 4 Continued
increased in Zn2 + -deficient mothers independent of the Zn2 + status of pups (Chi square test) (Supplementary Fig. 6G). (E) Maternal
behaviour (licking, change in nursing position, nest building) of both groups of mothers was increased in response to calls of
Zn2 + -deficient pups and to the call model (Chi square test) (Supplementary Fig. 6H and I). (F) In the same test for maternal behaviour
(A–E) conducted with mice that suffered from prenatal Zn2 + -deficiency, over-responsiveness and hyperactivity were not detected.
However, prenatal Zn2 + -deficient mice (n = 6, 15 weeks of age) showed significantly less maternal behaviour in response to the natural
calls of pups compared to control mice (n = 7, Chi square test) (F). (G) A pup retrieval test revealed that prenatal Zn2 + -deficient mice
(n = 24, 14 weeks of age with pups post-natal Day 4–5) responded significantly more to the neutral 20 kHz sound stimulus and thus
significantly less to the biologically meaningful 50 kHz sound compared with control mice (n = 42). Data of both groups were compared
with the sign test according to Dixon and Mood. The comparison of the number of choices between the two tones was done using
Chi-square test. Moreover, prenatal Zn2 + -deficient male mice (n = 7, 15 weeks of age) showed impaired ultrasonic vocalization compared
with control mice (n = 13) (H–L). The number of calls per minute (H) was significantly decreased (t-test), the loudness was significantly
reduced (U-test) (I) and the latency to start calling significantly increased (U-test) (J) compared with control animals. Prenatal Zn2 + -
deficient males additionally showed a trend towards a reduced call duration (#P = 0.073, t-test) (K) and significantly less vocalizations with
overtones and harmonics (L) compared with control animals (Chi-square test). (M) A maternal resident intruder test revealed that,
although the time a mother spent in contact with an intruding male was not significantly different between prenatal Zn2 + -deficient and
control mothers (U-test) (M), the number of attacks was significantly increased in prenatal Zn2 + -deficient mothers (n = 5, t-test) (N).
*P 5 0.05; **P 5 0.01.
Zn deficiency dysregulates ProSAP/Shank Brain 2014: 137; 137–152 | 149

Figure 5 Zn2 + deficiency is associated with an increased rate of seizures, attention or hyperactivity issues, hypotonia and possible signs of
primary immunodeficiency in patients with Phelan-McDermid syndrome. (A and B) Using atomic absorption spectroscopy, Cu2 + and
Zn2 + -levels in blood samples of 38 participants diagnosed with Phelan-McDermid syndrome were evaluated. (A) From 37 participants,
30 were found to have normal blood Zn2 + -levels, four showed a mild Zn2 + -deficiency and three a severe Zn2 + -deficiency. (B) A copper
overload was detected in four participants. (A and B) The measured values are shown in per cent compared with age matched control
reference values. (C) A total of 26 categories were analysed based on the participant’s data provided by the Phelan-McDermid Syndrome
Registry. The results show that 61% of participants suffering from seizures have Zn2 + -deficiency compared with only 12% of participants
without seizures (participants with mild and severe Zn2 + -deficiency combined) (n = 32 total). Sixty per cent of participants with attention
deficit disorder/attention deficit hyperactivity disorder (ADD/ADHD) or other attention of hyperactivity issues were Zn2 + -deficient and
27% of participants with hypotonia compared with 15% and 0%, respectively, of participants without the symptom (participants with
mild and severe Zn2 + -deficiency combined; n = 17 and 24 total, respectively). In the group of participants showing possible signs of
primary immunodeficiency (four or more ear infections within 1 year, two or more serious sinus infections within 1 year, two or more

(continued)
150 | Brain 2014: 137; 137–152 S. Grabrucker et al.

The present results raise a number of questions concerning the instinctive behaviour, which might reflect cerebellar action, were
source of postsynaptic free Zn2 + , the dynamics of the ProSAP/ not observed. Because autism spectrum disorders are developmen-
Shank scaffold and the pathomechanism of autism spectrum dis- tal disorders, induction of Zn2 + -deficiency in adulthood is not
order and related disorders. Knockout mice for ZnT3, the major expected to generate autism in mice. Nevertheless, the results
and possibly sole synaptic vesicular Zn2 + -transporter, were not show that Zn2 + -deficiency might be responsible for co-morbidities
reported yet to display autism-like features, although they show seen in autism spectrum disorders such as increased anxiety and
an absence of free Zn2 + in synaptic vesicles (Cole et al., 1999). sensory over-responsiveness, which is underlined by the increased
Therefore the question arises whether presynaptically released rate of hyperactivity issues in patients with Phelan-McDermid syn-
Zn2 + is the sole source of Zn2 + ions involved in the synaptic drome and Zn2 + -deficiency. For instance, in addition to the core
targeting of ProSAP/Shank. Alternatively, local elevation of diagnostic features for autism spectrum disorder, atypical stimuli-
Zn2 + -concentrations could arise by activation of intracellular evoked responses have been reported in up to 95% of children
Zn2 + -stores through NO. In the brain, NO has been shown to with autism spectrum disorder (Lane et al., 2012).
displace Zn2 + from protein-binding sites (Cuajungco and Lee, Intriguingly, our data from pups from Zn2 + -deficient mice indi-
1998; Aravindakumar et al., 1999), causing free or weakly cate that maternal Zn2 + -deficiency is also able to induce changes
bound Zn2 + to be present in the cytoplasm that could play a in ProSAP/Shank levels in the offspring, similar to those reported
role in intracellular signalling (Bossy-Wetzel et al., 2004; Lin in adult Zn2 + -deficient mice here. Although Zn2 + -deficiency led to
et al., 2007). Our in vitro experiments indicate that MT-3 might the death of pups on post-natal Day 20 hinting towards a devel-
be a candidate for postsynaptic activity-dependent Zn2 + -release opmental milestone that Zn2 + -deficient pups were unable to
due to activity-dependent nitrosylation or oxidation of the thiol achieve, provision of adequate Zn2 + supply by control mothers
ligands by NO (Maret, 2000). In line with this, NO antagonists interestingly led to survival and a rescue of the observed alter-
prevented the stimulation-induced upregulation of Zn2 + -binding ations in most, but not all postsynaptic density proteins. At the
ProSAP/Shank proteins at the synapse. Overexpression of MT-3, age of 17–22 days, pups usually start eating solid food and during
because of spontaneous synaptic activity or more so after HiK + this weaning period, the mothers begin to withdraw from their
stimulation, increased synaptic ProSAP2/Shank3 levels, whereas litter to reduce suckling attempts by pups (Koenig and Markl,
MT-3 knockdown prevented an activity-dependent increase. The 1987). Zn2 + -deficiency is known to have an influence on motor
slight increase in ProSAP2/Shank3 by MT-3 knockdown in development (Black, 1998) and a possible reason for the death of
absence of stimulation might be because of a higher baseline pups on post-natal Day 20 might be an inability to reach sufficient
availability of free Zn2 + that would have been bound to MT-3 amounts of food and water at the top of the cage.
otherwise. Intriguingly, mice lacking MT-3 show a reduction in Prenatal Zn2 + -deficient mice, in contrast to acute
brain Zn2 + -concentrations as a result of the absence of Zn2 + Zn2 + -deficient mice, did not show signs of increased hyperactiv-
bound to MT-3. However, the histochemically-reactive ity-like and over-responsive behaviour. We observed significantly
Zn2 + -content is unaffected, providing evidence that MT-3 does decreased maternal behaviour along with impairment in auditory
not influence presynaptic vesicular Zn2 + (Erickson et al., 1995, discrimination of sound stimuli, whereas sound perception per se
1997). MT-3 knockout mice show abnormalities in behaviour was not altered. Most prominent, we could detect significant dif-
(Koumura et al., 2009) and have been suggested as suitable sub- ferences in the vocalization of prenatal Zn2 + -deficient males.
jects to investigate psychological disorders (Koumura et al., 2009). Intriguingly, a reduction in ultrasonic vocalizations has been re-
Interestingly, previous studies have reported a high rate of ported previously for heterozygous ProSAP2/Shank3 b (Bozdagi
Zn2 + -deficiency in autistic children (Walsh et al., 1997; Jen and et al., 2010) and homozygous ProSAP1/Shank2 knockout mice
Yan, 2010; Yasuda et al., 2011), suggesting that infantile (Schmeisser et al., 2012; Won et al., 2012). Additionally, prenatal
Zn2 + -deficiency might contribute to the pathogenesis of autism Zn2 + -deficient mice showed alterations in the social context of a
(Yasuda et al., 2011). Along with a reduction in synaptic maternal resident intruder test. We observed a significant increase
ProSAP1/Shank2 and ProSAP2/Shank3 levels, Zn2 + -deficient in attacks towards the intruder. Increased aggressiveness has been
animals developed behavioural abnormalities. reported in prenatal Zn2 + -deficient animals (Sandstead et al.,
Acute Zn2 + -deficiency led to increased hyperactivity-like spon- 1977). Taken together, prenatal Zn2 + -deficient animals indeed
taneous behaviour and over-responsiveness to acoustic stimuli. display some behavioural features observed in mouse models for
This suggests lability of behavioural coordination in the highly af- autism.
fected striatum (see above) as changes both in sound perception, Patients with Phelan-McDermid syndrome already lack one
which could be related to cortical processing, and in goal-directed functional copy of ProSAP2/Shank3, so that they should be

Figure 5 Continued
pneumonias within 1 year, persistent thrush in mouth or fungal infection of skin, or the need of intravenous antibiotics to clear infections),
38% of participants were found with mild or severe Zn2 + -deficiency compared to 7% of participants without any of these symptoms
(n = 31 total). Differences between patients with normal Zn2 + -levels/mild Zn2 + -deficiency and patients with severe Zn2 + -deficiency are
significant for the occurrence of seizures (P 5 0.001), attention deficit disorder/attention deficit hyperactivity disorder or other attention or
hyperactivity issues (P 5 0.01), possible signs of immunodeficiency (P 5 0.01), while only a trend can be observed for hypotonia
(P = 0.058) (chi-square Goodness of fit).
Zn deficiency dysregulates ProSAP/Shank Brain 2014: 137; 137–152 | 151

more susceptible to changes in body Zn2 + -levels. Although, spectrum disorder and mental retardation. Nat Genet 2010; 42:
489–91.
given the low incidence rate of Phelan-McDermid syndrome
Bitanihirwe BK, Cunningham MG. Zinc: the brain’s dark horse. Synapse
with 2.5–10 per million births, the number of participants in our 2009; 63: 1029–49.
study was rather low and serum Zn2 + -levels are only indicative of Black MM. Zinc deficiency and child development. Am J Clin Nutr 1998;
brain Zn2 + -levels, the results complement those from our 68: 464–9.
Zn2 + -deficient mouse model with regard to increased rate of Boeckers TM, Bockmann J, Kreutz MR, Gundelfinger ED. ProSAP/Shank
attention and hyperactivity issues compared with patients without proteins-a family of higher order organizing molecules of the postsy-
naptic density with an emerging role in human neurological disease.
these symptoms. Similarly, we found a higher proportion of
J Neurochem 2002; 81: 903–10.
Zn2 + -deficient participants in the group of patients suffering Bonaglia MC, Giorda R, Borgatti R, Felisari G, Gagliardi C, Selicorni A,
from hypotonia and/or seizures and signs of primary immunodefi- et al. Disruption of the ProSAP2 gene in a t(12;22)(q24.1;q13.3) is
ciency. Immune system abnormalities have been linked to associated with the 22q13.3 deletion syndrome. Am J Hum Genet
Zn2 + -deficiency before and both have been reported as risk 2001; 69: 261–8.
Bossy-Wetzel E, Talantova MV, Lee WD, Schölzke MN, Harrop A,
factors for the development of autism (Grabrucker, 2012).
Mathews E, et al. Crosstalk between nitric oxide and zinc pathways
Although an alternative explanation would be that patients with to neuronal cell death involving mitochondrial dysfunction and p38-
more severe clinical presentations are more susceptible to activated K + channels. Neuron 2004; 41: 351–65.
Zn2 + -deficiency, it is likely that Zn2 + -deficiency is influencing Bozdagi O, Sakurai T, Papapetrou D, Wang X, Dickstein DL,
the severity of these symptoms. Takahashi N, et al. Haploinsufficiency of the autism-associated
Shank3 gene leads to deficits in synaptic function, social interaction,
In conclusion, our data predict that autistic children on
and social communication. Mol Autism 2010; 1: 15.
Zn2 + -therapy should improve regarding attention, hyperactivity, Cole TB, Wenzel HJ, Kafer KE, Schwartzkroin PA, Palmiter RD.
seizures, sound and tactile sensitivity, among others. Recent data Elimination of zinc from synaptic vesicles in the intact mouse brain
confirm this therapeutic outlook (Russo and Devito, 2011). by disruption of the ZnT3 gene. Proc Natl Acad Sci USA 1999; 96:
1716–21.
Coyle P, Zalewski PD, Philcox JC, Forbes IJ, Ward AD, Lincoln SF, et al.
Measurement of zinc in hepatocytes by using a fluorescent probe,
Acknowledgements zinquin: relationship to metallothionein and intracellular zinc.
Biochem J 1994; 303 (Pt 3): 781–6.
The authors gratefully acknowledge the professional technical Cuajungco MP, Lees GJ. Nitric oxide generators produce accumulation of
assistance of Elisabeth Pica and thank Geraldine Bliss, Megan chelatable zinc in hippocampal neuronal perikarya. Brain Res 1998;
O’Boyle and the Phelan McDermid Foundation for their help as 799: 118–29.
well as the Phelan-McDermid syndrome families for participating Curtis LT, Patel K. Nutritional and environmental approaches to prevent-
ing and treating autism and attention deficit hyperactivity disorder
in blood donations.
(ADHD): a review. J Altern Complement Med 2008; 14: 79–85.
Durand CM, Betancur C, Boeckers TM, Bockmann J, Chaste P,
Fauchereau F, et al. Mutations in the gene encoding the synaptic
Funding scaffolding protein SHANK3 are associated with autism spectrum
disorders. Nat Genet 2007; 39: 25–7.
This work was supported by Baustein 3.2 (L.SBN.0083) and the Erickson JC, Hollopeter G, Thomas SA, Froelick GJ, Palmiter RD.
DAAD (to A.M.G). T.B. was funded by the ANR (ANR-08-MNPS- Disruption of the metallothionein-III gene in mice: analysis of brain
zinc, behavior, and neuron vulnerability to metals, aging, and seizures.
037-01 – SynGen), Neuron-ERANET (EUHF-AUTISM), Fondation
J Neurosci 1997; 17: 1271–81.
Orange and the Fondation FondaMentale. M.R.K by the DFG (SFB Erickson JC, Masters BA, Kelly EJ, Brinster RL, Palmiter RD. Expression of
779/TPB8; Kr1879/3-1). S.G. is a member of the International human metallothionein-III in transgenic mice. Neurochem Int 1995;
Graduate School in Molecular Medicine at Ulm University. 27: 35–41.
Faber S, Zinn GM, Kern JC, Kingston HM. The plasma zinc/serum copper
ratio as a biomarker in children with autism spectrum disorders.

Supplementary material Biomarkers 2009; 14: 171–80.


Frederickson CJ. Neurobiology of zinc and zinc-containing neurons. Int
Rev Neurobiol 1989; 31: 145–238.
Supplementary material is available at Brain online. Frederickson CJ, Koh JY, Bush AI. The neurobiology of zinc in health and
disease. Nat Rev Neurosci 2005; 6: 449–462.
Frederickson CJ, Moncrieff DW. Zinc-containing neurons. Biol Signals
References 1994; 3: 127–39.
Frederickson CJ, Suh SW, Koh JY, Cha YK, Thompson RB, LaBuda CJ,
Aravindakumar CT, Ceulemans J, De Ley M. Nitric oxide induces Zn2 + et al. Depletion of intracellular zinc from neurons by use of an
release from metallothionein by destroying zinc-sulphur clusters without extracellular chelator in vivo and in vitro. J Histochem Cytochem
concomitant formation of S-nitrosothiol. Biochem J 1999; 344: 253–8. 2002; 50: 1659–1662.
Bangash MA, Park JM, Melnikova T, Wang D, Jeon SK, Lee D, et al. Gauthier J, Spiegelman D, Piton A, Lafrenière RG, Laurent S, St-Onge J,
Enhanced polyubiquitination of Shank3 and NMDA receptor in a et al. Novel de novo SHANK3 mutation in autistic patients. Am J Med
mouse model of autism. Cell 2011; 145: 758–72. Genet B Neuropsychiatr Genet 2009; 150B: 421–4.
Baron MK, Boeckers TM, Vaida B, Faham S, Gingery M, Sawaya MR, Grabrucker AM. A role for synaptic zinc in ProSAP/Shank PSD scaffold
et al. An architectural framework that may lie at the core of the malformation in autism spectrum disorders. Dev Neurobiol 2013,
postsynaptic density. Science 2006; 311: 531–5. in press.
Berkel S, Marshall CR, Weiss B, Howe J, Roeth R, Moog U, et al. Grabrucker AM. Environmental factors in autism. Front Psychiatry 2012;
Mutations in the SHANK2 synaptic scaffolding gene in autism 3: 118.
152 | Brain 2014: 137; 137–152 S. Grabrucker et al.

Grabrucker AM, Knight MJ, Proepper C, Bockmann J, Joubert M, Moessner R, Marshall CR, Sutcliffe JS, Skaug J, Pinto D, Vincent J, et al.
Rowan M, et al. Concerted action of zinc and ProSAP/Shank in synap- Contribution of SHANK3 mutations to autism spectrum disorder.
togenesis and synapse maturation. EMBO J 2011a; 30: 569–81. Am J Hum Genet 2007; 81: 1289–97.
Grabrucker AM, Schmeisser MJ, Udvardi PT, Arons M, Schoen M, Outten CE, O’Halloran TV. Femtomolar sensitivity of metalloregulatory
Woodling NS, et al. Amyloid beta protein-induced zinc sequestration proteins controlling zinc homeostasis. Science 2001; 292: 2488–92.
leads to synaptic loss via dysregulation of the ProSAP2/Shank3 Peça J, Feliciano C, Ting JT, Wang W, Wells MF, Venkatraman TN, et al.
scaffold. Mol Neurodegener 2011b; 6: 65. Shank3 mutant mice display autistic-like behaviours and striatal
Grabrucker A, Vaida B, Bockmann J, Boeckers TM. Synaptogenesis of dysfunction. Nature 2011; 472: 437–42.
hippocampal neurons in primary cell culture. Cell Tissue Res 2009; Phelan MC, Rogers RC, Saul RA, Stapleton GA, Sweet K, McDermid H,
338: 333–41. et al. 22q13 deletion syndrome. Am J Med Genet 2001; 101: 91–9.
Gundelfinger ED, Boeckers TM, Baron MK, Bowie JU. A role for zinc in Pinto D, Pagnamenta AT, Klei L, Anney R, Merico D, Regan R, et al.
postsynaptic density asSAMbly and plasticity? Trends Biochem Sci Functional impact of global rare copy number variation in autism
2006; 31: 366–73. spectrum disorders. Nature 2010; 466: 368–72.
Jaarsma D, Korf J. A novel non-perfusion Timm method for human brain Rükgauer M, Klein J, Kruse-Jarres JD. Reference values for the trace
tissue. J Neurosci Methods 1990; 35: 125–31. elements copper, manganese, selenium, and zinc in the serum/
Jan HH, Chen IT, Tsai YY, Chang YC. Structural role of zinc ions bound plasma of children, adolescents, and adults. J Trace Elem Med Biol
to postsynaptic densities. J Neurochem 2002; 83: 525–534. 1997; 11: 92–8.
Jen M, Yan AC. Syndromes associated with nutritional deficiency and Russo AJ, Devito R. Analysis of copper and zinc plasma concentration
excess. Clin Dermatol 2010; 28: 669–85. and the efficacy of zinc therapy in individuals with Asperger’s
Krezel A, Maret W. Zinc-buffering capacity of a eukaryotic cell at physio- Syndrome, Pervasive Developmental Disorder not otherwise specified
logical pZn. J Biol Inorg Chem 2006; 11: 1049–62. (PDD-NOS) and Autism. Biomark Insights 2011; 6: 127–33.
Koenig B, Markl H. Maternal care in house mice. Behav Ecol Sociobiol Sandstead HH, Fosmire GJ, Halas ES, Jacob RA, Strobel DA, Marks EO.
1987; 20: 1–9. Zinc deficiency: effects on brain and behavior of rats and rhesus
Koumura A, Kakefuda K, Honda A, Ito Y, Tsuruma K, Shimazawa M, monkeys. Teratology 1977; 16: 229–34.
et al. Metallothionein-3 deficient mice exhibit abnormalities of psycho- Sato D, Lionel AC, Leblond CS, Prasad A, Pinto D, Walker S, et al.
logical behaviors. Neurosci Lett 2009; 467: 11–4. SHANK1 Deletions in Males with Autism Spectrum Disorder. Am
Lakshmi Priya MD, Geetha A. Level of trace elements (copper, zinc, J Hum Genet 2012; 90: 879–87.
magnesium and selenium) and toxic elements (lead and mercury) in Schmeisser MJ, Ey E, Wegener S, Bockmann J, Stempel AV, Kuebler A,
the hair and nail of children with autism. Biol Trace Elem Res 2011; et al. Autistic-like behaviours and hyperactivity in mice lacking
142: 148–58. ProSAP1/Shank2. Nature 2012; 486: 256–60.
Lane SJ, Reynolds S, Dumenci L. Sensory overresponsivity and anxiety in Tsuriel S, Geva R, Zamorano P, Dresbach T, Boeckers T,
typically developing children and children with autism and attention Gundelfinger ED, et al. Local sharing as a predominant determinant
deficit hyperactivity disorder: cause or coexistence? Am J Occup Ther of synaptic matrix molecular dynamics. PLoS Biol 2006; 4: e271.
2012; 66: 595–603. Walsh WJ, Isaacson HR, Rehman F, Hall A. Elevated blood copper/zinc
Leblond CS, Heinrich J, Delorme R, Proepper C, Betancur C, Huguet G, ratios in assaultive young males. Physiol Behav 1997; 62: 327–9.
et al. Genetic and functional analyses of SHANK2 mutations suggest a Wang H, Li H, Cai B, Huang ZX, Sun H. The effect of nitric oxide on
multiple hit model of autism spectrum disorders. PLoS Genet 2012; 8: metal release from metallothionein-3: gradual unfolding of the protein.
e1002521. J Biol Inorg Chem 2008; 13: 411–9.
Lee JY, Kim JH, Palmiter RD, Koh JY. Zinc released from metallothionein- Wang X, McCoy PA, Rodriguiz RM, Pan Y, Je HS, Roberts AC, et al.
iii may contribute to hippocampal CA1 and thalamic neuronal death Synaptic dysfunction and abnormal behaviors in mice lacking major
following acute brain injury. Exp Neurol 2003; 184: 337–47. isoforms of Shank3. Hum Mol Genet 2011; 20: 3093–108.
Lin DD, Cohen AS, Coulter DA. Zinc-induced augmentation of excitatory Wei G, Hough CJ, Li Y, Sarvey JM. Characterization of extracellular
synaptic currents and glutamate receptor responses in hippocampal accumulation of Zn2 + during ischemia and reperfusion of hippocam-
CA3 neurons. J Neurophysiol 2001; 85: 1185–96. pus slices in rat. Neuroscience 2004; 125: 867–77.
Lin W, Mohandas B, Fontaine CP, Colvin RA. Release of intracellular Wilson HL, Wong AC, Shaw SR, Tse WY, Stapleton GA, Phelan MC,
Zn(2 + ) in cultured neurons after brief exposure to low concentrations et al. Molecular characterisation of the 22q13 deletion syndrome sup-
of exogenous nitric oxide. Biometals 2007; 20: 891–901. ports the role of haploinsufficiency of SHANK3/PROSAP2 in the major
Manning MA, Cassidy SB, Clericuzio C, Cherry AM, Schwartz S, neurological symptoms. J Med Genet 2003; 40: 575–84.
Hudgins L, et al. Terminal 22q deletion syndrome: a newly recognized Won H, Lee HR, Gee HY, Mah W, Kim JI, Lee J, et al. Autistic-like social
cause of speech and language disability in the autism spectrum. behaviour in Shank2-mutant mice improved by restoring NMDA
Pediatrics 2004; 114: 451–7. receptor function. Nature 2012; 486: 261–5.
Maret W. The function of zinc metallothionein: a link between cellular Yasuda H, Yoshida K, Yasuda Y, Tsutsui T. Infantile zinc deficiency:
zinc and redox state. J Nutr 2000; 130: 1455–8. association with autism spectrum disorders. Sci Rep 2011; 1: 129.

S-ar putea să vă placă și