Sunteți pe pagina 1din 13

Biological

Psychiatry Archival Report

Rett-like Severe Encephalopathy Caused by a


De Novo GRIN2B Mutation Is Attenuated by
D-serine Dietary Supplement

ED
David Soto, Mireia Olivella, Cristina Grau, Judith Armstrong, Clara Alcon, Xavier Gasull,
Macarena Gómez de Salazar, Esther Gratacòs-Batlle, David Ramos-Vicente,
Víctor Fernández-Dueñas, Francisco Ciruela, Àlex Bayés, Carlos Sindreu, Anna López-Sala,
Àngels García-Cazorla, and Xavier Altafaj

ABSTRACT

CT
BACKGROUND: N-Methyl-D-aspartate receptors (NMDARs) play pivotal roles in synaptic development, plasticity,
neural survival, and cognition. Despite recent reports describing the genetic association between de novo mutations
of NMDAR subunits and severe psychiatric diseases, little is known about their pathogenic mechanisms and potential
therapeutic interventions. Here we report a case study of a 4-year-old Rett-like patient with severe encephalopathy
carrying a missense de novo mutation in GRIN2B(p.P553T) coding for the GluN2B subunit of NMDAR.
METHODS: We generated a dynamic molecular model of mutant GluN2B-containing NMDARs. We expressed the
mutation in cell lines and primary cultures, and we evaluated the putative morphological, electrophysiological, and
synaptic plasticity alterations. Finally, we evaluated D-serine administration as a therapeutic strategy and
translated it to the clinical practice.
RESULTS: Structural molecular modeling predicted a reduced pore size of mutant NMDARs. Electrophysiological
RA
recordings confirmed this prediction and also showed gating alterations, a reduced glutamate affinity associated with
a strong decrease of NMDA-evoked currents. Moreover, GluN2B(P553T)-expressing neurons showed decreased
spine density, concomitant with reduced NMDA-evoked currents and impaired NMDAR-dependent insertion of
GluA1 at stimulated synapses. Notably, the naturally occurring coagonist D-serine was able to attenuate
hypofunction of GluN2B(p.P553T)-containing NMDARs. Hence, D-serine dietary supplementation was initiated.
Importantly, the patient has shown remarkable motor, cognitive, and communication improvements after
17 months of D-serine dietary supplementation.
CONCLUSIONS: Our data suggest that hypofunctional NMDARs containing GluN2B(p.P553T) can contribute to Rett-
like encephalopathy and that their potentiation by D-serine treatment may underlie the associated clinical
improvement.
Keywords: De novo mutation, D-serine, Glutamatergic neurotransmission, Neuropsychiatric disorders, NMDA
T

receptor, Severe encephalopathy


http://dx.doi.org/10.1016/j.biopsych.2017.05.028
RE

Rett syndrome (RTT) (Online Mendelian Inheritance in Man mutations of RTT-associated genes and/or different causative
[OMIM]: 312750) is a neurodevelopmental disorder (NDD) genes.
affecting 1 in 10,000 live female births (1,2). Girls with RTT A growing number of genetic and functional studies are
seem to normally develop until 6 to 18 months and regress unraveling the complex scenario and molecular players
thereafter. Clinical manifestations include microcephaly, loss involved in NDD and, in particular, in Rett-like syndrome.
of achieved psychomotor abilities, intellectual disability (ID), Notably, it has been shown that the dysregulation of synaptic
and autistic behaviors (3). While most typical RTT cases harbor proteins can lead to NDD (6–8). Genes encoding for the
loss-of-function mutations in X-linked gene encoding methyl- N-methyl-D-aspartate receptor (NMDAR) could play critical
CpG binding protein 2 (MECP2) (4), mutations in CDKL5 and roles in the dysfunction of glutamatergic transmission associ-
FOXG1 genes have been identified in RTT variants (5). Rett-like ated with RTT. NMDARs belong to the ionotropic group of
syndrome mostly affects patients exhibiting symptoms that are glutamate receptors (iGluRs). Functionally, NMDARs play
similar to those seen in RTT. However, the genetic and mo- critical roles in neurogenesis, synaptogenesis, and synaptic
lecular etiology of this rare disease pointed out different plasticity processes. Early in development, NMDAR subunit

160 ª 2017 Society of Biological Psychiatry.


Biological Psychiatry January 15, 2018; 83:160–172 www.sobp.org/journal ISSN: 0006-3223
Biological
D-serine Treatment of Pathogenic GRIN2B De Novo Mutation Psychiatry

GluN2B expression is particularly high (9). Accordingly, it has impairment of social interaction, and no interest in the envi-
been proposed that GRIN2B gene disturbance might dramat- ronment. Along with these alterations, the patient showed high
ically compromise critical steps of neuronal, synaptic, and irritability together with sleeping disturbances. Considering
brain circuitry development. In agreement with this, animal these symptoms, together with the presence of handwashing
models with altered Grin2b expression exhibit neuronal stereotypies, the girl was tentatively diagnosed with RTT

ED
dysfunction (10). Moreover, discrete de novo mutations of phenotype, but further studies and disease progression lead to
GRIN2B gene have been recently associated with NDD the diagnosis of a Rett-like phenotype. Cytogenetic analysis,
(11–13), including early infantile epileptic encephalopathy-27 brain magnetic resonance imaging, and neurometabolic anal-
(EIEE27) (OMIM: 616139) (14) and autosomal dominant mental ysis did not show alterations. At 2.5 years old, she was less
retardation (MRD6) (OMIM: 613970) (15–17). Therefore, irritable and developed the capacity to hold up her head.
understanding the functional impact of discrete mutations Behaviorally, she slightly improved in social interaction, and 1
leading to NDD is fundamental to the development of precise year later her sleeping pattern was ameliorated. At that age, the
therapeutic strategies. electroencephalogram indicated the presence of epileptiform
In the current work, we report the identification of a de novo alterations of brain activity, and she was treated with valproic
missense mutation in the GRIN2B gene in a Rett-like patient acid; later, the treatment was changed to levetiracetam to
with severe encephalopathy. Functional studies showed that prevent increase in irritability. At 5 years old, the patient’s

CT
channel gating is altered in mutant NMDARs and that this adaptive behavior was assessed by the Vineland test, with
mutation drastically reduces synaptic NMDA-mediated cur- scores indicative of a mental age below 1 year old (Table 1).
rents. Importantly, dietary supplementation with D-serine—an
NMDAR coagonist (18,19)—during 17 months ameliorated Genetic Studies
the intellectual, communication, and motor deficits of the The patient was tentatively diagnosed with RTT-like pheno-
patient. These results support the pathogenicity of GRIN2B type. However, no mutations of RTT candidate genes (MECP2,
mutation and suggest that enhancing NMDAR activity using CDKL5, and FOXG1) were detected, and whole-exome
D-serine dietary supplement can have therapeutic benefits in sequencing was performed from the trio (21). Following ge-
certain NDDs. netic data filtering (Supplement), a missense de novo hetero-
zygous mutation of GRIN2B(p.P553T), coding for the GluN2B
RA
METHODS AND MATERIALS subunit of NMDARs, was identified (Figure 1A).

Patient Neurodevelopmental Assessment/Adaptive Molecular Modeling of the Mutant (GluN1)2–


Behavior and Dietary D-serine Supplement (GluN2B(P553T))2 Receptor
The study and D-serine dietary supplement (500 mg/kg/day; To identify the structural changes induced by
three divided doses during the day) was approved by the GluN2B(p.P553T) mutation, the molecular models of wild-type
appropriate informed consent of the patient’s parents. The and mutant receptors were generated by homology modeling
Vineland Adaptive Behavior Scale-II (20) semistructured inter- and molecular dynamics simulation. The GluN2B(P553) res-
view, allowing the assessment of four domains of adaptive idue is located at the beginning of the transmembrane helix 1
behavior (communication, daily living skills, socialization, and (TMH1; Figure 1B). This model suggested a role for P553 in
motor skills), was conducted by a trained neuropsychologist breaking TMH1 and bending TMH1 toward TMH3 (Figure 2C,
T

before (at 5 years 10 months old) and after (at 7 years 3 months upper panel), hence allowing P553 interaction with F653
old) 17 months of D-serine supplementation. (TMH3) of the GluN2B subunits. In addition, according to this
model, the residue P557 (TMH1) is also bending TMH1 to
In Silico, Biochemical, Cellular, and
Electrophysiological Studies
RE

In silico studies and experiments in cell lines and primary Table 1. Clinical Manifestations of the Patient Harboring De
Novo GRIN2B(p.P553T) Mutation Before and After 17
neuronal cultures were performed as fully detailed in the
Months of D-serine Dietary Supplement
Supplement, which also includes a description of the statistical
methods. Clinical Manifestations
After D-serine Dietary
Initial Assessment Supplement
RESULTS (5 Years 10 Months Old) (7 Years 3 Months Old)
Patient Clinical Information Behavior Irritability Reactive smiling
Poor visual contact Establishment of visual contact
The patient is a girl who was born after an uneventful preg- No gestural communication Some gestural communication
nancy. Prior to the patient’s birth, the familial history was Disconnected and lost look Interested in surroundings
negative for NDD, with nonconsanguineous parents and a Motor Inability to sit down Autonomy to sit down
healthy sister. She was referred to our clinic at 1 year old, and Clasping hands Walking with orthopedic
the primary clinical examination showed a psychomotor delay Severe hypotonia walker
with severe hypotonia (with the presence of osteotendinous Sleep Altered sleeping pattern Improved sleeping pattern
reflexes and devoid of pyramidal signs), inability to hold the EEG Epileptiform alterations Epileptiform alterations
head and to be seated with no support. Behaviorally, she had without seizures without seizures
an overall absent affect, as manifested by poor visual contact, EEG, electroencephalogram.

Biological Psychiatry January 15, 2018; 83:160–172 www.sobp.org/journal 161


Biological
Psychiatry D-serine Treatment of Pathogenic GRIN2B De Novo Mutation

Figure 1. Identification of GRIN2B(p.P553T) mu-


tation associated with the case study and predicted
structural consequences. (A) (Left panel) Pedigree of
the family showing the absence of clinical manifes-
tations of the progenitors and the sister of the

ED
patient. (Right panels) Chromatograms of the com-
plementary DNA sequence of GRIN2B showing the
GRIN2B(c.1657C.A) mutated nucleotide (indicated
by an arrow) using forward and reverse oligonucle-
otides. (B) Structure of heterotetrameric (GluN1)2–
(GluN2B)2 N-methyl-D-aspartate receptor (Protein
Data Bank ID: 4PE5), according to Karakas and
Furukawa (41), showing the N-terminal domain
(NTD), the ligand binding domain (LBD), and the
transmembrane domain (TMD, containing the
mutated amino acid P553T). (Lower panel) Magnifi-
cation of the TMD, showing the topological position
of Pro553 residue at the beginning of trans-

CT
membrane helix 1 (TMH1, in green) of the TMD,
facing TMH3 (in blue). (C) (Upper panel) Top view of
the TMD structural molecular model of wild-type
(GluN1)2–(GluN2B)2 receptor (from the extracellular
domain). The model predicts that P553 residue (in
orange) of TMH1–GluN2B (in green) is bending
TMH1 toward TMH3 (in dark blue), allowing the
interaction with F653 residue (in light blue) located in
TMH3. The interaction between TMH3 and TMH1
brings TMH3 out of the center of the pore channel.
(Lower panel) Top view of the TMD structural mo-
lecular model of mutant (GluN1)2–(GluN2B(P553T))2
RA
receptor (from the extracellular domain). The model
predicts that T553 residue (in green) of TMH1–
GluN2B is not able to bend TMH1, abolishing the
interaction with F653 (located in TMH3, light blue).
According to this model, the breaking of the inter-
action between TMH3 and TMH1 brings TMH3
toward the center of the pore, inducing a reduction of
the channel pore. (D) GluN2B protein sequence
alignment around residues P553 and F653. Representative sequences from a larger alignment containing 147 proteins from 12 metazoan species spanning
seven phyla are shown. Displayed protein sequences are from the following species: Homo sapiens GluN2B (Uniprot Ref. Q13224), Mus musculus GluN2B
(Uniprot Ref. Q01097), Danio rerio GluN2Bb (Ensembl Ref. ENSDARG00000030376), Branchiostoma belcheri 254360R.t1 (from the database B.belcher-
i_v18h27.r3_ref_protein included in LanceletDB Genome browser, Sun Yat-sen University), Strigamia maritima SmarNMDAR2b, Apis melifera GB48097,
Capitela teleta CapteT179505, and Lottia gigantea LotgiT137890 (all from Ensemble Metazoa).
T

TMH3 through its proline kink, allowing the interaction with This high conservation strongly supports a critical role of
F654 (TMH3) of the GluN1 subunit. Because the TMH3 of GluN GluN2B(P553) in NMDAR channel activity. These data
subunits contributes to the ion channel pore, our molecular prompted us to further assess the molecular and biophysical
RE

model strongly suggests that the TMH3–TMH1 interaction consequences of the GluN2B(P553T) mutation.
would bring TMH3 out of the center of the channel pore.
Importantly, this model predicts that GluN2B(P553T) mutation
would prevent TMH1 breaking and bending, Consequently, Heteromerization and Trafficking of GluN1–
TMH1–TMH3 interaction would be disrupted (Figure 2C, lower GluN2B(P553T) Receptors
panel), which in turn would bring TMH3 closer to the center of To assess the effects of GluN2B(P553T) on NMDAR oligo-
the pore and potentially reduce its size or alter the gating merization and trafficking, we cotransfected HEK-293T cells
properties of mutant receptor. with GluN1 subunit and either green fluorescent protein (GFP)–
Based on this structural model, we investigated the evolu- GluN2Bwt or GFP–GluN2B(P553T). Biochemical analysis
tionary conservation of GluN2B proline 553 and its predicting showed that GluN2B(P553T) protein levels were similar to
interacting residue GluN2B phenylalanine 653. Indeed, the those of GluN2Bwt (Figure 2A). Furthermore, coimmunopreci-
GluN2B(F653) and GluN1(F654) residues belong to the pitation experiments showed the presence of GluN1 and
SYTANLAAF motif, the most highly conserved motif among hemagglutinin–GluN2A subunits in anti-GFP pulled-down
mammalian iGluRs (22). Multiple sequence alignment of 147 complexes, indicating that GluN2B(P553T) subunit interacts
metazoan iGluRs showed extremely high conservation of these with GluN1 and/or GluN2A, similarly to GluN2Bwt (Figure 2B).
residues (Figure 1D). Indeed, the proline in position 553 was Surface expression of wild-type or mutant GFP–GluN2B was
observed in 144 of the 147 iGluR sequences investigated. also evaluated in COS-7 cells and in mouse primary cortical
Similarly, phenylalanine 653 is conserved in 131 of all iGluRs. neurons. Immunofluorescent analysis showed the ability of

162 Biological Psychiatry January 15, 2018; 83:160–172 www.sobp.org/journal


Biological
D-serine Treatment of Pathogenic GRIN2B De Novo Mutation Psychiatry

Figure 2. Protein interactions and cellular


trafficking of GluN2Bwt- and GluN2B(P553T)-
containing N-methyl-D-aspartate receptors. (A)
Western blot analysis of heterologous expression
of wild-type (first lane) and (P553T) mutant green

ED
fluorescent protein (GFP)–GluN2B (second lane) in
equal amounts of proteins (actin loads) extracted
from transiently transfected HEK-293T cells. (B)
Representative Western blot analysis of coimmu-
noprecipitation (co-IP) experiments from HEK-293T
cells transiently cotransfected with hemagglutinin
(HA)–GluN2A, GluN1, and either GFP–GluN2Bwt or
GFP–GluN2B(P553T). The coimmunoprecipitation
was performed either with anti-GFP (pulling
down GFP–GluN2B construct products and their
interacting proteins) or immunoglobulin G (IgG)
(negative control). Interaction studies showed the
ability of GFP-tagged GluN2B(P553T) subunit to

CT
interact with HA–GluN2A and GluN1 subunits,
similarly to GluN2Bwt. Bar graph represents the
densitometric analysis of immunoprecipitated pro-
teins (relative to immunoprecipitated GFP–GluN2B
for GluN2A and GluN1 subunits; relative to GFP–
GluN2B load for GluN2B subunit) [mean 6 SEM
densitometric of three independent experiments;
white bars: GluN2Bwt condition; red bars:
GluN2B(P553T) condition]. (C) Left panel: Immu-
nofluorescent analysis of cell surface expression
(red channel) and total expression (green channel)
of wild-type (left panels) and mutant GFP–GluN2B
RA
(right panels) in COS-7 cells. Right panel: Bar
graph representing the relative surface to total
expression of GFP–GluN2B constructs in COS-7
cells (n = 35–38 cells per condition, from three
independent experiments; **p = .007, Student’s
t test). (D) Time course analysis of GFP–GluN2B
cell surface expression (green channel) and intra-
cellular expression (red channel) of wild-type
(left panels) and mutant GFP–GluN2B (right
panels) in primary cortical neuronal cultures
transiently transfected. Primary cultures were
transfected at day in vitro 4 (DIV4) or DIV7, and
immunofluorescence analysis of surface (green
channel) and intracellular (red channel) expression
T

was performed at DIV6, DIV11, and DIV16.


Bottom: Bar graph showing the ability of mutant
GluN2B(P553T)-containing N-methyl-D-aspartate
receptors to reach the plasma membrane of the
dendritic processes (insets) of transfected cortical
neurons (n = 25–62 cells per condition, from three or four independent experiments; *p = .026; ns, nonsignificant; Student’s t test). Scale bar = 40 mm
RE

(low magnification) or 3 mm (dendrites).

mutant GFP–GluN2B(P553T) to reach COS-7 plasma transfected HEK-293T cells. Current amplitudes were drasti-
membrane (100 6 10.3 vs. 146.6 6 13.1 in GFP–GluN2Bwt/ cally reduced in cells expressing GluN1–GluN2B(P553T)
P553T-expressing cells; Figure 2C). In a neuronal context, receptors compared with GluN1–GluN2B-expressing cells
GluN2B(P553T) dendritic surface/intracellular ratio expression [280.56 6 14.25 pA/pF for GluN2Bwt vs. 215.49 6 3.16
was normal at day in vitro 7 (DIV7) and DIV11, with a slight pA/pF for GluN2B(P553T); Figure 3A–C]. The GluN2B(p.P553T)
decrease at DIV16 (100 6 2.6 vs. 92.0 6 1.6 GFP–GluN2Bwt/ mutation spared the voltage-dependent channel block by
P553T-transfected primary cortical neurons; Figure 2D). extracellular Mg21 (Figure 3D–F). Because GluN2B(P553T)
mutation is located in the vicinity of the agonist binding site
and the channel pore, we investigated possible effects of the
Electrophysiological Changes in GluN2B(P553T) mutation on channel kinetics. Electrophysiological recordings
Subunit-Containing NMDARs showed a significantly faster deactivation rate in mutant
Patch-clamp experiments were performed to evaluate the receptors [0.60 6 0.06 seconds for GluN2Bwt vs. 0.26 6 0.03
biophysical properties of GluN2B(P553T)-containing NMDARs. seconds for GluN2B(P553T); Figure 4A, B] and faster desen-
Following fast application of glutamate (1 mM) and glycine (50 sitization quantified on 5-second-duration jumps [1.87 6 0.33
mM), whole-cell NMDA-mediated currents were evoked in seconds for GluN2Bwt vs. 0.79 6 0.22 seconds for

Biological Psychiatry January 15, 2018; 83:160–172 www.sobp.org/journal 163


Biological
Psychiatry D-serine Treatment of Pathogenic GRIN2B De Novo Mutation

ED
CT
RA
Figure 3. GluN2B(P553T) mutation reduces N-methyl-D-aspartate receptor (NMDAR)-mediated whole-cell currents without affecting magnesium block.
(A) Representative whole-cell currents evoked by rapid application of 1 mM glutamate plus 50 mM glycine (0.5 seconds duration; 260 mV) for HEK-293T cells
expressing GluN1–GluN2B (black trace) or GluN1–GluN2B(P553T) NMDARs (red trace). Note that currents for GluN2B(P553T)-containing receptors were notably
smaller even when both cells displayed a similar membrane capacitance (5.5 and 7.0 pF). (B) Average of raw peak currents for GluN2B- and GluN2B(P553T)-
expressing cells with single experiment values superimposed (open circles; p = .0003, Mann-Whitney U test, n = 19 and 21, respectively). (C) Normalized peak
currents (2pA/pF) for GluN2B- and GluN2B(P553T)-expressing cells with single experiment values superimposed (open circles; p , .0001, Mann-Whitney U test,
n = 19 and 21, respectively). (D) Traces recorded at 260 mV for a GluN2B(P553T)-expressing cell showing the characteristic magnesium block of NMDARs. (E)
Percentage of current block at 260 mV by Mg21 (1mM) for GluN2Bwt- and GluN2B(P553T)-containing receptors. Single-value experiments are denoted as open
circles for each condition [p = .0732, Mann-Whitney U test, n = 5 and 7 for GluN2B and GluN2B(P553T) respectively]. (F) Current–voltage relationship for GluN2B-
and GluN2B(P553T)-containing receptors where it can be observed that Mg21 blocks similarly both NMDARs at the whole range of negative potentials. ***p , .001;
****p , .0001; n.s., nonsignificant.
T

GluN2B(P553T); Figure 4C, D]. Moreover, in agreement GluN1–GluN2B(P553T)-expressing cells (14.9 6 3.25% at 10
with aforementioned modeling predictions, nonstationary mM, 43.4 6 11.08% at 100 mM, and 32.3 6 7.12% at 300 mM,
fluctuation analysis (23) showed a reduction of the single- ps = .006, .004, and .001, respectively; Figure 5) (p = .022 at
channel conductance in mutant receptors [49.73 6 2.61 pS 100 mM D-serine between mutant and wild-type receptors;
for GluN2Bwt vs. 39.15 6 2.60 pS for GluN2B(P553T);
RE

Figure 5B). This differential potency of D-serine might result


Figure 4E, F]. Together with biochemical data indicating normal from a reduced affinity for GluN1–GluN2B(P553T), leading to
expression and oligomerization, we conclude that GluN1– the enhanced potentiation and faster deactivation/desensiti-
GluN2B(P553T) receptors are intrinsically hypofunctional. zation rates at higher concentrations. Alternatively, because
glutamate binding increases the dissociation rate of glycine/
D-serine coagonist with NMDARs (24), seeming changes in
D-serine Administration Enhances NMDA Currents D-serine potency might be explained by an altered glutamate
in GluN1–GluN2B(P553T) Receptors affinity. Concentration–response experiments showed no
Next, we sought to enhance activity of mutant NMDARs by changes in D-serine EC50 [0.51 mM for GluN2Bwt vs. 0.66 mM
using compounds with unlikely side effects or impaired as a for GluN2B(P553T); Figure 5C], while the EC50 for glutamate
result of the GluN2B(P553T) mutation. D-serine, a naturally increased by sevenfold in GluN1-GluN2B(P553T) receptors
occurring coagonist of the NMDAR, may fulfill both criteria. compared with wild-type receptors [3.09 mM for GluN2Bwt vs.
In agreement with previous reports (19), D-serine administra- 20.94 mM for GluN2B(P553T); Figure 5D].
tion dose-dependently increased GluN1–GluN2Bwt-mediated
current density in HEK-293T cells (13.3 6 4.61% increase at Biophysical Alterations in Triheteromeric GluN1–
100 mM and 16.9 6 3.97% increase at 300 mM, ps = .018 and GluN2A–GluN2B(P553T) Receptors
.002, respectively; Figure 5). By comparison, NMDA-evoked GluN2B subunit interacts with different GluN subunits, forming
currents were more strongly potentiated by D-serine in both (GluN1)2–(GluN2B)2 heterodimers (prominent NMDARs at

164 Biological Psychiatry January 15, 2018; 83:160–172 www.sobp.org/journal


Biological
D-serine Treatment of Pathogenic GRIN2B De Novo Mutation Psychiatry

Figure 4. Faster deactivation and lower single-


channel conductance contribute to GluN2B(P553T)
loss of function. (A) Peak-scaled representative re-
sponses to 1 mM glutamate plus 50 mM glycine (0.5
second agonist application; 260 mV) for GluN2B

ED
(black trace) and GluN2B(P553T) (red trace), showing
a faster inactivation of GluN2B(P553T)-containing N-
methyl-D-aspartate receptors following coagonist
removal. (B) Average deactivation time constant
(fitted to a double exponential) fitted from tail cur-
rents for GluN2B and GluN2B(P553T). P553T muta-
tion accelerates channel closure after coagonists
unbind [0.60 6 0.06 seconds for GluN2B vs. 0.26 6
0.03 seconds for GluN2B(P553T), p , .0001; Mann-
Whitney U test; n = 16 and 17, respectively]. Single-
experiment values are shown as open circles for
each condition. (C) Peak-scaled representative re-
sponses to 1 mM glutamate plus 50 mM glycine (long

CT
jumps of 5 seconds duration; 260 mV) for GluN2B
(black trace) and GluN2B(P553T) (red trace), where it
can be observed the faster desensitization of the
current mediated by GluN2B(P553T) in the presence
of coagonists. (D) Desensitization weighted time
constant for GluN2Bwt and GluN2B(P553T), showing
enhanced desensitization in GluN2B(P553T)-
containing N-methyl-D-aspartate receptors. Single-
experiment values are shown as open circles for
each condition [1.87 6 0.33 seconds for GluN2B vs.
0.79 6 0.22 seconds for GluN2B(P553T), p = .0054,
Mann-Whitney U test, n = 14 for both). (E) Whole-cell
RA
current activated by rapid application of 1 mM
glutamate plus 50 mM glycine (0.5 second; 260 mV)
from a cell expressing GluN2Bwt (left panel) or
GluN2B(P553T) (right panel). Gray traces represent
single responses, and black lines are the averages of
69 and 33 responses for wild-type and mutant
GluN2B, respectively. Insets denote the current
variance versus mean current plot calculated from
the deactivating tail current for this cell. The
weighted mean single-channel conductance (g)
values estimated from nonstationary fluctuation
analysis for these recording cells were 53.9 pS (GluN2Bwt) and 36.8 pS [GluN2B(P553T)]. (F) Bar graph showing that single-channel conductance values are
decreased in GluN2B(P553T)-containing N-methyl-D-aspartate receptors [49.73 6 2.61 pS for GluN1–GluN2B vs. 39.15 6 2.60 pS for GluN1–GluN2B(P553T),
p = .0209, Mann-Whitney U test; n = 12 and 9, respectively]. Single cells are shown as open circles superimposed to bar graph. *p , .05; **p , .01;
T

****p , .0001.

initial developmental stages) and (GluN1)2–GluN2A–GluN2B effects observed in the mutant (GluN1)2–(GluN2B(P553T))2
triheteromers. Both (GluN1)2–(GluN2B)2 and (GluN1)2–
RE

diheterodimers.
GluN2A–GluN2B receptor complexes exist in adult hippo-
campus and cortex (25). Thus, we explored whether the
GluN2B(P553T) Overexpression Decreases Spine
GluN2B(P553T) mutation may also impair the subpopulation
of triheteromeric receptors or, instead, its deleterious ef- Density and Reduces Excitatory Postsynaptic
fects might be mitigated when coassembled with GluN2A Currents
subunit. To record triheteromeric-mediated NMDA currents, To evaluate the effect(s) of the GluN2B(P553T) mutation in
HEK-293T cells were transfected with GluN constructs central neurons, we overexpressed GluN2B(P553T) in primary
harboring ectopic retention signals (26). NMDA current hippocampal neurons and measured both morphological
amplitudes were not significantly reduced in parameters and NMDAR-mediated synaptic currents. Sholl
GluN2B(P553T)-containing triheteromers (Figure 6A, B). analysis of dendrites labeled with GFP–GluN2B(P553T) or
However, administration of 100 mM D-serine potentiated GFP–GluN2Bwt indicated similar distributions of the subunit
GluN2B(P553T)-containing triheteromers more strongly than across the dendritic arbor (F = 0.71, p = .884; Figure 7A).
wild-type ones (30.59 6 4.20% for GluN2A–GluN2Bwt vs. Interestingly, however, the spine density was significantly
54.76 6 10.10% for GluN2A–GluN2B(P553T); Figure 6A–C). reduced in neurons expressing GluN2B(P553T) [21.1 6 1.6
Likewise, the desensitization and deactivation kinetics of vs. 13.1 6 1.0 spines/10 mm dendrite for GluN2Bwt and
triheteromeric mutants were increased compared with GluN2B(P553T), respectively, p = .0002; Figure 7A]. The
control subjects (Figure 6D–H), recapitulating some of the reduction in spine density mainly resulted from a decrease in

Biological Psychiatry January 15, 2018; 83:160–172 www.sobp.org/journal 165


Biological
Psychiatry D-serine Treatment of Pathogenic GRIN2B De Novo Mutation

ED
CT
T RA
RE

Figure 5. N-Methyl-D-aspartate receptor coactivation by D-serine is enhanced in GluN2B(P553T)-containing receptors. (A) Representative traces evoked by
physiological concentrations of 1 mM glutamate plus 1 mM glycine (0.5 second; 260mV) from GluN2Bwt- or GluN2B(P553T)-expressing HEK-293T cells, either in the
absence (black traces) or in the presence (red traces) of D-serine. The coagonist D-serine was applied at different concentrations (1 mM: upper panel; 10 mM: second
panel from top; 100 mM: second panel from bottom; 300 mM: lower panel). Note that GluN2B(P553T)-mediated currents are smaller and have faster deactivation
kinetics than GluN2B-mediated currents. (B) Average of normalized peak currents evoked by 1 mM glutamate 1 1 mM glycine at different D-serine concentrations
(gray scale colored bars) compared with absence of D-serine (white bar: 100% of the response). Although D-serine was able to enhance evoked currents for both
GluN2B and GluN2B(P553T) at high concentrations, the modulatory effect was greater for GluN2B(P553T)-containing receptors, whose currents were significantly
increased at 10 mM D-serine compared with GluN2B. (*p , .05 and **p , .01 for comparisons between different D-serine concentrations within same receptor type;
#
p , .05 for comparisons between GluN2B and GluN2B(P553T) at the same D-serine concentration). Numbers inside the bars denote the number of recordings for
each condition. (C) D-serine concentration–response curves constructed from responses to 1 mM glutamate plus the specified D-serine concentration in the absence
of glycine. GluN2Bwt- and GluN2B(P553T)-containing N-methyl-D-aspartate receptors expressed in HEK-293T cells showed similar EC50 values [0.51 mM for
GluN2B vs. 0.66 mM for GluN2B(P553T); n = 5 for each construct]. (D) Glutamate concentration–response curves constructed from responses to 1 mM D-serine plus
the specified glutamate concentration in the absence of glycine of GluN2Bwt- and GluN2B(P553T)-containing N-methyl-D-aspartate receptors. GluN2B(P553T)
mutation produces an approximately sevenfold increase in EC50 value for glutamate [3.09 mM for GluN2B vs. 20.94 mM for GluN2B(P553T); n = 5 and 7, respectively].

166 Biological Psychiatry January 15, 2018; 83:160–172 www.sobp.org/journal


Biological
D-serine Treatment of Pathogenic GRIN2B De Novo Mutation Psychiatry

ED
CT
RA
Figure 6. Altered biophysical properties of triheteromeric GluN1–GluN2A–GluN2B(P553T) N-methyl-D-aspartate receptors (NMDARs). (A) Representative
traces evoked by 1 mM glutamate plus 1 mM glycine (0.5 second; 260 mV) in HEK-293T cells expressing triheteromeric GluN1–GluN2A–GluN2Bwt (2A-2B, left
traces) or GluN1–GluN2A-2B(P553T) [2A-2B(P553T), right traces] in the absence (black traces) or presence (red traces) of 100 mM D-serine. (B) Normalized
peak currents (2pA/pF) for GluN2A-2B- and GluN2A-2B(P553T)-expressing cells with single-experiment values superimposed (open circles; p . .05, Mann-
Whitney U test, n = 18 and 13, respectively). (C) Percentage of current increment in the presence of 100 mM D-serine for GluN2A-2B- and GluN2A-2B(P553T)-
expressing cells with single-experiment values superimposed (open circles; p , .05, Mann-Whitney U test, n = 14 and 13, respectively). (D) Representative
whole-cell currents evoked by 1 mM glutamate plus 1 mM glycine from HEK-293T cells expressing GluN2A-2B (black trace) or GluN2A-2B(P553T) (red trace),
showing the faster kinetics of triheteromeric NMDARs containing GluN2B(P553T) subunit. Responses have been peak scaled for comparison purposes.
(E) Magnification of the tail-deactivating currents (shown in panel D) after glutamate and glycine removal. Traces (aligned at the coagonist removal time point)
showed a faster deactivating kinetics of mutant triheteromeric NMDARs (0.14 second for depicted cell, red trace) compared with GluN2Bwt-containing tri-
heteromers (0.29 second for depicted cell, black trace). Fits are shown overlapped in green. (F) Magnification of triheteromeric NMDAR desensitization (shown
in panel D). Both currents have been aligned at the peak current. GluN2B(P553T)-containing receptors show a faster desensitization time constant
(0.12 second for depicted cell, red trace) compared with GluN2Bwt-containing triheteromers (0.38 second for depicted cell, black trace). Fits are shown in
T

green. (G) Bar graph representing the deactivation time constant fitted from tail currents for triheteromeric GluN2A-2B and GluN2A-2B(P553T). GluN2B(P553T)
mutation accelerates channel closure after coagonist removal [0.30 6 0.02 second for GluN2A-2B vs. 0.21 6 0.02 second for GluN2A-2B(P553T), p , .01,
Mann-Whitney U test, n = 17 and 14, respectively]. Single-experiment values are shown as open circles for each condition. (H) Bar graph representing the
average desensitization time constant for GluN2A-2B- and GluN2A-2B(P553T)-expressing cells, measured in the presence of the agonists (as shown in panel
F). P553T mutation accelerates desensitization of triheteromeric NMDARs [0.30 6 0.02 second for GluN2A-2B vs. 0.15 6 0.02 second for GluN2A-2B(P553T),
RE

p , .0001, Mann-Whitney U test, n = 17 and 14, respectively]. Single-experiment values are shown as open circles for each condition. *p , .05; **p , .01;
****p , .0001; n.s., nonsignificant.

the number of thin and mushroom spines [8.6 6 1.0 vs. GluN2B(P553T) respectively, p = .0002; Supplemental
3.5 6 0.5 for thin spines/10 mm dendrite and 5.8 6 0.4 vs. Figure S1). Overall, these morphological and molecular
3.5 6 0.3 for mushroom spines/10 mm dendrite for GluN2Bwt changes indicate deficient spine development in hippocampal
and GluN2B(P553T), ps = .0001 and .0002, respectively; neurons expressing GluN2B(P553T).
Figure 7A], while the number of stubby spines remained un- Patch-clamp recordings revealed a decrease in the ampli-
affected [6.7 6 0.5 vs. 6.1 6 0.5/10 mm dendrite for tude of spontaneous excitatory postsynaptic currents (EPSCs)
GluN2Bwt and GluN2B(P553T), respectively; Figure 7A]. mediated by NMDARs in neurons overexpressing mutant
Together with the reduction in spine density, immunofluo- GluN2B(P553T) compared with GluN2Bwt [2593.62 6 59.58
rescence analysis of GluA1 (alpha-amino-3-hydroxy-5- pA for GluN2Bwt vs. 2212.98 6 48.27 pA for GluN2B(P553T);
methyl-4-isoxazole propionic acid receptor [AMPAR] Figure 7B, top traces, and Figure 7C, left bar graph], directly
subunit) expression [overexpressed in RTT murine models demonstrating an effect of this mutation in the synaptic pop-
(27)] revealed a significant increase of GluA1 at DIV11 in ulation of NMDARs. Because D-serine administration
neurons overexpressing GluN2B(P553T) mutation (10.5 6 enhanced the activity of GluN2B(P553T)-containing NMDARs
1.7 vs. 23.3 6 2.6 arbitrary units (A.U.) for GluN2Bwt and in heterologous cells (see above), we assessed its effect

Biological Psychiatry January 15, 2018; 83:160–172 www.sobp.org/journal 167


Biological
Psychiatry D-serine Treatment of Pathogenic GRIN2B De Novo Mutation

Figure 7. GluN2B(P553T) mutation alters spine


morphology and decreases synaptic N-methyl-D-
aspartate receptor (NMDAR)-mediated currents in
hippocampal neurons. (A) Representative images of
primary hippocampal neurons transfected at day

ED
in vitro 7 with green fluorescence protein (GFP)–
GluN2Bwt (top image) or GFP–GluN2B(P553T) (bot-
tom image). Immunofluorescent detection of GFP–
GluN2B allowed the analysis of the dendritic arbori-
zation of GFP–GluN2Bwt- or GFP–GluN2B(P553T)-
expressing hippocampal neurons (upper and lower
panels, respectively). GFP–GluN2B immunode-
tection allowed the quantification of dendritic
complexity by Sholl analysis (upper left panel), which
is similar between neurons expressing GFP–
GluN2Bwt and GFP–GluN2B(P553T) (F = 0.71, p =
0.884, repeated-measures two-way analysis of vari-
ance and Bonferroni posttest, n = 16–18 transfected

CT
neurons per condition, from three independent
experiments). Immunolabeling of GFP–GluN2B
transfected neurons was used to visualize spines
(insets, indicated by yellow arrows) and to quantify
spine density and morphology (bar graph, bottom
right). Compared with GFP–GluN2Bwt, the expres-
sion of GFP–GluN2B(P553T) reduced spine density,
resulting from a significant reduction of mushroom-
and thin-type spines, while stubby spines were un-
affected (n = 11–14 dendrites per condition, from two
independent experiments; ***p , .001; ns, nonsig-
nificant; Student’s t test). (B) Representative traces
RA
from spontaneous activity-dependent NMDAR-
mediated excitatory postsynaptic current (EPSC)
recordings from hippocampal neuronal cultures.
EPSCs from neurons transfected with either wild-
type GluN2B (left traces) or mutant GluN2B(P553T)
(right traces) were recorded at 270 mV in the pres-
ence of NBQX (50 mM) and picrotoxin (10 mM) and in
the absence of TTX. The addition of 100 mM D-serine
(bottom traces) produced an increase in the fre-
quency of EPSCs (decrease in the interevent inter-
val). (C) Left: Mean amplitudes of NMDAR-mediated
EPSCs for GluN2Bwt and GluN2B(P553T) before
(basal) and after D-serine addition (n = 8 and 6
neurons, respectively). Right: Mean time of inter-
T

event intervals of NMDA-mediated EPSCs in primary


hippocampal neurons transfected with GFP–
GluN2Bwt or GFP–GluN2B(P553T), before and after
addition of D-serine (**p , .01 GluN2Bwt vs.
GluN2B(P553T), #p , .05 baseline vs. D-serine
RE

administration).

in primary neurons. The addition of 100 mM D-serine desensitization at higher frequencies, thereby masking a pu-
similarly increased EPSC frequency in GluN2Bwt- and tative effect on EPSC amplitudes. Indeed, the rate of recovery
GluN2B(P553T)-overexpressing neurons (measured as a from desensitization for NMDARs is quite slow, spanning
shortening of the inter-event interval; Figure 7B, bottom traces, several seconds (28).
and Figure 7C, right bar graph). Furthermore, the charge
transfer (pA*ms) selectively increased in GluN2B(P553T)-
D-serine Rescues Altered Activity-Induced
overexpressing neurons (1687 pA*ms before D-serine vs.
2210 pA*ms after D-serine, i.e., a 34% change) but not in Recruitment of AMPARs in GluN2B(P553T)-
neurons overexpressing GluN2Bwt (4089 pA*ms before D- Expressing Neurons
serine vs. 3979 pA*ms after D-serine, a 20.1% change) (data Sustained activation of functional NMDARs can lead to the
not shown). In contrast to heterologous cell line data, the insertion of surface AMPARs at synapses and consequently
addition of 100 mM D-serine did not increase the EPSC am- induce long-term potentiation (LTP) (29). Therefore, we
plitudes in either group (Figure 7C, left bar graph), perhaps due assessed the ability of neurons expressing GluN2B(P553T)-
to the recruitment of new synapses or to increased containing NMDARs to support this form of synaptic

168 Biological Psychiatry January 15, 2018; 83:160–172 www.sobp.org/journal


Biological
D-serine Treatment of Pathogenic GRIN2B De Novo Mutation Psychiatry

Figure 8. Glycine-induced chemical long-term


potentiation (Gly-cLTP) deficits of primary hippo-
campal neurons expressing GluN2B(P553T) muta-
tion are rescued by D-serine administration.
(A) Immunofluorescent analysis of surface expres-

ED
sion of the alpha-amino-3-hydroxy-5-methyl-4-
isoxazole propionic acid receptor GluA1 subunit in
murine primary hippocampal neurons at day in vitro
16, transiently transfected at day in vitro 11 with
green fluorescent protein (GFP)–GluN2Bwt (left
panels) and GFP–GluN2B(P553T) (right panels).
Transfected neurons (green channel) were analyzed
for the detection of surface GluA1 in dendritic pro-
cesses (red channel), under basal conditions (left),
after Gly-cLTP (middle), or following Gly-cLTP in the
presence of 100 mM D-serine (right). (B) Bar graph
representing GluA1 surface fluorescence intensity
(arbitrary units, compared with basal conditions) in

CT
murine primary hippocampal neurons transfected
with GFP–GluN2Bwt (light boxes) and GFP–
GluN2B(P553T) (dark boxes). Histograms represent
the average 6 SEM. Measurements were performed
with 14 to 40 dendrites per condition (30–40 spines
per dendrite; 3–7 neurons per condition; **p , .05,
***p , .001, Student’s t test).
RA
plasticity. Primary hippocampal cultures transfected with either months). Before treatment, the patient had few communication
GFP–GluN2Bwt or GFP–GluN2B(P553T) were stimulated with skills; had poor eye contact; did not pay attention to the
200 mM glycine in the absence of Mg21 (glycine-mediated activities of parents, teachers, and schoolmates; did not have
chemical LTP [Gly-cLTP]) or simultaneously with 200 mM verbal language (any word/sounds) but was able to wave
glycine plus 100 mM D-serine. Immunofluorescent analysis bye-bye (although with no pointing); did not engage in sym-
showed that Gly-cLTP significantly increased surface GluA1 bolic play; could sit without support but could not move from
levels at spine-like structures in GluN2Bwt-transfected neu- prone lying to the sitting position. The Vineland test scores
rons (100 6 5.6 A.U. baseline vs. 121.8 6 8.4 A.U. Gly-cLTP; (where standard score [SS] is mean = 100, SD = 15) before
T

Figure 8A, left panels, and Figure 8B). In contrast, the sur- treatment were as follows: communication, 42 SS; daily living
face levels of GluA1 were not significantly increased in neurons skills, 36 SS; socialization, 49 SS; motor skills, 31 SS.
expressing GluN2B(P553T) (100 6 10.6 A.U. baseline vs. After 17 months of treatment, the patient remarkably
112 6 9.5 A.U. Gly-cLTP; Figure 8A, right panels, and improved communication, social, and motor skills (Table 1).
RE

Figure 8B). Importantly, the defects observed in AMPAR Behaviorally, she is interested in faces and has persistent eye
recruitment in GluN2B(P553T)-expressing neurons could be contact, follows the surrounding human activities with interest,
rescued by simultaneous administration of glycine and is able to stretch the arms in order to be held and turn her head
D-serine (100.0 6 4.3 A.U. baseline vs. 127.0 6 6.0 A.U. when called, laughs at funny situations, and is in general
glycine 1 D-serine cLTP; Figure 8A, right panels, and happier. She can also imitate toys and animal sounds, creep
Figure 8B). These results indicate that D-serine can also on the floor and move from prone lying to the sitting position,
facilitate a major mechanism of NMDAR-dependent plasticity and move from sitting to standing position with support. She
in the context of GluN2B(P553T) mutation. has also improved her sleeping patterns, is able to sit down
without help, and (importantly) can walk with the assistance of
an orthopedic walker. The electroencephalogram shows
Dietary Supplement of D-serine Improves Motor epileptiform alterations, but clinical seizures were not present.
and Communication Skills of the Patient Carrying Despite pharmacological interventions (valproate first and
GluN2B(P553T) Mutation levetiracetam later), the epileptiform alterations detected in the
The partial restoration of mutant NMDAR currents by D-serine, electroencephalogram are still present; thus, these treatments
together with the lack of side effects of D-serine treatment (30), have been stopped. In summary, the Vineland test (devoid of
prompted us to start a dietary supplement treatment of the motor score that cannot be performed beyond 7 years old)
patient. At 5 years 10 months of age, the patient was admin- after D-serine treatment resulted in the following test scores:
istered D-serine (500 mg/kg/day) and continued to date (i.e., 17 communication, 48 SS; daily living skills, 48 SS; socialization,

Biological Psychiatry January 15, 2018; 83:160–172 www.sobp.org/journal 169


Biological
Psychiatry D-serine Treatment of Pathogenic GRIN2B De Novo Mutation

50 SS. Overall, the clinical assessment indicates attenuation of alterations of glutamatergic synapses might be involved in the
both cognitive and motor impairments. pathophysiology of the classical RTT condition.
In agreement with the proposed functional impact of de
novo mutations of GluN2B subunit, previous studies have
DISCUSSION associated de novo mutations of GRIN2B with severe pheno-

ED
NMDARs are critical players of glutamatergic neurotransmis- typic alterations (6,14,15,33). Lemke et al. elegantly described
sion and are fundamental actors in neuritogenesis, synapto- the functional consequences of GRIN2B mutations in patients
genesis, and synaptic plasticity processes. Currently, on the with West syndrome as well as in individuals with ID with focal
development of next-generation sequencing, there is a growing epilepsy (14). In the latter study, the patient with ID and focal
body of data implicating de novo mutations of ionotropic epilepsy had a missense mutation in the extracellular
glutamate receptors (7,8) in mental and behavioral disorders. glutamate-binding domain. Interestingly, de novo GRIN2B
Indeed, several mutations in subunits of the NMDAR have been mutations lead to a gain of function, either significantly
related to neurodevelopmental diseases (11–13). However, reducing Mg21 block and increasing Ca21 permeability (N615I
these data require functional validation to unveil whether these and V618G mutations affecting the M2 domain in the pore of
mutations are really pathogenic. In the current study, we iden- the channel) or increasing the apparent glutamate binding
tified a GRIN2B(p.P553T) missense de novo mutation of the affinity (R540H mutation within the extracellular S1 domain).

CT
GluN2B subunit of the NMDAR in a Rett-like patient with severe These gain-of-function mutations point out the important role
encephalopathy. This mutation resulted in the exchange of of facilitated NMDAR signaling in epileptogenesis, with further
highly evolutionary conserved proline into threonine. Impor- therapeutic strategies involving the selective blockade of
tantly, a mutation affecting the same amino acid position mutant leak/hyperactive channels. On the contrary, loss-of-
GluN2B(p.P553L) was previously described in a patient from a function mutations might be causing a hypoglutamatergic
cohort of individuals with ID (16). This patient exhibited function that could be potentially rescued by increasing
phenotypic alterations similar to those of the case in the current NMDAR activity with a therapeutic purpose.
study, including severe ID, hypotonia, and no speech. The The glutamatergic synapse is an extremely sophisticated
phenotypic similarity between these two cases provides strong system where a plethora of molecular actors reside and
evidence for a pathogenic role of GluN2B(P553) mutations in interact to finely tune neurotransmission. However, under
RA
these neurodevelopmental conditions. pathological conditions, the dysregulation of critical players
Electrophysiological studies confirmed the in silico struc- might compromise glutamatergic neurotransmission, resulting
tural studies predicting the hypofunctionality of mutant in an enhancement or a reduction of glutamate signaling (8). In
NMDARs. In addition to a reduction of the channel conduc- the current work, in silico and in vitro experiments concluded
tance, we found a significant increase in both the desensiti- that GluN2B(P553T)-containing NMDARs are hypofunctional.
zation and deactivation rates of GluN1–GluN2B(P553T) Thus, we envisioned that enhancing NMDAR activity might
receptors. Whether the P553T mutation located far from the recover normal glutamatergic neurotransmission and attenuate
binding site is altering deactivation kinetics, an intrinsic prop- clinical manifestations. To this end, we evaluated the effect of
erty of the receptor, is unknown, but electrophysiological D-serine, a coagonist of the GluN1 subunit of the NMDAR,
concentration–response experiments indicate decreased coapplied with physiological glycine concentrations (34).
glutamate binding affinity of mutant NMDARs, triggering Interestingly, our findings indicate an enhancement of mutant
decreased receptor efficacy that seems to be physiologically GluN1–GluN2B receptor activity, suggesting that the structural
T

relevant. NMDAR function is largely determined by the high changes induced by the mutation are not transduced to GluN1
amount of Ca21 influx, which is mostly dependent on channel ligand-binding domain. Noteworthy, because D-serine acti-
kinetics, in particular the rates of desensitization and deacti- vates all NMDAR subtypes (35,36), it should have a general
vation. Thus, we can speculate that kinetics changes detected effect on glutamatergic function. Recently, the beneficial effect
on GluN2B(P553T) mutant receptors might be limiting Ca21
RE

of D-serine in healthy individuals was described in a clinical


influx, which in turn would alter Ca21-mediated signaling trial (37). In their work, Heresco-Levy et al. showed the pro-
pathways and synaptic plasticity. cognitive effects of D-serine throughout NMDAR function and,
Overall, these changes dramatically reduce NMDAR- as we proposed in the current study, intended the develop-
mediated currents and might be underlying the severe ment of NMDAR–glycine site strategies for treating synapt-
phenotype of the patient. Indeed, GluN2B subunits are highly opathy conditions. In agreement with this, D-serine deficits
expressed at embryonic and initial postnatal stages, playing a have been associated with aging in rats, with a functional
critical role in neurodevelopmental processes (31). Conse- rescue observed following exogenous D-serine administration
quently, the hypofunctionality of this major subtype of (38,39). In addition, serine deficiency disorders also provoke
NMDARs might certainly affect neuritogenesis and synapto- neurological phenotypes (psychomotor retardation, micro-
genesis, leading to altered synaptic transmission. This hy- cephaly, and seizures in newborns and children) that can be
pothesis is supported by the morphological and biochemical safely treated by serine oral replenishment (40). Taken
findings, showing a significant decrease of spine density together, these works and ours indicate that, independent of
together with increased levels of GluA1 subunit of AMPARs. the molecular etiology (serine racemase deficit, NMDAR
Interestingly, Pozzo-Miller’s group has reported similar syn- hypofunctionality), D-serine-potentiated NMDAR activity can
aptic outcomes in patients with RTT and in primary murine partially rescue hypoglutamatergic function. Noteworthy, in
neuronal cultures deficient in or harboring mutations of MeCP2 addition to the beneficial effect of D-serine dietary supple-
(27,32). Therefore, our data suggest that NMDAR-induced mentation, the improved condition of the patient might also be

170 Biological Psychiatry January 15, 2018; 83:160–172 www.sobp.org/journal


Biological
D-serine Treatment of Pathogenic GRIN2B De Novo Mutation Psychiatry

influenced by the developmental changes in expression of Floor, 08908 L’Hospitalet de Llobregat, Barcelona, Catalonia, Spain; E-mail:
GluN2 subunits, increasing the GluN2A/GluN2B subunit ratio xaltafaj@idibell.cat.
Received Sep 20, 2016; revised Apr 26, 2017; accepted May 17, 2017.
along development (9). Indeed, our findings indicate that
Supplementary material cited in this article is available online at http://dx.
mutant triheteromeric NMDARs are less affected than the doi.org/10.1016/j.biopsych.2017.05.028.
diheteromeric mutant receptors. Therefore, in addition to the

ED
NMDAR-potentiating effect of D-serine, the developmental
switch of GluN2 subunits might also contribute to improve the REFERENCES
clinical symptoms of the patient to some extent. 1. Rett A (1966): [On an unusual brain atrophy syndrome in hyper-
In summary, our data represent a proof-of-concept study to ammonemia in childhood]. Wien Med Wochenschr 116:723–726.
identify the pathogenicity of de novo mutations of NMDARs 2. Chahrour M, Zoghbi HY (2007): The story of Rett syndrome: From
and the development of precision therapeutic strategies. clinic to neurobiology. Neuron 56:422–437.
Indeed, the methodological pipeline developed in this study 3. Hagberg B, Aicardi J, Dias K, Ramos O (1983): A progressive syn-
drome of autism, dementia, ataxia, and loss of purposeful hand use in
might be further implemented to functionally stratificate de
girls: Rett’s syndrome: Report of 35 cases. Ann Neurol 14:471–479.
novo iGluR mutations associated with synaptic dysfunctions 4. Amir RE, Van den Veyver IB, Wan M, Tran CQ, Francke U, Zoghbi HY
and to define therapeutic strategies. Moreover, it supports the (1999): Rett syndrome is caused by mutations in X-linked MECP2,
use of D-serine as a dietary supplement for the enhancement encoding methyl-CpG-binding protein 2. Nat Genet 23:185–188.

CT
of glutamatergic neurotransmission and/or excitatory/inhibi- 5. Neul JL, Zoghbi HY (2004): Rett syndrome: A prototypical neuro-
tory neurotransmitter imbalance affected in a large spectrum of developmental disorder. Neuroscientist 10:118–128.
6. Hamdan FF, Gauthier J, Araki Y, Lin D-T, Yoshizawa Y, Higashi K, et al.
neurological disorders.
(2011): Excess of de novo deleterious mutations in genes associated
with glutamatergic systems in nonsyndromic intellectual disability. Am
ACKNOWLEDGMENTS AND DISCLOSURES J Hum Genet 88:306–316.
7. Soto D, Altafaj X, Sindreu C, Bayés À (2014): Glutamate receptor
This work was supported by the Grant No. PI16/00851 (National Institute mutations in psychiatric and neurodevelopmental disorders. Commun
of Health Carlos III [ISCIII]), La Marató (Project No. 20140210), PCIN-2014- Integr Biol 7:e27887.
105 (Ministry of Economy and Competitiveness [MINECO]) and Miguel 8. Volk L, Chiu S-L, Sharma K, Huganir RL (2015): Glutamate synapses in
Servet Program (Grant No. CPII16/00021, ISCIII) (all to XA); Grant No. human cognitive disorders. Annu Rev Neurosci 38:127–149.
BFU2014-57562-P (Ministerio de Ciencia e Innovacion, to DS); Grant No. 9. Monyer H, Burnashev N, Laurie DJ, Sakmann B, Seeburg PH (1994):
RA
PI15/01082 (ISCIII) to AG-C); Grant No. SAF2016-77830-R (to MO); Grant Developmental and regional expression in the rat brain and functional
Nos. BFU2012-34398 and BFU2015-69717-P (MINECO), a Career Inte- properties of four NMDA receptors. Neuron 12:529–540.
gration Grant (Ref. 304111), a Marie Curie Intra-European Fellowship 10. Kutsuwada T, Sakimura K, Manabe T, Takayama C, Katakura N,
(Ref. 221540), and a Ramón y Cajal Fellowship (Ref. RYC-2011-08391p) Kushiya E, et al. (1996): Impairment of suckling response, trigeminal
(all to AB); Grant No. SAF2012-40102 (MINECO/European Regional neuronal pattern formation, and hippocampal LTD in NMDA receptor
Development Fund [FEDER]), an FP7 Marie Curie CIG Grant (Ref. 631035), epsilon 2 subunit mutant mice. Neuron 16:333–344.
and Ramón y Cajal (Grant No. RYC-2011-08026) (all to CS); and Grant 11. Burnashev N, Szepetowski P (2015): NMDA receptor subunit mutations
Nos. PI14/00141 and RETIC RD16/0008/0014 (to XG). CA received an FPI in neurodevelopmental disorders. Curr Opin Pharmacol 20:73–82.
contract (MINECO). XA, CS, and AB also benefit from the financial support 12. Hu C, Chen W, Myers SJ, Yuan H, Traynelis SF (2016): Human
of Agency for Management of University and Research Grants (Ref. No. GRIN2B variants in neurodevelopmental disorders. J Pharmacol Sci
SGR14-297). 132:115–121.
We thank Sandra Jurado for guidance in chemical long-term potentiation 13. Platzer K, Yuan H, Schütz H, Winschel A, Chen W, Hu C, et al. (2017):
experiments and the Medicinal Computational Laboratory from Universitat GRIN2B encephalopathy: Novel findings on phenotype, variant clus-
T

Autònoma de Barcelona for providing computing facilities. We also tering, functional consequences and treatment aspects. J Med Genet
acknowledge Pierre Paoletti (Ecole Normale Supérieure, Paris) for kindly 54:460–470.
providing the plasmids allowing the studies with triheteromeric N-methyl- 14. Lemke JR, Hendrickx R, Geider K, Laube B, Schwake M, Harvey RJ,
D-aspartate receptors. et al. (2014): GRIN2B mutations in West syndrome and intellectual
The authors report no biomedical financial interests or potential conflicts disability with focal epilepsy. Ann Neurol 75:147–154.
of interest. 15. Endele S, Rosenberger G, Geider K, Popp B, Tamer C, Stefanova I,
RE

et al. (2010): Mutations in GRIN2A and GRIN2B encoding regulatory


subunits of NMDA receptors cause variable neurodevelopmental
ARTICLE INFORMATION phenotypes. Nat Genet 42:1021–1026.
From the Institute of Neurosciences (DS, CA, XG, EG-B, CS, FC, VF-D), 16. de Ligt J, Willemsen MH, van Bon BWM, Kleefstra T, Yntema HG,
Department of Biomedicine (DS, XG, EG-B), Department of Pathology and Kroes T, et al. (2012): Diagnostic exome sequencing in persons with
Experimental Therapeutics (FC, VF-D), Bellvitge Biomedical Research severe intellectual disability. N Engl J Med 367:1921–1929.
Institute–Unit of Neuropharmacology and Pain Group (CG, MGdS, VF-D, FC, 17. O’Roak BJ, Deriziotis P, Lee C, Vives L, Schwartz JJ, Girirajan S, et al.
XA), and Department of Clinical Foundations (CA, CS), University of Bar- (2011): Exome sequencing in sporadic autism spectrum disorders
celona, August Pi i Sunyer Biomedical Research Institute (DS, XG, EG-B), identifies severe de novo mutations. Nat Genet 43:585–589.
Bioinfomatics and Medical Statistics Group (MO), University of Vic–Central 18. Mothet J-P, Parent AT, Wolosker H, Brady RO, Linden DJ, Ferris CD,
University of Catalonia, Genetics and Molecular Medicine Service (JA, AG-C) et al. (2000): D-serine is an endogenous ligand for the glycine site of
and Department of Neurology (AL-S, AG-C), Neurometabolic Unit, Hospital the N-methyl-D-aspartate receptor. Proc Natl Acad Sci U S A
Sant Joan de Déu and Centro de Investigación Biomédica en Red de 97:4926–4931.
Enfermedades Raras, Molecular Physiology of the Synapse Laboratory 19. Papouin T, Ladepeche L, Ruel J, Sacchi S, Labasque M, Hanini M,
(DR-V, AB), Biomedical Research Institute Sant Pau, and Autonomous et al. (2012): Synaptic and extrasynaptic NMDA receptors are gated by
University of Barcelona, Bellaterra (DR-V, AB), Barcelona, Spain. different endogenous coagonists. Cell 150:633–646.
DS, MO, and CG contributed equally to this work. 20. Sparrow SS, Cicchetti DV (1985): Diagnostic uses of the Vineland
AG-C and XA contributed equally to this work. Adaptive Behavior Scales. J Pediatr Psychol 10:215–225.
Address correspondence to Xavier Altafaj, Ph.D., Neuropharmacology 21. Lucariello M, Vidal E, Vidal S, Saez M, Roa L, Huertas D, et al. (2016):
and Pain Unit–IDIBELL, Hosp. Duran Reynals, Av. Gran Via 199-203, 3rd Whole exome sequencing of Rett syndrome-like patients reveals

Biological Psychiatry January 15, 2018; 83:160–172 www.sobp.org/journal 171


Biological
Psychiatry D-serine Treatment of Pathogenic GRIN2B De Novo Mutation

the mutational diversity of the clinical phenotype. Hum Genet 32. Chapleau CA, Calfa GD, Lane MC, Albertson AJ, Larimore JL, Kudo S,
135:1343–1354. et al. (2009): Dendritic spine pathologies in hippocampal pyramidal
22. Traynelis SF, Wollmuth LP, McBain CJ, Menniti FS, Vance KM, neurons from Rett syndrome brain and after expression of Rett-
Ogden KK, et al. (2010): Glutamate receptor ion channels: Structure, associated MECP2 mutations. Neurobiol Dis 35:219–233.
regulation, and function. Pharmacol Rev 62:405–496. 33. O’Roak BJ, Vives L, Girirajan S, Karakoc E, Krumm N, Coe BP, et al.

ED
23. Gratacòs-Batlle E, Yefimenko N, Cascos-García H, Soto D (2014): (2012): Sporadic autism exomes reveal a highly interconnected protein
AMPAR interacting protein CPT1C enhances surface expression of network of de novo mutations. Nature 485:246–250.
GluA1-containing receptors. Front Cell Neurosci 8:469. 34. Harsing LG, Matyus P (2013): Mechanisms of glycine release, which
24. Regalado MP, Villarroel A, Lerma J (2001): Intersubunit cooperativity in build up synaptic and extrasynaptic glycine levels: The role of
the NMDA receptor. Neuron 32:1085–1096. synaptic and non-synaptic glycine transporters. Brain Res Bull 93:
25. Tovar KR, McGinley MJ, Westbrook GL (2013): Triheteromeric NMDA 110–119.
receptors at hippocampal synapses. J Neurosci 33:9150–9160. 35. Chen PE, Geballe MT, Katz E, Erreger K, Livesey MR, O’Toole KK,
26. Stroebel D, Carvalho S, Grand T, Zhu S, Paoletti P (2014): Controlling et al. (2008): Modulation of glycine potency in rat recombinant NMDA
NMDA receptor subunit composition using ectopic retention signals. receptors containing chimeric NR2A/2D subunits expressed in Xen-
J Neurosci 34:16630–16636. opus laevis oocytes. J Physiol 586:227–245.
27. Li W, Xu X, Pozzo-Miller L (2016): Excitatory synapses are stronger in 36. Dravid SM, Burger PB, Prakash A, Geballe MT, Yadav R, Le P, et al.
the hippocampus of Rett syndrome mice due to altered synaptic (2010): Structural determinants of D-cycloserine efficacy at the NR1/
trafficking of AMPA-type glutamate receptors. Proc Natl Acad Sci U S A NR2C NMDA receptors. J Neurosci 30:2741–2754.

CT
113:E1575–E1584. 37. Levin R, Dor-Abarbanel AE, Edelman S, Durrant AR, Hashimoto K,
28. Benveniste M, Clements J, Vyklický L, Mayer ML (1990): A kinetic anal- Javitt DC, Heresco-Levy U (2015): Behavioral and cognitive effects of
ysis of the modulation of N-methyl-D-aspartic acid receptors by glycine the N-methyl-D-aspartate receptor co-agonist D-serine in healthy
in mouse cultured hippocampal neurones. J Physiol 428:333–357. humans: Initial findings. J Psychiatr Res 61:188–195.
29. Liao D, Scannevin RH, Huganir R (2001): Activation of silent synapses 38. Turpin FR, Potier B, Dulong JR, Sinet P-M, Alliot J, Oliet SHR, et al.
by rapid activity-dependent synaptic recruitment of AMPA receptors. (2011): Reduced serine racemase expression contributes to age-
J Neurosci 21:6008–6017. related deficits in hippocampal cognitive function. Neurobiol Aging
30. D’Souza DC, Radhakrishnan R, Perry E, Bhakta S, Singh NM, Yadav R, 32:1495–1504.
et al. (2013): Feasibility, safety, and efficacy of the combination of 39. Billard J-M (2015): D-serine in the aging hippocampus. J Pharm Bio-
D-serine and computerized cognitive retraining in schizophrenia: An med Anal 116:18–24.
international collaborative pilot study. Neuropsychopharmacology 40. van der Crabben SN, Verhoeven-Duif NM, Brilstra EH, Van
38:492–503. Maldergem L, Coskun T, Rubio-Gozalbo E, et al. (2013): An update on
RA
31. Paoletti P, Bellone C, Zhou Q (2013): NMDA receptor subunit diversity: serine deficiency disorders. J Inherit Metab Dis 36:613–619.
Impact on receptor properties, synaptic plasticity and disease. Nat 41. Karakas E, Furukawa H (2014): Crystal structure of a heterotetrameric
Rev Neurosci 14:383–400. NMDA receptor ion channel. Science 344:992–997.
T
RE

172 Biological Psychiatry January 15, 2018; 83:160–172 www.sobp.org/journal

S-ar putea să vă placă și