ArticlePDF Available

Subunit-Specific Regulation of NMDA Receptor Endocytosis

Authors:

Abstract and Figures

At excitatory synapses, both NMDA and AMPA receptors are localized to the postsynaptic density (PSD). However, unlike AMPA receptors, synaptic NMDA receptors are stable components of the PSD. Even so, surface-expressed NMDA receptors undergo endocytosis, which is more robust early in development and declines during synaptic development. We investigated the subunit-specific contributions to NMDA receptor endocytosis, specifically defining the endocytic motifs and endocytic pathways preferred by the NR2A and NR2B subunits. We find that NR2A and NR2B have distinct endocytic motifs encoded in their distal C termini and that these interact with clathrin adaptor complexes with differing affinities. We also find that NR2A and NR2B sort into different intracellular pathways after endocytosis, with NR2B preferentially trafficking through recycling endosomes. In mature cultures, we find that NR2B undergoes more robust endocytosis than NR2A, consistent with previous studies showing that NR2A is more highly expressed at stable synaptic sites. Our findings demonstrate fundamental differences between NR2A and NR2B that help clarify developmental changes in NMDA receptor trafficking and surface expression.
Content may be subject to copyright.
Cellular/Molecular
Subunit-Specific Regulation of NMDA Receptor Endocytosis
Gabriela Lavezzari, Jennifer McCallum, Colleen M. Dewey, and Katherine W. Roche
National Institute of Neurological Disorders and Stroke, National Institutes of Health, Bethesda, Maryland 20892
At excitatory synapses, both NMDA and AMPA receptors are localized to the postsynaptic density (PSD). However, unlike AMPA
receptors, synaptic NMDA receptors are stable components of the PSD. Even so, surface-expressed NMDA receptors undergo endocyto-
sis, which is more robust early in development and declines during synaptic development. We investigated the subunit-specific contri-
butions to NMDA receptor endocytosis, specifically defining the endocytic motifs and endocytic pathways preferred by the NR2A and
NR2B subunits. We find that NR2A and NR2B have distinct endocytic motifs encoded in their distal C termini and that these interact with
clathrin adaptor complexes with differing affinities. We also find that NR2A and NR2B sort into different intracellular pathways after
endocytosis, with NR2B preferentially trafficking through recycling endosomes. In mature cultures, we find that NR2B undergoes more
robust endocytosis than NR2A, consistent with previous studies showing thatNR2A is more highly expressed at stable synaptic sites. Our
findings demonstrate fundamental differences between NR2A and NR2B that help clarify developmental changes in NMDA receptor
trafficking and surface expression.
Key words: internalization; clathrin; endocytosis; glutamate receptors; trafficking; endocytic sorting
Introduction
Glutamate receptors mediate the majority of excitatory neuro-
transmission in the CNS and are essential for synaptic develop-
ment and plasticity. Although the synaptic localization of gluta-
mate receptors is tightly regulated, it is becoming clear that the
targeting and anchoring of the various subtypes of glutamate recep-
tors are differentially regulated. For example, synaptic AMPA recep-
tors undergo rapid and robust cycling at the postsynaptic membrane
(Luscher et al., 1999; Sheng, 2001; Malinow and Malenka, 2002),
whereas synaptic NMDA receptors are relatively stable (Luscher et
al., 1999; Wenthold et al., 2003). Although synaptic NMDA recep-
tors are tightly anchored to the postsynaptic density (PSD), it is clear
that NMDA receptors are quite mobile within neurons, via intracel-
lular pathways and lateral diffusion in the membrane (Rao and
Craig, 1997; Tovar and Westbrook, 1999; Standley et al., 2000; Scott
et al., 2001; Snyder et al., 2001; Xia et al., 2001; Washbourne et al.,
2002). In addition, surface-expressed NMDA receptors have been
shown to internalize both in primary neuronal cultures and when
expressed in heterologous cells (Roche et al., 2001). The regulation of
NMDA receptor endocytosis is therefore likely to be a critical deter-
minant of NMDA receptor surface expression, both at synaptic and
extrasynaptic sites.
NMDA receptors are tetramers comprising NR1, NR2
(NR2A–NR2D), and NR3 (NR3A–NR3B) subunits. In the fore-
brain, endogenous NMDA receptors are thought to be composed
of NR1 subunits combined with NR2B early in development and
heteromeric combinations of NR1, NR2B, and NR2A in mature
neurons (Kew et al., 1998; Li et al., 1998; Vicini et al., 1998;
Rumbaugh and Vicini, 1999; Tovar and Westbrook, 1999).
NMDA receptor endocytosis is developmentally regulated, with
robust endocytosis early in development that declines as neurons
mature and synapses form (Roche et al., 2001). Interestingly, the
decline in NMDA receptor endocytosis correlates well with the
known changes in NMDA receptor subunit composition during
development. Previous studies have defined an endocytic motif
encoded within the NR2B subunit. This motif, YEKL, is located
just upstream of the PSD-95–Discs large–zona occludens-1
(PDZ)-binding domain on the extreme C terminus of NR2B
(Roche et al., 2001). However, it is not known whether NR2A also
contains endocytic motifs within its C terminus.
In the current study, we have characterized the specific con-
tributions of the NR2B and NR2A subunits to NMDA receptor
endocytosis using a variety of approaches. We find that NR2A
and NR2B contain different sorting motifs within their distal C
termini that regulate trafficking to distinct intracellular pathways
after endocytosis. In neurons, we find that NR2B preferentially
sorts to recycling endosomes, consistent with the more robust
internalization of NR2B-containing NMDA receptors. NR2B ex-
pression drives NMDA receptor endocytosis in both immature
and mature neurons, whereas NR2A endocytosis is most notable
at early stages of development. These results are consistent with
the presence of stable NR2A-containing NMDA receptor com-
plexes at mature synapses. Our findings demonstrate a strong
subunit-specific contribution to NMDA receptor endocytosis
and stability on the plasma membrane.
Materials and Methods
Antibodies and DNA constructs. The primary antibodies Tac 7G7 (Amer-
ican Type Culture Collection, Manassas, VA) and FLAG M2 (Sigma, St.
Received Feb. 6, 2004; accepted May 28, 2004.
WethankDaynaHayesforcontributiontothepreparationoftheprimaryneuronalcultures,theNationalInstitute
of Neurological Disorders and Stroke (NINDS) Light Imaging Facility, in particular the facility manager Dr. Carolyn
Smith, for advice and expertise in the collection and analysis of the confocal images, and the NINDS Sequencing
Facility for automated DNA sequence analysis.
Correspondence should be addressed to K. W. Roche, National Institute of Neurological Disorders and Stroke,
National Institutes of Health, Building 36, Room 5B20, Bethesda, MD 20892. E-mail: rochek@ninds.nih.gov.
DOI:10.1523/JNEUROSCI.1890-04.2004
Copyright © 2004 Society for Neuroscience 0270-6474/04/246383-09$15.00/0
The Journal of Neuroscience, July 14, 2004 24(28):6383– 6391 6383
Louis, MO) were used for immunofluorescence. All secondary antibod-
ies were obtained from Molecular Probes (Eugene, OR), except for cya-
nine 5 (Cy5)-conjugated anti-rabbit, which was obtained from Jackson
ImmunoResearch (West Grove, PA). The following cDNA constructs
were obtained as gifts: FLAGNR2B (F. A. Stephenson, School of Phar-
macy, University of London, London, UK), green fluorescent protein
(GFP)Rab5, GFPRab7, GFPRab9,
1,
2,
3a, and
4 in pACTII,
and the trans-Golgi network (TGN)38 full C terminus in pGBT9 (Juan
Bonifacino, National Institute of Child Health and Human Develop-
ment, National Institutes of Health, Bethesda, MD); and GFPRab11
(James Goldenring, Vanderbilt University, Nashville, TN). The TGN38
C terminus was subcloned into the pBHA vector using EcoRI and PstI
restriction sites. Both the NR2A and NR2B C termini were amplified by
PCR. The amplified tails included amino acids 13151482 (NR2B) or
1304 1464 (NR2A) and were subcloned using EcoRI restriction sites into
pBHA vector or with EcoRV and XbaI into Tac vector. The TacNR2A
truncations, TacNR2A7 and TacNR2A11, were obtained using PCR.
FLAG-NR2A was constructed in the laboratory using PCR with the fol-
lowing sequence: 5-GAACTTCGAAATCTGGACTACAAGGACGA-
CGATGACAAGTGGGGCCCAGAGCAG-3 and 3-CTGCTCTGGGC-
CCCACTTGTCATCGTCGTCCTTGTAGTCCAGATTTCGAAGTTC-5,
inserting the FLAG epitope between amino acids 54 and 55 of NR2A. The
bold sequence indicates the FLAG peptide. FLAGNR2A11 L1320A was
constructed in the laboratory using site-directed mutagenesis. All mutations
were confirmed using automated sequence analysis.
Yeast two-hybrid direct-interaction assays. Direct-interaction assays of
NR2B (amino acids 13151482), NR2A (1304 1464), or the TGN38 C
terminus binding to adaptor medium chains were performed as de-
scribed previously (Kim et al., 1995). Briefly, NR2B (amino acids 1315
1482), NR2A (13041464), or TGN38 in the LexA DNA binding domain
vector pBHA and medium chains in the GAL4 activation domain vector
pACTII were cotransformed into L40 yeast cells. For each condition,
one-half of the yeast cells were plated on dextrose plates containing his-
tidine but lacking leucine and tryptophan (His-containing), and the
other half were plated on dextrose plates lacking histidine, leucine, and
tryptophan (His-deficient). Nutritional selection was used as a measure
of a direct interaction. For nutritional selection, growth on His-deficient
plates was scored on a to ⫹⫹⫹⫹ scale for conditions with equivalent
growth on His-containing plates.
Immunofluorescent internalization assay in HeLa cells. HeLa cells
(American Type Culture Collection) grown on coverslips were trans-
fected with the cDNAs indicated (4
g of DNA per well in a six well dish)
using the calcium phosphate coprecipitation method, and experiments
were performed 48 hr later. Transfected cells were washed in PBS, incu-
bated with anti-Tac antibody for 1 hr on ice, washed with PBS, and
returned to conditioned media at 37°C for 15 min to allow internaliza-
tion. The cells were washed in PBS, fixed in 4% paraformaldehyde in PBS
for 15 min, washed in PBS, and permeabilized in 0.25% Triton X-100 in
PBS for 5 min. The coverslips were washed in PBS, incubated with Alexa
568-conjugated anti-mouse secondary antibodies for 30 min at room
temperature, washed, and mounted with ProLong Antifade Kit (Molec-
ular Probes). When double labeling [early endosome autoantigen 1
(EEA1); PSD-95], the cells were incubated with the appropriate primary
antibody for 1 hr at room temperature after permeabilization. The cells
were washed in PBS, incubated with both anti-mouse and anti-rabbit
secondary antibodies (1:500) for 30 min at room temperature, washed in
PBS, and mounted. Images were collected with a 63 objective on a Zeiss
(Oberkochen, Germany) LSM 510. Series of optical sections were col-
lected at intervals of 0.34
m. Figures show maximum projections. For
the statistical analysis, images from five different HeLa cells in three
independent experiments were collected at a 63 objective with a Zeiss
Axioplan 2 microscope and analyzed with the OpenLab colocalization
module (Improvision, Lexington, MA). Values represent the mean
SEM of Pearsons correlation.
Quantitative assay of internalization in HeLa cells. HeLa cells were
grown on 12 well dishes and transiently transfected with TacNMDA
receptor chimeras. The cells were incubated with anti-Tac antibody for 1
hr on ice to label surface-expressed protein. Cells were either left on ice or
washed and returned to conditioned media for 15 min at 37°C to allow
internalization. Cells were then fixed for 15 min in 4% paraformaldehyde
without detergent permeabilization. After fixation, the cells were washed
and incubated for 45 min with [
125
I]-labeled anti-mouse antibody (25
Ci) at room temperature. After extensive washing, the cells were incu-
bated with 1
M NaOH for 15 min at room temperature, and the lysate was
collected and analyzed. Each condition was assayed in triplicate. For each
experiment, background antibody binding was measured on cells trans-
fected with empty vector (also in triplicate), and the average background
value was subtracted from the average experimental counts. Quantitative
assessment of total surface labeling was calculated by measuring the
amount of anti-Tac antibody present on cells maintained on ice and not
subjected to the 15 min, 37°C incubation. Quantitative assessment of
internalization was calculated by subtracting the amount of anti-Tac
antibody remaining on cells after the 15 min, 37°C incubation from the
total surface counts. Results are expressed as a ratio of internalization to
total. The graph in Figure 2B represents the average of five independent
experiments.
Transfected hippocampal neurons. Preparation of primary hippocam-
pal cultures was performed as described previously (Roche and Huganir,
1995) with minor changes. Briefly, cells were dissected from embryonic
day 18 Sprague Dawley rats (Harlan, Indianapolis, IN), dissociated in
0.25% trypsin, and incubated with 0.01% DNase. Dispersed neurons
were plated onto polyornithinefibronectin-coated glass coverslips at a
density of 100,000 cells per well of a 12 well dish. The neurons were grown
in serum-free media (Neurobasal; Invitrogen, Gaithersburg, MD) with
B27 supplement (Invitrogen). The cultures were maintained in a 5% CO
2
incubator until transfected and processed for immunofluorescence. Hip-
pocampal neurons [3 or 10 d in vitro (DIV)] were transfected with the
appropriate cDNA using the calcium phosphate coprecipitation method.
Six micrograms of total DNA were used per well of the 12 well dish.
Precipitation was allowed to form on cells for 45 min, and the cells were
washed twice with serum-free media. The cells were returned to condi-
tioned media and maintained at 37°Cina5%CO
2
incubator for an
additional 48 hr. The use and care of animals used in this study followed
the guidelines of the National Institutes of Health Animal Research Ad-
visory Committee.
Internalization and immunocytochemistry of neurons. Transfected neu-
rons were washed in PBS and incubated with polyclonal FLAG antibody
for 45 min at room temperature to label surface-expressed protein. The
antibody was removed, and the coverslips were returned to conditioned
media for 15 or 30 min at 37°C. The cells were washed in PBS and fixed
with 4% paraformaldehyde 4% sucrose in PBS for 15 min. The cells
were incubated for 30 min at room temperature with Alexa 568-
conjugated (red) anti-rabbit secondary antibody (Molecular Probes) for
labeling the surface population. The cells were washed and permeabilized
in 0.25% Triton X-100 in PBS for 5 min. After a 30 min incubation with
10% normal goat serum (Vector Laboratories, Burlingame, CA), the cells
were then incubated with Alexa 488-conjugated (green) anti-rabbit sec-
ondary antibody for 30 min to specifically label the internalized popula-
tion. The cells were washed in PBS and mounted with a ProLong Antifade
kit (Molecular Probes). When neurons were cotransfected with GFP
constructs, the surface population was labeled with Alexa 568-
conjugated (red) anti-rabbit secondary antibody, and after permeabili-
zation, the internalized population was specifically labeled with Cy5-
conjugated (blue) anti-rabbit secondary antibody from Jackson
ImmunoResearch. Images were collected with a 63 objective on a Zeiss
LSM 510. Series of optical sections were collected at intervals of 0.34
m.
Figures show maximum projections. For quantitative analysis, images
from three dendrites per neuron (three neurons per experiment) were
collected, and colocalization analysis was measured based on the data
collected in three to five independent experiments with Volocity 2 soft-
ware (Improvision). Values represent mean SEM; n 5.
Results
The NR2A C terminus contains an internalization motif
Endogenous NMDA receptors undergo robust endocytosis that
decreases as synapses develop and neurons mature. We have
demonstrated previously that NR2B contains important regula-
tory motifs that influence NMDA receptor internalization and
6384 J. Neurosci., July 14, 2004 24(28):6383– 6391 Lavezzari et al. Regulation of NMDA Receptor Endocytosis
stability on the plasma membrane (Roche et al., 2001; Lavezzari et
al., 2003). Whereas NR2B is expressed early in development,
NR2A is only expressed in mature neurons. To determine
whether this developmental change in NR2 subunit expression
influences the surface stability of NMDA receptors, we have be-
gun to identify sequences within NR2A that might regulate its
surface expression. Like NR2B, NR2A contains a PDZ-binding
domain motif at the extreme C terminus (ESDV) (Fig. 1 A,B).
In addition, NR2A contains several potential internalization mo-
tifs, including a putative tyrosine-based endocytic motif and sev-
eral dileucine motifs. Both of these are consensus endocytic mo-
tifs that can regulate clathrin-mediated endocytosis (Ohno et al.,
1995, 1996; Bonifacino and Traub, 2003). It is known that NR2B
contains a tyrosine-based endocytic motif (YEKL) that binds di-
rectly to the AP-2 adaptor complex (Lavezzari et al., 2003) and
regulates endocytosis. Although the tyrosine residue is conserved
in both NR2A and NR2B, the surrounding residues in each sub-
unit are unique (YKKM vs YEKL) (Fig. 1A,B).
To determine whether any of these putative endocytic motifs
within the NR2A C terminus regulate surface expression, we have
created a chimera of the distal portion of NR2A (amino acids
1304 1464) and the stable surface integral membrane protein
Tac (TacNR2A) (Fig. 1B), and evaluated endocytosis of this chi-
mera compared with wild-type Tac and the TacNR2B chimera
(NR2B amino acids 13151482) (Fig. 1 B). We were unable to
compare the distal C terminus to the entire C terminus, because
Tac chimeras using the entire C termini of NR2A or NR2B are
retained by the endoplasmic reticulum (ER) (data not shown)
(Hawkins et al., 2004). Transfected HeLa cells were surface-
labeled on ice for 1 hr with Tac antibody and returned to condi-
tioned media for 15 min to allow internalization. Whereas Tac
alone was expressed almost exclusively on
the plasma membrane, TacNR2A was in-
ternalized into vesicular structures similar
to TacNR2B (Fig. 1C). These data show
that the C terminus of NR2A contains a
sequence that is sufficient to induce inter-
nalization of Tac. Within the NR2A C ter-
minus there is also the PDZ domain-
binding motif (ESDV) that is completely
conserved with NR2B and binds to the
PSD-95 family of proteins (Kim et al.,
1995; Kornau et al., 1995; Sheng, 2001). To
examine the role of PSD-95 in regulating
surface expression, we coexpressed TacNR2A
with PSD-95 in HeLa cells and evaluated
internalization of TacNR2A. As we had
observed previously in our examination of
NR2B-mediated endocytosis (Roche et al.,
2001; Lavezzari et al., 2003), coexpression
of PSD-95 blocked the internalization of
TacNR2A and clustered TacNR2A into
patches on the cell surface (Fig. 1D). Co-
expression of PSD-95 with TacNR2A7,
which does not contain the PDZ-binding
motif, did not affect internalization of the
chimera (data not shown), demonstrating
that the direct interaction with PSD-95 is
essential.
To determine whether the conserved ty-
rosine within the YXXA endocytic motif in
NR2A regulates endocytosis, we truncated
the last 11 amino acids of the TacNR2A C
terminus to remove this putative motif (TacNR2A11) (Fig. 1 B).
However, we found that this mutation did not suppress internaliza-
tion (Fig. 2). This result is in sharp contrast to previous findings
demonstrating the importance of the analogous tyrosine (1472) on
the endocytosis of TacNR2B (Roche et al., 2001; Lavezzari et al.,
2003). This result led us to investigate whether the dileucine motifs
in the NR2A C terminus were critical for internalization. We used
site-directed mutagenesis and mutated the three dileucine motifs
from LL to LA (TacNR2A11 L1320A; TacNR2A11 L1337A;
TacNR2A11 L1358A). Both the immunofluorescence assay for in-
ternalization (Fig. 2 A) and an independent quantitative measure
of endocytosis (Fig. 2B) demonstrated that endocytosis of
TacNR2A11 L1320A was significantly less compared with the
other TacNR2A chimeras. This therefore indicates that leucine 1320
regulates endocytosis of TacNR2A.
Differential binding of NR2A and NR2B to adaptor
medium chains
The medium chain of AP-2,
2, binds directly to internalized
cargo proteins, thereby linking them to clathrin and regulating
endocytosis (Ohno et al., 1995, 1996; Bonifacino and Traub,
2003). Although we have shown previously that the C terminus of
NR2B interacts directly with
2 (Lavezzari et al., 2003), we
wanted to determine whether the conserved tyrosine in the NR2A
C terminus also binds to
2. Using the yeast two-hybrid assay, we
found that unlike NR2B, the NR2A C terminus did not interact
with
2 (Fig. 3). In addition to AP-2, there are other intracellular
adaptor complexes (AP-1, AP-3, and AP-4), each of which pos-
sesses a unique medium chain and each of which regulates dis-
tinct transport steps in the endocytic pathway. Therefore, we
examined the ability of NR2A and NR2B to interact with different
Figure 1. The Cterminus ofNR2A contains anendocytic motifthat is inhibited byPSD-95 binding. A, Theamino acid sequence
of the distal C terminus of NR2A. Potential endocytic motifs (LL and YXXA) and the PDZ ligand (ESDV) are indicated in bold. B,
Schematicdiagrams oftheTac–NMDA receptor chimeras.The distalCterminus ofNR2B (aminoacids1315–1482) orNR2A (amino
acids 1304 –1464) was appended to the plasma membrane reporter molecule Tac (TacNR2B; TacNR2A). TacNR2A7 (NR2A
amino acids 1304 –1457) is a truncation removing the PDZ-binding domain. TacNR2A11 (amino acids 1304 –1453) is a trun-
cation removing both the PDZ-binding domain and the putative tyrosine-based endocytic motif. The transmembrane domain is
depicted as a striped rectangle, and the putative tyrosine-based motif and the PDZ-binding domain are depicted in bold. C, The
NR2B and NR2A C termini are both sufficient to induce internalization of the surface protein Tac into endosomal structures. HeLa
cells weretransiently transfected withTac, TacNR2B,or TacNR2A. Cells were incubatedwith Tac monoclonal antibody onice for 1
hr and then returned to conditioned media and incubated at 37°C for 15 min. The cells were fixed and permeabilized, and the
immunoreactivitywas visualizedusing Alexa 568-conjugated(red)anti-mouse secondaryantibodies. Images were collectedwith
a63 objective onaZeissLSM. Seriesof opticalsections werecollected atintervals of0.34
m.Maximum projectionsareshown.
D, PSD-95 inhibits internalization of TacNR2A andclusters the chimera onthe cell surface. HeLacells were transiently transfected
with PSD-95 and TacNR2A. Cells were incubated with Tac monoclonal antibody on ice for 1 hr and then returned to conditioned
media and incubated at 37°C for 15 min. The cells were fixed, permeabilized, and incubated with PSD-95 polyclonal antibodies.
Immunoreactivity was visualized using a combination of Alexa 568-conjugated (red) anti-mouse and Alexa 488-conjugated
(green) anti-rabbit secondary antibodies. Images were collected with a 63 objective on a Zeiss LSM. Series of optical sections
were collected atintervals of 0.34
m. Maximum projections are shown.
Lavezzari et al. Regulation of NMDA Receptor Endocytosis J. Neurosci., July 14, 2004 24(28):6383– 6391 6385
adaptor medium chains in the yeast two-hybrid assay. The well
characterized binding between the adaptor medium chains and
the TGN38 C terminus was used as a positive control (Aguilar et
al., 2001). Most notable were the interactions with
1 and
2.
Interestingly, we found that the NR2A and NR2B C termini bind
equally well to
1, the adaptor subunit that plays a role in clathrin
vesicle formation at the TGN. However, the plasma membrane
adaptor subunit
2 interacted robustly with NR2B but not
NR2A. This is consistent with the YKKM motif in NR2A not
playing a critical role in plasma membrane endocytosis but po-
tentially being important for other intracellular sorting steps.
This correlates quite well with results of our endocytosis assays
(Fig. 2), which also revealed that the YKKM motif does not play
an important role in endocytosis.
The NR2A and NR2B C termini regulate trafficking to distinct
intracellular pathways
After we determined that the C terminus of NR2A was sufficient
to induce endocytosis of the stable surface protein Tac, we began
to characterize the intracellular compartments that contained in-
ternalized TacNR2A. Previous data demonstrated that TacNR2B
is colocalized with internalized transferrin (Roche et al., 2001),
revealing its presence in early endosomes. To determine whether
TacNR2A follows the same intracellular trafficking pathway as
TacNR2B, we followed the same approach as before, but ob-
served that although TacNR2B and TacNR2A both traffic to early
endosomes initially, they subsequently traffic to distinct com-
partments over time. To characterize the intracellular sorting, we
analyzed TacNR2A and TacNR2B over time using immunofluo-
rescence microscopy to examine their subcellular localization.
We incubated the cells on ice for 1 hr with Tac antibody and then
returned the cells to 37°C for various times to allow internaliza-
tion. After 5 min, TacNR2A colocalized completely with the early
endosome marker GFPRab5 (Fig. 4 A). At 15 min, we saw that
the amount of colocalization with GFPRab5 decreased and
TacNR2A trafficked into a different compartment (data not
shown). To identify this compartment we coexpressed the late
endosome marker GFPRab9 in these cells. At 15 min, we were
able to see partial colocalization of TacNR2A with GFPRab9-
positive late endosomes, and by 30 min, we found almost com-
plete colocalization (Fig. 4B–D).
We then quantitated the colocalization of TacNR2A with the
early endosomal marker EEA1 and the late endosomal marker
GFPRab9 over time (5, 15, and 30 min) (Fig. 4E). At 5 min,
TacNR2A colocalized well with EEA1 but not well with GFP
Rab9. At 15 min, there was moderate colocalization with both
endosomal markers, and at 30 min, TacNR2A strongly colocal-
ized with GFPRab9, demonstrating that the NR2A C terminus
regulates sorting from early to late endosomes.
We had originally observed strong colocalization of TacNR2B
with transferrin at both 5 and 15 min (data not shown) (Roche et
al., 2001). In contrast to TacNR2A, we found very little colocal-
ization of TacNR2B with GFPRab9 after 30 min, indicating that
the NR2B C terminus did not regulate sorting to late endosomes.
At this time point, most of the internalized TacNR2B was distrib-
uted throughout the cell, with some accumulation at the cell
edges. This expression pattern was very different from the strong
perinuclear distribution of GFPRab9 that colocalized with in-
ternalized TacNR2A after 30 min. To define the compartment to
which TacNR2B trafficked at later time points, we coexpressed
cells with GFPRab11, a marker for recycling endosomes. After
30 min of internalization, we observed that the majority of inter-
nalized TacNR2B colocalized with GFPRab11 (Fig. 5A). These
data are consistent with the idea that the NR2B C terminus reg-
Figure 3. The NR2A C terminus and NR2B C terminus differentially bind to the medium
chains of adaptor complexes. The NR2A C terminus (amino acids 1304 –1464), the NR2B C
terminus (amino acids 1315–1482), or the TGN38 C terminus in the LexA DNA binding domain
wascotransformedwith
1,
2,
3
,or
4intheGAL4activation domain as follows: (1)
1
and NR2A, (2)
2 and NR2A, (3)
3
and NR2A, (4)
4 and NR2A, (5)
1 and NR2B, (6)
2
and NR2B, (7)
3
and NR2B, (8)
4 and NR2B, (9)
1 and TGN38, (10)
2 and TGN38, (11)
3
and TGN38, and(12)
4 and TGN38.Growth onHis-deficient plates was scoredon ascale
of to ⫹⫹⫹⫹ for conditions with equivalent growth on His-containing plates.
Figure 2. TacNR2A internalization is dependent on a dileucine motif. A, HeLa cells were
transiently transfected with TacNR2A11 or TacNR2A11 L1320A. Cells were incubated with
Tac monoclonalantibody on icefor 1 hrand thenreturned to conditionedmedia and incubated
at 37°C for 15 min. The cells were fixed and permeabilized, and the immunoreactivity was
visualized using Alexa 568-conjugated (red) anti-mouse secondary antibodies. Images were
collected with a 63 objective on a Zeiss LSM. Series of optical sections were collected at
intervals of 0.34
m. Maximum projections are shown. B, HeLa cells were transiently trans-
fected with TacNR2A, TacNR2A11, TacNR2A11 L1320A, TacNR2A11 L1337A, or
TacNR2A11 L1358A. The amount of internalization was determined using an independent
quantitative internalization assay as described in Materials and Methods. Values are expressed
as internalization per total (the total represents initial surface labeling; mean SEM; n 5).
Statistical comparison was performed using ANOVA. *p 0.01.
6386 J. Neurosci., July 14, 2004 24(28):6383– 6391 Lavezzari et al. Regulation of NMDA Receptor Endocytosis
ulates sorting from early endosomes into recycling endosomes.
We then quantitated the colocalization of TacNR2B with the
early endosomal marker EEA1 and the recycling endosomal
marker GFPRab11 over time (5, 15, and 30 min) (Fig. 5B),
demonstrating that TacNR2B strongly colocalized with EEA1 at 5
min and that colocalization with Rab11 increased over time. The
relatively high colocalization of TacNR2B with both EEA1 and
GFPRab11 at all time points is consistent with continual cycling
through this endocytic pathway.
Developmental regulation of NR2A and NR2B trafficking
in neurons
Having characterized the trafficking of NR2A and NR2B chime-
ras in HeLa cells, we set out to determine whether NMDA recep-
tors in neurons also displayed differential endocytosis dependent
on subunit composition. We expressed full-length NR2A or
NR2B proteins containing an extracellular FLAG epitope in hip-
pocampal neurons at different stages of development. The use of
the FLAG epitope tag allows us to specifically label surface-
expressed NR2A or NR2B proteins on living cells. We transfected
hippocampal neurons at 10 DIV with FLAGNR2A or FLAG
NR2B. At 12 DIV, we labeled the surface-expressed receptors
with anti-FLAG antibody and returned the cells to 37°C for 15
min to allow internalization. The cells were then fixed and stained
with Alexa 568 (red)-conjugated secondary antibody to label the
surface population. The stained cells were then permeabilized,
and Alexa 488 (green)-conjugated secondary antibody was added
to specifically label the internalized population. Both NR2A and
NR2B internalized in these neurons (Fig. 6A); however, the in-
Figure 4. The NR2A C terminus regulates trafficking from early endosomes to late endo-
somes. A, After endocytosis, TacNR2A is localized to early endosomes at early time points. HeLa cells
were transiently transfected with TacNR2A and GFP–Rab5. Cells were incubated with Tac polyclonal
antibody onice for1 hr andthen returnedto conditionedmedia for 5min at37°C. Thecells were fixed
and permeabilized, and the immunoreactivitywas visualized using Alexa 568-conjugated (red) anti-
mouse. B–D, Over time, TacNR2A traffics to late endosomes. HeLa cells were transiently transfected
with TacNR2A and GFP–Rab9. Cells were incubated with Tac polyclonal antibody on ice for 1 hr and
then returned to conditioned media for 5 min ( B), 15 min ( C), or 30 min ( D) at 37°C. The cells were
fixed and permeabilized, andthe immunoreactivity was visualized using Alexa568-conjugated (red)
anti-mouse. Images were collected with a 63 objective on a Zeiss LSM. Series of optical sections
were collectedat intervalsof 0.34
m. Maximumprojections areshown. E,Analysis ofthe amountof
colocalization betweenTacNR2A andEEA1 orGFP–Rab9. HeLacells weretransiently transfected with
TacNR2A. Cells were incubated with Tac polyclonal antibody on ice for 1 hr and then returned to
conditionedmediafor5, 15,or30minat37°C.The cellswerefixed,permeabilized,andincubatedwith
antibody for early endosomal marker EEA1. Immunoreactivity was visualized using a combination of
Alexa568-conjugated(red) anti-mouseIgG2AandAlexa488-conjugated(green) anti-mouseIgG1.In
separate experiments, HeLa cells were transiently transfected with TacNR2A and GFP–Rab9. Cells
were incubated with Tac polyclonal antibody on ice for 1 hr and then returned to conditioned media
for 5, 15, or 30 min at 37°C. The cells were fixed and permeabilized, and the immunoreactivity was
visualizedusingAlexa 568-conjugated(red)anti-mousesecondary antibodies.Forthestatisticalanal-
ysis, imagesfrom five differentHeLa cells inthree independent experiments werecollected at a63
objective with a Zeiss Axioplan 2 microscope and analyzed with the OpenLab colocalization module
(Improvision). Values represent the mean SEM of Pearson’s correlation.
Figure 5. The NR2B C terminus regulates trafficking to recycling endosomes. A, HeLa cells
were transiently transfected with TacNR2B and GFP–Rab11. Cells were incubated with Tac
polyclonal antibody on ice for 1 hr and then returned to conditioned media for 30 min at 37°C.
The cellswere fixed and permeabilized,and the immunoreactivityof internalized TacNR2B was
visualizedusing Alexa568-conjugated(red) anti-mouse.GFP–Rab11is visualizedasgreen,and
the merged image shows the level of colocalization of internalized TacNR2B with Rab11. B,
Analysis of the amount of colocalization between TacNR2B and EEA1 or GFP–Rab11 was per-
formed as described in the legend to Figure 4 E. For the statistical analysis, images from five
different HeLa cellsin threeindependent experimentswere collectedat a63 objective witha
Zeiss Axioplan 2 microscope and analyzed with the OpenLab colocalization module (Improvi-
sion). Values represent the mean SEM of Pearson’s correlation.
Lavezzari et al. Regulation of NMDA Receptor Endocytosis J. Neurosci., July 14, 2004 24(28):6383– 6391 6387
ternalization of FLAGNR2B was significantly greater than that
of FLAGNR2A (36 vs 20%) (Fig. 6B).
It is possible that the NR2B subunit internalizes more effi-
ciently than NR2A because of the extrasynaptic localization of
NR2B at sites separate from the scaffolding protein PSD-95.
NR2A is only expressed later in development and is known to be
synaptically localized, whereas NR2B is highly expressed at both
synaptic and extrasynaptic sites (Li et al., 2002). Therefore, we
evaluated the internalization of NR2A and NR2B at earlier devel-
opmental times, when the cells are less mature, when synapses are
still developing, and when PSD-95 is known to be less abundant.
We transfected hippocampal neurons at 3 DIV with FLAG
NR2A or FLAGNR2B and performed the immunofluorescence
2 d later as described above. Although FLAGNR2A consistently
internalized less well than FLAGNR2B, the difference was not
significant, and NR2A and NR2B displayed similar levels of en-
docytosis (26 vs 31%) (Fig. 6). We subsequently analyzed
whether the same mutations that inhibited the internalization of
TacNR2A in HeLa cells had the same effect in hippocampal neu-
rons. We transfected neurons at 3 DIV with FLAGNR2A and
FLAGNR2A11 L1320A but found no significant difference in
the internalization rate between the two constructs (data not
shown), suggesting that heteromeric complexes of NR2A in neu-
rons may use this motif differently or less efficiently than the
monomeric chimera in HeLa cells. Unfortunately, with the high
expression of NR2B in primary neuronal cultures at all develop-
mental stages, it is impossible for us to specifically evaluate sub-
populations of heteromeric NR2A in neurons (for example,
NR1NR2A vs NR1NR2BNR2A).
Differential sorting of NR2A and NR2B in
hippocampal neurons
Because both NR2A and NR2B internalize efficiently in hip-
pocampal neurons at 5 DIV, we decided to use this developmen-
tal time to analyze the endocytic sorting and intracellular distri-
bution of the internalized subunits. We coexpressed FLAG
NR2A or FLAGNR2B with various endosomal marker proteins
(GFPRab5, GFPRab9, or GFPRab11) and analyzed colocal-
ization at different time points. At 15 min, FLAGNR2A colocal-
izes well with the early endosome marker GFPRab5 (Fig. 7A)
but to a lesser degree with the late endosome marker GFPRab9
(Fig. 7D) and the recycling endosome marker GFPRab11 (Fig.
7D). At 30 min, the colocalization of FLAGNR2A with all three
markers was similar (Fig. 7D). In contrast, at 15 min, FLAG
NR2B colocalizes with all three markers (Fig. 8AC), but most
prominently with GFPRab5. At 30 min, the amount of colocal-
ization with the late endosome marker GFPRab9 decreased sub-
stantially (Fig. 8 BD), whereas colocalization of FLAGNR2B
with the recycling endosome marker GFPRab11 almost doubled
(Fig. 8CD), indicating the preferential sorting of NR2B through
recycling endosomes. Quantitative analyses revealed that
whereas only 25% of FLAGNR2A colocalized with GFPRab11
after 30 min of internalization, almost 50% of FLAGNR2B co-
localized with the recycling endosome marker under these con-
ditions (Fig. 8 D), demonstrating that NMDA receptors contain-
ing NR2B rather than NR2A have a clear preference for the
recycling pathway. Together, these data extend our studies of
NMDA receptor trafficking in heterologous cells to primary neu-
ronal cultures and demonstrate distinct endocytic sorting motifs
encoded within NR2A and NR2B.
Discussion
Recent studies have demonstrated that NMDA receptors are
quite mobile, including intracellular pools of NMDA receptors
(Standley et al., 2000; Scott et al., 2001; Xia et al., 2001; Wash-
bourne et al., 2002), plasma membrane-expressed NMDA recep-
tors, and even synaptic NMDA receptors (Rao and Craig, 1997;
Figure 6. Differential internalization of NR2A and NR2B in hippocampal neurons during
development. A, Hippocampal neurons (3 or 10 DIV) were transfected with FLAG–NR2B or
FLAG–NR2A using thecalcium phosphatecoprecipitation method.After 2d (at5 or12 DIV),the
cells were incubated with FLAG polyclonal antibody for 45 min at room temperature. The anti-
bodywas removed,andthe cellswere returnedtoconditioned mediafor15 minat 37°Ctoallow
internalization. Neuronswere fixedand incubated withAlexa 568-conjugated (red) anti-rabbit
secondary antibody to visualize the surface pool. Cells were then washed, permeabilized, and
incubated with Alexa 488-conjugated (green) anti-rabbit secondary antibody to specifically
visualize theinternalized pool. Green fluorescenceindicated internalization compared withthe
total (red plus green). Images were collected with 63 objectives on a Zeiss LSM. Series of
optical sections were collected at intervals of 0.34
m. Maximum projections are shown. B,
Histogram of the amount of internalization of FLAG–NR2A or FLAG–NR2B at 5 or12 DIV. Series
of optical sections were collected at intervals of 0.34
m. Colocalization was measured with
Volocity 2 software (Improvision). At 5 DIV, there is no significant difference between the
amount ofinternalization ofNR2A andNR2B, whereasat 12DIV, thereis astatistical difference
between the two constructs. Data represent means SEM; n 9; *p 0.01; Student’s
unpaired t test.
6388 J. Neurosci., July 14, 2004 24(28):6383– 6391 Lavezzari et al. Regulation of NMDA Receptor Endocytosis
Rumbaugh and Vicini, 1999; Snyder et al., 2001; Grosshans et al.,
2002; Prybylowski et al., 2002; Tovar and Westbrook, 2002). Fur-
thermore, endocytosis of native NMDA receptors is develop-
mentally regulated such that endocytosis is robust early in devel-
opment but declines as neurons mature and synapses form
(Roche et al., 2001). Interestingly, this change in endocytosis par-
allels changes in NMDA receptor subunit composition. In the
forebrain early in development, NMDA receptors are thought to
be NR1NR2B heteromers that switch to heteromeric combina-
tions of NR1, NR2A, and NR2B in the adult brain (Kew et al.,
1998; Li et al., 1998; Rumbaugh and Vicini, 1999; Tovar and
Westbrook, 1999). In the present study, we have probed the pos-
sibility that NR2A and NR2B differentially regulate plasma mem-
brane expression and endocytic sorting of NMDA receptors. We
found that NR2A and NR2B do indeed possess distinct endocytic
motifs and sorting information within their distal C termini. In
addition, we found that the NR2 subunit content of NMDA re-
Figure 7. Internalized NR2A traffics through early, late, and recycling endosomes in hip-
pocampal neurons.Hippocampal neurons(3 DIV)were transfectedwith FLAG–NR2Aand GFP–
Rab5 ( A), FLAG–NR2A and GFP–Rab9 ( B), or FLAG–NR2A and GFP–Rab11 ( C) using the cal-
cium phosphate coprecipitation method. At 5 DIV, the cells were incubated with FLAG
polyclonal antibody for 45min atroom temperature,antibody wasremoved, andthe cellswere
returned to conditioned media for 15 or 30 min at 37°C to allow internalization. Neurons were
then fixed and incubated with Alexa 568-conjugated (red) anti-rabbit secondary antibody to
visualize the surface pool. Cells were then washed, permeabilized, and incubated with Cy5-
conjugated (blue)anti-rabbit secondary antibody to specifically visualize theinternalized pool.
Blue fluorescence indicated internalization compared with the total (red plus blue). Images
were collected with 63 objectives on a Zeiss LSM. Maximum projections are shown. D, Sta-
tistical analysis of the amount of colocalization between FLAG–NR2A and GFP–Rab5, GFP–
Rab9, or GFP–Rab11. Series of optical sections were collected at intervals of 0.34
m. Images
from three dendrites per neuron (3 neuronsper experiment) were collected,and colocalization
analysis was measured based on the data collected in three to five independent experiments
with Volocity 2 software (Improvision). Values represent mean SEM; n 5.
Figure 8. NR2B preferentially traffics through recycling endosomes in hippocampal neu-
rons. Hippocampal neurons (3 DIV) were transfected with FLAG–NR2B and GFP–Rab5 ( A),
FLAG–NR2B and GFP–Rab9 ( B), or FLAG–NR2B and GFP–Rab11 ( C) using the calcium phos-
phatecoprecipitationmethod.At5 DIV, the cells were incubated with FLAG polyclonal antibody
for 45 min at room temperature, antibody was removed, and the cells were returned to condi-
tioned media for 15 or 30 min at 37°C to allow internalization. Neurons were then fixed and
incubated with Alexa 568-conjugated (red) anti-rabbit secondary antibody to visualize the
surface pool. Cells were then washed, permeabilized, and incubated with Cy5-conjugated
(blue) anti-rabbit secondary antibody to specifically visualize the internalized pool. Blue fluo-
rescence indicated internalization compared with the total (red plus blue). Images were col-
lected with 63 objectives on a Zeiss LSM. Series of optical sections werecollected at intervals
of 0.34
m. Maximum projections are shown. D, Statistical analysis of the amount of colocal-
ization between FLAG–NR2B and GFP–Rab5, GFP–Rab9, or GFP–Rab11. Series of optical sec-
tionswere collectedat intervalsof0.34
m.Images fromthree dendritesperneuron(3neurons
per experiment) were collected, and colocalization analysis was measured based on the data
collected in three to five independent experiments with Volocity 2 software (Improvision).
Values represent mean SEM; n 5.
Lavezzari et al. Regulation of NMDA Receptor Endocytosis J. Neurosci., July 14, 2004 24(28):6383– 6391 6389
ceptors in hippocampal neurons regulates the amount of NMDA
receptor endocytosis and determines the intracellular sorting of
NMDA receptors after endocytosis. This demonstrates the im-
portant influence of subunit composition of NMDA receptors on
trafficking, plasma membrane expression, and endocytosis.
NMDA receptors, like all integral membrane proteins expressed
on the cell surface, must undergo regulated transport to the plasma
membrane, where they can be selectively stabilized, undergo endo-
cytosisdegradation, or undergo endocytosis followed by recycling
to the plasma membrane. Protein trafficking through distinct intra-
cellular pathways is tightly regulated primarily by specific protein
protein interactions between cargo proteins and the trafficking ma-
chinery. One of our primary goals is to define motifs and molecular
binding sites on NMDA receptors critical for regulating trafficking,
surface expression, and endocytosis. Although in recent years there
have been numerous studies published on glutamate receptor traf-
ficking and endocytosis, it is interesting to note that AMPA recep-
tors, the best-characterized subtype of glutamate receptor thus far,
do not contain any of the strong consensus sequences known to play
a role in clathrin-mediated endocytosis. In sharp contrast, the cyto-
solic domains of NMDA receptors have numerous consensus endo-
cytic motifs. Thus it is likely that the mechanisms regulating the
surface expression and synaptic targeting of AMPA receptors and
NMDA receptors are distinct. In addition, NMDA receptors possess
a variety of well characterized protein-binding motifs that regulate
interactions with components of the PSD (Hung and Sheng, 2002).
These proteins are also likely to mediate synaptic expression of
NMDA receptors.
In this study, we have focused on elucidating differences be-
tween NR2A and NR2B that are important for NMDA receptor
trafficking and developmental changes in surface expression.
Both NR2 subunits need to associate with NR1 to get to the
surface (McIlhinney et al., 1998). Similarly, Tac chimeras includ-
ing the entire NR2A or NR2B C terminus are retained in the ER
(data not shown) (Hawkins et al., 2004). Therefore, we focused
on the distal portion of the NR2 C terminus for our studies and
found that the NR2A C terminus, like that of NR2B, contains a
dominant endocytic motif. Previous studies have characterized a
tyrosine-based endocytic motif on NR2B, YEKL, which binds
directly to the medium chain of AP-2 and can regulate endocy-
tosis in heterologous cells and in neurons (Roche et al., 2001;
Lavezzari et al., 2003). Although the tyrosine in NR2B (Tyr 1472)
is conserved in NR2A (Tyr 1454), the surrounding residues are
different, suggesting that the interaction of these proteins with
clathrin-associated adaptors might also differ. We found that this
tyrosine-based motif within the C terminus of NR2A did not
regulate endocytosis; in this case, a dileucine motif (Leu 1319,
Leu 1320) plays a critical role in NR2A-mediated internalization.
Interestingly, as described previously for NR2B, we still found
that coexpression with PSD-95 inhibited NR2A-mediated endo-
cytosis. Thus for both NR2A and NR2B, binding of PSD-95 in-
hibits clathrin-mediated endocytosis.
Our evidence that endocytic motifs in the distal C termini of
NMDA receptors were different in the individual NR2 subunits
prompted us to perform binding assays of the NR2A and NR2B C
termini with the medium chains of the various clathrin adaptor
complexes (
1
4). Interestingly, we found that although both
NR2A and NR2B C termini could directly interact with adaptor
medium chains, the specificity varied. For example, the binding
to
1, the medium chain for AP-1 that regulates trafficking at the
TGN, was similar for NR2A and NR2B, whereas the binding to
2 was quite different. We found that NR2B binds very well to
2
(Fig. 3) (Lavezzari et al., 2003); however, the NR2A C terminus
displays almost no binding. This is consistent with the YEKL
motif within NR2B being important in plasma membrane endo-
cytosis, unlike the YKKM motif of NR2A. However, our findings
still clearly demonstrate that the C termini of NR2A and NR2B
both directly interact with medium chains of clathrin adaptor
proteins and are both likely to interact with the clathrin machin-
ery at various intracellular sorting steps. It is important to note
that the preference of NR2A for
1 versus
2 is somewhat rare. It
has even been proposed in the literature (Ohno et al., 1996; Boni-
facino and Traub, 2003) that
2 binding is almost a default in-
teraction for any sequence that interacts with clathrin adaptor
medium chains. Specificity for a particular medium chain is more
common for
1, for example. These data suggest that the lack of
2 binding to the NR2A C terminus is of particular importance.
In addition to the distal C terminus of NR2A and NR2B that
we have characterized in the present study, there is good evidence
that a region adjacent to transmembrane domain 4 (TM4) of the
NMDA receptor subunits is important for surface expression and
functional regulation of NMDA receptors (Vissel et al., 2001).
Interestingly, there is a conserved tyrosine in all NMDA receptor
subunits adjacent to TM4 that appears to be sensitive to phos-
phorylation, and mutation of this residue of NR2A influences
channel function and rundown in rat hippocampal pyramidal
neurons and heterologous cells (Vissel et al., 2001). It is possible
that these conserved tyrosines form a ring near the plasma mem-
brane of NMDA receptors and may regulate plasma membrane
expression or trafficking through intracellular pathways. One of
the challenges for the future will be to determine how these motifs
in different regions of NMDA receptors work together to mediate
trafficking, surface expression, and/or endocytosis of endoge-
nous NMDA receptors.
Our evidence indicates that NR2B undergoes more robust endo-
cytosis than NR2A. This could be attributable to several factors.
Many groups have shown that NR2A is highly synaptic, whereas
NR2B is expressed at both synaptic and extrasynaptic sites (Stocca
and Vicini, 1998; Tovar and Westbrook, 1999; Li et al., 2002). This is
consistent with NR2A being strictly colocalized with components of
the PSD that are likely to stabilize synaptic receptors, most notably
PSD-95. Additional support for this hypothesis is that NR2A expres-
sion and PSD-95 expression are coordinately increased during de-
velopment. Conversely, NR2B is expressed not only early in devel-
opment during synaptogenesis but also in mature neurons at
extrasynaptic sites. We also demonstrated that the NR2A and NR2B
subunits direct differential sorting into distinct intracellular path-
ways after endocytosis. The NR2A and NR2B C termini both initially
regulate internalization into early endosomes; however, over time,
these proteins diverge and NR2A traffics to late endosomes, whereas
NR2B preferentially sorts into recycling endosomes. These data fit
well with the described higher level of NMDA receptor endocytosis
early in development (Roche et al., 2001), with NR2B preferring the
recycling pathway (Figs. 5, 8), and with high expression of NR2B at
extrasynaptic sites in mature neurons. Our finding that NR2B is
more likely to recycle seems quite logical, because NR2B is highly
expressed early in development, when endocytosis is more robust.
Given the relatively long half-life of both surface and total pools of
glutamate receptors (Hall and Soderling, 1997; Mammen et al.,
1997; Huh and Wenthold, 1999) and the fact that NR2B endocytosis
occurs at a rapid rate (3136% of NR2B-containing receptors in 15
min) (Fig. 6), if recycling of NR2B were not an ongoing process, the
neuron would rapidly deplete the surface pool of NMDA receptors.
Our findings support a model of NMDA receptor endocytosis
that is more robust early in development and at extrasynaptic sites,
conditions in which PSD-95 expression is limited and is therefore
6390 J. Neurosci., July 14, 2004 24(28):6383– 6391 Lavezzari et al. Regulation of NMDA Receptor Endocytosis
not available to stabilize plasma membrane-expressed NMDA.
When NMDA receptors are expressed at the plasma membrane at
nonsynaptic sites, their C termini are free to bind to the endocytic
machinery and the receptors undergo endocytosis. In support of our
model, other studies of NMDA receptor function at synaptic versus
extrasynaptic sites demonstrate a role for endocytosis regulated by
tyrosine phosphorylation of NMDA receptor and subsequent regu-
lation of rundown (Wang et al., 1996; Li et al., 2002). These data fit
well with our findings on a differential contribution of NR2A and
NR2B to NMDA receptor endocytosis and the differential sorting of
distinct NMDA receptor complexes. Our evidence of the direct in-
teraction of NR2A and NR2B with clathrin adaptor medium chains
is also supported by recent findings that NMDA receptors coimmu-
noprecipitate with adaptor complexes (Nong et al., 2003), demon-
strating the importance of clathrin-mediated endocytosis and
NMDA receptor regulation.
Our studies elucidated critical differences between the NR2
subunits that are expressed in the hippocampus and cortex. Us-
ing a variety of approaches, we demonstrate that the NR2A and
NR2B subunits have dominant endocytic motifs that regulate
sorting into distinct intracellular pathways. These differences are
also seen when NR2A and NR2B are expressed and their endocy-
tosis is monitored in primary neuronal culture. These findings
provide a framework for understanding developmental changes
in NMDA receptor subunit expression and how it regulates
NMDA receptor trafficking and synaptic expression.
References
Aguilar RC, Boehm M, Gorshkova I, Crouch RJ, Tomita K, Saito T, Ohno H,
Bonifacino JS (2001) Signal-binding specificity of the
4 subunit of the
adaptor protein complex AP-4. J Biol Chem 276:1314513152.
Bonifacino JS, Traub LM (2003) Signals for sorting of transmembrane pro-
teins to endosomes and lysosomes. Annu Rev Biochem 72:395447.
Grosshans DR, Clayton DA, Coultrap SJ, Browning MD (2002) LTP leads to
rapid surface expression of NMDA but not AMPA receptors in adult rat
CA1. Nat Neurosci 5:2733.
Hall RA, Soderling TR (1997) Quantitation of AMPA receptor surface ex-
pression in cultured hippocampal neurons. Neuroscience 78:361371.
Hawkins LM, Prybylowski K, Chang K, Moussan C, Stephenson FA,
Wenthold RJ (2004) Export from the endoplasmic reticulum of assem-
bled NMDA receptors is controlled by a motif in the C terminus of the
NR2 subunit. J Biol Chem, in press.
Huh KH, Wenthold RJ (1999) Turnover analysis of glutamate receptors iden-
tifies a rapidly degraded pool of the N-methyl-D-aspartate receptor subunit,
NR1, in cultured cerebellar granule cells. J Biol Chem 274:151157.
Hung AY, Sheng M (2002) PDZ domains: structural modules for protein
complex assembly. J Biol Chem 277:56995702.
Kew JN, Richards JG, Mutel V, Kemp JA (1998) Developmental changes in
NMDA receptor glycine affinity and ifenprodil sensitivity reveal three
distinct populations of NMDA receptors in individual rat cortical neu-
rons. J Neurosci 18:19351943.
Kim E, Niethammer M, Rothschild A, Jan YN, Sheng M (1995) Clustering
of Shaker-type K channels by interaction with a family of membrane-
associated guanylate kinases. Nature 378:8588.
Kornau HC, Schenker LT, Kennedy MB, Seeburg PH (1995) Domain inter-
action between NMDA receptor subunits and the postsynaptic density
protein PSD-95. Science 269:17371740.
Lavezzari G, McCallum J, Lee R, Roche KW (2003) Differential binding of
the AP-2 adaptor complex and PSD-95 to the C-terminus of the NMDA
receptor subunit NR2B regulates surface expression. Neuropharmacol-
ogy 45:729 737.
Li B, Chen N, Luo T, Otsu Y, Murphy TH, Raymond LA (2002) Differential
regulation of synaptic and extra-synaptic NMDA receptors. Nat Neurosci
5:833 834.
Li JH, Wang YH, Wolfe BB, Krueger KE, Corsi L, Stocca G, Vicini S (1998)
Developmental changes in localization of NMDA receptor subunits in
primary cultures of cortical neurons. Eur J Neurosci 10:17041715.
Luscher C, Xia H, Beattie EC, Carroll RC, von Zastrow M, Malenka RC, Nicoll
RA (1999) Role of AMPA receptor cycling in synaptic transmission and
plasticity. Neuron 24:649 658.
Malinow R, Malenka RC (2002) AMPA receptor trafficking and synaptic
plasticity. Annu Rev Neurosci 25:103126.
Mammen AL, Huganir RL, OBrien RJ (1997) Redistribution and stabiliza-
tion of cell surface glutamate receptors during synapse formation. J Neu-
rosci 17:73517358.
McIlhinney RA, Le Bourdelles B, Molnar E, Tricaud N, Streit P, Whiting PJ
(1998) Assembly intracellular targeting and cell surface expression of the
human N-methyl-
D-aspartate receptor subunits NR1a and NR2A in
transfected cells. Neuropharmacology 37:13551367.
Nong Y, Huang YQ, Ju W, Kalia LV, Ahmadian G, Wang YT, Salter MW
(2003) Glycine binding primes NMDA receptor internalization. Nature
422:302307.
Ohno H, Stewart J, Fournier MC, Bosshart H, Rhee I, Miyatake S, Saito T, Gal-
lusser A, Kirchhausen T, Bonifacino JS (1995) Interaction of tyrosine-based
sorting signals with clathrin-associated proteins. Science 269:18721875.
Ohno H, Fournier MC, Poy G, Bonifacino JS (1996) Structural determi-
nants of interaction of tyrosine-based sorting signals with the adaptor
medium chains. J Biol Chem 271:2900929015.
Prybylowski K, Fu Z, Losi G, Hawkins LM, Luo J, Chang K, Wenthold RJ,
Vicini S (2002) Relationship between availability of NMDA receptor
subunits and their expression at the synapse. J Neurosci 22:89028910.
Rao A, Craig AM (1997) Activity regulates the synaptic localization of the
NMDA receptor in hippocampal neurons. Neuron 19:801812.
Roche KW, Huganir RL (1995) Synaptic expression of the high-affinity kai-
nate receptor subunit KA2 in hippocampal cultures. Neuroscience
69:383393.
Roche KW, Standley S, McCallum J, Dune Ly C, Ehlers MD, Wenthold RJ
(2001) Molecular determinants of NMDA receptor internalization. Nat
Neurosci 4:794 802.
Rumbaugh G, Vicini S (1999) Distinct synaptic and extrasynaptic NMDA
receptors in developing cerebellar granule neurons. J Neurosci
19:1060310610.
Scott DB, Blanpied TA, Swanson GT, Zhang C, Ehlers MD (2001) An
NMDA receptor ER retention signal regulated by phosphorylation and
alternative splicing. J Neurosci 21:30633072.
Sheng M (2001) Molecular organization of the postsynaptic specialization.
Proc Natl Acad Sci USA 98:70587061.
Snyder EM, Philpot BD, Huber KM, Dong X, Fallon JR, Bear MF (2001)
Internalization of ionotropic glutamate receptors in response to mGluR
activation. Nat Neurosci 4:10791085.
Standley S, Roche KW, McCallum J, Sans N, Wenthold RJ (2000) PDZ do-
main suppression of an ER retention signal in NMDA receptor NR1 splice
variants. Neuron 28:887 898.
Stocca G, Vicini S (1998) Increased contribution of NR2A subunit to syn-
aptic NMDA receptors in developing rat cortical neurons. J Physiol
(Lond) 507:1324.
Tovar KR, Westbrook GL (1999) The incorporation of NMDA receptors
with a distinct subunit composition at nascent hippocampal synapses in
vitro. J Neurosci 19:4180 4188.
Tovar KR, Westbrook GL (2002) Mobile NMDA receptors at hippocampal
synapses. Neuron 34:255264.
Vicini S, Wang JF, Li JH, Zhu WJ, Wang YH, Luo JH, Wolfe BB, Grayson DR
(1998) Functional and pharmacological differences between recombi-
nant N-methyl-
D-aspartate receptors. J Neurophysiol 79:555566.
Vissel B, Krupp JJ, Heinemann SF, Westbrook GL (2001) A use-dependent
tyrosine dephosphorylation of NMDA receptors is independent of ion
flux. Nat Neurosci 4:587596.
Wang YT, Yu XM, Salter MW (1996) Ca
2
-independent reduction of
N-methyl-
D-aspartate channel activity by protein tyrosine phosphatase.
Proc Natl Acad Sci USA 93:17211725.
Washbourne P, Bennett JE, McAllister AK (2002) Rapid recruitment of
NMDA receptor transport packets to nascent synapses. Nat Neurosci
5:751759.
Wenthold RJ, Prybylowski K, Standley S, Sans N, Petralia RS (2003) Traf-
ficking of NMDA receptors. Annu Rev Pharmacol Toxicol 43:335358.
Xia H, Hornby ZD, Malenka RC (2001) An ER retention signal explains
differences in surface expression of NMDA and AMPA receptor subunits.
Neuropharmacology 41:714 723.
Lavezzari et al. Regulation of NMDA Receptor Endocytosis J. Neurosci., July 14, 2004 24(28):6383– 6391 6391
... 37 In addition, LTD with downregulation of postsynaptic GABA-A receptors (GABA-ARs) occurs in concert with LTP and trafficking of glutamate receptors at excitatory synapses. 38 Receptor trafficking may involve multiple steps that include lateral diffusion between synaptic and extrasynaptic sites, exocytosis with delivery of new receptors to the plasma membrane, and endocytosis with removal of receptors that subsequently may be recycled or targeted for proteosomal degradation [39][40][41][42][43][44][45] (Figure 1). Lateral diffusion is largely regulated by receptor subunit-specific affinities with synaptic scaffolding proteins, [46][47][48] and an increase in the ability of synapses to accommodate new receptor entry also is important because augmented surface expression alone may change the character of synapses but will not increase their size. ...
... NMDAR location and function is strongly regulated within minutes to hours by neuronal activity 37,65,159,171,172 through constitutive endocytosis with recycling endosomes, 43,173 vesicular exocytosis to the plasma membrane 30,[174][175][176] and lateral diffusion between synaptic and extrasynaptic sit es, 31,42,144,147,149,[177][178][179][180][181][182] and this trafficking is subunit specific 43,159,173,183,184 (Figure 1). The phosphorylation status at several serine/threonine and tyrosine residues, particularly at the distal C-terminus of NMDAR subunits involved with protein-protein interactions, 185,186 regulates several important functions related to synaptic plasticity by influencing the lateral diffusion and endocytosis of re ceptors. ...
... NMDAR location and function is strongly regulated within minutes to hours by neuronal activity 37,65,159,171,172 through constitutive endocytosis with recycling endosomes, 43,173 vesicular exocytosis to the plasma membrane 30,[174][175][176] and lateral diffusion between synaptic and extrasynaptic sit es, 31,42,144,147,149,[177][178][179][180][181][182] and this trafficking is subunit specific 43,159,173,183,184 (Figure 1). The phosphorylation status at several serine/threonine and tyrosine residues, particularly at the distal C-terminus of NMDAR subunits involved with protein-protein interactions, 185,186 regulates several important functions related to synaptic plasticity by influencing the lateral diffusion and endocytosis of re ceptors. ...
Article
Full-text available
Status epilepticus (SE) remains a significant cause of morbidity and mortality and often is refractory to standard first‐line treatments. A rapid loss of synaptic inhibition and development of pharmacoresistance to benzodiazepines (BZDs) occurs early during SE, while NMDA and AMPA receptor antagonists remain effective treatments after BZDs have failed. Multimodal and subunit‐selective receptor trafficking within minutes to an hour of SE involves GABA‐A, NMDA, and AMPA receptors and contributes to shifts in the number and subunit composition of surface receptors with differential impacts on the physiology, pharmacology, and strength of GABAergic and glutamatergic currents at synaptic and extrasynaptic sites. During the first hour of SE, synaptic GABA‐A receptors containing γ2 subunits move to the cell interior while extrasynaptic GABA‐A receptors with δ subunits are preserved. Conversely, NMDA receptors containing N2B subunits are increased at synaptic and extrasynaptic sites, and homomeric GluA1 (“GluA2‐lacking”) calcium permeant AMPA receptor surface expression also is increased. Molecular mechanisms, largely driven by NMDA receptor or calcium permeant AMPA receptor activation early during circuit hyperactivity, regulate subunit‐specific interactions with proteins involved with synaptic scaffolding, adaptin‐AP2/clathrin‐dependent endocytosis, endoplasmic reticulum (ER) retention, and endosomal recycling. Reviewed here is how SE‐induced shifts in receptor subunit composition and surface representation increase the excitatory to inhibitory imbalance that sustains seizures and fuels excitotoxicity contributing to chronic sequela such as “spontaneous recurrent seizures” (SRS). A role for early multimodal therapy is suggested both for treatment of SE and for prevention of long‐term comorbidities.
... In addition to serine/threonine phosphorylation, NMDAR contains different phosphotyrosine sites in the CTD, which are regulated by protein tyrosine kinases, including Src and Fyn. NMDAR2B is the predominant subunit phosphorylated at tyrosine residues [14][15][16][17][18]. NMDAR2B contains three major tyrosine phosphorylation sites (Y1252, Y1336, and Y1472). ...
... NMDAR2B is the predominant subunit phosphorylated at tyrosine residues [14][15][16][17][18]. NMDAR2B contains three major tyrosine phosphorylation sites (Y1252, Y1336, and Y1472). Tyrosine 1472 is localized inside the tyrosine-based internalization motif (YEKL), which binds the AP2 adaptor, a protein involved in clathrin-coated endocytic vesicle formation [16]. Y1472 phosphorylation interferes with AP2 binding, resulting in the inhibition of NMDAR endocytosis. ...
Article
Full-text available
Glutamate is a key player in excitatory neurotransmission in the central nervous system (CNS). The N-methyl-D-aspartate receptor (NMDAR) is a glutamate-gated ion channel which presents several unique features and is involved in various physiological and pathological neuronal processes. Thanks to great efforts in neuroscience, its structure and the molecular mechanisms controlling its localization and functional regulation in neuronal cells are well known. The signaling mediated by NMDAR in neurons is very complex as it depends on its localization, composition, Ca2+ influx, and ion flow-independent conformational changes. Moreover, NMDA receptors are highly diffusive in the plasma membrane of neurons, where they form heterocomplexes with other membrane receptors and scaffold proteins which determine the receptor function and activation of downstream signaling. Interestingly, a recent paper demonstrates that NMDAR signaling is involved in epithelial cell competition, an evolutionary conserved cell fitness process influencing cancer initiation and progress. The idea that NMDAR signaling is limited to CNS has been challenged in the past two decades. A large body of evidence suggests that NMDAR is expressed in cancer cells outside the CNS and can respond to the autocrine/paracrine release of glutamate. In this review, we survey research on NMDAR signaling and regulation in neurons that can help illuminate its role in tumor biology. Finally, we will discuss existing data on the role of the glutamine/glutamate metabolism, the anticancer action of NMDAR antagonists in experimental models, NMDAR synaptic signaling in tumors, and clinical evidence in human cancer.
... Among them, STEP 61, is only expressed in the hippocampus and cortex and enriched in the extrasynaptic membrane [24][25][26]. STEP 61 dephosphorylates the 1472 tyrosine residue site on GluN2B, leading to NR2B binding to the clathrin-associated adapter protein complex (AP-2), thereby promoting endocytosis and transfer of NR2B [27]. The dynamic regulation of NR2B phosphorylation enables NR2B to rapidly reallocate and express within synaptic and extrasynaptic membranes. ...
... The increased expression could potentially be attributed to the functional disruption of a di-leucine motif positioned at residues 1319-1320, situated in close proximity to the K1339Ter mutation site. This motif has been established to play a regulatory role in the endocytosis of NMDARs containing GluN2A 31 . All GRIN2A missense variants tested, except G784A, were expressed at comparable or higher levels than wild-type GluN2A (Fig. 1C, D). ...
Article
Full-text available
Human genetic studies have revealed rare missense and protein-truncating variants in GRIN2A, encoding for the GluN2A subunit of the NMDA receptors, that confer significant risk for schizophrenia (SCZ). Mutations in GRIN2A are also associated with epilepsy and developmental delay/intellectual disability (DD/ID). However, it remains enigmatic how alterations to the same protein can result in diverse clinical phenotypes. Here, we performed functional characterization of human GluN1/GluN2A heteromeric NMDA receptors that contain SCZ-linked GluN2A variants, and compared them to NMDA receptors with GluN2A variants associated with epilepsy or DD/ID. Our findings demonstrate that SCZ-associated GRIN2A variants were predominantly loss-of-function (LoF), whereas epilepsy and DD/ID-associated variants resulted in both gain- and loss-of-function phenotypes. We additionally show that M653I and S809R, LoF GRIN2A variants associated with DD/ID, exert a dominant-negative effect when co-expressed with a wild-type GluN2A, whereas E58Ter and Y698C, SCZ-linked LoF variants, and A727T, an epilepsy-linked LoF variant, do not. These data offer a potential mechanism by which SCZ/epilepsy and DD/ID-linked variants can cause different effects on receptor function and therefore result in divergent pathological outcomes.
... Neurotransmitter receptors expressed at the plasma membrane of immature neurons are highly diffusive and poorly confined when compared to mature neurons 3,11,12 . Their cycling between intracellular and membrane pools is also upregulated at early stages 13 . Clustering the highly-diffusive NMDARs at early synaptic contacts would thus require some active and potent processes. ...
Article
Full-text available
Direct interactions between receptors at the neuronal surface have long been proposed to tune signaling cascades and neuronal communication in health and disease. Yet, the lack of direct investigation methods to measure, in live neurons, the interaction between different membrane receptors at the single molecule level has raised unanswered questions on the biophysical properties and biological roles of such receptor interactome. Using a multidimensional spectral single molecule-localization microscopy (MS-SMLM) approach, we monitored the interaction between two membrane receptors, i.e. glutamatergic NMDA (NMDAR) and G protein-coupled dopamine D1 (D1R) receptors. The transient interaction was randomly observed along the dendritic tree of hippocampal neurons. It was higher early in development, promoting the formation of NMDAR-D1R complexes in an mGluR5- and CK1-dependent manner, favoring NMDAR clusters and synaptogenesis in a dopamine receptor signaling-independent manner. Preventing the interaction in the neonate, and not adult, brain alters in vivo spontaneous neuronal network activity pattern in male mice. Thus, a weak and transient interaction between NMDAR and D1R plays a structural and functional role in the developing brain.
... Zeng et al. [15] identified the elevated expression of the NMDAR2B subunit in breast-to-brain metastasis. They found that tyrosine-phosphorylated NMDAR2B, which is crucial for the activation of cell-surface NMDARs [16][17][18], was detected at higher levels in breast-to-brain metastasis compared to primary breast tumors. The inhibition of NMDAR2B in these cells resulted in a reduced brain tumor size and increased survival in mice, indicating a potential role of NMDAR in promoting the growth of invasive cancer cells in the brain. ...
Article
Full-text available
The involvement of the N-methyl-D-aspartate receptor (NMDAR), a glutamate-gated ion channel, in promoting the invasive growth of cancer cells is an area of ongoing investigation. Our previous findings revealed a physical interaction between NMDAR and MET, the hepatocyte growth factor (HGF) receptor. However, the molecular mechanisms underlying this NMDAR/MET interaction remain unclear. In this study, we demonstrate that the NMDAR2B subunit undergoes proteolytic processing, resulting in a low-molecular-weight form of 100 kDa. Interestingly, when the NMDAR2B and MET constructs were co-transfected, the full-size high-molecular-weight NMDAR2B form of 160 kDa was predominantly observed. The protection of NMDAR2B from cleavage was dependent on the kinase activity of MET. We provide the following evidence that MET opposes the autophagic lysosomal proteolysis of NMDAR2B: (i) MET decreased the protein levels of lysosomal cathepsins; (ii) treatment with either an inhibitor of autophagosome formation or the fusion of the autophagosome and lysosome elevated the proportion of the NMDAR2B protein’s uncleaved form; (iii) a specific mTOR inhibitor hindered the anti-autophagic effect of MET. Finally, we demonstrate that MET coopts NMDAR2B to augment cell migration. This implies that MET harnesses the functionality of NMDAR2B to enhance the ability of cancer cells to migrate.
... The dephosphorylation of GluN2B leads to its internalization and contributes to synaptic weakening (Barria and Malinow 2002;Lussier et al. 2015). Overactivity of STEP61 will lead to an internalization of NMDAr for a pronounced period, disturbing the regulatory cycle and process of memory formation (Carty 2012;Roche et al. 2001;Paul et al. 2003;Lavezzari et al. 2004;Hee et al. 2004;Snyder et al. 2005;Furukawa et al. 2005;Gee et al. 2006;Lau and Zukin 2007;Xu et al. 2009;Sanz-Clemente et al. 2010;Poddar et al. 2010;Paoletti et al. 2013;Jang et al. 2015;Morisot and Ron 2017;Iacobucci and Popescu 2017;Lind et al. 2017;Carvajal et al. 2021). ...
Article
Full-text available
STEP (STriatal-Enriched Protein Tyrosine Phosphatase) is a brain-specific phosphatase that plays an important role in controlling signaling molecules involved in neuronal activity and synaptic development. The striatum is the main location of the STEP enzyme. An imbalance in STEP61 activity is a risk factor for Alzheimer’s disease (AD). It can contribute to the development of numerous neuropsychiatric diseases, including Parkinson’s disease (PD), schizophrenia, fragile X syndrome (FXS), Huntington’s disease (HD), alcoholism, cerebral ischemia, and stress-related diseases. The molecular structure, chemistry, and molecular mechanisms associated with STEP61’s two major substrates, Alpha-amino-3-hydroxy-5-methyl-4-isoxazole propionic acid receptors (AMPAr) and N-methyl-d-aspartate receptors (NMDARs), are crucial in understanding the relationship between STEP61 and associated illnesses. STEP’s interactions with its substrate proteins can alter the pathways of long-term potentiation and long-term depression. Therefore, understanding the role of STEP61 in neurological illnesses, particularly Alzheimer’s disease-associated dementia, can provide valuable insights for possible therapeutic interventions. This review provides valuable insights into the molecular structure, chemistry, and molecular mechanisms associated with STEP61. This brain-specific phosphatase controls signaling molecules involved in neuronal activity and synaptic development. This review can aid researchers in gaining deep insights into the complex functions of STEP61.
Article
Full-text available
N-methyl-d-aspartate receptors (NMDARs) play a critical role in normal brain function, and variants in genes encoding NMDAR subunits have been described in individuals with various neuropsychiatric disorders. We have used whole-cell patch-clamp electrophysiology, fluorescence microscopy and in-silico modeling to explore the functional consequences of disease-associated nonsense and frame-shift variants resulting in the truncation of GluN2A or GluN2B C-terminal domain (CTD). This study characterizes variant NMDARs and shows their reduced surface expression and synaptic localization, altered agonist affinity, increased desensitization, and reduced probability of channel opening. We also show that naturally occurring and synthetic steroids pregnenolone sulfate and epipregnanolone butanoic acid, respectively, enhance NMDAR function in a way that is dependent on the length of the truncated CTD and, further, is steroid-specific, GluN2A/B subunit-specific, and GluN1 splice variant-specific. Adding to the previously described effects of disease-associated NMDAR variants on the receptor biogenesis and function, our results improve the understanding of the molecular consequences of NMDAR CTD truncations and provide an opportunity for the development of new therapeutic neurosteroid-based ligands. Supplementary Information The online version contains supplementary material available at 10.1007/s00018-023-05062-6.
Article
The pre- and post-synaptic compartments contain a variety of molecules that are known to recycle between the plasma membrane and intracellular organelles. The recycling steps have been amply described in functional terms, with, for example, synaptic vesicle recycling being essential for neurotransmitter release, and postsynaptic receptor recycling being a fundamental feature of synaptic plasticity. However, synaptic protein recycling may also serve a more prosaic role, simply ensuring the repeated use of specific components, thereby minimizing the energy expenditure on the synthesis of synaptic proteins. This type of recycling has been recently described for components of the extracellular matrix, which undergo long-loop recycling (LLR), to and from the cell body. Here we suggest that the energy-saving recycling of synaptic components may be more widespread than is generally acknowledged, potentially playing a role in both synaptic vesicle protein usage and postsynaptic receptor metabolism.
Article
SNARE-mediated membrane fusion plays a crucial role in presynaptic vesicle exocytosis and also in postsynaptic receptor delivery. The latter is considered particularly important for synaptic plasticity and learning and memory, yet the identity of the key SNARE proteins remains elusive. Here, we investigate the role of neuronal synaptosomal-associated protein-23 (SNAP-23) by analyzing pyramidal-neuron specific SNAP-23 conditional knockout (cKO) mice. Electrophysiological analysis of SNAP-23 deficient neurons using acute hippocampal slices showed normal basal neurotransmission in CA3-CA1 synapses with unchanged AMPA and NMDA currents. Nevertheless, we found theta-burst stimulation-induced long-term potentiation (LTP) was vastly diminished in SNAP-23 cKO slices. Moreover, unlike syntaxin-4 cKO mice where both basal neurotransmission and LTP decrease manifested changes in a broad set of behavioral tasks, deficits of SNAP-23 cKO are more limited to spatial memory. Our data reveal that neuronal SNAP-23 is selectively crucial for synaptic plasticity and spatial memory without affecting basal glutamate receptor function.
Article
Full-text available
In rat cerebellar granule neurons, mRNA and protein levels of the NR2A and NR2C subunits of the NMDA receptor increase during the second postnatal week. At this time, mRNA and protein levels of the NR2B subunit begin to fall. To investigate targeting of NMDA receptor subunits, we performed whole-cell recordings from rat cerebellar granule neurons at different times during development and investigated the pharmacological and biophysical properties of mossy fiber-evoked NMDA EPSCs. Isolated NMDA EPSCs from newly formed synapses in the first postnatal week exhibited partial block by the NR2B subunit-specific antagonist (1 S ,2 S )-1-(4-hydroxyphenyl)-2-(4-hydroxy-4-phenylpiperidino)-1-propanol (CP 101,606). By the end of the second postnatal week, NMDA EPSCs were virtually unaffected by the NR2B antagonist. In parallel, NMDA EPSC decay times decreased over a similar developmental time course. We compared properties of synaptic NMDA receptors with extrasynaptic receptors that are present on the cell body with rapid application of glutamate to excised nucleated patches. Deactivation of patch responses accelerated with development and closely resembled evoked NMDA EPSCs in rats of the same age. However, patch responses were highly sensitive to CP 101,606 through the second postnatal week, and sensitivity was seen in some neurons up to the fourth postnatal week. Spermine potentiated peak NMDA patch responses from postnatal days 10–14 rats but had little effect on evoked NMDA EPSCs. Our data suggest selective targeting of a distinct NMDA receptor subtype to synaptic receptor populations in cerebellar granule neurons. Later in development, similar changes occur in the extrasynaptic receptor population.
Article
Full-text available
Tyrosine-based signals within the cytoplasmic domain of integral membrane proteins mediate clathrin-dependent protein sorting in the endocytic and secretory pathways. A yeast two-hybrid system was used to identify proteins that bind to tyrosine-based signals. The medium chains (mu 1 and mu 2) of two clathrin-associated protein complexes (AP-1 and AP-2, respectively) specifically interacted with tyrosine-based signals of several integral membrane proteins. The interaction was confirmed by in vitro binding assays. Thus, it is likely that the medium chains serve as signal-binding components of the clathrin-dependent sorting machinery.
Article
Full-text available
Regulation of ion channel function by intracellular processes is fundamental for controlling synaptic signaling and integration in the nervous system. Currents mediated by N-methyl-D-aspartate (NMDA) receptors decline during whole-cell recordings and this may be prevented by ATP. We show here that phosphorylation is necessary to maintain NMDA currents and that the decline is not dependent upon Ca2+. A protein tyrosine phosphatase or a peptide inhibitor of protein tyrosine kinase applied intracellularly caused a decrease in NMDA currents even when ATP was included. On the other hand, pretreating the neurons with a membrane-permeant tyrosine kinase inhibitor occluded the decline in NMDA currents when ATP was omitted. In inside-out patches, applying a protein tyrosine phosphatase to the cytoplasmic face of the patch caused a decrease in probability of opening of NMDA channels. Conversely, open probability was increased by a protein tyrosine phosphatase inhibitor. These results indicate that NMDA channel activity is reduced by a protein tyrosine phosphatase associated with the channel complex.
Article
Full-text available
Many integral membrane proteins contain tyrosine-based signals within their cytoplasmic domains that mediate internalization from the cell surface and targeting to lysosomal compartments. Internalization depends on an interaction of the tyrosine-based signals with the clathrin-associated adaptor complex AP-2 at the plasma membrane, whereas lysosomal targeting involves interaction of the signals with an analogous complex, AP-1, at the trans-Golgi network. Recent studies have identified the medium chains μ2 of AP-2 and μ1 of AP-1 as the recognition molecules for tyrosine-based signals. We have now investigated the structural determinants for interaction of the signals with μ2 and μ1. The position of the signals was found to be an important determinant of interactions with μ2 and μ1; signals were most effective when present at the carboxyl terminus of a polypeptide sequence. Another important determinant of interactions was the identity of residues surrounding the critical tyrosine residue. Mutation of some residues affected interactions with μ2 and μ1 similarly, whereas other mutations had differential effects. These observations suggest that both the position and the exact sequence of tyrosine-based sorting signals are major determinants of selectivity in their interaction with clathrin-associated adaptor complexes.
Article
The N-methyl-D-aspartate (NMDA) receptor subserves synaptic glutamate-induced transmission and plasticity in central neurons. The yeast two-hybrid system was used to show that the cytoplasmic tails of NMDA receptor subunits interact with a prominent postsynaptic density protein PSD-95. The second PDZ domain in PSD-95 binds to the seven-amino acid, COOH-terminal domain containing the terminal tSXV motif (where S is serine, X is any amino acid, and V is valine) common to NR2 subunits and certain NR1 splice forms. Transcripts encoding PSD-95 are expressed in a pattern similar to that of NMDA receptors, and the NR2B subunit co-localizes with PSD-95 in cultured rat hippocampal neurons. The interaction of these proteins may affect the plasticity of excitatory synapses.
Article
Pharmacologically isolated miniature NMDA receptor‐mediated excitatory postsynaptic currents (mN‐EPSCs) were recorded in large visual cortical neurons in layer V of rat cortical slices. Haloperidol (100 μ m ) and CP101,606 (10 μ m ), two specific blockers of NMDA receptors comprising NR1/NR2B subunits, were tested on mN‐EPSCs in rats at postnatal days 7 and 8 (P7–P8) and P13–P15. At both ages tested, no significant effects of these drugs were seen in the whole population of neurons, although in few neurons at both ages changes in amplitude were observed with haloperidol. Other dopamine receptor antagonists, spiperone and clozapine, failed to decrease mN‐EPSCs in cortical neurons at P13–P15. CP101,606 (10 μ m ) significantly decreased the amplitude of evoked N‐EPSCs (eN‐EPSCs) in visual cortical slices from rats at P3–P5, a developmental stage at which mRNA studies have indicated the virtual absence of NR2A mRNA. CP101,606 failed to significantly change evoked AMPA‐mediated EPSCs at P5 and eN‐EPSCs at P7–P8 and P13–P15. NMDA receptor‐mediated currents were also studied in somatic outside‐out patches at P13–P15 with fast application of l ‐glutamate (1 m m ). Haloperidol (50 μ m ) and CP101,606 (10 μ m ) blocked these currents in all patches tested. The effect of CP101,606 was concentration dependent. We suggest that rather early in development synaptic receptors comprising NR1/NR2B subunits could be associated with other subunits so that blockade by haloperidol and CP101,606 is prevented. Moreover, the consistent blockade seen in outside out patches might be ascribed to the confinement of NR1/NR2B receptors to an extrasynaptic population.
Article
ANCHORING of ion channels at specific subcellular sites is critical for neuronal signalling, but the mechanisms underlying channel localization and clustering are largely unknown (reviewed in ref. 1). Voltage-gated K+ channels are concentrated in various neuronal domains, including presynaptic terminals, nodes of Ranvier and dendrites, where they regulate local membrane excitability. Here we present functional and biochemical evidence that cell-surface clustering of Shaker-subfamily K+ channels is mediated by the PSD-95 family of membrane-associated putative guanylate kinases, as a result of direct binding of the carboxy-terminal cytoplasmic tails to the K+ channel subunits to two PDZ (also known as GLGF or DHR) domains in the PSD-95 protein. The ability of PDZ domains to function as independent modules for protein-protein interaction, and their presence in other junction-associated molecules (such as ZO-1 (ref. 3) and syntrophin), suggest that PDZ-domain-containing polypeptides may be widely involved in the organization of proteins at sites of membrane specialization.
Article
The N-methyl-D-aspartate (NMDA) receptor subserves synaptic glutamate-induced transmission and plasticity in central neurons. The yeast two-hybrid system was used to show that the cytoplasmic tails of NMDA receptor subunits interact with a prominent postsynaptic density protein PSD-95. The second PDZ domain in PSD-95 binds to the seven-amino acid, COOH-terminal domain containing the terminal tSXV motif (where S is serine, X is any amino acid, and V is valine) common to NR2 subunits and certain NR1 splice forms. Transcripts encoding PSD-95 are expressed in a pattern similar to that of NMDA receptors, and the NR2B subunit co-localizes with PSD-95 in cultured rat hippocampal neurons. The interaction of these proteins may affect the plasticity of excitatory synapses.
Article
Non-N-methyl-D-aspartate glutamate receptors are responsible for fast excitatory neurotransmission in the mammalian CNS. These receptors are rapidly activated and desensitized in the presence of glutamate, and are often further subdivided into alpha-amino-3-hydroxy-5-methyl-4-isoxazole proprionic acid and kainate receptors based on their selective agonists. Non-NMDA glutamate receptors are composed of multiple subunits which recently have been cloned, and studies on the recombinant glutamate receptors have helped clarify the distinctions between AMPA and kainate-preferring glutamate receptors. Although the subunits which make up both AMPA and kainate receptors have a widespread distribution, most currents recorded in vivo are characteristic of recombinant AMPA receptors. To help clarify the functional role of high-affinity kainate receptors, we have characterized the expression of a high-affinity kainate receptor subunit, KA2, in cultured hippocampal neurons. Using immunocytochemistry, we found that KA2 was expressed in hippocampal neurons at all times during the development of the cells in culture, and the subunit was enriched in dendritic spines after about 14 days. The subcellular distribution of KA2 paralleled that of the AMPA receptor subunit GluR1, with the AMPA and kainate subunits being colocalized at all times in culture. The enriched KA2 immunoreactivity co-localized with the synaptic vesicle protein synaptophysin at the resolution of light microscopy, indicating synaptic localization of KA2. Although the kainate subunit KA2 co-localized with the AMPA subunit GluR1, co-immunoprecipitation experiments demonstrated a direct interaction between the AMPA receptor subunits GluR1 and GluR2/3, but not between GluR1 and the kainate subunits GluR6/7 or KA2. We therefore, conclude that both AMPA and kainate receptor subunits are enriched in the same dendritic spines, yet do not combine to form receptor complexes.