You are on page 1of 15

Molecular Psychiatry (2021) 26:5097–5111

https://doi.org/10.1038/s41380-020-0796-3

ARTICLE

Cell-type specific modulation of NMDA receptors triggers


antidepressant actions
Santosh Pothula 1 Taro Kato1 Rong-Jian Liu1 Min Wu1 Danielle Gerhard1,2 Ryota Shinohara 1
● ● ● ● ● ●

Alexa-Nicole Sliby1 Golam M. I. Chowdhury1 Kevin L. Behar1 Gerard Sanacora1 Pradeep Banerjee3
● ● ● ● ●

Ronald S. Duman 1

Received: 30 April 2020 / Revised: 13 May 2020 / Accepted: 18 May 2020 / Published online: 2 June 2020
© The Author(s), under exclusive licence to Springer Nature Limited 2020

Abstract
Both the NMDA receptor (NMDAR) positive allosteric modulator (PAM), and antagonist, can exert rapid antidepressant
effects as shown in several animal and human studies. However, how this bidirectional modulation of NMDARs causes
similar antidepressant effects remains unknown. Notably, the initial cellular trigger, specific cell-type(s), and subunit(s) of
NMDARs mediating the antidepressant-like effects of a PAM or an antagonist have not been identified. Here, we used
1234567890();,:
1234567890();,:

electrophysiology, microdialysis, and NMR spectroscopy to evaluate the effect of a NMDAR PAM (rapastinel) or NMDAR
antagonist, ketamine on NMDAR function and disinhibition-mediated glutamate release. Further, we used cell-type specific
knockdown (KD), pharmacological, and behavioral approaches to dissect the cell-type specific role of GluN2B, GluN2A,
and dopamine receptor subunits in the actions of NMDAR PAM vs. antagonists. We demonstrate that rapastinel directly
enhances NMDAR activity on principal glutamatergic neurons in medial prefrontal cortex (mPFC) without any effect on
glutamate efflux, while ketamine blocks NMDAR on GABA interneurons to cause glutamate efflux and indirect activation
of excitatory synapses. Behavioral studies using cell-type-specific KD in mPFC demonstrate that NMDAR-GluN2B KD on
Camk2a- but not Gad1-expressing neurons blocks the antidepressant effects of rapastinel. In contrast, GluN2B KD on Gad1-
but not Camk2a-expressing neurons blocks the actions of ketamine. The results also demonstrate that Drd1-expressing
pyramidal neurons in mPFC mediate the rapid antidepressant actions of ketamine and rapastinel. Together, these results
demonstrate unique initial cellular triggers as well as converging effects on Drd1-pyramidal cell signaling that underlie the
antidepressant actions of NMDAR-positive modulation vs. NMDAR blockade.

Introduction neurobiology of depression is unknown and currently


available medications have significant limitations, including
Depression is a widespread, debilitating illness with a time-lag of weeks to months for a therapeutic response,
devastating personal and economic consequences, and is a low rates of response and remission, and a significant
leading cause of disability world-wide [1]. In addition, the subpopulation of depressed patients considered treatment
resistant [2, 3]. The discovery that a single sub-anesthetic
dose of ketamine, an NMDA receptor (NMDAR) antago-
nist, exerts rapid and sustained antidepressant effects in
Supplementary information The online version of this article (https://
doi.org/10.1038/s41380-020-0796-3) contains supplementary treatment-resistant patients represents a fundamental
material, which is available to authorized users. breakthrough and major advance for the treatment of
depression, including difficult to treat and suicidal patients
* Santosh Pothula
santosh.pothula@yale.edu
[4, 5].
However, ketamine produces dissociative and psycho-
1
Department of Psychiatry, Yale School of Medicine, New Haven, tomimetic effects and is a drug of abuse [6, 7]. Recent
CT 06511, USA studies have shown that NMDAR positive allosteric mod-
2
Department of Psychiatry, Weill Cornell Medicine, New York, ulators (PAMs) also exerts rapid and sustained anti-
NY 10065, USA depressant effects without the dissociative and
3
Allergan Inc., Madison, NJ, USA psychotomimetic-like side effects of ketamine [8–10].
5098 S. Pothula et al.

Due to these attractive properties, there is immense interest Gad1-cre (male; obtained from Dr. Marina Picciotto, Yale
in developing NMDAR PAMs as rapid acting anti- University) mice, and WT littermates bred on a C57BL/6J
depressants. Preclinical studies demonstrate that while background (Jackson Laboratories, Bar Harbor ME). Sst-
ketamine and rapastinel have opposing effects on tdTomato male mice were used for NMDA- or AMPA-
NMDARs, they both require activation of glutamate AMPA inward currents after rapastinel treatment. Sst-Cre mice
receptors, and have convergent downstream signaling cas- were bred with Ai9(RCL-tdT) mice (catalog 007909;
cades, including mTORC1, BDNF release, and synaptic Jackson Laboratories) to obtain Sst-tdTomato mice. Adult
plasticity in medial prefrontal cortex (mPFC) [11–13]. The male Sprague Dawley (SD) rats (Charles River, Raleigh,
actions of ketamine are mediated by blockade of NMDAR NC, USA) of 180–220 g weight range were used for
on GABA interneurons, resulting in disinhibition of gluta- microdialysis and NMR spectroscopy analysis of 13C
mate transmission [14–17], but the cellular targets of incorporation. Animals were housed under 12 h light–dark
NMDAR PAMs have not been determined. Further, it is cycle with ad libitum access to water and rodent chow.
unclear how NMDAR antagonists and PAMs exert similar Animals were group housed until surgery. All experiments
antidepressant effects, and the cellular trigger and neuronal were performed during the 12-h light cycle. Animal use and
cell-types involved in the actions of NMDAR PAMs remain procedures were in accordance with the National Institutes
elusive. Here, we used ketamine and rapastinel as tool of Health Guide for the Care and Use of Laboratory Ani-
compounds of NMDAR antagonists and PAMs, respec- mals and approved by the Yale University Animal Care and
tively, to address these questions. Use Committees. Sample sizes for this study were deter-
In the current study, we investigate the role of NMDAR mined based on our previous experience [17] with the types
subunits on Camk2a-excitatory neurons vs. Gad1-inhibitory of viral infusion studies conducted in this project.
interneurons in the mPFC. We target GluN2B, one of the
major NMDAR subunits expressed on both inhibitory and Drugs
excitatory neurons in mPFC, as preclinical and clinical stu-
dies have reported that selective GluN2B antagonists pro- Rapastinel was procured from Sai Life Sciences (India).
duce rapid antidepressant effects [11, 18]. In addition, we (S)-ketamine and ketamine were purchased from Sigma-
further investigate a subtype of excitatory neurons, char- Aldrich (St. Louis, MO) and Tocris Biosciences (USA).
acterized by expression of dopamine receptor Drd1, as well SCH39166 and L741,626 were purchased from Tocris
as the role of Drd1 and Drd2 dopamine receptors (Drd1R Biosciences (USA). Rapastinel and (S)-ketamine or keta-
and Drd2R) in the actions of rapastinel [19–23]. mine were dissolved in sterile saline (0.9% NaCl). For
The results demonstrate that GluN2B knockdown on microdialysis and NMR spectroscopy studies in rats, drugs
glutamatergic, but not GABAergic neurons in mPFC blocks were administered via s.c. (rapastinel) or i.p. ((S)-ketamine
the antidepressant effects of rapastinel. Conversely, the or ketamine) routes, respectively. For behavioral studies in
actions of ketamine are blocked by GluN2B KD on mice, rapastinel and ketamine were administered via i.v.
GABAergic but not glutamatergic neurons, in agreement and i.p. routes.
with a recent study [17], indicating unique initial cellular
triggers for the actions of NMDAR PAM vs. NMDAR Electrophysiology
blockade. The results also demonstrate a role for Drd1-
expressing excitatory neurons in the actions of rapastinel, as The mPFC brain slices were prepared from male mice
well as ketamine, indicating a convergent mechanism for (8–12 weeks old). Coronal slices of 300-μm thickness
these two types of rapid antidepressants. containing the mPFC were prepared and placed in artificial
cerebrospinal fluid (ACSF) (pH 7.35–7.38) equilibrated
with 95% O2/5% CO2 before transferring them to recording
Materials and methods chamber fixed with an Olympus BX50WI scope for elec-
trophysiology recordings. The chamber was continuously
Animals perfused with normal ACSF at a rate of 2–3 ml/min and
its temperature maintained at 33 ± 0.5 °C. Patch pipettes
Rodents used in the current study included adult (3–5 MΩ) were pulled from glass tubing with a Flaming-
(8–12 week old) male mice. Wild-type C57BL/6 male mice Brown Horizontal Puller. The pipette solution contained the
were used (Jackson Laboratories) for electrophysiology, following: 115 mM K gluconate, 5 mM KCl, 2 mM MgCl2,
and Drd1R and Drd2R antagonist studies. For viral- 2 mM Mg-ATP, 2 mM Na2ATP, 10 mM Na2-phospho-
mediated knockdown and behavioral experiments, creatine, 0.4 mM Na2GTP, and 10 mM Hepes, pH 7.33.
Camk2a-cre (male; obtained from Dr. Günter Schütz, Pyramidal neurons and interneurons in mPFC were visua-
German Cancer Research Center, Heidelberg, Germany), lized by video microscopy using microscope (40× IR lens)
Cell-type specific modulation of NMDA receptors triggers antidepressant actions 5099

with infrared differential interference contrast (IR/DIC). acquired at 11.7 T (1 H frequency of 500.13 MHz; Bruker
Whole-cell recordings were made with an Axoclamp-2B AVANCE; Bruker Instruments, Billerica, MA, USA) to
amplifier. NMDA- and AMPA-induced inward currents determine total concentrations, and 13C enrichments of
were measured in magnesium free ACSF upon bath appli- amino acids and metabolites in mPFC as previously
cation of 5 or 10 µM NMDA, or 5 µM AMPA alone, before described [27].
and after application of rapastinel or ketamine.
Surgical procedures for viral/drug infusions
Immunohistology
Mice were anesthetized with a cocktail of ketamine/xyla-
Brains were collected from all virus infused mice after zine (100/10 mg/kg, i.p.), head fixed in a stereotaxic appa-
transcardiac perfusion with sterile PBS and 4% paraf- ratus (David Kopf, Tujunga, CA) and the incision site was
ormaldehyde (PFA). They were postfixed in 4% PFA for sterilized. Ophthalmic ointment was applied on eyes. A pair
24 h and incubated in 30% sucrose for an additional 24 h. of craniotomies were made at following coordinates from
30-µm thick sections were made from postfixed frozen bregma: +1.9 mm AP, ±0.4 mm ML, and –2.8 mm DV,
brains using a Microm HM550 cryostat and mounted on and bilateral viral infusions into the mPFC (0.5 μl per side;
slides for further immunofluorescence visualization. Images 0.1 μl/min) were performed using Hamilton syringe (Reno,
were acquired using confocal laser scanning microscope NV) fitted with a 30 gauge needle. Incision sites were
(Olympus FV1000). closed with sutures, an antibiotic was applied, and carprofen
(5 mg/kg, i.p.) was injected immediately after surgery and
Microdialysis daily for the next 2 days. We followed the same procedure
for drug infusions, where bilateral stainless steel guide
Microdialysis studies were performed as previously cannulae (0.8 mm center-to-center, 26 gauge, Plastics One,
described [24]. Using coordinates: +3.2 mm anterior- Roanoke, VA) were placed in the mPFC (coordinates from
posterior (AP); +0.5 mm medial–lateral (ML); −1.5 mm bregma: +1.9 AP, ±0.4 ML, −2.5 DV), fixed to the skull
dorsal–ventral (DV) from bregma, guide cannulae were using dental cement and skull screws. Mice were indivi-
implanted to target mPFC. A microdialysis probe (3 mm dually housed to prevent dislodging of cannula, resulting in
membrane length; 50 kDa cutoff) was inserted via guide a social isolation period. Infusion needles (2.8 mm long)
cannula to dialyze mPFC region (V, −4.5 mm) with ACSF were inserted via guide cannulae to infuse drugs into
(148 NaCl, 4 KCl, 0.8 MgCl2, 1.4 CaCl2, 1.2 Na2HPO4, the mPFC.
and 0.3 NaH2PO4; in mM; pH 7.2) at a flow rate of 1 μl/
min. After a 3-h stabilization period, samples were collected Viral constructs
at every 20 min intervals for 1 h for baseline and up to 4 h
after drug administration. Glutamate concentrations were The following shRNA sequences were designed to
measured by ultra high-performance liquid chromatography target GluN2B or GluN2A subunits. 5′-Tgtaccaacagg
tandem mass spectrometry as previously described [25]. tctcaccttaaacTTCAAGAGAgtttaaggtgagacctgttggtacTTTT
TTC-3′ (GluN2B) or 5′-TtgacagaacgcgaacttcgaaatcTTCA
[1,6-13C2]glucose infusions and NMR spectroscopy AGAGAgatttcgaagttcgcgttctgtcaTTTTTTC-3′ (GluN2A;
analysis Integrated DNA Technologies). They were ligated into a
TATAloxP-flanked CMV-EGFP cassette containing pSico
[1,6-13C2]glucose infusions and NMR spectroscopy ana- plasmid (from Tyler Jacks, Massachusetts Institute of
lysis were performed as previously described [26]. Rats Technology, Boston, Massachusetts, USA; Addgene
were briefly anethesized with isoflurane and catheters were plasmid catalog 11578), designed to restrict shRNA
placed into tail vein. Drugs/vehicle were injected following expression to cells that express Cre-recombinase. The
recovery for at least 30 min after anesthesia. Ten minute resulting pGluN2BshRNA or pGluN2AshRNA constructs
after administration of ketamine or rapastinel or vehicle, a were subcloned into pAAV-mCherry (Virovek) to gen-
solution of [1,6-13C2]glucose (99 atom%; Cambridge Iso- erate the plasmid, which allows ubiquitous mCherry
topes, Andover, MA, USA) dissolved in water (0.75 mol expression and conditional EGFP expression driven by
l−1 per 200 g body weight) was infused for 8 min. Imme- floxed CMV cassette. These constructs were packaged into
diately following the 8 min infusion of [1,6-13C2]glucose, adeno-associated virus 2 (AAV2). AAV-DIO-EmGFP-
the rats were quickly sacrificed and brains were flash fro- D1miR was used for cell-type-specific knockdown of
zen. Metabolites in the mPFC were extracted from frozen Drd1Rs as previously described [28]. AAV-Camk2a-
tissue (65–85 mg) and brain samples were loaded into EGFP was used a control for Drd1R knockdown
5 mm tubes for NMR analysis. 1H-[13C]-NMR spectra were experiment.
5100 S. Pothula et al.

Behavioral tests Locomotor activity (LMA)

All tests were conducted at least three weeks after virus Mice were placed in clean home cages (25 × 15 × 12 cm) for
infusion or one week after cannulation surgeries. During 30 min. LMA was tracked using an automated infrared
this time mice were single housed, resulting in social iso- beam break system and monitored with Med-PC software
lation. Time lines of experimental approaches were detailed (Med Associates). The number of beam breaks were
in figures. Animals were habituated at least 30 min to recorded as a measure of locomotion.
experimental conditions prior to behavioral tests. Animals
were randomly assigned to the treatment groups and Statistics
observers were blinded to treatments and genotypes during
the experiments and behavioral analysis. Data are expressed as the mean ± standard error of mean
and analyzed using Graphpad prism v8. For multi-group
Forced swim test (FST) comparisons and to determine treatment or genotype effects
or interaction, one-way ANOVA or two-way ANOVA post
Mice were placed in a clear cylinder filled with water (24 ± hoc Tukey’s multiple comparison tests were used to deter-
1 °C, 18 cm depth) for 6 min. Tests were video recorded and mine statistical significance. For comparisons between two
scored for immobility time, defined as a lack of activity groups, either unpaired or paired Student’s t test (two-tailed)
except the minimal amount of effort required to keep head was performed to determine statistical significance. p < 0.05
above water, by an experimenter blinded to treatment groups. was considered as statistically significant. The details of
Immobility time during 2–6 min window was scored. Pre- statistical tests and their outcomes were presented in the
swim is the FST conducted prior to drug administrations. figure legends. The sampling distribution of the data was
assumed to be normal and the variances were assumed to be
Female urine sniffing test (FUST) equal, but this was not formally tested for every analysis.
Any value >2 standard deviation from the group mean was
FUST, a validated test of reward-seeking behavior in considered as an outlier (a criteria decided on prior to
rodents, was performed as previously described [29]. Each initiating the experiments) and excluded from the analysis.
mouse was placed in individual novel cage for acclimati-
zation (30 min) followed by habituation to a cotton-tipped
applicator for 1 h. Subsequently, a fresh water-soaked cot- Results
ton-tipped applicator was placed on the inner wall of the
cage, and video recorded for 5 min. The cotton-tipped Rapastinel, but not ketamine, directly enhances
applicator was removed. After 45 min, a cotton-tipped NMDA-inward current, but has no effect on
applicator with fresh female urine was placed in the cage, glutamate release
and video recorded for 5 min. Only the time spent sniffing
water- or female urine-soaked cotton-tipped applicator were We first determined the effects of rapastinel and ketamine on
scored. NMDAR activity by patch clamp recordings of layer V
pyramidal neurons in slices of mPFC. Incubation with
Novelty-suppressed feeding test (NSFT) rapastinel enhanced NMDA-induced inward currents and
displayed an inverted-U-dose response relationship: 0.1 µM
Mice were food deprived for 18 h prior to exposure to a rapastinel significantly enhanced the NMDA-inward current
novel open arena (40 × 40 × 25 cm) with chow pellet in the in layer V neurons and 1 µM produced a relatively smaller,
center. The latency to feed was assessed. Later, home cage nonsignificant enhancement, while a higher concentration
feeding was assessed as a feeding control, where mice were (10 µM) had no effect compared with vehicle (Fig. 1a, b).
allowed ad libitum access to a preweighed chow pellet in Analysis of GABA interneurons, tested in Sst-reporter mice
their home cages. revealed that rapastinel (0.1 µM) produced a nonsignificant
trend for increase in NMDA-induced inward currents (Sup-
Open field test (OFT) plementary Fig. 1a, b), with no effect on AMPA-induced
inward currents. In line with its NMDAR antagonistic
Each mouse was placed in the center of a plexiglass test activity, ketamine produced a near complete blockade of the
apparatus (40 × 40 × 25 cm) and video recorded for 10 min. NMDA-induced inward current (Fig. 1c, d).
The time spent in the center zone and total distance traveled Ketamine has been shown to increase glutamate release
in the open field were analyzed using an automated analysis via disinhibition, which is required for its downstream
software (ANY-maze, Stoelting). signaling and antidepressant behavioral actions [15, 17, 30].
Cell-type specific modulation of NMDA receptors triggers antidepressant actions 5101

Fig. 1 Rapastinel directly enhances NMDAR activity without any rapastinel or (S)-ketamine administration up to 240 min post-treatment.
changes in glutamate release. NMDA-inward currents were recorded (S)-ketamine (30 mg/kg) significantly increased the % change in basal
from layer V pyramidal neurons of mPFC before and after bath levels of glutamate in mPFC at 60 min and remained elevated up to
application of NMDA alone or in combination with rapastinel or 180 min. No significant differences in glutamate levels were observed
ketamine. a, c Representative traces showing inward currents in with 3, 10, and 30 mg/kg doses of rapastinel (n = 8–9 SD rats per
response to NMDA (5 µM) before and after bath application of group, one-way ANOVA and post hoc Dunnett’s multiple comparison
0.1–10 µM rapastinel or 10 µM ketamine. b Rapastinel increased the tests, *p < 0.05). f NMR spectroscopy was performed to analyze 13C
NMDA-inward current at 0.1 and 1 µM, respectively, while there was fraction enrichment of glutamate-C4, GABA-C2, and glutamine-C4 in
no effect observed at 10 µM compared with baseline; results are the mPFC of rats 30 min after ketamine or rapastinel administration. 13C
mean ± SEM, n = 14–23 neurons (One-way ANOVA and post hoc fraction enrichment of glutamate-C4, GABA-C2, and glutamine-C4
Tukey’s test, F3,74 = 6.09, p < 0.01; **p < 0.01). d Ketamine sig- significantly increased in mPFC after ketamine (10 mg/kg) adminis-
nificantly reduced the inward currents in response to NMDA (10 µM) tration, but no changes were observed with 3, 10, and 30 mg/kg doses
compared with baseline (n = 6 neurons, paired t-test, **p < 0.01). of rapastinel (n = 7–13 SD rats per group, one-way ANOVA and post
e Microdialysis was performed in rats and dialysate samples were hoc Dunnett’s test, *p < 0.05; **p < 0.01).
collected from mPFC at 20 min intervals starting 40 min before

To determine if rapastinel enhances glutamate release, we GluN2B knockdown on glutamatergic, but not
used in vivo microdialysis and 1H-[13C]-nuclear magnetic GABAergic, neurons blocks the antidepressant-like
resonance (NMR) spectroscopy. For comparison in micro- effects of rapastinel
dialysis studies, we used (S)-ketamine, which has higher
affinity for the NMDAR than (R)-ketamine [31]. The results To identify the initial cellular trigger and specific NMDAR
show that (S)-ketamine (10 mg/kg) significantly increased subunit which mediates the antidepressant-like behavioral
glutamate efflux as measured by percent change of basal actions of rapastinel, we used an AAV driven shRNA-
levels of glutamate in rat mPFC (Fig. 1e), consistent with targeting NMDAR subunits and cell-specific Cre-
previous reports [14, 15]. In contrast, rapastinel at 3, 10, and recombinase (Cre+) transgenic mouse lines to achieve
30 mg/kg, doses previously shown to exert antidepressant- cell-type-specific knockdown. Based on reports that selec-
like effects in rodents [13, 32, 33], had no significant effects tive GluN2B modulators produce rapid antidepressant
on extracellular glutamate (Fig. 1e). In line with the actions in rodent models and in clinical trials [30], we first
microdialysis results, ketamine but not rapastinel sig- focused on knockdown of this subunit in Camk2a-expres-
nificantly increased the percent change in glutamate-C4, sing pyramidal neurons. The GluN2B shRNA construct was
GABA-C2 and glutamine-C4 fractional enrichments of 13C packaged into AAV-2 (referred to as AAV2GluN2BshRNA); the
as measured by 1H-[13C]-NMR spectroscopy in rat PFC viral construct included dsRed and EGFP and upon Cre-
(Fig. 1f). These findings highlight the differential effects of mediated recombination, EGFP is excised, resulting in the
rapastinel and ketamine on glutamate release. expression of only dsRed as a marker of recombination
5102 S. Pothula et al.

a GluN2B knockdown in Camk2a-neurons: b


TATA-lox TATA-lox

CMV dsRed U6 CMV EGFP Sh-GluN2B hGH polyA

+ Camk2a-Cre recombinase
CMV dsRed U6 Sh-GluN2B hGH polyA
TATA-lox GluN2B
c WT d NMDA +Rps 0.1 μM e ACSF Rps 100nM Glutamatergic
neuron
2500 **

NMDA- inward current


WT 2000

dsRED/EGFP 1500
#

(pA)
Camk2a cre+
GluN2B KD

1000

500 pA
500
1 min
0
dsRED/EGFP WT GluN2B KD
f
WTAAV2B-Sal
WTAAV2B-Rps or Ket
Camk2aCre+/AAV2B-Sal
Camk2aCre+/AAV2B-Rps or Ket

g FST-Rps h FUST-Rps i NSFT-Rps j FST-Ket

** ** * ** 250 *
80
*

Immobility time (sec)


200 1000
Latency to feed (sec)
Immobility time (sec)

** *
Sniffing time (sec)

60 800 200
150
600 150
40
100
400 100
20
50 200 50
0
0 0 0
Water Urine

Fig. 2 Knockdown of GluN2B in the mPFC of Camk2a-Cre mice #p < 0.05); rapastinel significantly enhanced NMDA-inward current in
blocks the antidepressant-like actions of rapastinel. a Schematic WTCre−/AAV2B but not in Camk2aCre+/AAV2B mice (n = 8–11 neurons,
showing pGluN2BshRNA construct before and after Cre-recombinase paired t-test, *p < 0.05). f Timeline for experimental approach.
to generate the active shRNA construct and b removal of NMDARs Rapastinel significantly reduced immobility time in the FST
(GluN2B) on glutamatergic neuron. c Representative images of (g), increased female urine sniffing time in the FUST (h), and reduced
AAV2GluN2BshRNA virus expression showing fluorescent labeling of latency to feed in the NSFT (i) in WTCre−/AAV2B but these effects were
WT neurons (yellow: colocalization of EGFP and dsRed fluorescence) blocked in Camk2aCre+/AAV2B mice. Data expressed as the Mean ±
and Cre-recombination dependent pyramidal neurons (subset of SEM and analyzed by two-way ANOVA with Tukey’s multiple
only dsRed-labeled neurons) in the mPFC of WTCre−/AAV2B and comparisons (n = 8–10 per group; FST: treatment: F1,34 = 7.18, p =
Camk2aCre+/AAV2B mice, respectively. d Representative traces of 0.01, genotype: F1,34 = 6.02, p = 0.02, interaction: F1,34 = 5.83,
NMDA (10 µM) induced inward currents in WT and GluN2B KD p = 0.02; NSFT: treatment: F1,34 = 9.01, p < 0.01, genotype:
glutamatergic neurons of WTCre−/AAV2B and Camk2aCre+/AAV2B mice F1,34 = 2.23, p = 0.14, interaction: F1,34 = 5.9, p = 0.02; *p < 0.05;
before and after rapastinel (100 nM) treatment. e NMDA-inward cur- **p < 0.01). j Ketamine significantly reduced immobility time in FST
rent was significantly reduced in neurons of Camk2aCre+/AAV2B in both WTCre−/AAV2B and Camk2aCre+/AAV2B mice (n = 9–10; FST:
compared with WTAAV2B mice (n = 8–11 neurons, unpaired t-test, treatment: F1,34 = 15.6, p < 0.001).

(Fig. 2a). We have recently validated this viral construct by antidepressant effects similar to systemic administration
demonstrating a reduction of GluN2B immunoreactivity and [32, 34]. AAV2GluN2BshRNA was bilaterally infused into the
reduction of NMDA-induced inward current in recombined mPFC of adult, littermate wild-type controls (WTCre−/AAV2B)
layer V pyramidal neurons of mPFC [17]. The mPFC was and Camk2aCre+ mice (Camk2aCre+/AAV2B) and three-weeks
chosen as a target region as prior studies have reported that post-surgery, to allow for viral expression and recombina-
infusion of rapastinel or ketamine into the mPFC exerts tion, behavioral testing was conducted (Fig. 2f).
Cell-type specific modulation of NMDA receptors triggers antidepressant actions 5103

Histological and electrophysiological approaches con- both WTCre−/AAV2B and Gad1Cre+/AAV2B mice, rapastinel
firmed AAV2GluN2BshRNA expression, recombination, and significantly decreased immobility time and increased
cell-type-specific GluN2B KD. NMDA-induced inward female urine sniffing time in the FST and FUST, respec-
currents were measured in virus transduced cells of tively (Fig. 3d, e). Rapastinel also significantly decreased
WTCre−/AAV2B (EGFP + dsRed-expressing cells) and the latency to feed in the NSFT in both WTCre−/AAV2B and
recombined cells of Camk2aCre+/AAV2B mice (dsRed only Gad1Cre+/AAV2B mice (Fig. 3f). There were no significant
expressing cells) (Fig. 2c–e). WTCre−/AAV2B mice displayed effects on water sniffing time (Fig. 3e), or home cage
cells expressing both EGFP and dsRed (labeled in yellow), feeding and LMA (Supplementary Fig. 2c, d). As described
while slices from Camk2aCre+/AAV2B mice displayed a above for the Camk2aCre+/AAV2B mice, after a washout
subset of cells expressing only dsRed (Fig. 2c), confirming period we also evaluated the effect of ketamine in the same
cell-type-specific recombination. Consistent with our recent mice. Ketamine significantly decreased the immobility time
report validating NMDAR KD [17], NMDA-induced in WTCre−/AAV2B mice, but this effect was blocked in
inward current was significantly decreased in GluN2B KD Gad1Cre+/AAV2B mice (Fig. 3g). Together, these results
cells in Camk2aCre+/AAV2B mice compared with non- demonstrate that GluN2B containing NMDARs on gluta-
recombined cells, and rapastinel enhancement of NMDA- matergic neurons act as the initial cellular trigger for
induced inward currents was partially blocked (Fig. 2d, e). rapastinel, while blockade of NMDAR-GluN2B on
WTCre−/AAV2B and Camk2aCre+/AAV2B mice were admi- GABAergic neurons triggers ketamine’s antidepressant-like
nistered with saline or rapastinel (day 0), and evaluated for effects.
rapid and sustained antidepressant-like behavioral actions. To demonstrate the specificity of the GluN2B shRNA
This included the FST (a measure of behavioral despair) on knockdown, we also tested an AAV-scrambled shRNA
day 1, LMA on day 2, the FUST (a measure of reward construct in WT littermate and Camk2a-Cre mice
behavior) on day 3, and the NSFT (a measure of anxiety- (WTCre−/AAV-Scr and Camk2aCre+/AAV-Scr mice) (Supple-
like behavior) on day 4. A 1 mg/kg (i.v.) dose of rapastinel mentary Fig. 3a, b). As expected, yellow-labeled (EGFP +
was chosen based on our previous work demonstrating dsRed) cells were observed in mPFC slices from
rapid and sustained antidepressant-like effects in mice [13]. WTCre−/AAV-Scr mice and a subset of dsRed-expressing cells
Rapastinel significantly decreased immobility time in the were found only in Camk2aCre+/AAV-Scr mice, indicating
FST in WTCre−/AAV2B mice, but the effect was completely Cre-dependent recombination, EGFP excision and expres-
blocked in Camk2aCre+/AAV2B mice (Fig. 2g). In FUST, sion of scrambled shRNA only in Camk2aCre+/AAV-Scr mice
rapastinel significantly increased female urine sniffing (Supplementary Fig. 3c). In both WTCre−/AAV-Scr and
time in WTCre−/AAV2B mice without altering water Camk2aCre+/AAV-Scr mice, rapastinel significantly decreased
sniffing time, and the effect was completely blocked in immobility time, increased female urine sniffing time and
Camk2aCre+/AAV2AB mice (Fig. 2h). Rapastinel significantly decreased latency to feed in the FST, FUST and NSFT,
decreased latency to feed in the NSFT in WTCre−/AAV2B respectively (Supplementary Fig. 3d–f). There were no
mice, while the effect was blocked in Camk2aCre+/AAV2B significant differences observed in home cage feeding or
mice (Fig. 2i). There were no significant differences LMA (Supplementary Fig 3g, h).
observed in home cage feeding or LMA (Supplementary Fig.
2a, b). For comparison, we tested the effects of ketamine (10 GluN2A knockdown on glutamatergic neurons does
mg/kg, i.p.) in these same mice after a 9-day washout period not block the antidepressant-like effects of
(Fig. 2f). In contrast to blockade of the rapastinel response, rapastinel
ketamine significantly decreased the immobility time in
both WTCre−/AAV2B and Camk2aCre+/AAV2B mice in the FST To determine if there is specificity for GluN2B vs. another
(Fig. 2j). major subunit, GluN2A on Camk2a neurons in the
To examine the role of GluN2B subunits on GABAergic antidepressant-like actions of rapastinel, we used the
interneurons, we used the same design as above, infusing same approach but infused AAV2GluN2AshRNA into WT
AAV2GluN2BshRNA into WTCre−/AAV2B littermate controls or littermates and Camk2a-cre mice (WTCre−/AAV2A and
Gad1-Cre mice (Gad1Cre+/AAV2B) (Fig. 3a–c). Sections of Camk2aCre+/AAV2A mice) (Fig. 4a–f). For this construct,
mPFC from WTCre−/AAV2B mice displayed cells expressing mCherry was substituted for dsRed to enhance the visuali-
both EGFP and dsRed (labeled in yellow), while slices from zation of recombined cells. Immunohistochemistry revealed
Gad1Cre+/AAV2B mice displayed a subset of cells expressing yellow (EGFP + mCherry) labeled cells in mPFC slices
only dsRed (Fig. 3b), confirming cell-type-specific recom- from WTCre−/AAV2A mice, while a subset of mCherry only
bination. We have also validated this construct on func- expressing cells were found in Camk2aCre+/AAV2A mice,
tional knockdown of NMDA-induced inward current in confirming recombination (Fig. 4c). Further, there was a
recombined GABA interneurons of mPFC slices [17]. In significant reduction in NMDA-inward current in
5104 S. Pothula et al.

a GluN2B knockdown in Gad1-neurons: b WT


TATA-lox TATA-lox

CMV dsRed U6 CMV EGFP Sh-GluN2B hGH polyA

+ Gad1-Cre recombinase
CMV dsRed U6 Sh-GluN2B hGH polyA

TATA-lox

c Gad1 cre+

dsRED/EGFP
WTAAV2B-Sal Gad1Cre+/AAV2B-Sal
WTAAV2B-Rps or Ket Gad1Cre+/AAV2B-Rps or Ket

d FST-Rps e FUST-Rps
f NSFT-Rps
g FST-Ket
100 800 250

Latency to feed (sec)


250
**

Immobility time (sec)


** **
Immobility time (sec)

**
Sniffing time (sec)

200 * 80 ** ** 200
600
150 60 150
400
100 40 100
200
50 20 50

0 0 0 0
Water Urine

Fig. 3 Knockdown of GluN2B in the mPFC of Gad1-cre mice does significantly reduced immobility time (d), increased female urine
not block the antidepressant-like actions of rapastinel. a Schematic sniffing time (e), and reduced latency to feed (f) in the FST, FUST, and
showing pGluN2BshRNA construct before and after Gad1-Cre− to NSFT, respectively, both in WTCre−/AAV2B and Gad1Cre+/AAV2B mice
generate the active construct in GABA neurons. b Representative (n = 8–10; FST: treatment: F1,30 = 15.1, p < 0.001, genotype:
images of AAV2GluN2BshRNA virus expression, demonstrating fluor- F1,30 = 0.5, p = 0.49; NSFT: treatment: F1,30 = 24.4, p < 0.001,
escent labeling of WT neurons (yellow: colocalization of EGFP and genotype: F1,30 = 0.38, p = 0.54; *p < 0.05; **p < 0.01). g Ketamine
dsRed) and Cre-recombined GABA neurons (subset of only dsRed- significantly reduced immobility time in FST in WTCre−/AAV2B but this
expressing cells) in the mPFC of WTCre−/AAV2B and Gad1Cre+/AAV2B effect was blocked in Gad1Cre+/AAV2B mice (n = 8–9; FST: treatment:
mice, respectively. c Timeline for experimental approach. Rapastinel F1,29 = 8.04, p < 0.01; interaction: F1,29 = 4.48, p = 0.04).

recombined, mCherry only expressing pyramidal neurons in Fig. 4e). These results demonstrate that GluN2A subunits
Camk2aCre+/AAV2A mice compared with yellow-labeled on glutamatergic neurons are not involved in the rapid and
cells in WTCre−/AAV2A mice, demonstrating functional sustained antidepressant-like effects of rapastinel.
knockdown of GluN2A. In contrast to the results from
GluN2B KD, rapastinel significantly increased the Drd1-expressing pyramidal neurons are required for
NMDA-inward current in virus infected glutamatergic the antidepressant-like effects of rapastinel and
neurons of Camk2aCre+/AAV2A as well as WTCre−/AAV2A ketamine
mice (Fig. 4d, e).
Baseline behavioral tests conducted before rapastinel Pyramidal neurons in the mPFC display distinct molecular,
administration revealed that there were no significant dif- cellular, and morphological characteristics, with two well-
ferences between WTCre−/AAV2A and Camk2aCre+/AAV2A characterized subtypes, referred to as types 1 and 2, which
mice in preswim immobility time or center or total distance express Drd2 and Drd1, respectively [19–23]. Chronic
traveled in an OFT (Supplementary Fig. 4a–c). Rapastinel unpredictable stress or social defeat stress alters the activity
administration resulted in significant antidepressant-like and morphology of Drd1-expressing neurons, indicating a
actions in both WTCre−/AAV2A and Camk2aCre+/AAV2A role for this pyramidal cell subtype in stress responses
mice, including decreased immobility time in the FST [28, 35, 36]. Here we examined the role of Drd1-expressing
(Fig. 4g) and latency to feed in the NSFT (Fig. 4h) without pyramidal neurons in the actions of rapastinel. In pre-
any significant change in home cage feeding (Supplemen- liminary studies we found that deletion of GluN2B on
tary Fig. 4d). As expected, we also found that GluN2A KD Drd1-expressing type 2 neurons altered baseline behaviors
in Camk2aCre+/AAV2A mice had no effect on the response to (not shown), so as an alternative approach we examined the
ketamine in the FST or NSFT (Fig. 4i, j); there was no role of Drd1-expressing neurons by knockdown of Drd1 in
effect on home cage feeding in either group (Supplementary Camk2a-expressing pyramidal neurons. We used a
Cell-type specific modulation of NMDA receptors triggers antidepressant actions 5105

GluN2A knockdown in Camk2a-neurons:


a
TATA-lox TATA-lox
b
CMV mCherry U6 CMV EGFP sh-GluN2A hGH polyA

+Camk2a-Cre recombinase
CMV mCherry U6 sh-GluN2A hGH polyA
TATA-lox
GluN2A
c WT
d NMDA +Rps 0.1 μM
e Glutamatergic
neuron

2500
*

NMDA- inward current


ACSF

WT
2000 * Rps 100nM
1500

(pA)
Camk2a cre+ #
1000

500
GluN2A-KD

500 pA
0
WTAAV2A Camk2aCre+/AAV2A
mCherry/EGFP 1 min

AAV2-
f GluN2A- WTAAV2A-Sal
shRNA WTAAV2A-Rps or Ket
infusion Camk2aCre+/AAV2A-Sal
Camk2aCre+/AAV2A-Rps or Ket

g FST-Rps h NSFT-Rps i FST-Ket NSFT-Ket


j ** *
250 ** ** * * * * 600
*
Immobility time (Sec)

Latency to feed (Sec)


600 250
Immobility time (Sec)
Latency to feed (Sec)

200
** **
200
400 400
150 150
100 100
200 200
50 50
0 0 0 0
WT Camk2a-Cre WT Camk2a-Cre WT Camk2a-Cre WT Camk2a-Cre

Fig. 4 Knockdown of GluN2A in the mPFC of Camk2a-Cre paired t-test, *p < 0.05). f Schematic showing time line for experi-
mice does not block the antidepressant-like actions of rapastinel. mental approach. Rapastinel significantly reduced immobility time
a, b Schematics showing pGluN2AshRNA construct and removal of (g) and latency to feed (h) in the FST and NSFT, respectively in both
NMDARs (GluN2A) on glutamatergic neuron. c Representative ima- WTCre−/AAV2A and Camk2aCre+/AAV2A mice compared with saline
ges of AAV2GluN2AshRNA virus expression showing fluorescent labeling controls. Data expressed as Mean ± SEM (n = 8–10 per group, two-
of WT neurons (yellow: colocalization of EGFP and mCherry fluor- way ANOVA with Tukey’s multiple comparisons; FST: treatment:
escence) and Cre-recombined pyramidal neurons (subset of only F1,32 = 20.6, p < 0.001, genotype: F1,32 = 0.185, p = 0.67, interac-
mCherry-labeled neurons) in the mPFC of WTCre−/AAV2A and tion: F1,32 = 0.183, p = 0.67; NSFT: treatment: F1,32 = 17.5, p <
Camk2aCre+/AAV2A mice, respectively. d Representative traces of 0.001, genotype: F1,32 = 0.21, p = 0.65, interaction: F1,32 = 0.025,
NMDA-inward currents in WT and GluN2A KD glutamatergic neu- p = 0.88; *p < 0.05; **p < 0.01). Ketamine significantly reduced
rons of WTCre−/AAV2A and Camk2aCre+/AAV2A mice before and after immobility time (i) and latency to feed (j) in the FST and NSFT,
rapastinel (100 nM) treatment. e NMDA-inward current was sig- respectively, in both WTCre−/AAV2A and Camk2aCre+/AAV2A mice (n =
nificantly reduced in neurons of Camk2aCre+/AAV2A compared with 8–9 per group, two-way ANOVA with Tukey’s multiple comparisons;
WTCre−/AAV2A mice (n = 10–11 neurons, unpaired t-test, #p < 0.05), FST: treatment: F1,31 = 26.9, p < 0.001; NSFT: treatment: F1,31 =
and rapastinel significantly enhanced NMDA-inward current in both 25, p < 0.001).
WTCre−/AAV2A and Camk2aCre+/AAV2A mice (n = 10–11 neurons,

previously described Cre-dependent Drd1 miRNA virus, of pyramidal neurons in Camk2aCre+/AAV-Drd1 mice (Fig. 5b,
AAV-DIO-EmGFP-Drd1amiR, infused into Camk2aCre+ c). Analysis of baseline behavior showed no significant
mice (referred to as Camk2aCre+/AAV-Drd1 mice) (Fig. 5a); differences in Camk2aCre+/AAV-Drd1 mice in preswim
functional knockdown of Drd1 using this virus was vali- immobility time or total distance traveled in the OFT,
dated in an earlier study [28]. Control, WT mice were although there was a significant reduction of time spent in
infused with AAV-Camk2a-EGFP (referred as WTCre−/AAV). center zone (Supplementary Fig. 5a–c). Rapastinel sig-
Immunohistochemistry shows Cre-independent EGFP nificantly reduced the immobility time and latency to feed
labeling in WTCre−/AAV and Cre-dependent EmGFP labeling in WTCre−/AAV mice, but these effects were completely
5106 S. Pothula et al.

a Drd1 knockdown in Camk2a-neurons: b WT Camk2a cre+


Double floxed inverted ORF (DIO)
miRNA
EmGFP
EF1a Drd1a-

lox2272 + Cre recombinase


loxP
EmGFP
Drd1a-
EF1a EmGFP
miRNA
c

WTAAVDrd1-Sal Camk2aCre+/AAVDrd1-Sal
WTAAVDrd1-Rps or Ket Camk2aCre+/AAVDrd1-Rps or Ket

d FST-Rps e NSFT-Rps f FST-Ket g NSFT-Ket


250 600
200 600
200
150 400
400 150
100
100
200 200
50 50

0 0
0 0

Fig. 5 Knockdown of Drd1 in the mPFC of Camk2a-Cre mice ANOVA with Tukey’s multiple comparisons (n = 7–12 per group;
blocks antidepressant-like actions of rapastinel. a Schematic show- FST: treatment: F1,32 = 6.03, p < 0.05, genotype: F1,32 = 5.06,
ing the viral construct of AAV-DIO-EmGFP-Drd1amiR. p = 0.03, interaction: F1,32 = 4.44, p = 0.04; NSFT: treatment:
b Representative images of AAV-Camk2a-EGFP and AAV-DIO- F1,32 = 15.3, p < 0.001, genotype: F1,32 = 3.4, p = 0.07, interaction:
EmGFP-Drd1amiR virus expression, showing EGFP and F1,32 = 2.12, p = 0.16; *p < 0.05; **p < 0.01; ***p < 0.001). Keta-
EmGFP fluorescent labeling of neurons in the mPFC of WTcre−/AAV mine significantly reduced immobility time (f) and latency to feed (g) in
and Camk2aCre+/AAV-Drd1 mice, respectively. c Schematic showing the the FST and NSFT, respectively, in WTcre−/AAV but these effects were
timeline for experimental approach. Rapastinel significantly reduced blocked in Camk2aCre+/AAV-Drd1 mice (FST: treatment: F1,32 = 18.3,
immobility time (d) and latency to feed (e) in the FST and NSFT, p < 0.001, genotype: F1,32 = 12.9, p = 0.001, interaction: F1,32 =
respectively, in WTcre−/AAV but these effects were blocked in Cam- 8.38, p = 0.007; NSFT: treatment: F1,32 = 11.4, p = 0.002, genotype:
k2aCre+/AAV-Drd1 mice. Data expressed as Mean ± SEM, two-way F1,32 = 3.87, p = 0.06, interaction: F1,32 = 0.92, p = 0.34).

blocked in Camk2aCre+/AAV-Drd1 mice (Fig. 5d, e); there in the actions of rapastinel using a pharmacological
were no significant differences in home cage feeding approach. Pre-infusion of a selective Drd1 (SCH39166,
(Supplementary Fig. 5d). After a two-week washout, we 500 ng per side), but not Drd2 (L741,626, 700 ng per side)
found that the antidepressant actions of ketamine on antagonist into mPFC blocked the antidepressant-like
immobility time in the FST and latency to feed in the NSFT behavioral actions of rapastinel in the FST and NSFT
were also completely blocked in the Camk2aCre+/AAV-Drd1 (Fig. 6a–e). There were no significant differences in home
mice (Fig. 5f, g); there were no significant differences in cage feeding or LMA in response to rapastinel with or
home cage feeding (Supplementary Fig. 5e). These findings without SCH39166 (Supplementary Fig. 6a, b) or L741,626
are consistent with recent optogenetic and chemogenetic (Supplementary Fig. 6c, d).
studies demonstrating that Drd1-expressing neurons are
necessary and sufficient for the rapid antidepressant actions
of ketamine [37]. Discussion
Previous studies report that rapastinel [38] as well as
ketamine increase dopamine efflux in the mPFC [14, 15], Research in the past decade has generated tremendous
and we have reported that the actions of ketamine are interest in the utility of NMDAR modulators in depression.
blocked by infusion of a Drd1 antagonist into the PFC [37]. Both positive and negative modulation of NMDARs by
Based on these findings, we further tested the role of Drd1 rapastinel and ketamine, respectively, exert rapid and
Cell-type specific modulation of NMDA receptors triggers antidepressant actions 5107

a Cannulaon
Drug treatment FST LMA NSFT
surgery
//
-15/-16 Day 1 2 Food 3
days SCH39166/L-741626 10-15 min Rapasnel (i.v)
deprivaon
(mPFC)

Sal SCH39166 / Sal L741,626 / Sal


Rps SCH39166 / Rps L741,626 / Rps
FST NSFT FST NSFT
b c d e * * *
0.1
200 600 ** ** 200 * ** 600

Latency to feed (Sec)


Latency to feed (Sec)

Immobility time (Sec)


Immobility time (Sec)

** *
150 150
400 400
100 100
200 200
50 50

0 0 0 0
Saline SCH39166 Saline SCH39166 Saline L741626 Saline L741626

Fig. 6 Infusion of dopamine Drd1R, but not Drd2R, antagonists NSFT, rapastinel significantly reduced latency to feed in mice pre-
prevent the antidepressant-like actions of rapastinel. a Schematic infused with either vehicle or L471,626 (treatment: F1,20 = 19.4, p <
for the timeline for experimental approach. Prior infusion of Drd1R 0.001, infusion: F1,20 = 0.04, p = 0.84, interaction: F1,20 = 0.035,
antagonist SCH39166 (500 ng per side) into mPFC blocked the p = 0.85). g Proposed model of direct vs. indirect actions of rapastinel
antidepressant-like effects of rapastinel on immobility time (b) and and ketamine. The NMDAR antagonist ketamine blocks NMDARs on
latency to feed (c) in the FST and NSFT, respectively. Data expressed GABAergic interneurons to induce a disinhibition-mediated glutamate
as Mean ± SEM; two-way ANOVA with Tukey’s multiple compar- burst and indirect activation of glutamatergic neurons in mPFC. In
isons (n = 6–8 mice per group; FST: treatment: F1,27 = 11.9, p < contrast, the NMDAR positive modulator rapastinel directly activates
0.01, infusion: F1,27 = 0.805, p = 0.38, interaction: F1,27 = 6.74, the NMDARs on glutamatergic neurons without causing a glutamate
p = 0.02; NSFT: treatment: F1,23 = 13.4, p = 0.001, infusion: burst, demonstrating distinct initial cellular triggers for these rapid
F1,23 = 4.75, p = 0.04, interaction: F1,23 = 5.81, p = 0.02; *p < 0.05; acting antidepressants. Indirect and direct activation of excitatory
**p < 0.01). d In the FST, rapastinel significantly reduced immobility mPFC-VTA projection neurons by NMDAR antagonists and PAMs,
time in mice pre-infused with vehicle and showed a strong reduction respectively, could likely induce dopamine efflux in mPFC, which
in mice pre-infused with Drd2R antagonist 741,626 (700 ng per side) could activate the Drd1-expressing pyramidal neurons (in mPFC), a
(n = 6 mice per group; treatment: F1,20 = 16.2, p < 0.001, infusion: point of convergence for rapastinel and ketamine, resulting in rapid
F1,20 = 2.58, p = 0.12, interaction: F1,20 = 0.29, p = 0.6). e In the and sustained antidepressant-like actions of these agents.
5108 S. Pothula et al.

sustained antidepressant effects by activating convergent lack of effect on glutamate release. Together, these results
downstream signaling cascades [11–13], but NMDAR are consistent with an indirect and direct hypothesis for the
antagonists and NMDAR PAMs with opposing actions on actions for ketamine and rapastinel [30], respectively
NMDARs exert similar antidepressant effects and the initial (Fig. 6g).
cellular target for NMDAR PAMs have not been deter- Previous studies have reported that rapastinel non-
mined. To address these questions, here we used ketamine selectively enhances the activity of NMDAR GluN2
and rapastinel as tool compounds for these two types of subunits (A–D) and blockade of NMDARs prevents the
compounds. The results of the current study demonstrate antidepressant activity of rapastinel [8, 32]. However, the
that NMDAR PAM, rapastinel enhances NMDAR inward specific GluN2 subunits and cell type (excitatory or inhi-
currents without effecting glutamate release in the mPFC bitory neurons) mediating the antidepressant actions of
and that GluN2B subunits on glutamatergic, but not rapastinel are unknown. Using a brain region- and cell-type-
GABAergic neurons in mPFC are required for the specific approach we show that knockdown of GluN2B
antidepressant-like effects of rapastinel (Fig. 6g). In con- subunits on glutamatergic but not GABAergic interneurons
trast, ketamine inhibits NMDAR activity and enhances in the mPFC blocks the antidepressant-like behavioral
glutamate release, and selective knockdown of GluN2B actions of rapastinel in the FST, FUST, and NSFT. In
subunits on GABAergic but not glutamatergic neurons in contrast, the results show that after a washout period in the
mPFC blocks the antidepressant-like effects of ketamine, in same mice the antidepressant actions of ketamine are
agreement with a recent report [17]. The results also show blocked by GluN2B KD in GABAergic, but not glutama-
that Drd1-expressing subtype pyramidal neurons, and Drd1 tergic neurons consistent with our recent study [17]. The
receptors are required for the actions of rapastinel, as well as lack of baseline reduction in immobility time in FST after
ketamine, indicating that Drd1 is a point of convergence for GluN2B knockdown in our study differs from earlier stu-
these two agents. dies [40, 41] reporting a reduction in baseline immobility
Here we show that rapastinel exhibits an inverted-U dose time in FST after constitutive deletion of GluN2B in the
response on NMDA-induced inward current in mPFC pyr- forebrain [40]. This could be due to the robust increase in
amidal neurons, with low concentrations (100 nM) enhan- locomotion reported in mice with constitutive forebrain
cing NMDA-inward current, indicating a direct activation of deletion of GluN2B [40], which would produce an
excitatory neurons. A recent study reported that rapastinel antidepressant-like decrease in immobility time in the FST
(1 µM), as well as ketamine disinhibits pyramidal neurons and TST. In contrast, in the current study there were no
by acting on GABAergic neurons in hippocampus [39]. The effects of GluN2B KD on LMA in Camk2a-Cre mice.
possible explanation for this discrepancy is the higher Another study reported that viral-mediated deletion of
concentration of rapastinel used in their study because GluN2B in thalamocortical prelimbic mPFC circuits, which
30–100 nM of rapastinel was previously shown to be differs from the region and circuits targeted in the current
detected in mPFC after systemic injection and is sufficient study, also results in a baseline antidepressant phenotype
to induce antidepressant effects [32]. We have reported a [41].
similar disinhibition by ketamine [34], but not rapastinel The results also demonstrate a lack of off-target effects of
(current study). In contrast, a trend for increase in NMDA AAV viral-GluN2B shRNA as infusion of an AAV-
current observed in Sst interneurons even at 10 μM (data not scrambled virus did not block the antidepressant-like
shown). Further, rapastinel concentrations ≥1 μM have been effects of rapastinel. In addition, our study demonstrates
shown to partially inhibit NMDAR activity [32], although that knockdown of GluN2A subunits in glutamatergic
in the current study there was no effect observed on neurons does not block the antidepressant-like behavioral
NMDAR activity of pyramidal neurons with 10 μM actions of rapastinel. We have also reported that the actions
rapastinel. of ketamine on GABAergic neurons are mediated by
Here we confirm that ketamine produces a rapid, but GluN2B but not GluN2A [17]. In support of these beha-
transient increase in extracellular glutamate release in vioral results, electrophysiology studies demonstrate that
mPFC [14] and also provide evidence that rapastinel, at knockdown of GluN2B, but not GluN2A partially blocks
doses which produce an antidepressant-like response in rapastinel enhancement of NMDA-induced inward currents
rodents, has no effect on extracellular glutamate. Consistent in layer V pyramidal neurons of mPFC. The reason for this
with microdialysis results, we also found that ketamine, but in vivo selectivity, in spite of nonselective GluN2 subunit
not rapastinel increased glutamate signaling measured by effects in vitro [32] is unknown but could be due to higher
NMR spectroscopy in mPFC. It has been suggested that expression levels of GluN2B and/or different subunit
disinhibition-mediated glutamate efflux underlies the psy- combinations or higher calcium influx mediated by GluN2B
chotomimetic and dissociative effects of ketamine [14], and or activity patterns of principal neurons in vivo. Previous
absence of these side effects for rapastinel may be due to its studies have reported reductions of GluN2B levels and
Cell-type specific modulation of NMDA receptors triggers antidepressant actions 5109

NMDAR binding in PFC of depressed individuals [42, 43]. excitability [48] and synaptic plasticity [49, 50]. These
In addition, previous studies report that rapastinel increases findings suggest that Drd1-signaling could contribute to
GluN2B levels and GluN2B-mediated metaplasticity in upregulation of GluN2B expression, activity, and plasticity
mPFC [8], providing further evidence for a specific role of observed after rapastinel, as well as ketamine administration
GluN2B in the sustained antidepressant-like effects of [8]. Together the results indicate that Drd1-signaling is a
rapastinel. It will be interesting in future studies to deter- point of convergence for the molecular and behavioral
mine how NMDAR PAMs specifically increases GluN2B actions of both NMDAR positive modulation (rapastinel)
levels and whether this increase occurs in a cell-type- and inhibition (ketamine) (Fig. 6g). It will be interesting in
specific manner. future studies to identify the role of the mesolimbic dopa-
Optogenetic activation of principal neurons in mPFC mine system and downstream signaling pathways in the
induces rapid and sustained antidepressant effects [34]. synaptic and antidepressant-like behavioral actions of these
More recent optogenetic and chemogenetic studies rapid acting agents.
demonstrate that activity of Drd1-, but not Drd2-expressing Previous studies report that NMDAR PAM, rapastinel
neurons in the mPFC is necessary and sufficient for the exerts rapid and sustained antidepressant effects in both
antidepressant actions of ketamine, presumably via disin- stress models and naïve, non-stressed animals
hibition and subsequent glutamate and dopamine burst [37] [8, 12, 13, 33]. Although the current study does not employ
which activate these neurons to induce synaptic plasticity. stress models per se, the mice were single housed/socially
In support of this possibility, we have recently reported that isolated (at least 2–3 weeks post-surgery), and social iso-
chemogenetic inhibition of Drd1-expressing neurons blocks lation is reported to cause depressive-like molecular and
the effects of ketamine [37]. To further characterize the behavioral alterations [51–54]. Future studies are needed to
mechanisms underlying the actions of NMDAR antagonists determine if GluN2B and/or Drd1 on glutamatergic neurons
and PAM, we also examined the role of the Drd1-expres- mediate the actions of rapastinel and ketamine in patholo-
sing, type 2 subtype pyramidal neurons in the mPFC. Here gical stress models, such as social defeat and chronic
we show that knockdown of Drd1 on Camk2a-neurons in unpredictable stress. Nevertheless, the current findings
the mPFC blocks the antidepressant-like behavioral actions provide crucial mechanistic insights into the role of differ-
of rapastinel, as well as ketamine. Projection targets of these ent neuronal cell-types and receptor subtypes in initiating
mPFC Drd1-expressing neurons, including the basolateral and mediating the antidepressant-like behavioral actions of
amygdala (BLA) and bed nucleus of stria terminalis, have NMDAR enhancing vs. blocking agents.
been implicated in regulation of anxiety and depression-like In conclusion, the findings of the current study provide
behaviors [37, 44]. Moreover, optogenetic activation of direct evidence for previously unknown mechanisms
mPFC Drd1-neurons projecting to BLA induces anti- underlying the initial cellular trigger, neuronal cell-types
depressant effects [37], consistent with the possibility that and specific NMDA and dopamine receptor subtypes
BLA projecting Drd1-pyramidal neurons could mediate the involved in the antidepressant-like actions of NMDAR
actions of rapastinel, as well as ketamine. PAMs. Given the significant unmet need for safe and effi-
Both ketamine and rapastinel increase dopamine efflux in cacious rapid acting antidepressants, ongoing clinical trials
mPFC [14, 38] and identification of a role for Drd1- of NMDAR PAMs like AGN-241751 which are more
expressing neurons in the current as well as earlier studies potent and longer acting than rapastinel, and the need for
[37] suggests that dopamine signaling is involved in the additional rapid acting antidepressants without the side
actions of ketamine and rapastinel. In support of this pos- effect profile of ketamine, it is important to understand the
sibility, we have recently reported that pharmacological cellular trigger and mechanisms underlying the actions of
blockade of Drd1 receptors blocks the antidepressant NMDAR PAMs. The results reveal both unique as well as
actions of ketamine [37]. Here we show that pharmacolo- convergent mechanisms underlying the antidepressant-like
gical blockade of Drd1, but not Drd2, in the mPFC blocks actions of NMDAR positive modulation vs. antagonism.
the antidepressant-like actions of rapastinel. The mechan- Previous reports of altered mPFC dopamine and
isms underlying the increase in dopamine efflux is unclear Drd1 synaptic transmission in chronic stress models com-
but it could likely be possible that ketamine indirectly bined with the current findings indicate that Drd1-expres-
activate excitatory mPFC-VTA neuronal projections, sing neurons could play a crucial role in both the
implicated in depression [45], via disinhibition in mPFC pathophysiology of depression, as well as the response to
and rapastinel could directly activate these projections rapid acting antidepressants. Further characterization of the
(Fig. 6g). Also, mechanisms underlying these Drd1R- mPFC neuronal cell-types and circuits using cellular and
mediated actions are unclear, but previous studies demon- molecular approaches will provide novel and promising
strate that Drd1-signaling increases GluN2B and AMPAR molecular targets for the development of safer and effica-
expression [46, 47] and increases mPFC pyramidal neurons cious rapid antidepressants.
5110 S. Pothula et al.

Acknowledgements This research was supported by National Institute 10. Preskorn S, Macaluso M, Mehra DO, Zammit G, Moskal JR,
of Mental Health grants MH093897 and MH105910 (RSD) and a Burch RM, et al. Randomized proof of concept trial of GLYX-13,
research grant from Allergan Inc. New Jersey. We thank Xiao Yuan Li an N-methyl-D-aspartate receptor glycine site partial agonist, in
for her help with genotyping of mouse lines. We thank Jan Kehr major depressive disorder nonresponsive to a previous anti-
(Pronexus Analytical AB, Stockholm, Sweden) for performing the depressant agent. J Psychiatr Pract. 2015;21:140–9.
microdialysis experiments. Dr. Duman passed away on February 1st 11. Li N, Lee B, Liu RJ, Banasr M, Dwyer JM, Iwata M, et al. mTOR-
2020. This article is dedicated to Dr. Duman in memory of his men- dependent synapse formation underlies the rapid antidepressant
torship and scientific leadership. effects of NMDA antagonists. Science. 2010;329:959–64.
12. Liu RJ, Duman C, Kato T, Hare B, Lopresto D, Bang E, et al.
Author contributions SP, TK, PB, and RSD designed the study. SP GLYX-13 produces rapid antidepressant responses with key
wrote the manuscript and RSD contributed to writing the manuscript. synaptic and behavioral effects distinct from ketamine. Neu-
SP, TK, RJL, and MW conducted experiments, analyzed data, and ropsychopharmacology. 2017;42:1231–42.
interpreted the results. GS, GMIC, and KLB designed glutamate 13. Kato T, Fogaca M, Deyama S, Li X, Fukumoto K, Duman R.
cycling experiment using MRS spectroscopy. GMIC and KLB con- BDNF release and signaling are required for the antidepressant
ducted glutamate cycling experiment. DG and RS are involved in actions of GLYX-13. Mol Psych. 2017;23:2007–17.
design and preparation of pGluN2BshRNA (dsRed version) and AAV- 14. Moghaddam B, Adams B, Verma A, Daly D. Activation of glu-
DIO-EmGFP-D1miR viruses. A-NS helped with experiments. All tamatergic neurotransmission by ketamine: a novel step in the
authors reviewed and approved the final manuscript. pathway from NMDA receptor blockade to dopaminergic and
cognitive disruptions associated with the prefrontal cortex. J
Neurosci. 1997;17:2921–7.
Compliance with ethical standards 15. Lorrain DS, Baccei CS, Bristow LJ, Anderson JJ, Varney MA.
Effects of ketamine and N-methyl-D-aspartate on glutamate and
Conflict of interest RSD has received consulting fees from Taisho, dopamine release in the rat prefrontal cortex: modulation by a
Johnson & Johnson, and Naurex, and grant support from Taisho, group II selective metabotropic glutamate receptor agonist
Johnson & Johnson, Naurex, Allergan, Navitor, Lundbeck, Relmada, LY379268. Neuroscience. 2003;117:697–706.
and Lilly. PB is an employee of Allergan Inc, New Jersey. All other 16. Homayoun H, Moghaddam B. NMDA receptor hypofunction
authors declare no competing interests. produces opposite effects on prefrontal cortex interneurons and
pyramidal neurons. J Neurosci. 2007;27:11496–500.
Publisher’s note Springer Nature remains neutral with regard to 17. Gerhard D, Pothula S, Liu RJ, Wu M, Li XY, Girgenti MJ, et al.
jurisdictional claims in published maps and institutional affiliations. GABA interneurons are the initial trigger for the rapid anti-
depressant actions of ketamine. J Clin Invest. 2020;130:1336–49.
18. Preskorn S, Baker B, Kolluri S, Menniti FS, Krams M, Landen
References JW. An innovative design to establish proof of concept of the
antidepressant effects of the NR2B subunit selective N-Methyl-D-
1. Hasin DS, Sarvet AL, Meyers JL, Saha TD, Ruan WJ, Stohl M, et al. Aspartate antagonist, CP-101,606, in patients with treatment-
Epidemiology of adult DSM-5 major depressive disorder and Its refractory major depressive disorder. J Clin Psychopharmacol.
specifiers in the United States. JAMA Psychiatry. 2018;75:336–46. 2008;28:631–7.
2. Fava M. Diagnosis and definition of treatment-resistant depres- 19. Yang CR, Seamans JK, Gorelova N. Electrophysiological and
sion. Biol Psychiatry. 2003;53:649–59. morphological properties of layers V-VI principal pyramidal cells
3. Trivedi M, Rush AJ, Wisniewski SR, Nierenberg AA, Warden D, in rat prefrontal cortex in vitro. J Neurosci. 1996;16:1904–21.
Ritz L, STAR*D Study Team, et al. Evaluation of outcomes with 20. Santana N, Mengod G, Artigas F. Quantitative analysis of the
citalopram for depression using measurement-based care in expression of dopamine D1 and D2 receptors in pyramidal and
STAR*D: implications for clinical practice. Am J Psychiatry. GABAergic neurons of the rat prefrontal cortex. Cereb Cortex.
2006;163:28–40. 2009;19:849–60.
4. Berman RM, Cappiello A, Anand A, Oren DA, Heninger GR, 21. Wei X, Ma T, Cheng Y, Huang CCY, Wang X, Lu J, et al.
Charney DS, et al. Antidepressant effects of ketamine in depressed Dopamine D1 or D2 receptor-expressing neurons in the central
patients. Biol Psychiatry. 2000;47:351–4. nervous system. Addict Biol. 2018;23:569–84.
5. Abdallah CG, Sanacora G, Duman RS, Krystal JH. Ketamine and 22. Anastasiades PG, Boada C, Carter AG. Cell-Type-Specific D1
rapid-acting antidepressants: a window into a new neurobiology dopamine receptor modulation of projection neurons and inter-
for mood disorder therapeutics. Annu Rev Med. 2015;66:509–23. neurons in the prefrontal cortex. Cereb Cortex 2018;29:3224–42.
6. Sleigh J, Harvey M, Voss L, Denny B. Ketamine—more 23. Gee S, Ellwood I, Patel T, Luongo F, Deisseroth K, Sohal VS.
mechanisms of action than just NMDA blockade. Trends Anaesth Synaptic activity unmasks dopamine D2 receptor modulation of a
Crit Care. 2014;4:76–81. specific class of layer V pyramidal neurons in prefrontal cortex. J
7. Morgan CJA, Curran HV. Ketamine use: a review. Addiction. Neurosci. 2012;32:4959–71.
2012;107:27–38. 24. Kehr J, Yoshitake T. Monitoring brain chemical signals by
8. Moskal JR, Burch R, Burgdorf JS, Kroes RA, Stanton PK, Dis- microdialysis. In: Grimes CA, Dickey EC, Pishko MV, editors.
terhoft JF, et al. GLYX-13, an NMDA receptor glycine site Encyclopedia of sensors, Vol. 6. USA: American Scientific Pub-
functional partial agonist enhances cognition and produces anti- lishers; 2006. p. 287–312.
depressant effects without the psychotomimetic side effects of 25. Kehr J, Yoshitake T. Derivatization chemistries for improved
NMDA receptor antagonists. Expert Opin Investig Drugs. 2014; detection of monoamine neurotransmitters and their metabolites in
23:243–54. microdialysis samples by liquid chromatography with fluores-
9. Burch R, Preskorn S, Bastin L, Yu W, Burgdorf J, Moskal J. cence detection and mass spectrometry. In: Wilson G, Michael A,
Adjunctive GLYX-13 induces prolonged efficacy in subjects with editors. Compendium of in-vivo monitoring in real-time molecular
major depressive disorder (MDD). Neuropsychopharmacology. neuroscience, Vol. 2, microdialysis and sensing of neural tissues.
2014;39:S335. Singapore: World Scientific Publishing; 2017. p. 193–216.
Cell-type specific modulation of NMDA receptors triggers antidepressant actions 5111

26. Chowdhury GM, Behar KL, Cho W, Thomas MA, Rothman DL, 39. Widman AJ, McMahon LL. Disinhibition of CA1 pyramidal cells
Sanacora G. (1)H-[(1)(3)C]-nuclear magnetic resonance spectro- by low-dose ketamine and other antagonists with rapid anti-
scopy measures of ketamine’s effect on amino acid neuro- depressant efficacy. Proc Natl Acad Sci USA. 2018;115:E3007–16.
transmitter metabolism. Biolo Psychiatry. 2012;71:1022–25. 40. Miller OH, Yang L, Wang CC, Hargroder EA, Zhang Y,
27. Chowdhury GM, Zhang J, Thomas M, et al. Transiently increased Delpire E. et al. GluN2B-containing NMDA receptors regulate
glutamate cycling in rat PFC is associated with rapid onset of depression-like behavior and are critical for the rapid anti-
antidepressant-like effects. Mol Psychiatry. 2017;22:120–6. depressant actions of ketamine. Elife. 2014;3:e03581. https://doi.
28. Shinohara R, Taniguchi M, Ehrlich AT, Yokogawa K, Deguchi Y, org/10.7554/eLife.03581.
Cherasse Y, et al. Dopamine D1 receptor subtype mediates acute 41. Miller OH, Bruns A, Ben Ammar I, Mueggler T, Hall BJ.
stress-induced dendritic growth in excitatory neurons of the Synaptic regulation of a thalamocortical circuit controls
medial prefrontal cortex and contributes to suppression of stress depression-related behavior. Cell Rep. 2017;20:1867–80.
susceptibility in mice. Mol Psychiatry. 2018;23:1717–30. 42. Feyissa AM, Chandran A, Stockmeier CA, Karolewicz B.
29. Wohleb ES, Wu M, Gerhard DM, Taylor SR, Picciotto MR, Reduced levels of NR2A and NR2B subunits of NMDA receptor
Alreja M, et al. GABA interneurons mediate the rapid and PSD-95 in the prefrontal cortex in major depression. Prog
antidepressant-like effects of scopolamine. J Clin Invest. Neuro-Psychopharmacol Biol Psychiatry. 2009;33:70–5.
2016;126:2482–94. 43. Nowak G, Ordway GA, Paul IA. Alterations in the N-methyl-d-
30. Duman RS, Shinohara R, Fogaça MV, Hare B. Neurobiology of asparatate (NMDA) receptor complex in the frontal cortex of
rapid-acting antidepressants: convergent effects on GluA1- suicide victims. Brain Res. 1995;675:157–64.
synaptic function. Mol Psychiatry. 2019;24:1816–32. 44. Land BB, Narayanan NS, Liu RJ, Gianessi CA, Brayton CE,
31. White PF, Schüttler J, Shafer A, Stanski DR, Horai Y, Trevor AJ. Grimaldi D, et al. Medial prefrontal D1 dopamine neurons control
Comparative pharmacology of the ketamine isomers: studies in food intake. Nat Neurosci. 2014;17:248–53.
volunteers. Br J Anaesth. 1985;57:197–203. 45. Chaudhury D, Walsh JJ, Friedman AK, Juarez B, Ku SM, Koo
32. Donello JE, Banerjee P, Li XY, Guo YX, Yoshitake T, Zhang XL, JW, et al. Rapid regulation of depression-related behaviors by
et al. Positive N-Methyl-D-Aspartate receptor modulation by control of midbrain dopamine neurons. Nature. 2013;493:532–6.
rapastinel promotes rapid and sustained antidepressant-like 46. Gao C, Wolf ME. Dopamine receptors regulate NMDA receptor
effects. Int J Neuropsychopharmacol. 2019;22:247–59. surface expression in prefrontal cortex neurons. J Neurochem.
33. Burgdorf J, Zhang XL, Nicholson KL, Balster RL, Leander JD, 2008;106:2489–501.
Stanton PK, et al. GLYX-13, an NMDA receptor glycine-site 47. Sun X, Zhao Y, Wolf ME. Dopamine receptor stimulation mod-
functional partial agonist, induces antidepressant-like effects ulates AMPA receptor synaptic insertion in prefrontal cortex
without ketamine-like side effects. Neuropsychopharmacology. neurons. J Neurosci. 2005;25:7342–51.
2013;38:729–42. 48. Lavin A, Grace AA. Stimulation of D1-type dopamine receptors
34. Fuchikami M, Thomas A, Liu R, Wohleb ES, Land BB, DiLeone enhances excitability in prefrontal cortical pyramidal neurons in a
RJ, et al. Optogenetic stimulation of infralimbic PFC reproduces state-dependent manner. Neuroscience. 2001;104:335–46.
ketamine’s rapid and sustained antidepressant actions. Proc Natl 49. Gurden H, Takita M, Jay TM. Essential role of D1 but not D2
Acad Sci USA. 2015;112:8106–11. receptors in the NMDA receptor‐dependent long‐term potentiation
35. Tanaka K, Furuyashiki T, Kitaoka S, Senzai Y, Imoto Y, Segi- at hippocampal‐prefrontal cortex synapses in vivo. J Neurosci.
Nishida E, et al. Prostaglandin E2-mediated attenuation of 2000;20:RC106.
mesocortical dopaminergic pathway is critical for susceptibility to 50. Jay TM. Dopamine: a potential substrate for synaptic plasticity
repeated social defeat stress in mice. J Neurosci. and memory mechanisms. Prog Neurobiol. 2003;69:375–90.
2012;32:4319–29. 51. Berry A, Bellisario V, Capoccia S, Tirassa P, Calza A, Alleva E,
36. Anderson EM, Gomez D, Caccamise A, McPhail D, Hearing M. et al. Social deprivation stress is a triggering factor for the
Chronic unpredictable stress promotes cell-specific plasticity in emergence of anxiety- and depression-like behaviours and leads to
prefrontal cortex D1 and D2 pyramidal neurons. Neurobiol Stress. reduced brain BDNF levels in C57BL/6J mice. Psychoneur-
2019;10:100152. oendocrinology. 2012;37:762–72.
37. Hare BD, Shinohara R, Liu RJ, Pothula S, DiLeone RJ, Duman 52. Ieraci A, Mallei A, Popoli M. Social isolation stress induces
RS. Optogenetic stimulation of medial prefrontal cortex Drd1 anxious-depressive-like behavior and alterations of
neurons produces rapid and long-lasting antidepressant effects. neuroplasticity-related genes in adult male mice. Neural Plast.
Nat Commun. 2019;10:223. 2016;2016:6212983.
38. Rajagopal L, Huang M, Li J, He W, Soni D, Banerjee P, et al. 53. Mumtaz F, Khan MI, Zubair M, Dehpour AR. Neurobiology and
Rapastinel, a novel NMDA receptor modulator, produces pro- consequences of social isolation stress in animal model-A com-
longed rescue of subchronic phencyclidine-induced deficits in prehensive review. Biomed Pharmacother. 2018;105:1205–22.
episodic memory as well as other beneficial effects on cognitive 54. Haj-Mirzaian A, Amini-Khoei H, Haj-Mirzaian A, Amiri S,
function in a rapamycin sensitive manner. 341.12 / VV3. 2017 Ghesmati M, Zahir M, et al. Activation of cannabinoid receptors
Neuroscience Meeting Planner. Washington, DC: Society for elicits antidepressant-like effects in a mouse model of social iso-
Neuroscience; 2017. lation stress. Brain Res Bull. 2017;130:200–10.

You might also like