ArticlePDF Available

The Prodrug 4-Chlorokynurenine Causes Ketamine-Like Antidepressant Effects, but Not Side Effects, by NMDA/GlycineB-Site Inhibition

Authors:

Abstract and Figures

Currently approved antidepressant drug treatment typically takes several weeks to be effective. The non-competitive N-methyl-D-aspartate (NMDA) receptor antagonist ketamine has shown efficacy as a rapid-acting treatment for depression, but its use is associated with significant side effects. We assessed effects following blockade of the glycineB co-agonist site of the NMDA receptor by the selective full antagonist 7-Cl-kynurenic acid (7-Cl-KYNA), delivered by systemic administration of its brain-penetrant prodrug 4-Cl-kynurenine (4-Cl-KYN) in mice. Following administration of 4-Cl-KYN, 7-Cl-KYNA was promptly recovered extracellularly in hippocampal microdialysate of freely-moving animals. The behavioral responses of the animals were assessed using measures of ketamine-sensitive antidepressant efficacy (including the 24-hour forced swim test, learned helplessness test, and novelty-suppressed feeding test). In these tests, distinct from fluoxetine, and similar to ketamine, 4-Cl-KYN administration resulted in rapid, dose-dependent and persistent antidepressant-like effects following a single treatment. The antidepressant effects of 4-Cl-KYN were prevented by pre-treatment with glycine or the α-amino-3-hydroxy-5-methyl-4-isoxazolepropionic acid (AMPA) receptor antagonist 2,3-dihydroxy-6-nitro-7-sulfamoyl-benzo[f]quinoxaline-2,3-dione (NBQX). 4-Cl-KYN administration was not associated with the rewarding and psychotomimetic effects of ketamine, and did not induce locomotor sensitization or stereotypic behaviors. Our results provide further support for antagonism of the glycineB site for the rapid treatment of treatment-resistant depression and indicate that the prodrug approach using 4-Cl-KYN holds promise for use in humans, without the negative side effects seen with ketamine or other channel blocking NMDA receptor antagonists. The American Society for Pharmacology and Experimental Therapeutics.
Content may be subject to copyright.
1521-0103/355/1/7685$25.00 http://dx.doi.org/10.1124/jpet.115.225664
THE JOURNAL OF PHARMACOLOGY AND EXPERIMENTAL THERAPEUTICS J Pharmacol Exp Ther 355:7685, October 2015
Copyright ª2015 by The American Society for Pharmacology and Experimental Therapeutics
The Prodrug 4-Chlorokynurenine Causes Ketamine-Like
Antidepressant Effects, but Not Side Effects, by
NMDA/Glycine
B
-Site Inhibitions
Panos Zanos, Sean C. Piantadosi, Hui-Qiu Wu, Heather J. Pribut, Matthew J. Dell,
Adem Can, H. Ralph Snodgrass, Carlos A. Zarate, Jr., Robert Schwarcz,
and Todd D. Gould
Department of Psychiatry (P.Z., S.C.P., H.-Q.W., H.J.P., M.J.D., A.C., R.S., T.D.G.), Maryland Psychiatric Research Center
(H.-Q.W., R.S.), Department of Pharmacology (R.S., T.D.G.), Department of Anatomy and Neurobiology (T.D.G.), University of
Maryland School of Medicine, Baltimore, Maryland; VistaGen Therapeutics, Inc., San Francisco, California (H.R.S.); Experimental
Therapeutics and Pathophysiology Branch, Intramural Research Program, National Institute of Mental Health, National Institutes
of Health, Bethesda, Maryland (C.A.Z.)
Received May 3, 2015; accepted July 29, 2015
ABSTRACT
Currently approved antidepressant drug treatment typically
takes several weeks to be effective. The noncompetitive
N-methyl-D-aspartate (NMDA) receptor antagonist ketamine has
shown efficacy as a rapid-acting treatment of depression, but its
use is associated with significant side effects. We assessed
effects following blockade of the glycine
B
co-agonist site of the
NMDA receptor, located on the GluN1 subunit, by the selective
full antagonist 7-chloro-kynurenic acid (7-Cl-KYNA), delivered
by systemic administration of its brain-penetrant prodrug
4-chlorokynurenine (4-Cl-KYN) in mice. Following administration
of 4-Cl-KYN, 7-Cl-KYNA was promptly recovered extracellularly
in hippocampal microdialysate of freely moving animals. The
behavioral responses of the animals were assessed using
measures of ketamine-sensitive antidepressant efficacy (in-
cluding the 24-hour forced swim test, learned helplessness
test, and novelty-suppressed feeding test). In these tests,
distinct from fluoxetine, and similar to ketamine, 4-Cl-KYN
administration resulted in rapid, dose-dependent and persistent
antidepressant-like effects following a single treatment. The
antidepressant effects of 4-Cl-KYN were prevented by
pretreatment with glycine or the a-amino-3-hydroxy-5-methyl-
4-isoxazolepropionic acid (AMPA) receptor antagonist 2,3-dihydroxy-
6-nitro-7-sulfamoyl-benzo[f]quinoxaline-2,3-dione(NBQX).4-Cl-KYN
administration was not associated with the rewarding and
psychotomimetic effects of ketamine, and did not induce
locomotor sensitization or stereotypic behaviors. Our results
provide further support for antagonism of the glycine
B
site for the
rapid treatment of treatment-resistant depression without the
negative side effects seen with ketamine or other channel-
blocking NMDA receptor antagonists.
Introduction
Although interventions for major depressive disorder
(MDD) such as pharmacotherapies and cognitive behavioral
psychotherapies are available, more than 30% of patients
remain treatment-resistant. Even when effective, currently
used monoaminergic drugs often take up to several months to
exert their full therapeutic effects (Rush et al., 2006). More-
over, compelling evidence suggests a key role for the gluta-
matergic system in mood disorders (Popoli et al., 2012). Tissue
glutamate levels are higher in the brain of depressed patients
compared with healthy individuals (Sanacora et al., 2012),
and evidence indicates that the N-methyl-D-aspartate
(NMDA) subtype of glutamate receptors can be successfully
targeted for the treatment of MDD (Pittenger et al., 2007).
Placebo-controlled trials have demonstrated rapid-acting
antidepressant effects, within hours, of subanesthetic doses
of the noncompetitive NMDA-receptor antagonist ketamine in
treatment-resistant depressed patients [see Duman (2014)].
Moreover, antidepressant effects of ketamine have been
demonstrated in several relevant tests in experimental ani-
mals [see Browne and Lucki (2013)]. However, ketamines
potential as a long-term antidepressant medication is limited
by its addictive properties and its anesthetic, cognitive, and
psychotomimetic side effects (Krystal et al., 1994).
This study was supported by the National Institutes of Health National
Institute of Mental Health (Grant No. MH099345). This work was supported
by the Intramural Research Program of the National Institutes of Health.
dx.doi.org/10.1124/jpet.115.225664.
sThis article has supplemental material available at jpet.aspetjournals.org.
ABBREVIATIONS: 4-Cl-KYN, 4-chlorokynurenine; 7-Cl-KYNA, 7-chlorokynurenic acid; AMPA, a-amino-3-hydroxy-5-methyl-4-isoxazolepropionic
acid; ANOVA, analysis of variance; FLX, fluoxetine; FST, forced-swim test; GLYX-13, (S)-N-[(2S,3R)-1-amino-3-hydroxy-1-oxobutan-2-yl]-1-[(S)-1-
((2S,3R)-2-amino-3-hydroxybutanoyl)pyrrolidine-2-carbonyl]pyrrolidine-2-carboxamide; MDD, major depressive disorder; MK-801, [5R,10S]-[1]-5-
methyl-10,11- dihydro-5H-dibenzo[a,d]cyclohepten-5,10-imine; NBQX, 2,3-dihydroxy-6-nitro-7-sulfamoyl-benzo[f]quinoxaline-2,3-dione; NMDA,
N-methyl-D-aspartate; NSF, novelty-suppressed feeding test; TST, tail-suspension test.
76
http://jpet.aspetjournals.org/content/suppl/2015/08/11/jpet.115.225664.DC1.html
Supplemental material to this article can be found at:
at ASPET Journals on April 4, 2016jpet.aspetjournals.orgDownloaded from
Activity of the NMDA receptor requires binding of glycine or
D-serine to an obligatory co-agonist (glycine
B
) site, which is
located on the GluN1 subunit (Leeson and Iversen, 1994;
Danysz and Parsons, 1998). Although efficacy of glycine
B
receptor blockade by a full antagonist has been demonstrated
in tests in mice sensitive to selective serotonin reuptake
inhibition (Przegalinski et al., 1998; Poleszak et al., 2007),
limited work has assessed actions in ketamine-sensitive tests
predictive of rapid-acting antidepressant efficacy. 7-Chloro-
kynurenic acid (7-Cl-KYNA) is a potent and highly selective
competitive glycine
B
receptor antagonist that has been widely
used as a pharmacological probe to study the biology of the
NMDA receptor (Kemp et al., 1988). Although the therapeutic
use of 7-Cl-KYNA is limited by its poor penetration of the
blood-brain barrier, it can be enzymatically formed from its
prodrug 4-chlorokynurenine [4-Cl-KYN; Salituro et al. (1994)],
which readily enters the brain after systemic administration
(Hokari et al., 1996) and is then converted to 7-Cl-KYNA
within astrocytes [Wu et al. (1997); Fig. 1A].
In the present study, we evaluated the antidepressant
efficacy and side effect profile of 4-Cl-KYN in comparison with
ketamine and, in some experiments, with the selective
serotonin reuptake inhibitor fluoxetine (FLX). Our results
provide direct support for targeting the glycine
B
site in the
treatment of MDD and indicate that the prodrug approach
using 4-Cl-KYN holds promise for use in humans, without the
negative side effects seen with ketamine or related NMDA
receptor antagonists.
Materials and Methods
Animals
Male CD-1 mice (seven-weeks old on arrival; Charles River
Laboratories, MA) were housed in groups of four or five per cage with
a 12-hour light/dark cycle (lights on at 07:00). Food and water were
available ad libitum. Mice acclimatized to the new environment for
7 days prior to the start of the experiments. All experimental procedures
were approved by the University of Maryland, Baltimore Animal
Care and Use Committee and were conducted in full accordance with
the National Institutes of Health Guide for the Care and Use of
Laboratory Animals.
Drugs
Ketamine-HCl and glycine (Sigma-Aldrich, St. Louis, MO), NBQX,
and FLX (National Institute of Mental Health Chemical Synthesis
and Drug Supply Program) were dissolved in 0.9% saline. 4-Cl-KYN
(provided by VistaGen Therapeutics, Inc., South San Francisco, CA)
and 7-Cl-KYNA (Tocris Bioscience, Ellisville, MO) were suspended in
0.1 N NaOH until fully dissolved and neutralized with 0.1 N HCl
before injection. All drugs were administered intraperitoneally in
a volume of 7.5 ml/kg of body mass.
Measurement of 7-Cl-KYNA in Brain Microdialysate
Microdialysis in freely-moving mice was carried out as previously
described (Potter et al., 2010). Animals were anesthetized with chloral
hydrate (360 mg/kg) and mounted in a stereotaxic frame. A guide
cannula (outer diameter: 0.65 mm) was positioned over the dorsal
hippocampus (AP: 2.2 mm posterior to bregma, L: 2.0 mm from the
midline, V: 1.1 mm below the dura) and secured to the skull with an
anchor screw and acrylic dental cement. A concentric microdialysis
probe (membrane length: 1 mm; SciPro, Sanborn, NY) was then
inserted, extending 1 mm beyond the tip of the guide cannula. The
probe was subsequently connected to a microinfusion pump set to
a speed of 1 ml/min and perfused with Ringer solution containing
144 mM NaCl, 4.8 mM KCl, 1.2 mM MgSO
4
, and 1.7 mM CaCl
2
,
pH 6.7. Microdialysate samples were collected every 30 minutes for
eight hours. 4-Cl-KYN or 7-Cl-KYNA was administered 2 hours after
the initial collection of baseline fractions. 7-Cl-KYNA levels were
measured by high-performance liquid chromatography, using fluores-
cence detection (excitation wavelength: 344 nm; emission wavelength:
398 nm), as described (Lee and Schwarcz, 2001). Briefly, 20 mlof
the microdialysate were subjected to a 3-mm C18 reverse phase column
(80 4.6 mm; Thermo Scientific, Waltham, MA). 7-Cl-KYNA was
isocratically eluted at a flow rate of 1 ml/min using a mobile phase
containing 250 mM zinc acetate, 50 mM sodium acetate, and 8%
acetonitrile, pH 6.2. The retention time of 7-Cl-KYNA was 8 minutes.
Pharmacological Screening
Binding profiles and K
i
determination data of 7-Cl-KYNA and
4-Cl-KYN for a broad panel of receptors and channels were provided
by the National Institute of Mental Health Psychoactive Drug Screen-
ing Program (University of North Carolina, Chapel Hill, NC). For all
radioligand receptor assay methods, see the Psychoactive Drug
Screening Program web site (http://pdsp.med.unc.edu) as well as
previously published protocols (Besnard et al., 2012). Experimental
details for the binding assays are provided in Supplemental Table S2.
Fig. 1. Extracellular levels of 7-chorokynurenic acid studied by micro-
dialysis. (A) Schematic representation of 7-chlorok ynurenic acid (7-Cl-KYNA)
production from 4-chlorokynurenine (4-Cl-KYN) in the brain. 4-Cl-KYN
readily enters the brain from the circulation and is then converted to
7-Cl-KYNA by kynurenine aminotransferase (KAT) in astrocytes. (B)
Hippocampal microdialysis in freely-moving mice. The arrow indicates
intraperitoneal injection of 7-Cl-KYNA (25 mg/kg) or 4-Cl-KYN (25 mg/kg).
Data are the mean 6S.E.M. (n= 6/group). Inset: area under the curve
(AUC); unpaired Studentst-test. ***p,0.001.
Fast-Onset Antidepressant Effects of 4-Chlorokynurenine 77
at ASPET Journals on April 4, 2016jpet.aspetjournals.orgDownloaded from
Behavioral Tests
All behavioral experiments were performed during the light phase.
Ketamine was used as a positive control in all of the behavioral
experiments. The dose used routinely (10 mg/kg) was previously
shown to exert antidepressant effects in mice (Maeng et al., 2008;
Ma et al., 2013; Antony et al., 2014).
Forced-Swim Test. Saline, FLX (20 mg/kg), ketamine (10 mg/kg),
or 4-Cl-KYN (0.2, 1, 5, 25, or 125 mg/kg) was injected, and mice were
tested in the forced-swim test (FST) 1 hour or 24 hours postinjection.
During the FST, mice were subjected to a 6-minute swim session in
clear Plexiglas cylinders (30 cm height 20 cm diameter) filled with
15 cm of water (23 61°C). The FST was performed in normal light
conditions (800 lux). Sessions were recorded using a digital video-
camera. Immobility time, defined as passive floating with no addi-
tional activity other than that necessary to keep the head above water,
was scored for the last 4 minutes of the 6-minute test by a trained
observer blind to the treatment. In a separate cohort of animals,
glycine [1.6 g/kg, i.p.; Evoniuk et al. (1991); Javitt et al. (1999)] was
administered 10 minutes prior to injection of saline or 4-Cl-KYN. Since
a leading hypothesis of ketamine antidepressant action involves the
activation of AMPA receptors (Maeng et al., 2008; Li et al., 2010; Autry
et al., 2011), we also assessed the role of this receptor in the antidepres-
sant effects of 4-Cl-KYN by pretreatment with the AMPA receptor
antagonist NBQX (10 mg/kg) 5 minutes prior to injection of saline,
ketamine, or 4-Cl-KYN, and assessed behavior 1 hour later in the FST.
Tail-Suspension Test. The TST was performed as previously
described by Can et al. (2012). Animals were tested 1 hour post-
injection of saline, ketamine (10mg/kg), or 4-Cl-KYN (5, 25, or 125mg/kg).
Mice were individually suspended from the tail in suspension boxes
(55 cm height 60 cm width 11.5 cm depth; Four-Hour Day,
Baltimore, MD) for 6 minutes using a 15-cm long tape attached to the
end of their tails. TST was performed under normal light conditions
(800 lux). The test session was recorded using a digital videocamera.
All videos were scored by a trained observer blinded to the treatment.
Immobility time was measured for the entire 6-minute session.
A mouse was considered immobile when it hung passively.
Novelty-Suppressed Feeding. The novelty-suppressed feeding
test (NSF) was performed as described (Warner-Schmidt and Duman,
2007), with minor modifications. Briefly, mice were singly housed and
food-deprived for twenty-four hours in freshly made home-cages. Two
normal chow diet pellets were placed on a square food platform (10
10 cm) in the center of an open-field arena (40 40 cm). Thirty
minutes after saline, ketamine, or 4-Cl-KYN administration, mice
were introduced into a corner of the arena for 10 minutes. The time
needed for the mice to take a bite of food was recorded by a trained
observer blind to the treatment groups. After the test, the mice were
returned to their home-cages containing preweighed food pellets, and
latency to bite the food as well as consumption was recorded for
a period of 10 minutes.
Learned Helplessness. As in previously published procedures
(Maeng et al., 2008), the learned helplessness (LH) paradigm con-
sisted of three different phases, i.e., inescapable shock training, LH
screening, and the LH test. For the inescapable shock portion of the
test (Day 1), the animals were placed in one side of two-chambered
shuttle boxes (34 cm height 37 cm width 18 cm depth; Coulbourn
Instruments, Whitehall, PA), with the door between the chambers
closed. Following a 5-minute adaptation period, 120 inescapable foot-
shocks (0.45 mA, 15-second duration, randomized average intershock
interval of 45 seconds) were delivered through the grid floor. During the
screening session (Day 2), the mice were placed in one of the two
chambers of the apparatus for 5 minutes. A shock (0.45 mA) was then
delivered, and the door between the two chambers was raised simulta-
neously. Crossing over into the second chamber terminated the shock. If
the animal did not cross over, the shock terminated after 3 seconds. A
total of 30 screening trials of escapable shocks were presented to each
mouse with an average of 30-second delay between each trial. Mice that
developed helplessness behavior (.5 escape failures during the last 10
screening shocks) (Malberg and Duman, 2003) were treated with either
saline (7.5 ml/kg), FLX (20 mg/kg), ketamine (10 mg/kg), 4-Cl-KYN (5, 25
or 125 mg/kg), or 7-Cl-KYNA (1 or 25 mg/kg) 24 hours following screening
(Day 3). During the LH test phase (Day 4), the animals were placed in the
shuttle boxes and, after a 5-minute adaptation period, a 0.45-mA shock
was delivered concomitantly with door opening for the first five trials,
followed by a 2-second delay for the next 40 trials. Crossing over to the
second chamber terminated the shock. If the animal did not cross over to
the other chamber, the shock was terminated after 24 seconds. A total of
45 trials of escapable shock was presented to each mouse with 30-second
intertrial intervals. The number of escape failures was recorded for each
mouse. To determine the prolonged efficacy of the drugs, mice were
retested for helpless behavior after 7 days (i.e., on Day 11).
Open-Field Test Behaviors. Experiments were performed at 30
lux ambient light levels within the arena. Three days prior to the
locomotor sensitization experiments (Days 13), mice were placed into
individual open-field arenas (50 cm length 50 cm width 38 cm
height; San Diego Instruments, San Diego, CA) for a 30-minute daily
habituation period. Saline was then injected, and mice were assessed
for another 90 minutes. For the next 3 consecutive days (Days 46),
mice were randomly divided into treatment groups and received
injections after a 30-minute habituation period. Locomotor responses
were then recorded for an additional period of 90 minutes. During
Days 712 and 1419, the animals received no injections. On Days 13
and 20, mice received a challenge treatment injection, and their
locomotor activity was recorded. On Day 21, all mice received a saline
injection to evaluate a possible contextual conditioning effect. Dis-
tance traveled was analyzed using TopScan v2.0 (CleverSys, Inc.,
Reston, VA) for a 30-minute period following the daily injection or 90
minutes for the acute effects of the drugs. Stereotypic rearing (vertical
activity) and circling behavior were scored for the 30 minutes following
the daily injections by a trained observer blind to the treatment
groups.
Conditioned-Place Preference. The conditioned-place prefer-
ence (CPP) apparatus consisted of a rectangular Plexiglas three-
chambered box (53 cm length 15 cm width 36 cm height) divided
into three compartments (two equal-sized end-chambers and a middle
waitingchamber) by two removable doors. One end-chamber (22 cm
length 15 cm width 36 cm height) was composed of a 1-cm-spaced
wired floor with four dark-gray walls (dark compartment), whereas
the other end-chamber (same dimensions) had a 0.3-cm-spaced wired
floor with white walls with black vertical stripes (1.5 cm; striped
compartment). The middle compartment (9 cm length 15 cm width
36 cm height) had white Plexiglas floor and walls. The CPP protocol
consisted of a habituation phase, a preconditioning test, six condi-
tioning sessions, and a postconditioning test. On Day 1, mice were
placed in the CPP apparatus for a 20-minute habituation period and
were allowed to freely explore all chambers. On Day 2 (preconditioning
phase), mice were placed in the CPP apparatus and were allowed to
explore all compartments for a period of 20 minutes. During the
conditioning phase, saline (7.5 ml/kg), ketamine (10 mg/kg), or 4-Cl-KYN
(25 or 125 mg/kg) was administered, and mice were placed in their
least-preferred compartment on alternating days (i.e., Days 3, 5, and
7). All groups of animals received saline (7.5 ml/kg) in their preferred
compartment on Days 4, 6, and 8. During the postconditioning test
session (i.e., Day 9), mice were placed in the CPP apparatus to freely
explore all three compartments for 20 minutes. Time spent in each
compartment was measured during the last 15 minutes of both pre- and
postconditioning sessions, as previously described (Zanos et al., 2014).
Prepulse Inhibition. The prepulse inhibition (PPI) paradigm
used was derived from previously published protocols (Chan et al.,
2008), with minor modifications. Mice were individually tested in
acoustic startle boxes (SR-LAB; San Diego Instruments, San Diego,
CA). The animals first received an injection of saline, ketamine, or
4-Cl-KYN and were placed in the startle chamber for a 30-minute
habituation period. The experiment started with a further 5-minute
adaptation period during which the mice were exposed to a constant
background noise (67 db), followed by five initial startle stimuli
(120 db, 40-millisecond duration each). Subsequently, animals were
78 Zanos et al.
at ASPET Journals on April 4, 2016jpet.aspetjournals.orgDownloaded from
exposed to five different trial types: pulse alone trials (120 db,
40-millisecond duration), three prepulse trials of 76, 81, and 86 db of
white noise bursts (20-millisecond duration) preceding a 120-db pulse
by 100 milliseconds, and background (67 db) no-stimuli trials. Each of
these trials was randomly presented five times. The percentage
prepulse inhibition (% PPI) was calculated using the following
formula: [(magnitude on pulse alone trial magnitude on prepulse 1
pulse trial)/magnitude on pulse alone trial] 100.
Statistics
All values are expressed as the mean 6S.E.M. For analyses of the
acute (i.e., 1-hour) and long-term (i.e., 24-hour) antidepressant-like
efficacy in the FST and TST, one-way ANOVA was performed. Glycine
pretreatment effects in the FST were assessed by a two-way ANOVA
with pretreatment(i.e., saline and glycine) and treatment(i.e., saline
and 4-Cl-KYN) as factors. Effects in the LH and NSF tests were analyzed
by one-way ANOVA. Acute and behavioral sensitization effects in the
open-field test were analyzed using two-wayrepeated measures anal-
ysis of variance (ANOVA) with treatmentand timeas factors. CPP data
were assessed by two-wayrepeated measures ANOVA for factors
treatmentand CPP phase(i.e., pre-Cond, post-Cond). The data of
startle amplitude and circling behavior were assessed by one-way
ANOVA. The % PPI data were analyzed by two-way ANOVA for factors
treatmentand intensity(i.e., 76 db, 81 db, and 86 db). A Studentst-test
was used to compare the area under the curve (AUC) following 4-Cl-KYN
or 7-Cl-KYNA administration, and to determine the effects of acute
(1 hour postinjection) FLX administration, in the FST. All ANOVAs were
followed by a Bonferroni post-hoc comparison when significance was
reached (i.e., p,0.05). All statistical analyses were performed using
STATISTICA 10 (StatSoft Inc., Tulsa, OK). ANOVA results are
presented in Table 1, while post-hoc comparisons are detailed in figures
and text.
Results
In Vivo Microdialysis. 7-Cl-KYNA levels were not mea-
surable in hippocampal microdialysates during the baseline
collection period (02 hours; Fig. 1B). Extracellular 7-Cl-KYNA
levels became detectable in mice that received 4-Cl-KYN or
7-Cl-KYNA (each 25 mg/kg), attaining peak concentrations of
100 616 nM 1.5 hours after the administration of 4-Cl-KYN
and 25 65 nM 1 hour after the administration of 7-Cl-KYNA.
Quantitative assessment of the AUC revealed approximately
10-times higher extracellular 7-Cl-KYNA levels for the
4-Cl-KYN-treated mice compared with 7-Cl-KYNA-treated
animals (inset, Fig. 1B). Notably, considering a typical
1020% recovery and time lag from the microdialysis pro-
cedure (Stahle et al., 1991), the peak extracellular concen-
tration of 7-Cl-KYNA following administration of 4-Cl-KYN
approximated its IC
50
at the glycine
B
site [560 nM; Kemp et al.
(1988)].
Pharmacological Screening. In vitro screening revealed
only two receptors with K
i
values of ,10 mM for 7-Cl-KYNA
(Supplemental Table S1 and Supplemental Fig. S1): b-adrenergic
receptor subtype 1 (K
i
:3.95mM) and muscarinic receptor subtype
5(K
i
:2.81mM). This contrasts with the IC
50
of 7-Cl-KYNA at the
glycine
B
site [560 nM; Kemp et al. (1988)] and indicates a lack of
off-target effects of the compound. 4-Cl-KYN showed no affinity
(,10 mM)foranyofthepossibletargets assessed (Supplemental
Table S1 and Supplemental Fig. S2).
Antidepressant-Like Effects of 4-Cl-KYN in the
Forced-Swim Test, Tail-Suspension Test, and Novelty-
Suppressed Feeding Tests. To assess antidepressant-like
properties in classic tests of antidepressant efficacy, mice were
tested in the FST or TST 1 hour postinjection. Both ketamine
(10 mg/kg) and 4-Cl-KYN (25 and 125 mg/kg) administration
significantly decreased immobility time in the FST, compared
with the saline-treated controls (Fig. 2A). A similar decrease
in immobility was also observed after acute FLX administra-
tion (Fig. 2A). To examine whether systemic 7-Cl-KYNA
administration itself elicits antidepressant-like effects in the
FST, we administered the compound at three different doses
(25, 75, or 225 mg/kg). Tested 1 hour later, only the highest
dose of 7-Cl-KYNA resulted in significant antidepressant-like
effects (saline: 157.4 614.9 seconds; 25 mg/kg: 116.1 618.5
seconds; 75 mg/kg: 125.4 612.9 seconds; 225 mg/kg (P,0.05):
81.3 613.0 seconds; n578/group). The effective doses of
ketamine and 4-Cl-KYN in the FST also significantly decreased
immobility time in the TST (Fig. 2D). Notably, 4-Cl-KYN, in
apparent contrast to ketamine (Li et al., 2010), elicited antide-
pressant actions across a rather broad dose range; however, our
studies did not determine the maximally effective dose in either
FST or TST.
To determine whether these antidepressant-like effects
of 4-Cl-KYN are attributable to glycine
B
site inhibition, we
pretreated mice with glycine at a dose previously shown to
reverse the behavioral effects of NMDA channel blockers in
rodents [1.6 g/kg, i.p.; Evoniuk et al. (1991); Javitt et al. (1999)].
Whereas glycine administration alone did not change immo-
bility time of saline-treated animals in the FST, it prevented
the antidepressant-like effects of 4-Cl-KYN (25 mg/kg) in this
paradigm (Fig. 2B). Moreover, to mechanistically assess
downstream mechanisms mediating the antidepressant
effects of 4-Cl-KYN, we administered the AMPA receptor antag-
onist NBQX (10 mg/kg) 5 minutes prior to treatment with 4-
Cl-KYN and assessed immobility time in the FST 1 hour later.
Pretreatment with NBQX prevented the antidepressant ef-
fects of both ketamine and 4-Cl-KYN (Fig. 2C).
To determine the capacity for rapid antidepressant action of
4-Cl-KYN, we assessed its effects in the NSF test, which is
sensitive to monoamine-acting antidepressants only following
chronic administration (Santarelli et al., 2003). Administra-
tion of both ketamine and 4-Cl-KYN rapidly decreased the
latency to feed in the NSF arena compared with saline-treated
mice (Fig. 2E). There were no significant differences be-
tween the amounts of food consumed by saline-, ketamine-,
or 4-Cl-KYN-treated mice in their home cage assessed immediately
following the NSF test (Fig. 2F).
Sustained Antidepressant-Like Effects of 4-Cl-KYN
in the 24-Hour FST. It has previously been reported that, in
contrast to monoamine-acting antidepressants, ketamine
elicits antidepressant actions in the FST 24 hours after
administration, when ketamine is no longer present [e.g., Li
et al. (2010); Autry et al. (2011)]. We therefore compared the
antidepressant-like effects of ketamine and 4-Cl-KYN in the
FST 24 hours following a single injection. Similar to ketamine
(10 mg/kg), 4-Cl-KYN (25 or 125 mg/kg) significantly de-
creased immobility time compared with saline-treated ani-
mals (Fig. 3A). Consistent with its delayed time course of
action in humans [e.g., Barr et al. (1997)], FLX (20 mg/kg) had
no significant effect when it was administered a single time
24 hours prior to testing (Fig. 3A).
Antidepressant Effects of 4-Cl-KYN in the Learned
Helplessness Paradigm. Mice that developed helpless be-
havior (50% of the mice having experienced inescapable
foot shocks) were randomly assigned to treatment groups,
Fast-Onset Antidepressant Effects of 4-Chlorokynurenine 79
at ASPET Journals on April 4, 2016jpet.aspetjournals.orgDownloaded from
receiving single injections of saline, ketamine (10 mg/kg),
4-Cl-KYN (25 mg/kg), FLX (20 mg/kg), or 7-Cl-KYNA (1 or
25 mg/kg). Both ketamine and 4-Cl-KYN significantly reduced
helpless behaviors as assessed by the number of escape
failures compared with saline-treated controls (Fig. 3, B and
D). Notably, the antidepressant-like effects of both ketamine
and 4-Cl-KYN persisted when the animals were retested 7 days
later (Fig. 3, C and E). In contrast, FLX- and 7-Cl-KYNA-treated
mice did not manifest antidepressant-like effects 24 hours or
7 days postinjection (Fig. 3, D and E).
4-Cl-KYN Does Not Affect Behaviors in the Open
Field. Acute administration of 4-Cl-KYN (25 or 125 mg/kg)
did not cause ketamine-like acute hyperlocomotor activity in
the open-field compared with saline-treated controls (Fig. 4B).
In the same mice, we assessed possible locomotor sensitization
effects following repeated saline, ketamine, or 4-Cl-KYN
injections. Administration of ketamine, but not of 4-Cl-KYN,
on Days 13 and 20 revealed a significant increase in locomotor
activity compared with the acute motor-enhancing effects
(Day 4), indicating locomotor sensitization (Fig. 4C). No effect
was seen when saline was administered on Day 21.
Similar to other compounds with stimulant properties [e.g.,
Morency et al. (1987)], administration of ketamine, but not of
4-Cl-KYN, caused significant circling behavior following acute
administration on Day 4, compared with saline controls (Fig.
4D). Animals did not manifest maintenance of this effect on
subsequent days (data not shown). Moreover, injection of
ketamine, but not of 4-Cl-KYN, induced a significant increase
in repetitive vertical activity (rearing) on the first day of drug
injection (Day 4; Fig. 4E). No sensitization to this effect was
observed (Fig. 4E).
4-Cl-KYN Does Not Elicit Conditioned Place Prefer-
ence. Ketamine-treated (10 mg/kg) mice exhibited a robust
CPP, as illustrated by an increase in the time spent in the
drug-paired compartment during the postconditioning phase
compared with the preconditioning phase (Fig. 5A). In con-
trast, administration of 4-Cl-KYN (25 or 125 mg/kg) did not
induce CPP (Fig. 5A). Additionally, analysis of the ratio of the
time mice spent in the drug-paired compartment during the
postconditioning phase and preconditioning session indicated
a significant increase in the preference of ketamine-treated
mice to spend their time in the drug-paired compartment
compared with saline-treated mice.
4-Cl-KYN Does Not Disrupt Prepulse Inhibition. Ket-
amine administration dose-dependently disrupted % PPI,
with 30 mg/kg found to be the lowest significantly effective
dose (Fig. 5B). In contrast, 4-Cl-KYN administration did not
induce significant alterations in % PPI at any dose tested (25,
TABLE 1
Statistical analyses
Factor Effect Interaction Effect
Overall effects for Fig. 2
Acute antidepressant effects Factor treatmentFactor pretreatmentFactor pretreatmenttreatment
Forced-swim test F
[6,83]
= 4.10 P,0.01
Glycine, forced-swim test F
[1,53]
= 7.57 P,0.001 F
[1,53,]
= 7.50 P,0.001 F
[1,53]
= 6.55 P = 0.01
NBQX, forced-swim test F
[2,65]
= 4.51 P,0.05 F
[1,65]
= 16.80 P,0.001 F
[2,65]
= 6.16 P,0.01
Tail-suspension test F
[4,74]
= 3.57 P,0.05
NSF latency to feed F
[2,27]
= 10.51 P,0.001
Home-cage food consumption F
[2,27]
= 0.53 P = 0.60
Overall effects for Fig. 3
Sustained antidepressant effects,
24-hour FST Factor treatment
Immobility time F
[4,45]
= 11.84 P,0.001
Sustained antidepressant effects,
LH
Effects of 4-Cl-KYN (24 hours) F
[2,28]
= 71.68 P,0.001
Effects of 4-Cl-KYN (7 days) F
[2,28]
= 43.78 P,0.001
Effects of 7-Cl-KYN (24 hours) F
[4,39]
= 11.47 P,0.001
Effects of 7-Cl-KYN (7 days) F
[4,39]
= 4.23 P,0.01
Overall effects for Fig. 4
Effects of 4-Cl-KYN in the
open-field test Factor treatmentFactor timeFactor treatmenttime
Acute locomotor effects F
[3,28]
= 0.35 P = 0.79 F
[23,644]
= 29.73 P,0.001 F
[69,644]
= 6.72 P,0.001
Locomotor sensitization F
[3,28]
= 16.08 P,0.001 F
[8,224]
= 8.87 P,0.001 F
[24,168]
= 11.34 P,0.001
Circling behavior F
[3,28]
= 8.04 P,0.001
Rearing activity F
[3,28]
= 3.39 P,0.05 F
[8,224]
= 7.86 P,0.001 F
[24,224]
= 2.87 P,0.001
Overall effects for Fig. 5
4-Cl-KYN effects in CPP Factor treatmentFactor CPP phaseFactor treatmentCPP phase
Time in drug-paired compartment F
[3,27]
= 1.71 P = 0.19 F
[1,27]
= 11.71 P,0.01 F
[3,27]
= 4.34 P,0.05
ratio (Post-Cond/Pre-Cond) F
[3,27]
= 4.40 P,0.05
PPI, Ketamine dose-response Factor treatmentFactor intensityFactor treatmentintensity
% PPI F
[4,148]
= 15.83 P,0.001 F
[2,148]
= 25.40 P,0.001 F
[8,148]
= 0.33 P = 0.95
Startle amplitude F
[4,51]
= 0.51 P = 0.73
PPI, Effects of 4-Cl-KYN Factor treatmentFactor intensityFactor treatmentintensity
% PPI F
[4,153]
= 23.33 P,0.001 F
[2,153]
= 16.57 P,0.001 F
[8,153]
= 0.74 P = 0.66
Startle amplitude F
[4,54]
= 0.99 P = 0.42
Effects of 7-Cl-KYNA in the FST
Antidepressant effects Factor treatment
Immobility time F
[3,27]
= 4.121 P,0.05
80 Zanos et al.
at ASPET Journals on April 4, 2016jpet.aspetjournals.orgDownloaded from
125, or 375 mg/kg; Fig. 5D). Neither ketamine nor 4-Cl-KYN
affected startle amplitude at any dose (Fig. 5, C and E).
Discussion
The present study demonstrated that systemic administra-
tion of 4-Cl-KYN, the brain-penetrant prodrug of the glycine
B
-
site antagonist 7-Cl-KYNA, results in ketamine-like
antidepressant-like effects in mice. In particular, we observed
acute, dose-dependent effects of 4-Cl-KYN in the FST and the
TST, and showed that antidepressant-like efficacy was main-
tained for 24 hours in the FST. Moreover, 4-Cl-KYN showed
remarkable efficacy in the LH paradigm, where the effect was
evident up to 7 days after a single treatment, at a time point
past which 7-Cl-KYNA had become undetectable in brain
microdialysate (Fig. 1B). In addition, 4-Cl-KYN administra-
tion rapidly decreased the latency to feed in the NSF test. All
these antidepressant-like effects were qualitatively identical,
Fig. 2. Antidepressant effects in the forced-swim, tail-suspension, and novelty-suppressed feeding tests. Mice received intraperitoneal injections of
saline (SAL), fluoxetine (FLX), ketamine (KET), or 4-chlorokynurenine (4-Cl-KYN) and were tested in the FST 1-hour post-treatment. (A) Acute
administration of FLX (n= 9/group; unpaired Studentst-test) as well as KET and 4-Cl-KYN (n=1216/group; one-way ANOVA followed by Bonferronis
multiple comparison) significantly reduced immobility in the FST. (B) Glycine pretreatment prevented the antidepressant -like effects of 4-Cl-KYNinthe
FST (n=1315/group; two-way ANOVA followed by Bonferronis multiple comparison). (C) Administration of NBQX prevented the antidepressant
effects of both KET and 4-Cl-KYN in the FST (n=1114/group, two-way ANOVA followed by Bonferronis multiple comparison). (D) In the TST,
administration of 4-Cl-KYN resulted in antidepressant-like effects 1 hour postinjection (n= 16/group; one-way ANOVA followed by Bonf erronis multiple
comparison). (E) Administration of KET and 4-Cl-KYN significantly reduced latency to feed in the NSF test, (F) without affecting home-cage food
consumption (n= 10/group; one-way ANOVA followed by Bonferronis multiple comparison). Data are the mean 6S.E.M. *p,0.05, **p,0.01, ***p,
0.001.
Fast-Onset Antidepressant Effects of 4-Chlorokynurenine 81
at ASPET Journals on April 4, 2016jpet.aspetjournals.orgDownloaded from
and quantitatively very similar, to those of ketamine, which
was tested in parallel using a well-established antidepressant
dose for mice [10 mg/kg; Maeng et al. (2008); Ma et al. (2013)].
We confirmed that the antidepressant-like action of 4-Cl-KYN
in the acute FST involves the glycine
B
site, since pretreatment
with glycine prevented the effects at the same dose shown to
increase concentrations of glycine in the brain (albeit by less
than a 2-fold difference) and to reverse the behavioral effects of
NMDA channel blockers (Trullas and Skolnick, 1990; Evoniuk
et al., 1991; Javitt et al., 1999). In support, no off-site receptor
targets were identified by the extensive receptor screening
performed (Supplemental Figs. S1 and S2 and Supplemental
Table S1). Moreover, in line with the hypothesis that ket-
amines antidepressant mechanism involves enhanced synap-
tic glutamate release and activation of postsynaptic AMPA
receptors (Li et al., 2010; Duman and Aghajanian, 2012),
administration of an AMPA receptor antagonist completely
prevented the behavioral effects of 4-Cl-KYN in the FST.
Notably, effective doses of 4-Cl-KYN did not produce reward-
ing or aversive properties in the CPP paradigm, and were not
associated with locomotor stimulation, stereotypic behaviors,
or disruption of PPI, all of which are typically induced by
ketamine and other NMDA receptor channel blockers.
A major drawback of monoamine-based antidepressant med-
ications that are currently in clinical use is that they require
sustained treatment of several weeks or longer before thera-
peutic benefits are achieved (Rush et al., 2006). The ineffective-
ness of acute FLX administration in the 24-hour FST and LH
paradigms markedly contrasts with the actions of ketamine,
which have been documented in recent animal studies (Maeng
et al., 2008; Li et al., 2010; Autry et al., 2011; Browne and Lucki,
2013). On the basis of the theoretical construct introduced by
Skolnick and collaborators (1996), this pharmacological dispar-
ity in the effects of acute dosing can be explained at least partly
by the fact that clinical effects of monoaminergic drugs do not
become apparent until NMDA receptors are downregulated
following a delay period. Subsequently, the reduction of NMDA
receptor function initiates a cascade of molecular and cellular
events that may cumulatively account for the benefits seen in
MDD patients [see Skolnick et al. (1996)]. This conceptual
framework reconciles the monoamine and glutamate hypothe-
ses of MDD pathophysiology and accounts for the current
interest in glutamatergic modulators as rapid-acting medica-
tions for the treatment of the disease.
In the present study, 4-Cl-KYN was used as a prodrug of
7-Cl-KYNA, which is frequently employed as an experimental
tool to selectively probe the glycine
B
receptor in vitro, or
following intracerebral application in vivo (Kemp et al., 1988;
Wu et al., 1997). After peripheral administration, 4-Cl-KYN
gains rapid access to the brain via the large neutral amino acid
transporter (Hokari et al., 1996) and is then promptly
accumulated by astrocytes. Subsequently, 4-Cl-KYN is irre-
versibly converted to 7-Cl-KYNA by kynurenine aminotrans-
ferase, and the newly produced glycine
B
receptor antagonist is
then rapidly released into the extracellular milieu (Kiss et al.,
2003) (cf., Fig. 1A). Unlike 4-Cl-KYN (Hokari et al., 1996),
7-Cl-KYNA crosses the blood-brain barrier very poorly, prob-
ably because of its polar nature and the lack of an active
transport system (Leeson and Iversen, 1994). In the present
study, we confirmed the formation of 7-Cl-KYNA from sys-
temically administered 4-Cl-KYN by in vivo microdialysis in
the hippocampus of freely-moving mice.
A recent double-blind, placebo-controlled trial demon-
strated that long-term (6-week) administration of the glycine
B
partial agonist D-cycloserine has antidepressant effects in
humans (Heresco-Levy et al., 2013). Antidepressant-like
effects of glycine
B
-modulating compounds have been pre-
viously reported in behavioral tests that are also sensitive to
monoamine-acting drugs (e.g., the FST and TST). For exam-
ple, Poleszak et al. (2011) showed antidepressant-like efficacy
of the brain-penetrating, specific glycine
B
antagonist
L-701,324 and of D-cycloserine in the FST in mice. However,
few studies have addressed effects in ketamine-sensitive
paradigms (i.e., 24-hour FST, LH, and NSF). This is important
Fig. 3. Sustained antidepressant effects in the 24-hour forced-swim test
and learned helplessness paradigms. Mice received intraperitoneal injections
of saline (SAL), fluoxetine (FLX), ketamine (KET), 7-chlorokynurenic acid
(7-Cl-KYNA), or 4-chlorokynurenine (4-Cl-KYN) and were tested in the
FST 1 or 24 hours post-treatment, and in the LH paradigm 24 hours and
7 days following treatment. (A) While FLX administration did not induce
antidepressant effects, both KET and 4-Cl-KYN significantly reduced
immobility time in the FST and decreased the number of escape failures in
the LH paradigm at (B) 24 hours or (C) 7 days (n=1011/group; one-way
ANOVA followed by Bonferronis multiple comparison). Injections of FLX
or 7-Cl-KYNA did not replicate the effects of 4-Cl-KYN in the LH paradigm
24 hours (D) or 7 days (E) post-treatment (n=89/group; one-way ANOVA
followed by Bonferronis multiple comparison). Data are the mean 6S.E.M.
*p ,0.05, **p,0.01, ***p,0.001. ns,notsignificant.
82 Zanos et al.
at ASPET Journals on April 4, 2016jpet.aspetjournals.orgDownloaded from
as ketamine represents a first-in-class new generation rapid-
acting antidepressant drug. A notable exception is GLYX-13,
an amidated tetrapeptide, which can act as a partial agonist of
the glycine
B
receptor and has rapid-acting antidepressant
effects in the 24-hour FST, LH, and NSF tests (Burgdorf et al.,
2013). Since the endogenous ligands of the glycine
B
receptor
glycine and D-serineare present at high levels in brain, it is
hypothesized that at high concentrations glycine
B
receptor
partial agonists act as functional antagonists in vivo by
reducing the facilitating effect of glycine and/or D-serine (Clos
et al., 1996). However, the involvement of the glycine
B
site of
the NMDA receptor in the antidepressant effects of GLYX-13
has not been directly demonstrated so far.
Maj et al. (1994) described antidepressant-like effects
following repeated, but not single, administration of 7-Cl-KYNA
(20 mg/kg) in the FST, and acute antidepressant-like effects
were seen after a single injection of low doses of 7-Cl-KYNA
in rats (Zhu et al., 2013; Liu et al., 2015). However, the latter
findings were not replicated in our experiments. In contrast,
i.p. administration of lower doses of 4-Cl-KYN, the brain-
penetrant bioprecursor of 7-Cl-KYNA, exerted rapid and
sustained ketamine-like effects in tests that distinguish ket-
amine from monoamine-acting antidepressants. Reversal of
antidepressant effects with glycine supported the concept that
the antidepressant-like actions of 4-Cl-KYN involved inhibi-
tion of the glycine
B
site of the NMDA receptor.
Possible side effects of 4-Cl-KYN were examined by assess-
ing locomotor activity, stereotypic behaviors, PPI, CPP, and
behavioral sensitization. Unlike ketamine, 4-Cl-KYN produced
none of these effects at any of the doses tested. This indicates
that antagonism of the glycine
B
site with 4-Cl-KYN does not
appear to elicit the detrimental side effects (e.g., psychotomi-
metic and rewarding) that are associated with the use of
ketamine or other NMDA channel blockers (Mansbach, 1991;
Papp and Moryl, 1994). The advantages of targeting the
glycine
B
site may be linked to the different mechanisms by
which inhibition of the co-agonist site and channel blockade
reduce NMDA receptor function (Danysz and Parsons, 1998).
Alternatively or in addition, the detrimental effects of
ketamine, as well as other NMDA channel blockers, including
Fig. 4. Open-field behaviors. (A) Behavioral sensitization protocol. Acute intraperitoneal administration of 4-chlorokynurenine (4-Cl-KYN) did not have
effects similar to intraperitoneal ketamine (KET) on (B) acute hyperlocomotion (n=79/group; two-way ANOVA followed by Bonferronis multiple
comparison) or (C) locomotor sensitization (n=79/group; two-way ANOVA followed by Bonferronis multiple comparison). Unlike KET, 4-Cl-KYN
administration was not associated with (D) increased stereotypic circling(n=79/group; one-way ANOVA fo llowed by Bonferronis multiple comparison) or (E)
rearing behavior (n=79/group; two-way ANOVA followed by Bonferronis multiple comparison). Data are the mean 6S.E.M. **p,0.01, ***p,0.001.
Fast-Onset Antidepressant Effects of 4-Chlorokynurenine 83
at ASPET Journals on April 4, 2016jpet.aspetjournals.orgDownloaded from
phencyclidine and MK-801, may be related to the fact that
these compounds have been reported to directly influence
dopaminergic transmission by reducing dopamine reuptake
(Nishimura and Sato, 1999) or facilitating dopamine release
(Uchihashi et al., 1992). Notably, we demonstrated here that
neither 4-Cl-KYN nor 7-Cl-KYNA have significant affinity for
a large number of monoaminergic and other possible targets.
In summary, the present study supports the hypothesis that
antagonism of the glycine
B
co-agonist site of the NMDA
receptor constitutes a promising approach for the treatment
of major depression. Specifically, our results highlight the
potential of 4-Cl-KYN as a next-generation, rapid-acting
antidepressant medication, with a safer side-effect profile
than ketamine. Since the prodrug uses astrocytes for delivery
of the active compound, the efficacy of our approach may be
further enhanced by the fact that it could specifically buttress
astrocyte function, which is impaired in individuals with MDD
(Etievant et al., 2013; Rajkowska and Stockmeier, 2013).
Importantly, in a Phase 1, randomized, dose-escalation study
evaluating safety and pharmacokinetics of single doses of
orally administered 4-Cl-KYN in healthy volunteers, the drug
did not cause any serious adverse events (ClinicalTrials.gov
Identifier: NCT01483846), indicating a potential for clinical
trials in MDD patients.
Acknowledgments
4-Cl-KYN was provided by VistaGen Therapeutics, Inc. Receptor
binding profiles and K
i
determinations were generously provided by
the NIMH Psychoactive Drug Screening Program, Contract No.
HHSN-271-2008-025C, directed by Dr. Bryan Roth (University of
North Carolina, Chapel Hill, NC) in conjunction with Ms. Jamie
Driscoll (NIMH, Bethesda, MD).
Authorship Contributions
Participated in research design: Zanos, Piantadosi, Zarate Jr.,
Schwarcz, Gould.
Conducted experiments: Zanos, Piantadosi, Wu, Pribut, Can.
Contributed new reagents or analytic tools: Snodgrass.
Performed data analysis: Zanos, Piantadosi, Dell.
Wrote or contributed to the writing of the manuscript: Zanos,
Piantadosi, Schwarcz, Gould.
References
Antony LJ, Paruchuri VN, and Ramanan R (2014) Antidepressant effect of ketamine
in sub anaesthetic doses in male albino mice. J Clin Diagn Res 8:HC05HC07.
Autry AE, Adachi M, Nosyreva E, Na ES, Los MF, Cheng PF, Kavalali ET,
and Monteggia LM (2011) NMDA receptor blockade at rest triggers rapid behav-
ioural antidepressant responses. Nature 475:9195.
Barr LC, Heninger GR, Goodman W, Charney DS, and Price LH (1997) Effects of
fluoxetine administration on mood response to tryptophan depletion in healthy
subjects. Biol Psychiatry 41:949954.
Besnard J, Ruda GF, Setola V, Abecassis K, Rodriguiz RM, Huang XP, Norval S,
Sassano MF, Shin AI, and Webster LA, et al. (2012) Automated design of ligands to
polypharmacological profiles. Nature 492:215220.
Fig. 5. Conditioned-place preference and prepulse inhibi-
tion. (A) Time spent in the drug-paired compartment
during the postconditioning test revealed place preference
conditioning to ketamine (KET) but not to 4-chlorokynur-
enine (4-Cl-KYN) (n=78/group; two-way ANOVA followed
by Bonferronis multiple comparison). KET caused a (B)
dose-dependent disruption of prepulse inhibition (% PPI;
two-way ANOVA followed by Bonferronis multiple com-
parison), (C) but did not alter the startle amplitude
(one-way ANOVA followed by Bonferronis multiple com-
parison). 4-Cl-KYN did not inhibit (D) % PPI (two-way
ANOVA followed by Bonferronis multiple comparison) or
(E) startle amplitude (one-way ANOVA followed by
Bonferronis multiple comparison) (n=1112/group). Data
are the mean 6S.E.M.; *p,0.05, **p,0.01, ***p,0.001;
ns, not significant.
84 Zanos et al.
at ASPET Journals on April 4, 2016jpet.aspetjournals.orgDownloaded from
Browne CA and Lucki I (2013) Antidepressant effects of ketamine: mechanisms
underlying fast-acting novel antidepressants. Front Pharmacol 4:161.
Burgdorf J, Zhang XL, Nicholson KL, Balster RL, Leander JD, Stanton PK, Gross
AL, Kroes RA, and Moskal JR (2013) GLYX-13, a NMDA receptor glycine-site
functional partial agonist, induces antidepressant-like effects without ketamine-
like side effects. Neuropsychopharmacology 38:729742.
Can A, Dao DT, Terrillion CE, Piantadosi SC, Bhat S, and Gould TD (2012) The tail
suspension test. J Vis Exp 59:3769.
Chan MH, Chiu PH, Sou JH, and Chen HH (2008) Attenuation of ketamine-evoked
behavioral responses by mGluR5 positive modulators in mice. Psychopharmacology
(Berl) 198:141148.
Clos MV, Garcia Sanz A, Trullas R, and Badia A (1996) Effect of 1-amino-
cyclopropanecarboxylic acid on N-methyl-D-aspartate-stimulated [3H]-noradrenaline
release in rat hippocampal synaptosomes. Br J Pharmacol 118:901904.
Danysz W and Parsons CG (1998) Glycine and N-methyl-D-aspartate receptors:
physiological significance and possible therapeutic applications. Pharmacol Rev 50:
597664.
Duman RS (2014) Pathophysiology of depression and innovative treatments:
remodeling glutamatergic synaptic connections. Dialogues Clin Neurosci 16:1127.
Duman RS and Aghajanian GK (2012) Synaptic dysfunction in depression: potential
therapeutic targets. Science 338:6872.
Etiévant A, Lambás-Señas L, Scarna H, Lucas G, and Haddjeri N (2013) Astrocytes
and gliotransmitters: new players in the treatment of major depression? Curr Drug
Targets 14:12951307.
Evoniuk GE, Hertzman RP, and Skolnick P (1991) A rapid method for evaluating the
behavioral effects of phencyclidine-like dissociative anesthetics in mice. Psycho-
pharmacology (Berl) 105:125128.
Heresco-Levy U, Gelfin G, Bloch B, Levin R, Edelman S, Javitt DC, and Kremer I
(2013) A randomized add-on trial of high-dose D-cycloserine for treatment-
resistant depression. Int J Neuropsychopharmacol 16:501506.
Hokari M, Wu HQ, Schwarcz R, and Smith QR (1996) Facilitated brain uptake of
4-chlorokynurenine and conversion to 7-chlorokynurenic acid. Neuroreport 8:1518.
Javitt DC, Balla A, Sershen H, and Lajtha A (1999) A.E. Bennett Research Award.
Reversal of phencyclidine-induced effects by glycine and glycine transport inhib-
itors. Biol Psychiatry 45:668679.
Kemp JA, Foster AC, Leeson PD, Priestley T, Tridgett R, Iversen LL, and Woodruff
GN (1988) 7-Chlorokynurenic acid is a selective antagonist at the glycine modu-
latory site of the N-methyl-D-aspartate receptor complex. Proc Natl Acad Sci USA
85:65476550.
Kiss C, Ceresoli-Borroni G, Guidetti P, Zielke CL, Zielke HR, and Schwarcz R (2003)
Kynurenate production by cultured human astrocytes. J Neural Transm 110:114.
Krystal JH, Karper LP, Seibyl JP, Freeman GK, Delaney R, Bremner JD, Heninger
GR, Bowers MB, Jr, and Charney DS (1994) Subanesthetic effects of the non-
competitive NMDA antagonist, ketamine, in humans. Psychotomimetic, percep-
tual, cognitive, and neuroendocrine responses. Arch Gen Psychiatry 51:199214.
Lee SC and Schwarcz R (2001) Excitotoxic injury stimulates pro-drug-induced
7-chlorokynurenate formation in the rat striatum in vivo. Neurosci Lett 304:185188.
Leeson PD and Iversen LL (1994) The glycine site on the NMDA receptor: structure-
activity relationships and therapeutic potential. J Med Chem 37:40534067.
Li N, Lee B, Liu RJ, Banasr M, Dwyer JM, Iwata M, Li XY, Aghajanian G,
and Duman RS (2010) mTOR-dependent synapse formation underlies the rapid
antidepressant effects of NMDA antagonists. Science 329:959964.
Liu BB, Luo L, Liu XL, Geng D, Liu Q and Yi LT (2015) 7-Chlorokynurenic acid
(7-CTKA) produces rapid antidepressant-like effects: through regulating hippo-
campal microRNA expressions involved in TrkB-ERK/Akt signaling pathways in
mice exposed to chronic unpredictable mild stress. Psychopharmacology (Berl) 232:
541550.
Ma XC, Dang YH, Jia M, Ma R, Wang F, Wu J, Gao CG, and Hashimoto K (2013)
Long-lasting antidepressant action of ketamine, but not glycogen synthase kinase-3
inhibitor SB216763, in the chronic mild stress model of mice. PLoS One 8:e56053.
Maeng S, Zarate CA, Jr, Du J, Schloesser RJ, McCammon J, Chen G, and Manji HK
(2008) Cellular mechanisms underlying the antidepressant effects of ketamine:
role of alpha-amino-3-hydroxy-5-methylisoxazole-4-propionic acid receptors. Biol
Psychiatry 63:349352.
Maj J, Rogóz Z, Skuza G, and Kołodziejczyk K (1994) Some central effects of
kynurenic acid, 7-chlorokynurenic acid and 5,7- dichloro-kynurenic acid, glycine
site antagonists. Pol J Pharmacol 46:115124.
Malberg JE and Duman RS (2003) Cell proliferation in adult hippocampus is de-
creased by inescapable stress: reversal by fluoxetine treatment. Neuro-
psychopharmacology 28:15621571.
Mansbach RS (1991) Effects of NMDA receptor ligands on sensorimotor gating in the
rat. Eur J Pharmacol 202:6166.
Morency MA, Stewart RJ, and Beninger RJ (1987) Circling behavior following uni-
lateral microinjections of cocaine into the medial prefrontal cortex: dopaminergic or
local anesthetic effect? J Neurosci 7:812818.
Nishimura M and Sato K (1999) Ketamine stereoselectively inhibits rat dopamine
transporter. Neurosci Lett 274:131134.
Papp M and Moryl E (1994) Rewarding properties of non-competitive and competitive
NMDA antagonists as measured by place preference conditioning in rats. Pol J
Pharmacol 46:7981.
Pittenger C, Sanacora G, and Krystal JH (2007) The NMDA receptor as a therapeutic
target in major depressive disorder. CNS Neurol Disord Drug Targets 6:101115.
Poleszak E, Wla
z P, Wróbel A, Dybała M, Sowa M, Fidecka S, Pilc A, and Nowak G
(2007) Activation of the NMDA/glutamate receptor complex antagonizes the
NMDA antagonist-induced antidepressant-like effects in the forced swim test.
Pharmacol Rep 59:595600.
Poleszak E, Wla
zP,SzewczykB,Wla
z A, Kasperek R, Wróbel A, and Nowak G (2011) A
complex interaction between glycine/NMDA receptors and serotonergic/noradrenergic
antidepressants in the forced swim test in mice. J Neural Transm 118:15351546.
Popoli M, Yan Z, McEwen BS, and Sanacora G (2012) The stressed synapse: the impact
of stress and glucocorticoids on glutamate transmission. Nat Rev Neurosci 13:2237.
Potter MC, Elmer GI, Bergeron R, Albuquerque EX, Guidetti P, Wu HQ,
and Schwarcz R (2010) Reduction of endogenous kynurenic acid formation
enhances extracellular glutamate, hippocampal plasticity, and cognitive behavior.
Neuropsychopharmacology 35:17341742.
Przegali
nski E, Tatarczy
nska E, and Chojnacka-Wójcik E (1998) Anxiolytic- and
antidepressant-like effects of an antagonist at glycineB receptors. Pol J Pharmacol
50:349354.
Rajkowska G and Stockmeier CA (2013) Astrocyte pathology in major depressive
disorder: insights from human postmortem brain tissue. Curr Drug Targets 14:
12251236.
Rush AJ, Trivedi MH, Wisniewski SR, Nierenberg AA, Stewart JW, Warden D,
Niederehe G, Thase ME, Lavori PW, and Lebowitz BD, et al. (2006) Acute and
longer-term outcomes in depressed outpatients requiring one or several treatment
steps: a STAR*D report. Am J Psychiatry 163:19051917.
Salituro FG, Tomlinson RC, Baron BM, Palfreyman MG, McDonald IA, Schmidt W,
Wu HQ, Guidetti P, and Schwarcz R (1994) Enzyme-activated antagonists of the
strychnine-insensitive glycine/NMDA receptor. J Med Chem 37:334336.
Sanacora G, Treccani G, and Popoli M (2012) Towards a glutamate hypothesis of
depression: an emerging frontier of neuropsychopharmacology for mood disorders.
Neuropharmacology 62:6377.
Santarelli L, Saxe M, Gross C, Surget A, Battaglia F, Dulawa S, Weisstaub N, Lee J,
Duman R, and Arancio O, et al. (2003) Requirement of hippocampal neurogenesis
for the behavioral effects of antidepressants. Science 301:805809.
Skolnick P, Layer RT, Popik P, Nowak G, Paul IA, and Trullas R (1996) Adaptation of N-
methyl-D-aspartate (NMDA) receptors following antidepr essant treatment: implica-
tions for the pharmacotherapy of depression. Phar macopsychiatry 29:2326.
Ståhle L, Segersvärd S, and Ungerstedt U (1991) A comparison between three
methods for estimation of extracellular concentrations of exogenous and endoge-
nous compounds by microdialysis. J Pharmacol Methods 25:4152.
Trullas R and Skolnick P (1990) Functional antagonists at the NMDA receptor
complex exhibit antidepressant actions. Eur J Pharmacol 185:110.
Uchihashi Y, Kuribara H, and Tadokoro S (1992) Assessment of the ambulation-
increasing effect of ketamine by coadministration with central-acting drugs in
mice. Jpn J Pharmacol 60:2531.
Warner-Schmidt JL and Duman RS (2007) VEGF is an essential mediator of the
neurogenic and behavioral actions of antidepressants. Proc Natl Acad Sci USA
104:46474652.
Wu HQ, Salituro FG, and Schwarcz R (1997) Enzyme-catalyzed production of the
neuroprotective NMDA receptor antagonist 7-chlorokynurenic acid in the rat brain
in vivo. Eur J Pharmacol 319:1320.
Zanos P, Georgiou P, Wright SR, Hourani SM, Kitchen I, Winsky-Sommerer R,
and Bailey A (2014) The oxytocin analogue carbetocin prevents emotional im-
pairment and stress-induced reinstatement of opioid-seeking in morphine-
abstinent mice. Neuropsychopharmacology 39:855865.
Zhu WL, Wang SJ, Liu MM, Shi HS, Zhang RX, Liu JF, Ding ZB, and Lu L (2013)
Glycine site N-methyl-D-aspartate receptor antagonist 7-CTKA produces rapid
antidepressant-like effects in male rats. J Psychiatry Neurosci 38:306316.
Address correspondence to: Dr. Todd D. Gould, Department of Psychiatry,
University of Maryland School of Medicine, Rm. 936 MSTF, 685 W. Baltimore
St., Baltimore, MD 21201. E-mail: gouldlab@me.com
Fast-Onset Antidepressant Effects of 4-Chlorokynurenine 85
at ASPET Journals on April 4, 2016jpet.aspetjournals.orgDownloaded from
... Av-101 (4-chlorokyneurine, 4-Cl-KYN) is a prodrug, rapidly converted to 7-chloro-kynurenic acid (7-Cl-KYNA) by kynurenine aminotransferase (Salituro et al., 1994). 7-Cl-KYNA exerts a neuroprotective action by a full antagonism activity on the glycine B co-agonist site of the NMDAR (Zanos et al., 2015). The effects of this drug are also linked to the metabolite 4-chloro-3-hydroxyanthranilic acid (4-Cl-3-HAA), which inhibits the quinolinic acid, a neurotoxic NMDAR agonist (Walsh et al., 1994). ...
... The effects of this drug are also linked to the metabolite 4-chloro-3-hydroxyanthranilic acid (4-Cl-3-HAA), which inhibits the quinolinic acid, a neurotoxic NMDAR agonist (Walsh et al., 1994). Considering the evidence implicating the kynurenine pathway in the aetiology of mood disorders (Brown et al., 2021), and the rapid, dose-dependent, and sustained antidepressant effect following a single administration in an animal study (Zanos et al., 2015), AV-101 was tested in humans; however, it failed to demonstrate benefits on depressive symptoms at week 2, according to two randomized clinical trials (RCTs) in patients with MDD (ClinicalTrials.gov Identifier NCT03078322 and NCT02484456). ...
... To further increase bioavailability, despite not overcoming the limitations reported in animal studies, the combination of AV-101 with probenecid (a uricosuric agent) has recently been investigated in rodents, with safe results . Despite the drug was not associated with an improvement in suicidal ideation and/or suicidal behaviour, it was hypothesized to be a promising option for suicidality treatment in emergency settings, because of its ketamine-like glutamatergic mechanism of action, and the lack of dissociative adverse events (Zanos et al., 2015). Additionally, its antisuicidality effect may be not limited to the acute phase. ...
Article
Mood disorders are recurrent/chronic diseases with variable clinical remission rates. Available antidepressants are not effective in all patients and often show a relevant response latency, with a range of adverse events, including weight gain and sexual dysfunction. Novel rapid agents were developed with the aim of overcoming at least in part these issues. Novel drugs target glutamate, gamma-aminobutyric acid, orexin, and other receptors, providing a broader range of pharmacodynamic mechanisms, that is, expected to increase the possibility of personalizing treatments on the individual clinical profile. These new drugs were developed with the aim of combining a rapid action, a tolerable profile, and higher effectiveness on specific symptoms, which were relatively poorly targeted by standard antidepressants, such as anhedonia and response to reward, suicidal ideation/behaviours, insomnia, cognitive deficits, and irritability. This review discusses the clinical specificity profile of new antidepressants, namely 4-chlorokynurenine (AV-101), dextromethorphan-bupropion, pregn-4-en-20-yn-3-one (PH-10), pimavanserin, PRAX-114, psilocybin, esmethadone (REL-1017/dextromethadone), seltorexant (JNJ-42847922/MIN-202), and zuranolone (SAGE-217). The main aim is to provide an overview of the efficacy/tolerability of these compounds in patients with mood disorders having different symptom/comorbidity patterns, to help clinicians in the optimization of the risk/benefit ratio when prescribing these drugs.
... In contrast, acute treatment with the antidepressants fluoxetine, a selective serotonin reuptake inhibitor, and imipramine, a tricyclic antidepressant, did not affect immobility time in the FST 24 h after treatment (Kruskal-Wallis nonparametric ANOVA followed by Bonferroni's test; H(3) = 2.73, p = 0.26, Fig. 1e). The doses of fluoxetine and imipramine were selected based on previous studies (Garcia et al. 2008;Zanos et al. 2015). These results suggest that psychedelics have antidepressantlike effects at the active dose in mice. ...
Article
Full-text available
Serotonergic psychedelics such as psilocybin, lysergic acid diethylamide, and DOI exert a hallucinatory effect through serotonin 5-HT2A receptor (5-HT2A) activation. Recent studies have revealed that serotonergic psychedelics have therapeutic potential for neuropsychiatric disorders, including major depressive and anxiety-related disorders. However, the involvement of 5-HT2A in mediating the therapeutic effects of these drugs remains unclear. In this study, we ethopharmacologically analyzed the role of 5-HT2A in the occurrence of anxiolytic- and antidepressant-like effects of serotonergic psychedelics such as psilocin, an active metabolite of psilocybin, DOI, and TCB-2 in mice 24 h post-treatment. Mice with acute intraperitoneal psychedelic treatment exhibited significantly shorter immobility times in the forced swimming test (FST) and tail-suspension test (TST) than vehicle-treated control mice. These effects were eliminated by pretreatment with volinanserin, a 5-HT2A antagonist. Surprisingly, the decreasing immobility time in the FST in response to acute psilocin treatment was sustained for at least three weeks. In the novelty-suppressed feeding test (NSFT), the latency to feed, an indicator of anxiety-like behavior, was decreased by acute administration of psilocin; however, pretreatment with volinanserin did not diminish this effect. In contrast, DOI and TCB-2 did not affect the NSFT performance in mice. Furthermore, psilocin, DOI, and TCB-2 treatment did not affect the spontaneous locomotor activity or head-twitch response, a hallucination-like behavior in rodents. These results suggest that 5-HT2A contributes to the antidepressant effects of serotonergic psychedelics rather than anxiolytic effects.
... suggest that treatments aimed at upregulation of the KYNA arm in the brain may be favourable for female MDD sufferers but also might assist managing suicidal behaviour. Drugs that increase cerebral KYNA and show rapid antidepressant effects in preclinical models [54,55] have recently moved to clinical trials but did not show favourable outcomes [56]. However, future trials need to consider the importance of subgroups when selecting the most suitable cohorts. ...
Article
Full-text available
Major depressive disorder (MDD) is a serious psychiatric disorder that in extreme cases can lead to suicide. Evidence suggests that alterations in the kynurenine pathway (KP) contribute to the pathology of MDD. Activation of the KP leads to the formation of neuroactive metabolites, including kynurenic acid (KYNA) and quinolinic acid (QUIN). To test for changes in the KP, postmortem anterior cingulate cortex (ACC) was obtained from the National Institute of Health NeuroBioBank. Gene expression of KP enzymes and relevant neuroinflammatory markers were investigated via RT-qPCR (Fluidigm) and KP metabolites were measured using liquid chromatography-mass spectrometry in tissue from individuals with MDD ( n = 44) and matched nonpsychiatric controls ( n = 36). We report increased IL6 and IL1B mRNA in MDD. Subgroup analysis found that female MDD subjects had significantly decreased KYNA and a trend decrease in the KYNA/QUIN ratio compared to female controls. In addition, MDD subjects that died by suicide had significantly decreased KYNA in comparison to controls and MDD subjects that did not die by suicide, while subjects that did not die by suicide had increased KYAT2 mRNA, which we hypothesise may protect against a decrease in KYNA. Overall, we found sex- and suicide-specific alterations in the KP in the ACC in MDD. This is the first molecular evidence in the brain of subgroup specific changes in the KP in MDD, which not only suggests that treatments aimed at upregulation of the KYNA arm in the brain may be favourable for female MDD sufferers but also might assist managing suicidal behaviour.
... Results showed that the compound was able to significantly reduce the immobility in the test, even up to 7 days after the last administration (fig 10b). It is interesting to note that BDNF by itself can induce plasticity after local central administration [51]. ...
Preprint
Full-text available
The introduction of anti-amyloid monoclonal antibodies against Alzheimer’s disease (AD) is of high importance. However, even though treated patients show very little amyloid pathology, there is only a modest effect on the rate of cognitive decline. Although this effect possibly can increase over time, there is still a need for alternative treatments that will improve the cognitive function in patients with AD. Therefore, the purpose of this study was to characterize the triazinetrione ACD856, a novel pan-Trk positive allosteric modulator, in multiple models to address its neuroprotective and potential disease modifying effects. The pharmacological effect of ACD856 was tested in recombinant cell lines, primary cortical neurons or animals. We demonstrate that ACD856 enhanced NGF-induced neurite outgrowth, increased the levels of the pre-synaptic protein SNAP25 in PC12 cells and increased the degree of phosphorylated TrkB in SH-SY5Y cells. In primary cortical neurons, ACD856 led to increased levels of phospho-ERK1/2, showed neuroprotective effect against amyloid-beta or energy-deprivation induced neurotoxicity, and increased the levels of brain derived neurotrophic factor (BDNF). Consequently, administration of ACD856 resulted in a significant increase of BDNF in the brains of 21-months old mice. Furthermore, repeated administration of ACD856 resulted in a sustained anti-depressant effect which lasted up to seven days, suggesting effects that go beyond merely symptomatic effects. In conclusion, the results confirm ACD856 as a cognitive enhancer, but more importantly, they provide substantial in vitro and in vivo evidence of neuroprotective and long-term effects that contribute to neurotrophic support and increased neuroplasticity. Presumably, the described effects of ACD856 may improve cognition, increase resilience, and promote neurorestorative processes, thereby leading to a healthier brain in patients with AD.
... Results showed that the compound was able to significantly reduce the immobility in the test, even up to 7 days after the last administration (Figure 10b). It is interesting to note that BDNF and activation of TrkB can induce increased plasticity [51,52]. ...
Article
Full-text available
The introduction of anti-amyloid monoclonal antibodies against Alzheimer’s disease (AD) is of high importance. However, even though treated patients show very little amyloid pathology, there is only a modest effect on the rate of cognitive decline. Although this effect can possibly increase over time, there is still a need for alternative treatments that will improve cognitive function in patients with AD. Therefore, the purpose of this study was to characterize the triazinetrione ACD856, a novel pan-Trk positive allosteric modulator, in multiple models to address its neuroprotective and potential disease-modifying effects. The pharmacological effect of ACD856 was tested in recombinant cell lines, primary cortical neurons, or animals. We demonstrate that ACD856 enhanced NGF-induced neurite outgrowth, increased the levels of the pre-synaptic protein SNAP25 in PC12 cells, and increased the degree of phosphorylated TrkB in SH-SY5Y cells. In primary cortical neurons, ACD856 led to increased levels of phospho-ERK1/2, showed a neuroprotective effect against amyloid-beta or energy-deprivation-induced neurotoxicity, and increased the levels of brain-derived neurotrophic factor (BDNF). Consequently, administration of ACD856 resulted in a significant increase in BDNF in the brains of 21 months old mice. Furthermore, repeated administration of ACD856 resulted in a sustained anti-depressant effect, which lasted up to seven days, suggesting effects that go beyond merely symptomatic effects. In conclusion, the results confirm ACD856 as a cognitive enhancer, but more importantly, they provide substantial in vitro and in vivo evidence of neuroprotective and long-term effects that contribute to neurotrophic support and increased neuroplasticity. Presumably, the described effects of ACD856 may improve cognition, increase resilience, and promote neurorestorative processes, thereby leading to a healthier brain in patients with AD.
Article
Full-text available
L-type amino acid transporter 1 (LAT1) is integral to the transport of large neutral amino acids across the blood–brain barrier (BBB), playing a crucial role in brain homeostasis and the delivery of therapeutic agents. This review explores the multifaceted role of LAT1 in neurological disorders, including its structural and functional aspects at the BBB. Studies using advanced BBB models, such as induced pluripotent stem cell (iPSC)–derived systems and quantitative proteomic analyses, have demonstrated LAT1’s significant impact on drug permeability and transport efficiency. In Alzheimer’s disease, LAT1-mediated delivery of anti-inflammatory and neuroprotective agents shows promise in overcoming BBB limitations. In Parkinson’s disease, LAT1’s role in transporting L-DOPA and other therapeutic agents highlights its potential in enhancing treatment efficacy. In phenylketonuria, studies have revealed polymorphisms and genetic variations of LAT1, which could be correlated to disease severity. Prodrugs of valproic acid, pregabalin, and gabapentin help use LAT1-mediated transport to increase the therapeutic activity and bioavailability of the prodrug in the brain. LAT1 has also been studied in neurodevelopment disorders like autism spectrum disorders and Rett syndrome, along with neuropsychiatric implications in depression. Its implications in neuro-oncology, especially in transporting therapeutic agents into cancer cells, show immense future potential. Phenotypes of LAT1 have also shown variations in the general population affecting their ability to respond to painkillers and anti-inflammatory drugs. Furthermore, LAT1-targeted approaches, such as functionalized nanoparticles and prodrugs, show promise in overcoming chemoresistance and enhancing drug delivery to the brain. The ongoing exploration of LAT1’s structural characteristics and therapeutic applications reiterates its critical role in advancing treatments for neurological disorders.
Article
Depression is a leading cause of disability and reduced work capacity worldwide. The monoamine theory of the pathogenesis of depression has remained dominant for many decades, however, drugs developed on its basis have limited efficacy. Exploring alternative mechanisms underlying this pathology could illuminate new avenues for pharmacological intervention. Targeting glutamatergic pathways in the CNS, particularly through modulation of NMDA and AMPA receptors, demonstrates promising results. This review presents some existing drugs with glutamatergic activity and novel developments based on it to enhance the efficacy of pharmacotherapy for depressive disorders.
Article
Full-text available
Substantial clinical evidence has unravelled the superior antidepressant efficacy of ketamine: in comparison to traditional antidepressants targeting the monoamine systems, ketamine, as an N-methyl-d-aspartate receptor (NMDAR) antagonist, acts much faster and more potently. Surrounding the antidepressant mechanisms of ketamine, there is ample evidence supporting an NMDAR-antagonism-based hypothesis. However, alternative arguments also exist, mostly derived from the controversial clinical results of other NMDAR inhibitors. In this article, we first summarize the historical development of the NMDAR-centred hypothesis of rapid antidepressants. We then classify different NMDAR inhibitors based on their mechanisms of inhibition and evaluate preclinical as well as clinical evidence of their antidepressant effects. Finally, we critically analyse controversies and arguments surrounding ketamine’s NMDAR-dependent and NMDAR-independent antidepressant action. A better understanding of ketamine’s molecular targets and antidepressant mechanisms should shed light on the future development of better treatment for depression. This article is part of a discussion meeting issue ‘Long-term potentiation: 50 years on’.
Chapter
Depression is prevalent in people living with epilepsy (PLWE) due to a reciprocal relationship between both conditions. Various depressive disorders can occur in PLWE, often linked to hyperactivity of the hypothalamic-pituitary-adrenal axis, neuroinflammation, and serotonin transmission disruptions, among other factors. Antiseizure medications with adverse psychotropic effects can also cause or exacerbate depression. Depressive symptoms in PLWE are classified into interictal and peri-ictal types. The peri-ictal classes are further subdivided into preictal, ictal, and postictal categories. The dysphoric mood is the most common preictal symptom. Ictal symptoms are usually short-lived, and postictal symptoms may not occur immediately. Depression profoundly impacts PLWE, increasing suicidality, stigma, pharmacoresistance to antiseizure medications, and unfavorable epilepsy surgery outcomes. It notably deteriorates their quality of life even more than the level of seizure control. Therefore, early depression detection is crucial, though the hectic epilepsy clinical environments pose challenges. Efficient use of brief time-taking screening tools can help overcome busy clinical environments. Treatment primarily involves antidepressants, especially selective serotonin reuptake inhibitors, due to their lower seizure risk and favorable side effects. Cognitive-behavioral therapy alongside antidepressants can also be beneficial. When choosing antiseizure medications for PLWE who have depression or are prone to depression, those with positive psychotropic effects should be considered primarily.
Article
Full-text available
4-Chlorokynurenine (4-Cl-KYN, AV-101) is a prodrug of a NMDA receptor antagonist and is in clinical development for potential CNS indications. We sought to further understand the distribution and metabolism of 4-Cl-KYN, as this information might provide a strategy to enhance the clinical development of this drug. We used excretion studies in rats, in vitro transporter assays, and pharmacogenetic analysis of clinical trial data to determine how 4-Cl-KYN and metabolites are distributed. Our data indicated that a novel acetylated metabolite (N-acetyl-4-Cl-KYN) did not affect the uptake of 4-Cl-KYN across the blood–brain barrier via LAT1. 4-Cl-KYN and its metabolites were found to be renally excreted in rodents. In addition, we found that N-acetyl-4-Cl-KYN inhibited renal and hepatic transporters involved in excretion. Thus, this metabolite has the potential to limit the excretion of a range of compounds. Our pharmacogenetic analysis found that a SNP in N-acetyltransferase 8 (NAT8, rs13538) was linked to levels of N-acetyl-4-Cl-KYN relative to 4-Cl-KYN found in the plasma and that a SNP in SLC7A5 (rs28582913) was associated with the plasma levels of the active metabolite, 7-Cl-KYNA. Thus, we have a pharmacogenetics-based association for plasma drug level that could aid in the drug development of 4-Cl-KYN and have investigated the interaction of a novel metabolite with drug transporters.
Article
Full-text available
Background: Depression is the most common mental disorder in community settings, and is a major cause of disability across the world. Antidepressants such as SSRI (Selective Serotonin Reuptake Inhibitor) and TCA (Tricyclic antidepressants) are used. These drugs affect the adrenergic and serotonergic pathways. These drugs have an unfavorable side effect profile, take longer time to act and are not very effective in resistant cases. Alternate pathways involving the glutamate receptors have also been linked with depression, hence Ketamine an NMDA antagonist was evaluated for the antidepressant effect. Aim: To study the antidepressant effect of ketamine in subanaesthetic doses in male albino mice. Study Design, Materials and Methods: The study was a randomized controlled animal study done on 30 male albino BALB/c mice divided into five groups with Imipramine (10mg/kg) as the standard drug and Ketamine in varying doses (5, 7.5, 10 mg/kg) as the test drug. The animal model used was the forced swim test. The reduction in immobility time was taken as the index of the antidepressant effect. Statistical analysis: The data were analysed with the one way ANOVA test using SPSS version 12. Results and Conclusion: The data analysis showed that Ketamine at a dose lower dose of Ketamine (5mg/kg) did not show a significant antidepressant effect in contrast to the higher doses (7.5 and 10mg/kg) which showed significant antidepressant effect (‘p’ < 0.05). The inference of this study is that Ketamine has significant antidepressant property at variable dose ranges and further studies can be done on these lines.
Article
Full-text available
Despite the complexity and heterogeneity of mood disorders, basic and clinical research studies have begun to elucidate the pathophysiology of depression and to identify rapid, efficacious antidepressant agents. Stress and depression are associated with neuronal atrophy, characterized by loss of synaptic connections in key cortical and limbic brain regions implicated in depression. This is thought to occur in part via decreased expression and function of growth factors, such as brain-derived neurotrophic factor (BDNF), in the prefrontal cortex (PFC) and hippocampus. These structural alterations are difficult to reverse with typical antidepressants. However, recent studies demonstrate that ketamine, an N-methyl-D-aspartate (NMDA) receptor antagonist that produces rapid antidepressant actions in treatment-resistant depressed patients, rapidly increases spine synapses in the PFC and reverses the deficits caused by chronic stress. This is thought to occur by disinhibition of glutamate transmission, resulting in a rapid but transient burst of glutamate, followed by an increase in BDNF release and activation of downstream signaling pathways that stimulate synapse formation. Recent work demonstrates that the rapid-acting antidepressant effects of scopolamine, a muscarinic receptor antagonist, are also associated with increased glutamate transmission and synapse formation. These findings have resulted in testing and identification of additional targets and agents that influence glutamate transmission and have rapid antidepressant actions in rodent models and in clinical trials. Together these studies have created tremendous excitement and hope for a new generation of rapid, efficacious antidepressants.
Article
Full-text available
Newer antidepressants are needed for the many individuals with major depressive disorder (MDD) that do not respond adequately to treatment and because of a delay of weeks before the emergence of therapeutic effects. Recent evidence from clinical trials shows that the NMDA antagonist ketamine is a revolutionary novel antidepressant because it acts rapidly and is effective for treatment-resistant patients. A single infusion of ketamine alleviates depressive symptoms in treatment-resistant depressed patients within hours and these effects may be sustained for up to 2 weeks. Although the discovery of ketamine's effects has reshaped drug discovery for antidepressants, the psychotomimetic properties of this compound limit the use of this therapy to the most severely ill patients. In order to develop additional antidepressants like ketamine, adequate preclinical behavioral screening paradigms for fast-acting antidepressants need to be established and used to identify the underlying neural mechanisms. This review examines the preclinical literature attempting to model the antidepressant-like effects of ketamine. Acute administration of ketamine has produced effects in behavioral screens for antidepressants like the forced swim test, novelty suppression of feeding and in rodent models for depression. Protracted behavioral effects of ketamine have been reported to appear after a single treatment that last for days. This temporal pattern is similar to its clinical effects and may serve as a new animal paradigm for rapid antidepressant effects in humans. In addition, protracted changes in molecules mediating synaptic plasticity have been implicated in mediating the antidepressant-like behavioral effects of ketamine. Current preclinical studies are examining compounds with more specific pharmacological effects at glutamate receptors and synapses in order to develop additional rapidly acting antidepressants without the hallucinogenic side effects or abuse potential of ketamine.
Article
Full-text available
The main challenge in treating opioid addicts is to maintain abstinence due to the affective consequences associated with withdrawal which may trigger relapse. Emerging evidence suggests a role of the neurohypophysial peptide oxytocin in the modulation of mood disorders as well as drug addiction. However, its involvement in the emotional consequences of drug abstinence remains unclear. We investigated the effect of 7-day opioid abstinence on the oxytocinergic system and assessed the effect of the oxytocin analogue carbetocin on the emotional consequences of opioid abstinence, as well as relapse. Male C57BL6J mice were treated with a chronic escalating-dose morphine regimen (20-100 mg/kg/day, i.p.). Seven days withdrawal from this administration paradigm induced a decrease of hypothalamic oxytocin levels and a concomitant increase of oxytocin receptor binding in the lateral septum and amygdala. While no physical withdrawal symptoms or alterations in plasma corticosterone levels were observed after seven days of abstinence, mice exhibited increased anxiety-like and depressive-like behaviors and impaired sociability. Carbetocin (6.4 mg/kg, i.p.) attenuated the observed negative emotional consequences of opioid withdrawal. Furthermore, in the conditioned place preference paradigm with 10 mg/kg morphine conditioning, carbetocin (6.4 mg/kg, i.p.) was able to prevent the stress-induced reinstatement to morphine-seeking following extinction. Overall, our results suggest that alterations of the oxytocinergic system contribute to the mechanisms underlying anxiety, depression and social deficits observed during opioid abstinence. This study also highlights the oxytocinergic system as a target for developing pharmacotherapy for the treatment of emotional impairment associated with abstinence and thereby prevention of relapse.Neuropsychopharmacology accepted article preview online, 15 October 2013; doi:10.1038/npp.2013.285.
Article
Full-text available
With a lifetime prevalence of more than 16% worldwide, major depressive disorder is one of the most common psychiatric disorders. Only one third of patients experience a complete therapeutic improvement with the use of current antidepressant drugs, with a therapeutic effect appearing only after several weeks of treatment. Hence, a better understanding of the mechanisms of action of current antidepressant treatments is needed to ultimately identify new targets and enhance beneficial effects. Given the intimate relationships between astrocytes and neurons at synapses and the ability of astrocytes to "sense" neuronal communication and release gliotransmitters, an attractive hypothesis is emerging stating that the effects of antidepressants on brain function could be, at least in part, mediated by direct influences of astrocytes on neuronal networks. This review aims at highlighting the involvement of astrocytes and gliotransmission in the antidepressant effects of both non- and pharmacological therapies.
Article
Full-text available
Background: Glutamate N-methyl-D-aspartate (NMDA) receptor antagonists exert fast-acting antidepressant effects, providing a promising way to develop a new classification of antidepressant that targets the glutamatergic system. In the present study, we examined the potential antidepressant action of 7-chlorokynurenic acid (7-CTKA), a glycine recognition site NMDA receptor antagonist, in a series of behavioural models of depression and determined the molecular mechanisms that underlie the behavioural actions of 7-CTKA. Methods: We administered the forced swim test, novelty-suppressed feeding test, learned helplessness paradigm and chronic mild stress (CMS) paradigm in male rats to evaluate the possible rapid antidepressant-like actions of 7-CTKA. In addition, we assessed phospho-glycogen synthase kinase-3β (p-GSK3β) level, mammalian target of rapamycin (mTOR) function, and postsynaptic protein expression in the medial prefrontal cortex (mPFC) and hippocampus. Results: Acute 7-CTKA administration produced rapid antidepressant-like actions in several behavioural tests. It increased p-GSK3β, enhanced mTOR function and increased postsynaptic protein levels in the mPFC. Activation of GSK3β by LY294002 completely blocked the antidepressant-like effects of 7-CTKA. Moreover, 7-CTKA did not produce rewarding properties or abuse potential. Limitations: It is possible that 7-CTKA modulates glutamatergic transmission, thereby causing enduring alterations of GSK3β and mTOR signalling, although we did not provide direct evidence to support this possibility. Thus, the therapeutic involvement of synaptic adaptions engaged by 7-CTKA requires further study. Conclusion: Our findings demonstrate that acute 7-CTKA administration produced rapid antidepressant-like effects, indicating that the behavioural response to 7-CTKA is mediated by GSK3β and mTOR signalling function in the mPFC.
Article
Background: To characterize further behavioral, cognitive, neuroendocrine, and physiological effects of subanesthetic doses of ketamine hydrochloride in healthy human subjects. Ketamine, a phencyclidine hydrochloride derivative, is a dissociative anesthetic and a noncompetitive antagonist of the N-methyl-D-aspartate subtype of excitatory amino acid receptor.Methods: Nineteen healthy subjects recruited by advertisements from the community participated in this randomized, double-blind, placebo-controlled study. Subjects completed three test days involving the 40-minute intravenous administration of placebo, ketamine hydrochloride (0.1 mg/kg), or ketamine hydrochloride (0.5 mg/kg). Behaviors associated with the positive and negative symptoms of schizophrenia were assessed by using the Brief Psychiatric Rating Scale. Changes in perception and behaviors associated with dissociative states were assessed by the Perceptual Aberration Subscale of the Wisconsin Psychosis Proneness Scale and the Clinician-Administered Dissociative States Scale. Cognitive function was assessed by using the (1) Mini-Mental State Examination; (2) tests sensitive to frontal cortical dysfunction, including a continuous performance vigilance task, a verbal fluency task, and the Wisconsin Card Sorting Test; and (3) tests of immediate and delayed recall. Plasma levels of cortisol, prolactin, homovanillic acid, and 3-methoxy-4-hydroxyphenethyleneglycol were measured.Results: Ketamine (1) produced behaviors similar to the positive and negative symptoms of schizophrenia; (2) elicited alterations in perception; (3) impaired performance on tests of vigilance, verbal fluency, and the Wisconsin Card Sorting Test; (4) evoked symptoms similar to dissociative states; and (5) preferentially disrupted delayed word recall, sparing immediate recall and postdistraction recall. Ketamine had no significant effect on the Mini-Mental State Examination at the doses studied. Ketamine also had no effect on plasma 3-methoxy-4hydroxyphenethyleneglycol levels, although it blunted a test day decline in plasma homovanillic acid levels at the higher dose. It also dose dependently increased plasma cortisol and prolactin levels. Ketamine produced small dose-dependent increases in blood pressure.Conclusions: These data indicate that N-methyl-Daspartate antagonists produce a broad range of symptoms, behaviors, and cognitive deficits that resemble aspects of endogenous psychoses, particularly schizophrenia and dissociative states.
Article
Rationale: 7-Chlorokynurenic acid (7-CTKA), a NMDA receptor antagonist, has been reported as a potential rapid antidepressant with poor understanding about the molecular mechanism of its therapeutic action. MicroRNAs (miRNAs) are emerging as critical regulators of central nervous system plasticity and may play an important role in depression. Objective: The objective of this study was to investigate the molecular mechanism of antidepressant action of 7-CTKA in chronic unpredictable mild stress (CUMS) animal model. Methods: K252a (tropomyosin-related kinase receptor B (TrkB) antagonist), U0126 (extracellular signal-regulated kinase (ERK) phosphorylation inhibitor), LY294002 (serine-threonine kinase (Akt) phosphorylation inhibitor), or vehicle was given intracerebroventricularly to mice in each group 30 min before 7-CTKA or vehicle intraperitoneal injection. Behavioral changes were observed by sucrose preference test and miRNA microarray was performed to examine hippocampal miRNAs levels in mice. Quantitative RT-PCR was conducted to further confirm results in microarray study. Results: 7-CTKA not only reversed the decrease in sucrose preference and multiple hippocampal miRNAs changes induced by CUMS but also mediated 15 common miRNAs via TrkB-ERK/Akt pathways. Among them, the expression levels of four miRNAs (miR-34a-5p, miR-200a-3p, miR-144-3p, miR-1894-5p) were validated by quantitative real-time PCR (qRT-PCR). The findings from qRT-PCR study support results from microarray analysis except for the non-significance of miR-1894-5p expression. Conclusions: This demonstrated that the 15 miRNA targets shared by TrkB-ERK/Akt pathways might participate in rapid-acting molecular mechanism of antidepressant 7-CTKA.
Article
Background: Phencycline (PCP, “angel dust”) and other noncompetitive antagonists of N-methyl-d-aspartate (NMDA)-type glutamatergic neurotransmission induce psychotic effects in humans that closely resemble positive, negative, and cognitive symptoms of schizophrenia. Behavioral effects of PCP in rodents are reversed by glycine (GLY) and other NMDA augmenting agents. In rodents, behavioral effects of PCP are mediated, in part, by secondary dysregulation of subcortical dopaminergic neurotransmission. This study evaluates effects of GLY and GLY transport antagonists on behavioral and neurochemical consequences of PCP administration in rodents.Methods: Two separate experiments were performed. In the first, effects of GLY on PCP-induced stimulation of dopaminergic neurotransmission in nucleus accumbens were evaluated using in vivo microdialysis in awake animals. In the second, effects of a series of GLY transport antagonists were evaluated for potency in inhibiting PCP-induced hyperactivity.Results: In microdialysis studies, GLY significantly inhibited PCP-induced stimulation of subcortical DA release in a dose-dependent fashion. In behavioral studies, the potency of a series of GLY transport antagonists for inhibiting PCP-induced hyperactivity in vivo correlated significantly with their potency in antagonizing GLY transport in vitro.Conclusions: These findings suggest, first, that GLY reverses not only the behavioral, but also the neurochemical, effects of PCP in rodents. Second, the findings suggest that GLY transport antagonists may induce similar effects to GLY, and may therefore represent an appropriate site for targeted drug development.