You are on page 1of 11

Neuropharmacology 112 (2017) 210e220

Contents lists available at ScienceDirect

Neuropharmacology
journal homepage: www.elsevier.com/locate/neuropharm

Ketamine modulates hippocampal neurogenesis and pro-


inflammatory cytokines but not stressor induced neurochemical
changes
Melanie Clarke, Sara Razmjou, Natalie Prowse, Zach Dwyer, Darcy Litteljohn, Rowan Pentz,
Hymie Anisman, Shawn Hayley*
Department of Neuroscience, Carleton University, 1125 Colonel By Drive, Ottawa, Ontario K1S 5B6, Canada

a r t i c l e i n f o a b s t r a c t

Article history: Considerable recent attention has focused on the rapid antidepressant effects observed in treatment
Received 5 January 2016 resistant patients produced by the NMDA receptor antagonist, ketamine. Surprisingly, the effects of
Received in revised form ketamine in the context of stressor exposure, as well as the consequences of its chronic use are unclear.
13 April 2016
Thus, we assessed the impact of acute and repeated ketamine treatment together with acute [restraint or
Accepted 17 April 2016
Available online 20 April 2016
lipopolysaccharide (LPS)] or chronic (unpredictable different psychogenic challenges) stressor exposure.
Importantly, acute ketamine treatment did provoke an antidepressant-like effect in a forced swim test
(FST) and this effect lasted for 8 days following repeated exposure to the drug. Although acute restraint
Keywords:
Depression
and LPS individually provoked the expected elevation of plasma corticosterone and brain-region specific
Ketamine monoamine variations, ketamine had no influence on corticosterone and had, at best, sparse effects on
Stressor the monoamine changes. Similarly, ketamine did not appreciably influence the stressor induced
LPS neurochemical and sucrose preference alterations, it did however, dose-dependently reverse the LPS
Cytokine induced elevation of the pro-inflammatory cytokines, interleukin-1b (IL-1b) and tumor necrosis factor-a
Neurogenesis (TNF-a). Likewise, repeated ketamine administration increased adult hippocampal neurogenesis. These
data indicate that repeated ketamine administration had greater behavioral consequences than acute
treatment and that the drug might be imparting antidepressant effects through its effects on neuro-
plasticity and inflammatory processes rather than the typical neurochemical/hormonal factors affected
by stressors.
This article is part of the Special Issue entitled ‘Ionotropic glutamate receptors’.
© 2016 Elsevier Ltd. All rights reserved.

1. Introduction ineffective in 30e40% of depressed patients, leaving a large


treatment-resistant population (Thase et al., 2005). Furthermore,
Although currently available antidepressant medications are even treatment responders often still display some degree of
widely used, they are not without substantial limitations. Tradi- symptomology and are at risk of relapse (Rush et al., 2006; Ghaemi,
tional pharmacological antidepressants typically require weeks to 2008). Hence, it is of considerable importance to develop novel
months to provide symptomatic relief (Trivedi et al., 2006) and are more efficacious and rapid treatment strategies. Such efforts should
undoubtedly look beyond conventional monoamine based drugs
and begin to consider alternate mechanisms of depression
pathology.
Abbreviations: 5-HIAA, 5-hydroxyindole acetic acid; 5-HT, serotonin; ANOVA, Emerging findings have indicated that ketamine, a non-
analysis of variance; DCX, doublecortin; FST, forced swim test; GM-CSF, gran-
competitive N-methyl-D-aspartate (NMDA) receptor antagonist
ulocyte-macrophage colony stimulating factor; HPA, hypothalamic-pituitary adre-
nal; HPLC, High-performance liquid chromatography; IFN, interferon; IL, widely used for its anesthetic properties, shows promise as a novel
interleukin; i.p., intraperitoneal; LPS, lipopolysaccharide; MHPG, 3-methoxy-4- treatment for depression (Murrough, 2011; Blier et al., 2012). When
hydroxyphenylglycol; mTOR, mammalian target of rapamycin; NE, norepineph- given at sub-anesthetic doses, a single ketamine infusion was re-
rine; NMDA, N-methyl-D-aspartate; PFC, prefrontal cortex; PBS, phosphate-buffered ported to not only promote fast-acting antidepressant effects, often
saline; TNF-a, tumor necrosis factor-a.
* Corresponding author.
within hours (Berman et al., 2000; Zarate et al., 2006; Phelps et al.,
E-mail address: shawn_hayley@carleton.ca (S. Hayley). 2009), but also to be effective in historically treatment-resistant

http://dx.doi.org/10.1016/j.neuropharm.2016.04.021
0028-3908/© 2016 Elsevier Ltd. All rights reserved.
M. Clarke et al. / Neuropharmacology 112 (2017) 210e220 211

patients (Liebrenz et al., 2007; Machado-Vieira et al., 2009; Mice were placed into a 4 L glass beaker (height 25.1 cm,
Mathew et al., 2010). Furthermore, the antidepressant effects diameter 15.9 cm) filled to a depth of 15 cm with 22 ± 1  C water.
following a single treatment have been reported to persist for Animals remained in the beaker for six minutes during which time
several days (Liebrenz et al., 2007) to even weeks (Irwin and their activity was videotaped. The last four minutes of each video
Iglewicz, 2010; Mathew et al., 2010). Hence, ketamine is particu- was scored for time spent immobile. Mobile behaviors were
larly unique in its clinical profile and is therefore of potentially defined any motion beyond which was required for the animal to
enormous therapeutic significance. remain afloat.
Given that stressor exposure is a leading risk factor in the
development of depression (Fava and Kendler, 2000; Harro, 2013), 2.3. Experiment 1b: forced swim test following repeated ketamine
it is important to evaluate the actions of ketamine in stressor-based
models of the disorder. Admittedly, a few recent studies have This experiment was identical to Expt. 1a, except that mice now
shown that ketamine can modify some of the effects of chronic received three separate injections of either ketamine (10 mg/kg,
stressor exposure (Garcia et al., 2009; Li et al., 2011; Ma et al., 2013), i.p.) or saline over the course of two weeks. Two separate cohorts
as well as that of the bacterial endotoxin, lipopolysaccharide (LPS) were used with mice being assessed in the forced swim test 2 and 8
(Walker et al., 2013). However, data are still sparse and the effects of days following their final injection.
ketamine using various administration regimens in the context of
different stressors are still largely unexplored. To this end, we 2.4. Experiment 2: acute ketamine and restraint stress
evaluated the impact of a ketamine in three different stressors
models. Firstly, we utilized an acute stressor in the form of a psy- Mice received a single i.p. injection of either saline or ketamine
chogenic stressor (restraint). Secondly, we assessed the effects of (5 mg/kg) 30 min prior to receiving a restraint stressor. The re-
ketamine in the context of the systemic stressor, LPS, and did so for straint involved placing mice snugly into a conical shaped plastic
two main reasons: 1. Antidepressants can have ant-inflammatory bag with an opening at the end to allow for breathing, and it lasted
consequences and 2. It has been argued that some the acute ef- for 30 min. Five minutes after the stressor, mice were rapidly
fects of LPS (including sickness and neurochemical changes) may decapitated (n ¼ 8/group) with trunk blood and brain tissue
reflect aspects of depression (e.g. neurovegetative symptoms) collected.
(Dantzer and Kelley, 2007; Anisman and Hayley, 2012). Thirdly, we
assessed whether repeated ketamine injections in the context of a 2.4.1. Corticosterone analyses
chronic unpredictable stressor paradigm might have long lasting Trunk blood (collected in tubes containing 10 mg of EDTA) was
antidepressant-like behavioral and neuronal outcomes. centrifuged at 3600 rpm for 8 min. Plasma was then collected and
Since it is possible that ketamine's antidepressant actions might immediately stored at 80  C until analyzed. Corticosterone levels
stem from rapid changes in synaptic plasticity (Li et al., 2011; were measured by a commercial radioimmunoassay kit (ICN Bio-
Duman et al., 2012) or possibly through inflammatory factors, we medicals, CA, USA). Inter-assay variability was avoided by assaying
assessed adult hippocampal neurogenesis and cytokine levels, in all samples (in duplicate) within a single run.
addition to plasma corticosterone and brain-region specific
monoamines. Our data indicated that ketamine had protracted 2.4.2. Monoamine detection
antidepressant-like behavioral effects that were associated with Brains were sliced into coronal sections using a chilled micro-
augmented neuroplasticity (enhanced neurogenesis), as well as dissecting block containing slots (spaced 0.5 mm apart) for single
anti-inflammatory consequences (cytokine reductions). However, edged razor blades. The hypothalamus, prefrontal cortex (PFC) and
ketamine generally did not modify stressor-induced corticoid or hippocampus were dissected from the sections using razor blades
central monoamine changes, suggesting that the effects of the drug and immediately placed on dry ice. The tissue was stored at 80  C
were distinct from that of “traditional” antidepressants that typi- until analyzed using high-performance liquid chromatography
cally antagonize stressor provoked neurochemical disturbances. (HPLC).
HPLC was performed as previously reported by Liu et al. (2014),
2. Experimental procedures to determine levels of norepinephrine (NE) and serotonin (5-HT),
and their metabolites, 3-methoxy-4-hydroxyphenylglycol (MHPG)
2.1. Animals and 5-hydroxyindole acetic acid (5-HIAA). Briefly, brain tissue was
homogenized in a solution of 0.3 M monochloroacetic acid, 0.1 mM
Male CD1 mice (8e10 weeks) from Charles River Laboratories NaEDTA, 1% methanol and H2O, and centrifuged at 10 000 rpm at
(Laprairie, Quebec, Canada) were acclimatized for 1 week before 4  C. Supernatants were then injected, at a flow rate of 1 ml/min,
experimental procedures began. Animals were singly housed in into the HPLC system (Agilent 1100) with electrochemical detector
standard polypropylene cages and maintained on a 12-h light/dark (DECADE SDC) and Eclipse XDB-C-8 (4.6e150 mm column). Each
cycle. Food and water were provided ad libitum and room tem- liter of the mobile phase used for the separation comprised 90 mM
perature was maintained at 21  C. All experiments were approved sodium dehydrogenate phosphate, 1.1 mM 1-octane sulfuric acid,
by the Carleton University Committee for Animal Care and adhered 50 mM EDTA, acetonitrile 10%, 50 mM citric acid 50, 5 mM potas-
to the guidelines outlined by the Canadian Council for the Use and sium chloride 5, and HPLC-grade water. Determination of the area
Care of Animals in Research. and height of the peaks was carried out with the aid of a Hewlett-
Packard integrator. The protein concentration of each sample was
2.2. Experiment 1a: forced swim test following acute ketamine determined using bicinchoninic acid with a protein analysis kit
(Pierce Scientific, Brockville, Ontario) and a spectrophotometer
To establish that ketamine is indeed having antidepressant-like (Brinkman, PC800 colorimeter).
consequences, immobility was assessed in a forced swim test
following exposure to ketamine or vehicle. To this end, mice 2.5. Experiment 3: acute ketamine and LPS treatment
(n ¼ 10/group) received a single intraperitoneal (i.p.) injection of
either saline or ketamine (10 mg/kg) and were subjected to the Mice (n ¼ 8/group) were given a single injection of either saline
forced swim test 1 h and 5 days later or 2 and 8 days later. or ketamine (5 or 10 mg/kg, i.p.) 30 min prior to an injection of
212 M. Clarke et al. / Neuropharmacology 112 (2017) 210e220

saline or the bacterial endotoxin, LPS (10 mg, i.p.). Two hours later, mesh divider was placed between the two for the duration of the
mice were decapitated, with trunk blood and brain tissue collected. session) (30 min); exposure to social stress by placing mice in a
congener's soiled cage (with the latter having been removed)
2.5.1. Sickness behavior (overnight); exposure to predator (rat) odor by placing mice in a
The overall appearance of each animal was rated to assess the cage containing rat feces and soiled bedding (overnight); wet
degree of sickness exhibited. Sickness measurements were scored bedding (overnight); tail hanging (60 s); placement in an empty
on a four-point scale (0 ¼ no symptom, 1 ¼ one symptom present, cage without sawdust or nesting (overnight); lights on during dark
2 ¼ two symptoms presents, 3 ¼ three or more symptoms) with phase (overnight); tail pinch by placing a small butterfly clamp
respect to absent exploration and locomotion, curled body posture, (lined with a gauze pad) over the tail (5 min); 30 cage tilt (over-
ptosis, ragged fur, lethargy, pilo erection, drooping eyelids, and night). Due to the nature of the stressor paradigm, animals
overall nonresponsiveness. We previously observed that this pro- receiving the chronic stressor regimen were housed in holding
cedure yielded better than 90% agreement between two raters rooms separate from, but otherwise identical to, their non-stressed
blind to the treatment mice received. counterparts.
Two weeks into the stressor regimen, mice were further sub-
2.5.2. Corticosterone and monoamine assays divided, and received either saline or ketamine (10 mg/kg, i.p.) on
Corticosterone analysis and monoamine detection were con- Days 14, 21 and 28 (paralleling the repeated ketamine injection
ducted as in Experiment 2. regimen used in Experiment 1b). Four cohorts were used (n ¼ 8/
group in all cases), with one used for behavioral measures and three
2.5.3. Plasma cytokine analysis for biological outcomes with one sacrificed two hours after the last
Levels of circulating cytokines [granulocyte-macrophage colony ketamine injection on Day 28, and the other two were sacrificed on
stimulating factor (GM-CSF), interferon (IFN)-g, interleukin (IL)-10, Day 29, twenty-four hours after the last ketamine injection. Mice
IL-1b, IL-6 and tumor necrosis factor (TNF)-a] were determined by sacrificed on Day 28 were rapidly decapitated and trunk blood was
multiplex analysis using the Luminex 100 suspension-based bead taken for corticosterone determination. For the mice sacrificed on
array system (Luminex Corp., Austin, TX) with a custom multiple Day 29, one cohort underwent perfusion for immunohistochemical
cytokine detection kit (MILLIPLEX MAP Mouse Cytokine/Chemo- analyses, while mice in the second cohort were rapidly decapitated
kine Kit, Millipore, Cat. #MPXMCYTO-70K). The assay was per- and trunk blood and brain tissue were collected for corticosterone
formed as previously reported by Gibb et al. (2011) according to the and monoamine determinations, respectively.
kit manufacturer's instructions (see www.millipore.com/
userguides). 2.6.1. Corticosterone and monoamine assays
Briefly, a standard curve was generated using different dilutions Corticosterone analysis and monoamine detection were con-
of a standard cocktail in order to determine protein concentrations ducted exactly as already described.
in plasma samples. Next, 25ml of prepared standards or controls was
added to a pre-wet 96-well filter plate. For unknown wells, 25ml of 2.6.2. Doublecortin immunohistochemistry
assay buffer and 25ml of plasma sample were added to each well. Animals were perfused with saline followed by 4% para-
The pre-mixed anti-mouse cytokine beads were vortexed and formaldehyde. Brains were post-fixed overnight in 4% para-
sonicated, and 25 ml of the bead solution was added to each well. formaldehyde and then cryoprotected in a 30% w/v sucrose
Plates were then incubated on a plate shaker overnight at 4  C. solution. Brains were then sliced on a cryostat into 40-mm coronal
Following incubation samples were washed twice with 200ml of sections containing the hippocampus. Hippocampal sections were
wash buffer and removed by vacuum filtration, and 25ml of detec- operationally defined as early/rostral (bregma 1.22 to 1.82),
tion antibody beads were added to each well and incubated, again middle (bregma 1.82 to 2.46) and late/caudal (bregma 2.46
on a plate shaker, for 90 min at room temperature. This was fol- to 2.92), as we previously reported (Seguin et al., 2009).
lowed by 25 ml of streptavidin-PE being added to each well con- Slides were washed with phosphate-buffered saline (PBS) and
taining the detection antibodies and samples were again incubated incubated overnight at 4  C with a polyclonal goat anti-
with agitation for 30 min. Finally, 25 ml of Beadlyte stop solution doublecortin (DCX) antibody (Santa Cruz) at a concentration of
was added and samples were then resuspended in sheath fluid. The 1:200. Slides were then rinsed with PBS and incubated overnight at
filter plate was analyzed using the Luminex 100 instrument and the 4  C with AlexaFluor-488 conjugated donkey anti-goat antibody
data fitted with a five-parameter logistic regression curve using (1:100; Invitrogen Life Technologies). Subsequently, slides were
Analyst software (Millipore). In cases where cytokine levels were rinsed in PBS and cover-slipped using Fluoromount.
too low to be detected, samples were assigned a value of one-half DCXþ staining was assessed using a Nikon C1 fluorescent mi-
the lower limit of quantification. croscope equipped with a Hamamatsu Orca camera. Stereology
Investigator software was used to capture and quantify images.
2.6. Experiment 4: repeated ketamine and chronic unpredictable Images of the dentate gyrus were taken at representative z levels
stress and exposure was limited to one minute. The images were taken at
a constant exposure of 100.161, a constant gain of 2, a constant
Mice were randomly assigned to either a stress (chronic un- offset of 1 and a constant magnification of 30. DCXþ neurons
predictable stress) or non-stress condition. The stressor regimen were counted using the Cell Counter extension of Image-J software
involved twice daily exposure (given at unpredictable times; one in (National Institutes of Health). Neurons were only counted if the
the morning and one in the afternoon) to a series of different soma was clearly visible and in line with the shape of the dentate
stressors for 28 days. The chronic stressor regimen included the gyrus and a portion of the axonal processes was visible.
following stressors: restraint in semicircular Plexiglas tubes
(4  12 cm) with tails taped to prevent mice from turning (30 min); 2.6.3. Behavioral assessments: sucrose preference
restraint in tight-fitting triangular bags with a nose-hole for Mice were given free access to both a 2% sucrose solution and
breathing (30 min); exposure to social stress by placing mice in the water for 2 days prior to baseline testing to acclimate them to the
cage of an aggressive retired breeder non-experimental mouse taste of sucrose. Initial baseline preference measures were taken
(after first hostile contact or submission by experimental mouse a the night before the start of the stress regimen. From 5 pm to 8 am,
M. Clarke et al. / Neuropharmacology 112 (2017) 210e220 213

free access was provided to both 2% sucrose solution and water, and
resulting consumption of both solutions was measured. Both su-
crose and water consumption were calculated as a function of an-
imal weight. Sucrose preference was measured starting at 5 pm on
the same day as injections (which were delivered at 9 am). Per-
centage sucrose preference was calculated as
Sucrose Consumption
ðSucrose ConsumptionþWater ConsumptionÞ
*100. Thereafter, the baseline
percent sucrose was subtracted from that calculated during weeks
4 and 5 of the experiment.

2.7. Statistical analyses

Data from Experiments 1a and 1b were analyzed by two-tailed


Student's t-test. For all other experiments, data were analyzed by
analysis of variance (ANOVA). For ANOVAs exhibiting a significant
interaction, post hoc analyses using Fisher's protected least signif-
icant difference were conducted to determine the exact nature of
the relationship. Data were evaluated using a StatView (version 6.0)
statistical software package available from the SAS Institute, Inc.

3. Results

3.1. Experiment 1a: acute ketamine forced swim test

The effect of a single ketamine injection on forced swim test


performance 1 h, 2 days, 5 days and 8 days after ketamine treat-
ment was assessed. As shown in Fig. 1, ketamine treatment reduced
forced swim time at the one-hour time point (t ¼ 2.45, df ¼ 14,
p < 0.05) whereas no significant effects of ketamine treatment were
found for any of the other time points (p > 0.05).

3.2. Experiment 1b: repeated ketamine forced swim test

Repeated ketamine treatment significantly reduced immobility


time at both 2 and 8 days after the last injection (t ¼ 2.66, df ¼ 18,
p < 0.05 and t ¼ 5.43, df ¼ 18, p < 0.001, respectively; Fig. 1),
indicating long-term antidepressant-like effects following three
injections (over a two-week interval) of the drug.

3.3. Experiment 2

3.3.1. Plasma corticosterone levels


The restraint stressor significantly increased plasma cortico-
sterone levels relative to non-stressed animals (F1, 28 ¼ 104.53,
p < 0.001; Fig. 2A). There was no influence of ketamine adminis-
tration on corticosterone levels. Fig. 1. The effects of a single and repeated ketamine injection (10 mg/kg, i.p.) on
immobility time in a forced swim test. A. A single injection of ketamine reduced
3.3.2. Monoamine variations immobility time one hour but not 5 days after the injection. B. A single injection of
ketamine had no effect on immobility time 2 or 8 days after the injection. C. Repeated
Although there were no significant interaction effects, hypo-
ketamine treatment (three injections over 2 weeks) reduced immobility time both 2
thalamic levels of 5-HT were increased by the stressor days and 8 days after the final injection. Data expressed as means ± SEM. #p < 0.05;
(F1,27 ¼ 12.86, p < 0.05; data not shown), whereas ketamine **p < 0.001.
treatment was without effect. In contrast, there were no significant
differences in hypothalamic NE, MHPG or 5-HIAA concentrations (Fs1,26 ¼ 5.47 and 5.46 respectively, p < 0.05) along with promoting
among the groups. elevations of hippocampal 5-HIAA levels (Fs1,27 ¼ 7.76 and 9.07
Within the PFC, a significant Stress  Ketamine interaction was respectively, p < 0.01; Fig. 2C). Finally, levels of 5-HT were increased
present with regard to 5-HT levels within the PFC (F1,26 ¼ 4.70, by the stressor (F1, 26 ¼ 5.43, p < 0.05; Fig. 2C) whereas no signif-
p < 0.05). As shown in Fig. 2B, the restraint stress decreased 5-HT icant variations in hippocampal MHPG were observed.
levels (p < 0.05) but this effect was attenuated by ketamine treat-
ment (p < 0.05). Furthermore, 5-HIAA levels were reduced by ke- 3.4. Experiment 3
tamine treatment (F1, 26 ¼ 5.63, p < 0.05). Finally, PFC MHPG was
increased by the stressor (F1,27 ¼ 7.89, p < 0.01; Fig. 2B), in the 3.4.1. Sickness behavior
absence of any effect of ketamine or any effects on NE. A significant main effect was observed with regards to the
In the absence of interaction effects, significant main effects for impact of LPS on sickness symptom presentation (F1, 42 ¼ 111.79,
the stressor and ketamine treatments were reported within the p < 0.001). The follow up comparisons confirmed that LPS markedly
hippocampus, with both treatments reducing NE levels promoted sickness behavior (P < 0.05), with average composite
214 M. Clarke et al. / Neuropharmacology 112 (2017) 210e220

Fig. 2. Effects of a single ketamine injection (5 mg/kg, i.p.) and restraint stress on plasma corticosterone and monoamine levels. A. Acute ketamine treatment did not influence the
restraint stress-induced increase in levels of plasma corticosterone. B. Ketamine treatment prevented the stress-induced decrease in prefrontal cortex (PFC) 5-HT while it reduced
levels of the metabolite, 5-HIAA. Restraint stress increased levels of MHPG. C. Hippocampal levels of norepinephrine (NE) were reduced by both ketamine and restraint stress, while
levels of the 5-HT metabolite, 5-HIAA, were increased by both treatments. Restraint stress increased hippocampal 5-HT. Data expressed as means ± SEM. #p < 0.05; *p < 0.01;
**p < 0.001.

sickness score among the non-LPS treated (saline alone, or keta- and F2,40 ¼ 3.46, p < 0.05, respectively; data not shown). In contrast,
mine alone at 5 or 10 mg/kg) mice being 1.1 ± 0.07, while that of the in the absence of any LPS effects or changes in 5-HT, both ketamine
LPS treated (LPS alone or LPS with the either of the two ketamine doses significantly reduced hypothalamic levels of 5-HIAA
doses) mice was 2.43 ± 0.13. However, there was no significant (Fs2,40 ¼ 3.71, p < 0.05; data not shown).
main effect for ketamine nor was there any significant interaction Within the PFC, a significant main effect was apparent for LPS
with LPS. In fact, the ketamine treated mice had scores virtually with respect to levels of MHPG (F1,42 ¼ 12.38) but not NE. As shown
identical to those that received saline. in Fig. 3C, LPS increased MHPG accumulation (p < 0.01), whereas
ketamine had no effect. No significant variations in PFC 5-HT or 5-
3.4.2. Plasma corticosterone levels HIAA were apparent for either of the treatments.
LPS treatment provoked a large corticoid elevation (F1, Finally, within the hippocampus, LPS reduced NE while it
42 ¼ 68.81, p < 0.001; Fig. 3A). However, no effect of ketamine increased levels of the metabolite, MHPG (F1,42 ¼ 7.73, p < 0.01 and
administration at either of the doses was observed. F1,42 ¼ 6.76, p < 0.05, respectively; Fig. 3D). However, hippocampal
5-HT levels varied as a function of an LPS  Ketamine interaction
3.4.3. Monoamine variations (F2,42 ¼ 3.34, p < 0.05). As shown in Fig. 3D, ketamine attenuated
Although no differences were evident for hypothalamic NE, LPS the LPS induced elevation of 5-HT (p < 0.01). Similarly, 5-HIAA
and ketamine both increased hypothalamic levels of the NE levels were significantly increased by LPS (F1,42 ¼ 14.38,
metabolite, MHPG, within the hypothalamus (F1,40 ¼ 8.73, p < 0.01 p < 0.001; Fig. 3D), though ketamine had no effect in this case.
M. Clarke et al. / Neuropharmacology 112 (2017) 210e220 215

Fig. 3. Effects of a single ketamine injection (5 or 10 mg/kg, i.p.) and lipopolysaccharide treatment (LPS; 10 mg, i.p.) on plasma corticosterone and monoamine levels. A. Acute
ketamine treatment did not influence the LPS-induced increase in levels of plasma corticosterone. B. The high dose of ketamine prevented the LPS-induced increase in IL-1b (top
panel). Ketamine dose-dependently prevented the LPS-induced increase in TNF-a (bottom panel). C. LPS increased levels of the norepinephrine (NE) metabolite, MHPG, within the
prefrontal cortex (PFC) while ketamine was without effect. D. Ketamine attenuated the LPS-induced increase in hippocampal 5-HT, but did not influence the LPS-induced increase in
NE, MHPG or 5-HIAA. Data expressed as means ± SEM. #p < 0.05; *p < 0.01; **p < 0.001.

3.4.4. Cytokines 3.5.2. Monoamine variations


The effects of ketamine on LPS induced cytokine changes were Within the hypothalamus, levels of the 5-HT metabolite, 5-
assessed for the following cytokines: GM-CSF, IFN-g, IL-10, IL-1b, IL- HIAA, were reduced by stressor treatment (F1,28 ¼ 4.32, p < 0.05)
6 and TNF-a. Levels of IFN-g were generally below detection, while they were increased by ketamine treatment (F1,28 ¼ 8.11,
however, LPS significantly increased circulating levels of GM-CSF, p < 0.05; data not shown). In contrast, there were no significant
IL-10 and IL-6 (F1,44 ¼ 34.03, F1,42 ¼ 45.07 and F1,43 ¼ 229.46, differences in hypothalamic 5-HT, NE or 5-HT.
respectively, p < 0.05; data not shown), whereas ketamine was Levels of 5-HIAA within the PFC were found to be significantly
without effect. However, a significant LPS  Ketamine interaction elevated by ketamine treatment (F1,28 ¼ 5.75, p < 0.05). However, as
was apparent for IL-1b and TNF-a (F2,43 ¼ 3.26 and F2,41 ¼ 5.00 depicted in Fig. 4B, the rise of PFC 5-HIAA was clearly confined to
respectively, p < 0.05). As shown in Fig. 3B, ketamine dose- the control (non-stressed) mice (p < 0.05) and the stressor regimen
dependently attenuated LPS-induced elevations of both IL-1b and appeared to ablate this effect. None of the other assayed neuro-
TNF-a levels (p < 0.05). transmitters (5-HT, NE, MHPG) were significantly affected by the
ketamine or stressor treatments.
Finally, repeated ketamine treatment also elevated levels of
hippocampal MHPG (F1,28 ¼ 5.64, p < 0.05; Fig. 4C). Yet, neither NE
3.5. Experiment 4
or 5-HT (or 5-HIAA) were significantly influenced by any of the
treatments.
3.5.1. Plasma corticosterone levels
There were no significant differences in plasma corticosterone
levels among the groups at either two or twenty-four hours after 3.5.3. Doublecortin immunohistochemistry
the last ketamine injection (Fig. 4A). No significant effects were apparent for the “early” (i.e.
216 M. Clarke et al. / Neuropharmacology 112 (2017) 210e220

Fig. 4. Effects of repeated ketamine treatment (10 mg/kg, i.p.) and chronic unpredictable stress on plasma corticosterone and monoamine levels. A. Plasma corticosterone levels
were not affected by repeated ketamine treatment or chronic unpredictable stress two hours (top panel) or twenty-four hours (bottom panel) after the final of three ketamine
injections. B. Ketamine treatment increased levels of the 5-HT metabolite, 5-HIAA, within the prefrontal cortex (PFC). C. Hippocampal levels of the norepinephrine (NE) metabolite,
MHPG, were increased by ketamine treatment. Data expressed as means ± SEM. #p < 0.05.

bregma 1.22 to 1.82) portion of the hippocampus. Yet, although reduced sucrose preference below that of controls on both of
the interaction missed significance, there were significant main Weeks 4 and 5 of the experiment (p < 0.05). However, there
effects for both ketamine injection and stressor exposure in the appeared to be some degree of “rebound” at Week 5, with
“middle” portion (i.e., bregma 1.82 to 2.46). Specifically, ketamine þ stress increasing sucrose preference above that of the
repeated ketamine injections significantly increased the number of previous week (p < 0.05). In fact, there was an overall significant
DCXþ neurons found in the “middle” region of the hippocampal increase in sucrose preference at Week 5 compared to Week 4
dentate gyrus (F1,28 ¼ 4.53, p < 0.05; Fig. 5) relative to the control (F3,37 ¼ 5.13; p < 0.05).
groups. In contrast, the stressor regimen significantly reduced the
number of “middle” hippocampal DCXþ neurons (F1,28 ¼ 5.33, 4. Discussion
p < 0.05; Fig. 5). Finally, in the “late/caudal” (i.e. bregma 2.46
to 2.92) hippocampal region, the stressor had no effect and the Current antidepressant treatments are seriously limited, with
ANOVA for ketamine injection missed significance. However, based remission rates usually only achieved after prolonged treatment,
on our a prior hypothesis, we conduced followed up comparisons often with multiple different drug trials and 15e30% of individuals
and found that the non-stressed ketamine treated mice had not responding at all (Trivedi et al., 2006; Berlim and Turecki,
significantly elevated DCXþ counts, compared to the remaining 2007). The failure of artificial reductions in monoamine levels to
groups (p < 0.05; Fig. 5). produce depressive symptoms (Berton and Nestler, 2006; Ruhe
et al., 2007), coupled with studies showing that antidepressants
3.5.4. Behavioral effects: sucrose preference modulate glutamatergic imbalances in depressed patients (e.g.
A significant Stress  Ketamine interaction was apparent for Mauri et al., 1998; Prikhozhan et al., 1990), have contributed to the
sucrose preference (F1,37 ¼ 4.01; p < 0.05; Fig. 5). Surprisingly, both shift in focus towards non-monoaminergic mechanisms of
the stressor regimen and ketamine injections each significantly depression. In this regard, ketamine has emerged as a specific
M. Clarke et al. / Neuropharmacology 112 (2017) 210e220 217

Fig. 5. Chronic unpredictable stressor and repeated ketamine (10 mg/kg) treatment time-dependently altered sucrose preference (left bar graph). In particular, ketamine and the
stressor regimen reduced preference at Weeks 4 and 5 relative vehicle treatment (and adjusted for baseline preference). As depicted on the right, repeated ketamine treatment also
increased neurogenesis (as indicated by doublecortin (DCX) staining) in the operationally defined “middle” (bregma  1.82 to  2.46) and “later” (bregma  2.46 to  2.92) portions
of the hippocampal dentate gyrus. The chronic stressor reduced DCX staining only in the middle region and this effect reversed by ketamine treatment. Data expressed as
means ± SEM. *p < 0.05.

NMDA antagonist with very rapid and lasting antidepressant con- general olfactory or gustatory deficits. It is important to underscore
sequences (Berman et al., 2000 Zarate et al., 2006), even in previ- that one recent study found that ketamine did not affect anhedonia,
ously treatment resistant patients (Zarate et al., 2006; Krystal, as measured using intracranial self-stimulation (Donahue et al.,
2007). 2014). Similarly, ketamine did not influence the consumption of
We presently found that while a single ketamine injection sugar pellets in chronically stressed rats (Rezin et al., 2009). Yet,
altered forced swim test (FST) performance one hour after treat- others found that ketamine did have anti-anhedonic effects in ro-
ment, repeated administration of ketamine provoked long-lasting dents exposed to a number of different stressors (Garcia et al.,
antidepressant-like effects that were still evident 8 days 2009; Dwyer and Duman, 2013; Lally et al., 2014, 2015). Although
following the third of three ketamine injections. While these it is unclear as to the source of the discrepancies in these ketamine
findings confirm that ketamine can produce antidepressant effects behavioral findings, the dosing schedule and species used (mice vs
in a FST (Garcia et al., 2008; Li et al., 2010; Parise et al., 2013; rats) are obvious points of consideration.
Akinfiresoye and Tizabi, 2013), they also further suggest that the Given that depressed individuals often display heightened
efficacy of ketamine might be augmented over time through cu- stressor sensitivity and/or elevated basal neurochemical or
mulative dosing, which of course has important clinical hypothalamic-pituitary adrenal (HPA) axis activity (Parker et al.,
ramifications. 2003), it was of interest to assess whether the drug might reduce
It was surprising that ketamine did not reverse the chronic basic biological stressor responses. In this regard, we found that
stressor induced reductions in sucrose consumption and actually both an acute restraint stressor and a single challenge with the
itself reduced sucrose preference, which of course makes inter- bacterial endotoxin, LPS, markedly elevated plasma corticosterone
pretation of this test difficult. It is possible that the drug induced levels, but neither of these elevations were affected by ketamine
some dissociative or other unexpected effects (although these treatment. Similarly, the various stressor induced brain-region
should be minimal at the present relatively low dose) that could specific monoamine changes were generally unaffected by keta-
interfere with sucrose consumption. Yet, the fact that ketamine did mine treatment; or certainly at the very least, there was no
not influence food or water intake, suggests that it was not causing apparent pattern of ketamine effects. Thus, ketamine was clearly
218 M. Clarke et al. / Neuropharmacology 112 (2017) 210e220

not affecting the immediate hormonal/neurochemical response ameliorate many of the central actions of ketamine (Li et al., 2010).
induced by two different categories of stressors, and hence, its Whatever the case, it is important to note that numerous studies
relatively rapid antidepressant-like effects do not appear to stem have shown a relationship between hippocampal neurogenesis and
from any changes to acute stress reactivity. However, a caveat of antidepressant efficacy (Malberg et al., 2000; Van Bokhoven et al.,
these analyses is the fact that they represent a single static time 2011) and that ablation of neurogenesis diminished the behav-
point obtained from post-mortem tissue and further studies should ioral effects of a variety of antidepressants (Santarelli et al., 2003;
utilize in vivo sampling approaches (e.g. microdialysis) to deter- Surget et al., 2008).
mine any changes in the kinetics of neurotransmission. Finally, it is important to note that we only assessed neuro-
Beyond the neurochemical changes, it has become increasingly genesis after the repeated (three injection) ketamine treatment and
clear that inflammatory processes are relevant for depression and such effects might not be apparent after a single dose. Interestingly,
that immunological or systemic stressors (much like psychogenic however, a just published study reported that a single ketamine
stressor) might promote the illness (Sperner-Unterweger et al., injection increased the proportion of functionally mature dentate
2014; Dantzer and Kelley, 2007). We presently chose an LPS gyrus neurons in adult rats (Soumier et al., 2016), raising the pos-
regimen consistent with our previous work (Hayley et al., 2001; sibility that its neuroplastic effects could have immediate clinical
Gibb et al., 2008) in order to assess whether ketamine would consequences. In effect, the drug could be acting to increase sur-
modulate the well known immediate sickness, neurochemical and vival of recently generated neurons and rapidly facilitate their
inflammatory effects of this systemic stressor. As expected LPS integration into existing circuitry. However, given that the clinical
induced marked sickness symptoms, including curled body effects of ketamine dissipate within several days to a couple of
posture, lethargy, ptosis and piloerection, however, ketamine had weeks, it would be useful for a future study to conduct parallel time
no effect whatsoever on any of these symptoms. courses to address whether the neurogenesis changes are really
Yet, one of the most striking findings of the present study was linked to clinical efficacy in the long run.
that ketamine dose-dependently antagonized the LPS induced rise
of IL-1b and TNF-a. This is not totally unprecedented (Chang et al., 5. Conclusions
2009; Ward et al., 2011), but the current study is the first to illus-
trate ketamine's highly selective actions, as it had no effect on the In summary, ketamine provoked long-lasting antidepressant-
LPS induced elevations of IL-6, IL-10, IFN-g or GM-CSF. This might like behavioral effects, as well as increased hippocampal neuro-
be particularly important given that IL-1b and TNF-a act primarily genesis. It is interesting that ketamine increased neurogenesis in
through nuclear factor kappa B signaling [which has been linked to the later but not the earlier portions of the dorsal dentate gyrus,
depressive outcomes (Koo et al., 2010)], whereas the other cyto- since the more caudal-ventral portions of the hippocampus are
kines are more aligned with the JAK-STAT signaling pathway more related to anxiety and emotional processes, as opposed to the
(Mercurio and Manning, 1999; Kiu and Nicholson, 2012; Hayley heavier memory role ascribed to more anterior-dorsal portions
et al., 2013; Lichtblau et al., 2013). (Fuster-Matanzo et al., 2011). Ketamine also had selective antago-
Recent data have also raised the possibility that cytokines might nistic effects for LPS induced IL-1b and TNF-a, suggesting its po-
be used as endogenous biomarkers of antidepressant efficacy, with tential importance in depressed individuals with altered pro-
one study showing that baseline levels of IL-6 but not TNF-a were inflammatory cytokines (Hayley et al., 2013). However, the fact
able predictive of who would be responders vs non-responders to that this ketamine did not affect LPS-induced sickness or other
ketamine treatment (Yang et al., 2015). Furthermore, a recent cytokines, indicates that ketamine's anti-inflammatory effects are
multiplex study looking at 66 different factors (including over a highly selective and the drug is not influencing sickness symptom
dozen cytokines, numerous stress factors and peptides) found that clusters that have previously been suggested to reflect neuro-
ketamine, in the absence of an inflammatory stimulus, selectively vegetative features of depression (Dantzer and Kelley, 2007). One
altered TNF-a, fibroblast growth factor-9 and the anti-inflammatory caveat here is that we did not test mice at a later time (24 h) when a
cytokine, IL-4 (Wesseling et al., 2015). Hence, it appears that ke- recent study reported LPS induced anhedonia which was modified
tamine can alter the basal inflammatory cytokine milieu, as well as by ketamine treatment (Walker et al., 2013).
modify cytokine signaling in the face of an immune stressor (eg Intriguingly, ketamine did not appreciably influence the acute
LPS). It remains to be determined whether specific cytokine profiles corticoid and central monoamine effects of a typical laboratory
might offer predictive potential for gauging antidepressant stressor (restraint), immunogenic stressor (LPS) or a chronic un-
response efficacy and disease trajectory. predictable stressor regimen. Furthermore, the fact that ketamine
Accumulating evidence has indicated that ketamine might exert itself reduced sucrose preference (much like our chronic stress
its effects by influencing neuroplasticity (Li et al., 2011; Duman regimen did), raises the possibility that the drug had unexpected
et al., 2012). Accordingly, we presently found that three injections dissociative or other “side” effects that could influence the moti-
of ketamine administered over the span of two weeks increased vation or proclivity for sucrose preference. Overall, our data sup-
adult hippocampal neurogenesis. Interestingly, however, this effect ports the contention that ketamine can have beneficial
was restricted to the “middle and later” portions of the dentate antidepressant effects with repeated treatment but that its clinical
gyrus. Similarly, the chronic stress reduced hippocampal neuro- effects do not stem from the obvious modulation of stressor-
genesis, but only in the “middle” portion of this brain region. These sensitive neurochemical pathways, but could be linked to the
data indicate the importance of the internal hippocampal anatomy modulation of neuroplastic and inflammatory processes.
in stressor and antidepressant sensitivity.
Ketamine's neuroplastic effects might come about through the Funding and disclosure
mammalian target of rapamycin (mTOR) pathway, which typically
integrates signals from various endogenous growth factors to pro- The authors declare no conflict of interest.
mote rapid synaptogenesis and neurogenesis (Li et al., 2010; Zhou
et al., 2014). Although not evaluated in the current study, we pre- Acknowledgements
viously found that the mTOR inhibitor, rapamycin, prevented the
antidepressant-like and neurogenic effects of erythropoietin Shawn Hayley and Hymie Anisman are Canada Research Chairs
(Osborn et al., 2013). Likewise, rapamycin was also reported to and this work was supported by grants from the Canadian Institutes
M. Clarke et al. / Neuropharmacology 112 (2017) 210e220 219

of Health Research. We thank Kris Hansen for his assistance with Duman, R.S., 2010. mTOR-dependent synapse formation underlies the rapid
antidepressant effects of NMDA antagonists. Science 329 (5994), 959e964.
the experiments.
Li, N., Liu, R., Dwyer, J.M., Banasr, M., Lee, B., Son, H., Li, X.Y., Aghajamian, G.,
Duman, R.S., 2011. Glutamate N-methyl-D-aspartate receptor antagonists
rapidly reverse behavioral and synaptic deficits caused by chronic stress
References exposure. Biol. Psychiatry 69 (8), 754e761.
Lichtblau, N., Schmidt, F.M., Schumann, R., Kirkby, K.C., Himmerich, H., 2013. Cy-
Akinfiresoye, L., Tizabi, Y., 2013. Antidepressant effects of AMPA and ketamine tokines as biomarkers in depressive disorder: current standing and prospects.
combination: role of hippocampal BDNF, synapsin, and mTOR. Psycho- Int. Rev. Psychiatry 25 (5), 592e603.
pharmacol. (Berl.) 230 (2), 291e298. Liebrenz, M., Borgeat, A., Leisinger, R., Stohler, R., 2007. Intravenous ketamine
Anisman, H., Hayley, S., 2012. Inflammatory factors contribute to depression and its therapy in a patient with a treatment-resistant major depression. Swiss Med.
comorbid conditions. Sci. Signal 5 (244), 2; pe45. Wkly. 137 (15e16), 234e236.
Berlim, M.T., Turecki, G., 2007. What is the meaning of treatment resistant/re- Liu, G., Rustom, N., Litteljohn, D., Bobyn, J., Rudyk, C., Anisman, H., Hayley, S., 2014.
fractory major depression (TRD)? A systematic review of current randomized Use of induced pluripotent stem cell derived neurons engineered to express
trials. Eur. Neuropsychoparmacol. 17 (11), 696e707. BDNF for modulation of stressor related pathology. Front. Cell Neurosci. 8, 316.
Berman, R.M., Cappiello, A., Anand, A., Oren, D.A., Heninger, G.R., Charney, D.S., Ma, X.C., Dang, Y.H., Jia, M., Ma, R., Wang, F., Wu, J., Gao, C.G., Hashimoto, K., 2013.
Krystal, J.H., 2000. Antidepressant effects of ketamine in depressed patients. Long-lasting antidepressant action of ketamine, but not glycogen synthase
Biol. Psychiatry 47 (4), 351e354. kinase-3 inhibitor SB216763, in the chronic mild stress model of mice. PLoS One
Berton, O., Nestler, E.J., 2006. New approaches to antidepressant drug discovery: 8 (2), e56053.
beyond monoamines. Nat. Rev. Neurosci. 7 (2), 137e151. Machado-Vieira, R., Yuan, P., Brutsche, N., DiazGranados, N., Luckenbaugh, D.,
Blier, P., Zigman, D., Blier, J., 2012. On the safety and benefits of repeated intrave- Manji, H.K., Zarate Jr., C.A., 2009. Brain-derived neurotrophic factor and initial
nous injections of ketamine for depression. Biol. Psychiatry 72 (4), e11ee12. antidepressant response to an N-methyl-D-aspartate antagonist. J. Clin. Psy-
Chang, Y., Lee, J.J., Hsieh, C.Y., Hsiao, G., Chou, D.S., Sheu, J.R., 2009. Inhibitory effects chiatry 70 (12), 1662e1666.
of ketamine on lipopolysaccharide-induced microglial activation. Mediat. Malberg, J.E., Eisch, A.J., Nestler, E.J., Duman, R.S., 2000. Chronic antidepressant
Inflamm. 2009, 705379. http://dx.doi.org/10.1155/2009/705379. treatment increases neurogenesis in adult rat hippocampus. J. Neurosci. 20 (24),
Dantzer, R., Kelley, K.W., 2007 Feb. Twenty years of research on cytokine-induced 9104e9110.
sickness behavior. Brain Behav. Immun. 21 (2), 153e160. Mathew, S.J., Murrough, J.W., aan het Rot, M., Collins, K.A., Reich, D.L., Charney, D.S.,
Donahue, R.J., Muschamp, J.W., Russo, S.J., Nestler, E.J., Carlezon Jr., W.A., 2014 Oct 1. 2010. Riluzole for the relapse prevention following intravenous ketamine in
Effects of striatal DFosB overexpression and ketamine on social defeat stress- treatment-resistant depression: a pilot randomized, placebo-controlled
induced anhedonia in mice. Biol. Psychiatry 76 (7), 550e558. continuation trial. Int. J. Neuropsychopharmacol. 13 (1), 71e82.
Duman, R.S., Li, N., Liu, R., Duric, V., Aghajanian, G., 2012. Signaling pathways un- Mauri, M.C., Ferrrara, A., Boscati, L., Bravin, S., Zamberlan, F., Alecci, M.,
derlying the rapid antidepressant actions of ketamine. Neuropharmacology 62 Invernizzi, G., 1998. Plasma and platelet amino acid concentrations in patients
(1), 35e41. affected by major depression and under fluvoxamine treatment. Neuro-
Dwyer, J.M., Duman, R.S., 2013 Jun 15. Activation of mammalian target of rapamycin psychobiology 37 (3), 124e129.
and synaptogenesis: role in the actions of rapid-acting antidepressants. Biol. Mercurio, F., Manning, A.M., 1999. Multiple signals converging on NF-kB. Curr. Opin.
Psychiatry 73 (12), 1189e1198. Cell Biol. 11 (2), 226e232.
Fava, M., Kendler, K.S., 2000. Major depressive disorder. Neuron 28 (2), 335e341. Murrough, J.W., 2011. Ketamine as a novel antidepressant: from synapse to
Fuster-Matanzo, A., Llorens-Martín, M., de Barreda, E.G., Avila,  ndez, F.,
J., Herna behavior. Clin. Pharmacol. Ther. 91 (2), 303e309.
2011. Different susceptibility to neurodegeneration of dorsal and ventral hip- Osborn, M., Rustom, N., Clarke, M., Litteljohn, D., Rudyk, C., Anisman, H., Hayley, S.,
pocampal dentate gyrus: a study with transgenic mice overexpressing GSK3b. 2013. Antidepressant-like effects of erythropoietin: a focus on behavioural and
PLoS One 6 (11), e27262. hippocampal processes. PLoS One 8 (9), e72813.
Garcia, L.S., Comim, C.M., Valvassori, S.S., Reus, G.Z., Andreazza, A.C., Stertz, L., Parise, E.M., Alcantara, L.F., Warrn, B.L., Wright, K.N., Hadad, R., Sial, O.K.,
Fries, G.R., Gavioli, E.C., Kapczinski, F., Quevedo, J., 2008. Chronic administration Kroeck, K.G., Iniguez, S.D., Bolanos-Guzman, C.A., 2013. Repeated ketamine
of ketamine elicits antidepressant-like effects in rats without affecting hippo- exposure induces an enduring resilient phenotype in adolescent and adult rats.
campal brain-derived neurotrophic factor protein levels. Basic Clin. Pharmacol. Biol. Psychiatry 74 (10), 750e759.
Toxicol. 103 (6), 502e506. Parker, K.J., Schatzberg, A.F., Lyons, D.M., 2003. Neuroendocrine aspects of hyper-
Garcia, L.S., Comim, C.M., Valvassori, S.S., Reus, G.Z., Stertz, L., Kapczinski, F., cortisolism in major depression. Horm. Behav. 43 (1), 60e66.
Gavioli, E.C., Quevedo, J., 2009. Ketamine treatment reverses behavioral and Phelps, L.E., Brutsche, N., Moral, J.R., Luckenbaugh, D.A., Manji, H.K., Zarate Jr., C.A.,
physiological alterations induced by chronic mild stress in rats. Prog. Neuro- 2009. Family history of alcohol dependence and initial antidepressant response
psychopharmacol. Biol. Psychiatry 33 (3), 450e455. to an N-methyl-D-aspartate antagonist. Biol. Psychiatry 65 (2), 181e184.
Ghaemi, S.N., 2008. Why antidepressants are not antidepressants: STEP-BD, Prikhozhan, A.V., Kovalev, G.I., Raevskii, K.S., 1990. Effects of antidepressive agents
STAR*D, and the return of neurotic depression. Bipolar Disord. 10 (8), 957e968. on glutamatergic autoregulatory presynaptic mechanism in the rat cerebral
Gibb, J., Hayley, S., Gandhi, R., Poulter, M.O., Anisman, H., 2008. Synergistic and cortex. Bull. Eksp. Biol. Med. 110 (12), 624e626.
additive actions of a psychosocial stressor and endotoxin challenge: circulating Rezin, G.T., Gonçalves, C.L., Daufenbach, J.F., Fraga, D.B., Santos, P.M., Ferreira, G.K.,
and brain cytokines, plasma corticosterone and behavioral changes in mice. Hermani, F.V., Comim, C.M., Quevedo, J., Streck, E.L., 2009 Aug 14. Acute
Brain Behav. Immun. 22 (4), 573e589. administration of ketamine reverses the inhibition of mitochondrial respiratory
Gibb, J., Hayley, S., Poulter, M.O., Anisman, H., 2011. Effects of stressors and immune chain induced by chronic mild stress. Brain Res. Bull. 79 (6), 418e421.
activating agents on peripheral and central cytokines in mouse strains that Ruhe, H.G., Mason, N.S., Schene, A.H., 2007. Mood is indirectly related to serotonin,
differ in stressor responsivity. Brain Behav. Immun. 25 (3), 468e482. norepinephrine and dopamine levels in humans: a meta-analysis of mono-
Harro, J., 2013. Animal models of depression vulnerability. Curr. Top. Behav. Neu- amine depletion studies. Mol. Psychiatry 12 (4), 331e359.
rosci. 14, 29e54. Rush, A.J., Trivedi, M.H., Wisniewski, S.R., Nierenberg, A.A., Stewart, J.W., Warden, D.,
Hayley, S., Scharf, J., Anisman, H., 2013. Central administration of murine interferon- Niederehe, G., Thase, M.E., Lavori, P.W., Lebowitz, B.D., McGrath, P.J.,
a induces depressive-like behavioral, brain cytokine and neurochemical alter- Rosenbaum, J.F., Sackeim, H.A., Kupfer, D.J., Luther, J., Fava, M., 2006. Acute and
ations in mice: a mini-review and original experiments. Brain Behav. Immun. longer-term outcomes in depressed outpatients requiring one or several
31, 115e127. treatment steps: a STAR*D report. Am. J. Psychiatry 163 (11), 1905e1917.
Hayley, S., Lacosta, S., Merali, Z., van Rooijen, N., Anisman, H., 2001 Mar. Central Santarelli, L., Saxe, M., Gross, C., Surget, A., Battaglia, F., Dulawa, S., Weisstaub, N.,
monoamine and plasma corticosterone changes induced by a bacterial endo- Lee, J., Duman, R., Arancio, O., Belzung, C., Hen, R., 2003. Requirement of hip-
toxin: sensitization and cross-sensitization effects. Eur. J. Neurosci. 13 (6), pocampal neurogenesis for the behavioral effects of antidepressants. Science
1155e1165. 301 (5634), 805e809.
Irwin, S.A., Iglewicz, A., 2010. Oral ketamine for the rapid treatment of depression Seguin, J.A., Brennan, J., Mangano, E., Hayley, S., 2009. Proinflammatory cytokines
and anxiety in patients receiving hospice care. J. Palliat. Med. 13 (7), 903e908. differentially influence adult hippocampal cell proliferation depending upon
Kiu, H., Nicholson, S.E., 2012. Biology and significance of the JAK/STAT signalling the route and chronicity of administration. Neuropsychiatr. Dis. Treat. 5, 5e14.
pathways. Growth Factors 30 (2), 88e106. Soumier, A., Carter, R.M., Schoenfeld, T.J., Cameron, H.A., 2016 Mar 31. New hip-
Koo, J.W., Russo, S.J., Ferguson, D., Nestler, E.J., Duman, R.S., 2010. Nuclear factor- pocampal neurons mature rapidly in response to ketamine but are not required
kappaB is a critical mediator of stress-impaired neurogenesis and depressive for its acute antidepressant effects on neophagia in rats. eNeuro 3 (2).
behavior. Proc. Natl. Acad. Sci. 107 (6), 2669e2674. Sperner-Unterweger, B., Kohl, C., Fuchs, D., 2014 Jan 3. Immune changes and neu-
Krystal, J.H., 2007. Ketamine and the potential role for rapid-acting antidepressant rotransmitters: possible interactions in depression? Prog. Neuro-
medications. Swiss Med. Wkly. 137 (15e16), 215e216. psychopharmacol. Biol. Psychiatry 48, 268e276.
Lally, N., Nugent, A.C., Luckenbaugh, D.A., Niciu, M.J., Roiser, J.P., Zarate Jr., C.A., 2015 Surget, A., Saxe, M., Leman, S., Ibarguen-Vargas, Y., Chalon, S., Griebel, G., Hen, R.,
May. Neural correlates of change in major depressive disorder anhedonia Belzung, Z., 2008. Drug-dependent requirement of hippocampal neurogenesis
following open-label ketamine. J. Psychopharmacol. 29 (5), 596e607. in a model of depression and of antidepressant reversal. Biol. Psychiatry 64 (4),
Lally, N., Nugent, A.C., Luckenbaugh, D.A., Ameli, R., Roiser, J.P., Zarate, C.A., 2014 Oct 293e301.
14. Anti-anhedonic effect of ketamine and its neural correlates in treatment- Thase, M.E., Haight, B.R., Richard, N., Rockett, C.B., Mitton, M., Modell, J.G.,
resistant bipolar depression. Transl. Psychiatry 4, e469. VanMeter, S., Harriett, A.E., Wang, Y., 2005. Remission rates following antide-
Li, N., Lee, B., Liu, R., Banasr, M., Dwyer, J.M., Iwata, M., Li, X.Y., Aghajanian, G., pressant therapy with bupropion or selective serotonin reuptake inhibitors: a
220 M. Clarke et al. / Neuropharmacology 112 (2017) 210e220

meta-analysis of original data from 7 randomized controlled trials. J. Clin. J. Trauma 70 (6), 1471e1479.
Psychiatry 66 (8), 974e981. Wesseling, H., Rahmoune, H., Tricklebank, M., Guest, P.C., Bahn, S., 2015. A targeted
Trivedi, M.H., Rush, A.J., Wisniewski, S.R., Nierenberg, A.A., Warden, D., Ritz, L., multiplexed proteomic investigation identifies ketamine-induced changes in
Norquist, G., Howland, R.H., Lebowitz, B., McGrath, P.J., Shores-Wilson, K., immune markers in rat serum and expression changes in protein kinases/
Biggs, M.M., Balasubramani, G.K., Fava, M., 2006. Evaluation of outcomes with phosphatases in rat brain. J. Proteome Res. 14 (1), 411e421.
citalopram for depression using measurement-based care in STAR*D: implica- Yang, J.J., Wang, N., Yang, C., Shi, J.Y., Yu, H.Y., Hashimoto, K., 2015. Serum
tions for clinical practice. Am. J. Psychiatry 163 (1), 28e40. interleukin-6 is a predictive biomarker for ketamine's antidepressant effect in
Van Bokhoven, P., Oomen, C.A., Hoogendijk, W.J., Smit, A.B., Lucassen, P.J., Spijker, S., treatment-resistant patients with major depression. Biol. Psychiatry 77 (3),
2011. Reduction in hippocampal neurogenesis after social defeat is long-lasting e19e20.
and responsive to late antidepressant treatment. Eur. J. Neurosci. 33 (10), Zarate Jr., C.A., Singh, J.B., Carlson, P.J., Brutsche, N.E., Ameli, R., Luckenbaugh, D.A.,
1833e1840. Charney, D.S., Manji, H.K., 2006. A randomized trial of an N-methyl-D-aspartate
Walker, A.K., Budac, D.P., Bisulco, S., Lee, A.W., Smith, R.A., Beenders, B., Kelley, K.W., antagonist in treatment-resistant major depression. Arch. Gen. Psychiatry 63
Dantzer, R., 2013. NMDA receptor blockade by ketamine abrogates (8), 856e864.
lipopolysaccharide-induced depressive-like behavior in C57BL/6J mice. Neuro- Zhou, W., Wang, N., Yang, C., Li, X.M., Zhou, Z.Q., Yang, J.J., 2014. Ketamine-induced
psychopharmacology 38 (9), 1609e1616. antidepressant effects are associated with AMPA receptors-mediated upregu-
Ward, J.L., Harting, M.T., Cox Jr., C.S., Mercer, D.W., 2011. Effects of ketamine on lation of mTOR and BDNF in rat hippocampus and prefrontal cortex. Eur. Psy-
endotoxin and traumatic brain injury induced cytokine production in the rat. chiatry 29 (7), 419e423.

You might also like