ArticlePDF Available

The Impact of Ketamine and AV-101 on the Kynurenine Pathway in Subjects With Treatment-Resistant Unipolar or Bipolar Depression

Authors:
Molecular Psychiatry
https://doi.org/10.1038/s41380-019-0589-8
ARTICLE
The kynurenine pathway and bipolar disorder: intersection of the
monoaminergic and glutamatergic systems and immune response
Bashkim Kadriu1Cristan A. Farmer1Peixiong Yuan1Lawrence T. Park 1Zhi-De Deng1Ruin Moaddel2
Ioline D. Henter1Bridget Shovestul1Elizabeth D. Ballard1Cristoph Kraus1Philip W. Gold3
Rodrigo Machado-Vieira1,4 Carlos A. Zarate Jr.1
Received: 6 June 2019 / Revised: 21 October 2019 / Accepted: 30 October 2019
This is a U.S. government work and not under copyright protection in the U.S.; foreign copyright protection may apply 2019
Abstract
Dysfunction in a wide array of systemsincluding the immune, monoaminergic, and glutamatergic systemsis implicated in
the pathophysiology of depression. One potential intersection point for these three systems is the kynurenine (KYN) pathway.
This study explored the impact of the prototypic glutamatergic modulator ketamine on the endogenous KYN pathway in
individuals with bipolar depression (BD), as well as the relationship between response to ketamine and depression-related
behavioral and peripheral inammatory markers. Thirty-nine participants with treatment-resistant BD (23 F, ages 1865)
received a single ketamine infusion (0.5 mg/kg) over 40min. KYN pathway analytesincluding plasma concentrations of
indoleamine 2,3-dioxygenase (IDO), KYN, kynurenic acid (KynA), and quinolinic acid (QA)were assessed at baseline (pre-
infusion), 230 min, day 1, and day 3 post-ketamine. General linear models with restricted maximum likelihood estimation and
robust sandwich variance estimators were implemented. A repeated effect of time was used to model the covariance of the
residuals with an unstructured matrix. After controlling for age, sex, and body mass index (BMI), post-ketamine IDO levels were
signicantly lower than baseline at all three time points. Conversely, ketamine treatment signicantly increased KYN and KynA
levels at days 1 and 3 versus baseline. No change in QA levels was observed post-ketamine. A lower post-ketamine ratio of QA/
KYN was observed at day 1. In addition, baseline levels of proinammatory cytokines and behavioral measures predicted KYN
pathway changes post ketamine. The results suggest that, in addition to having rapid and sustained antidepressant effects in BD
participants, ketamine also impacts key components of the KYN pathway.
Introduction
The etiology of bipolar depression (BD) remains unknown,
in part due to both the heterogeneous nature of the disease
and its complex underlying neuropathology [1]. In addition,
the lack of reliable biomarkers substantially complicates
treatment of the disorder, worsens prognosis, and increases
treatment refractoriness. Recent studies have implicated
both altered glutamatergic neurotransmission and excessive
immune activation in the neurobiology of BD as well as
treatment resistance [2,3]. In particular, evidence from
preclinical and clinical studies suggests that the abnormal
activation of the kynurenine (KYN) pathway may underlie
BD [3,4]. Neuroactive byproducts of the KYN pathway are
involved in the interface between inammatory/immune
response [5,6] and serotoninergic neurotransmission via
catabolism of tryptophan to KYN [7], ultimately altering
downstream synaptic glutamatergic neurotransmission
[8]. The KYN pathway also facilitates inter-organ
*Bashkim Kadriu
bashkim.kadriu@nih.gov
1Section on the Neurobiology and Treatment of Mood Disorders,
National Institute of Mental Health, National Institutes of Health,
Bethesda, MD, USA
2National Institute on Aging, National Institutes of Health,
Baltimore, Maryland, USA
3Clinical Neuroendocrinology Branch, National Institute of Mental
Health, National Institutes of Health, Bethesda, MD, USA
4Department of Psychiatry and Behavioral Sciences, McGovern
Medical School, University of Texas Science Center,
Houston, TX, USA
Supplementary information The online version of this article (https://
doi.org/10.1038/s41380-019-0589-8) contains supplementary
material, which is available to authorized users.
1234567890();,:
1234567890();,:
Content courtesy of Springer Nature, terms of use apply. Rights reserved
communication between the brain and the immune system
by affecting neural afferents and circulating immune med-
iators that activate brain endothelial and innate immune
cells (microglia) [9,10]. This communication is primarily
achieved via two rate-limiting enzymes: tryptophan 2,3-
dioxygenase (TDO) and indoleamine 2,3-dioxygenase
(IDO) [11,12] (see Fig. 1), both of which are implicated in
the catabolism of tryptophan into KYN. Of the two
enzymes, TDO is mostly intra-hepatic and regulated by
glucocorticoid induction [13], whereas IDO is extra-hepatic,
highly expressed in the brain, and tightly upregulated in
response to proinammatory mediators, glucocorticoids,
and psychosocial stress [14].
In the brain, KYN can be differentially processed by
either astrocytes or microglia to produce distinct neuroac-
tive compounds. During homeostasis, KYN aminotransfer-
ase converts KYN to kynurenic acid (KynA), a metabolite
of the astrocytic process [15], which clears accumulated
KYN in the central nervous system (CNS). KynA binds at
the glycine co-agonist site of the NR2B N-methyl-D-
aspartate receptor (NMDAR) and also has antagonist
properties at the α7-nicotinic acetylcholine receptor; both of
these receptors are neuroprotective when activated
physiologically [16,17] and are closely linked to synaptic
plasticity and cognitive processes [18], partly via their anti-
inammatory properties [19] and ability to clear glutamate
spillover in the brain [2,17].
Conversely, the microglial processing of KYN results in
the formation of quinolinic acid (QA), a byproduct that
exerts powerful excitotoxic effects and promotes neuronal
apoptosis [3,19,20] (Fig. 1). Evidence suggests that during
depressive episodes, proinammatory components of the
KYN system in the brain are either directly or indirectly
activated, altering the neuroprotective/neurotoxic balance of
the KYN pathway and leading to the overproduction of
neurotoxic microglial byproducts (such as QA) that
potentiate NMDA activation [3,5,2123]. This increased
activation of inammatory circuitry within the brain, in
turn, contributes to the pathological activation of the glu-
tamatergic system [5,24], which leads to decreased neu-
rotrophic support, synaptic dysregulation, oxidative stress,
excitotoxicity, and loss of glial tissue in multiple sites in the
CNS [17,25].
The KYN pathway is best known for regulating the
interaction between the immune and stress pathways (see
Fig. 1)[26]. Psychosocial stress stimulates inammatory
Fig. 1 The impact of depression on the kynurenine (KYN) pathway in
brain and periphery. The gure depicts KYN metabolites and their
related effects on neuronal cells (inside boxes) and the enzymes that
metabolize them (arrows). The impact of inammation or stress-related
conditions on key rate-limiting enzymes such as indoleamine 2,3-
dioxygenase (IDO) shifts KYN metabolism towards microglial
byproducts such as 3-hydroxykynurenine (3-HK) and quinolinic acid
(QA). This metabolic change is associated with elevated oxidative
stress (3-HK and QA) and glutamate excitotoxicity that could
contribute to depressive symptoms (right panel). Conversely, during
homeostasis, substantial amounts of KYN are converted to kynurenic
acid (KynA), a process mediated by kynurenine aminotransferase II
(KAT II) in astrocytes (left panel). At physiologic levels, KynA is an
N-methyl-D-aspartate receptor (NMDAR) antagonist and contributes
to the clearance of glutamate spillover in the brain. TNF-αtumor
necrosis factor alpha, IFN-gamma interferon gamma, IL-6 interleukin-
6, α7nAChR alpha-7-nicotinic acetylcholine receptor
B. Kadriu et al.
Content courtesy of Springer Nature, terms of use apply. Rights reserved
mediators in the periphery and brain via multiple mechan-
isms; for example, peripheral immune cells, including T-
cells, can gain access to the brain [27], resulting in increased
IDO production. This, in turn, triggers the activation of
resident microglial cells that secrete the proinammatory
cytokine tumor necrosis factor alpha (TNF-α), with neuro-
nal injury as the net result (see Fig. 1). Postmortem human
data support this result, with studies showing that proin-
ammatory cytokines (such as interleukin-6 (IL-6) and
TNF-α) and other byproducts of microglial activation
(mainly QA) were signicantly upregulated in the frontal
cortex of individuals with BD [28]. Positive correlations
have also been found between QA and specic proin-
ammatory immunologic variables, such as IL-6, in the
CSF of individuals with previous suicide attempts [29]. In
contrast, KynA levels in the CSF were found to be inversely
correlated with the severity of depressive symptoms [30], as
well as signicantly downregulated in suicidal individuals
[29]. Another study found that the QA/KynA ratio was
signicantly elevated in the CSF of suicide attempters
compared with healthy participants [31], suggesting that net
positive QA levels in the brain might result in detrimental
structural and functional decits, likely due to enhanced
activation of microglial cells and a corresponding upsurge
in NMDAR agonism [32,33].
Previous studies found that a single infusion of
subanesthetic-dose ketamine leads to rapid (within hours),
robust, and sustained antidepressant effects in individuals
with BD [34,35]. Preclinical evidence also suggests that,
within this time frame, ketamine reverses decits in neu-
roplasticity and neurogenesis [36,37]. Ketamine has also
been found to reduce levels of key proinammatory med-
iators [38] as well as bone biomarkers and adipokines
[39,40] within hours to days following acute treatment. In
addition, ketamine abolished lipopolysaccharide (LPS)-
induced depressive-like behaviors in preclinical mouse
models [41]. Previous work from our laboratory also found
that ketamines antidepressant effects may depend on its
ability to affect KYN pathway metabolites [42].
This study explores the complex interactions between
ketamine and relevant behavioral and biological immune/
inammatory markers that affect the KYN pathway in
depression. The study had four main goals: (1) to explore
the impact of a single dose of ketamine on specic proin-
ammatory KYN pathway metabolites in participants with
treatment-resistant BD; (2) to assess whether baseline
values of specic KYN pathway analytes predicted change
in depressive symptom ratings post-ketamine; (3) to
investigate whether baseline levels of inammatory markers
predicted change in KYN pathway analytes post-ketamine
infusion; and (4) to determine whether inammatory mar-
kers were longitudinally associated with KYN pathway
analytes post-ketamine infusion.
Materials and methods
Study design and participants
The current data were drawn from the ketamine condition of
a randomized, placebo (saline)-controlled, crossover study
designed to assess the antidepressant efcacy of adjunctive
ketamine administered intravenously at 0.5 mg/kg over 40
min in participants with BD; all participants were receiving
adjunctive treatment with a mood stabilizer (either lithium
or valproic acid). Details regarding study design have been
previously published [34,35]. Clinical and demographic
data are presented in Table 1, and additional information
about the participant sample can be found in the Supple-
mentary Materials.
Psychiatric rating scales included the Beck Depression
Inventory [43], the Hamilton Depression Rating Scale [44],
the MontgomeryAsberg Depression Rating Scale [45], and
the SnaithHamilton Pleasure Scale [46]. For the purposes
of this analysis, empirically derived unidimensional scores
comprising items from all four scales were used [47]. The
three largest subscalesDepressed Mood,Negative Cog-
nition, and Anhedoniawere selected for use, though
results for all subscales are available upon request.
Table 1 Participant demographics
N(%) Mean ± SD
Age 39 (100) 45.92 ± 10.52
Sex
Male 16 (41)
Female 23 (59)
Race
Caucasian 32 (82)
African-American 4 (10)
Other 2 (5)
Not reported 1 (3)
BMI 29.84 ± 5.83
Age of illness onset 17.51 ± 6.88
MADRS Total 33.00 ± 4.39
Substance use disorder 18 (46%)
Alcohol dependence 14 (36%)
Generalized anxiety disorder 6 (15%)
Mood stabilizer
Lithium 26 (67)
Valproic acid 13 (33)
BMI data were missing for one participant; age of onset was missing
for two participants; substance/alcohol abuse data were missing for
one participant; and comorbid psychiatric diagnosis data were missing
for ve participants
BMI body mass index, MADRS MontgomeryAsberg Depression
Rating Scale
The kynurenine pathway and bipolar disorder: intersection of the monoaminergic and glutamatergic. . .
Content courtesy of Springer Nature, terms of use apply. Rights reserved
ELISA and multiplex immunoassay
Plasma samples were collected 60 min prior to ketamine
infusion (baseline) and at 230 min, day 1, and day 3 post-
ketamine infusion. Levels of human KYN, KynA, QA, and
IDO were measured using specic ELISA kits. Circulating
levels of several inammatory cytokines (TNF-α, soluble
tumor necrosis factor receptor 1 (sTNFR1), interferon
gamma (IFN-γ), IL-2, IL-5, IL-6, IL-8, and IL-10) were
measured in plasma using a high-sensitivity multiplex
Luminex immunoassay. For additional details, see the Sup-
plementary Materials.
Statistical analysis
General linear models with restricted maximum likelihood
estimation and robust sandwich variance estimators were
implemented using PROC MIXED in SAS/STAT Version
9.3. The syntax for all models is provided in the Supple-
mentary Materials. A repeated effect of time was used to
model the covariance of the residuals with an unstructured
matrix. Mood stabilizer (lithium or valproic acid), age, and
body mass index (BMI) were entered as covariates in all
analyses. The rst research questionwhich sought to
assess changes to the KYN pathway in response to ketamine
was investigated via prespecied contrasts between
baseline and follow-up points (230 min, day 1, day 3). The
second research questionwhether baseline KYN pathway
values predicted change in depressive symptom ratings post-
ketaminewas assessed by estimating the simple slopes for
KYN pathway member-by-time interactions. The third
research questionwhether baseline levels of inammatory
markers predicted KYN pathway changes post-ketamine
was similarly assessed by estimating the simple slope for
each baseline-to-follow-up contrast (i.e., simple slopes for
the inammatory marker-by-time interactions in predicting
KYN pathway changes post-ketamine). Finally, the fourth
research questionwhether inammatory markers were
longitudinally associated with KYN pathway analytes post-
ketamine infusionwas evaluated by determining whether
the inammatory markers changed over time (a mixed
model predicting inammatory marker from time and cov-
ariates); if so, whether that change correlated with changes
in the KYN pathway was then assessed (Spearman corre-
lations). Prior to the analyses, plasma data (for the KYN
pathway and for inammatory cytokines) were natural-log
transformed after adding 1 to all values. Ratios (e.g., KYN/
KynA) are the ratios of the transformed variables.
It should be noted that with regard to the third research
question, high intercorrelations were observed among the
pro- and anti-inammatory circulating markers (i.e., TNF-α,
sTNFR1, IFN-γ, IL-2, IL-5, IL-6, IL-8, and IL-10), and a
principal components analysis was therefore implemented.
Two components with eigenvalues greater than 1.0
explained 55% of the variance in the inammatory markers
(see Supplementary Table S1); the rst component included
mainly proinammatory markers and represented TNF-α,
sTNFR1, IFN-γ, IL-5, IL-6, and IL-8. The second compo-
nent, which included mainly anti-inammatory markers,
represented IL-2, IL-8 (reverse loading), and IL-10. These
pro- and anti-inammatory component scores were used in
the analysis rather than the values of the individual circu-
lating inammatory cytokines.
In addition, because this was a secondary analysis of
participants from a clinical trial, an aprioripower analysis
was not performed. Given the exploratory nature of this
study, we also did not adjust alpha or p-values for multi-
plicity. All parameter estimates with standard errors, as well
as exact p-values, are shown in Supplementary Tables S1S8.
Results
Changes in KYN pathway metabolites post-
ketamine infusion
After controlling for type of mood stabilizer, age, and BMI,
post-ketamine levels of IDO were signicantly lower than
baseline at all time points (Table 2, Fig. 2). Both KYN and
KynA levels were signicantly higher at days 1 and 3, as
was their ratio (KYN/KynA). No changes in QA levels were
observed post-ketamine administration, though the ratio of
QA to KYN was signicantly lower from baseline to day 1.
No post-ketamine difference was observed for KynA/QA
ratio, a measure of NMDA agonist/antagonist balance.
Model results appear in Table 2and Supplementary
Table S2.
Members of the KYN pathway as predictors of
change in depressive symptoms post-ketamine
infusion
Baseline concentrations of IDO, KYN, QA, and the KynA/
QA ratio were evaluated as predictors of Depressed Mood,
Negative Cognition, and Anhedonia subscale scores. Higher
baseline IDO levels (t(33) =3.84, p=0.0005) and lower
baseline QA levels (t(33) =2.35, p=0.02) were associated
with less severe baseline Depressed Mood scores, and lower
QA/KYN ratio was associated with less severe baseline
Negative Cognition scores (t(33) =2.58, p=0.01) (see
Supplementary Table S3 for slopes).
Baseline IDO levels were unrelated to antidepressant
response to ketamine on any subscale, and lower baseline
KYN levels were nominally related to improved Anhedonia
scores at day 3 (t(33) =2.09, p=0.04) (Fig. 3b, Supple-
mentary Table S4). Higher baseline QA levels predicted
B. Kadriu et al.
Content courtesy of Springer Nature, terms of use apply. Rights reserved
improvement in Depressed Mood scores at 230 min (t(33)
=2.34, p=0.03) and at day 3 (t(33) =3.21, p=0.003)
and improvement in Negative Cognition scores at day 3 (t
(33) =2.79, p=0.009) (Supplementary Table S4). A
lower baseline ratio of KynA/QA (a measure of NMDA
antagonist/agonist balance) predicted improvement in
Depressed Mood score at 230 min (t(33) =2.53, p=0.02)
and in Anhedonia score at day 3 (t(33) =2.29, p=0.02)
(Supplementary Table S4).
Baseline inammatory markers as predictors of
change in the KYN pathway post-ketamine infusion
Ten participants who were missing inammatory marker
concentrations were excluded from this analysis. The base-
line values of the two inammatory cytokine principal
component scores (the proinammatory cytokine component
score, representing TNF-α, sTNFR1, IFN-γ, IL-5, IL-6, and
IL-8, and the anti-inammatory cytokine component score,
representing IL-2, reverse loading IL-8, and IL-10) were
entered as predictors of IDO, KYN, and KynA levels, as
well as KYN/KynA and QA/KYN ratios. Baseline proin-
ammatory component score was not related to baseline
KYN pathway values (Supplementary Table S5). Nominal
trends were observed for higher baseline proinammatory
cytokine component levels to predict larger increases in
KYN levels and in the KYN/KynA ratio. These were
strongest at the 230-min time point (t(24) =2.46, p=0.02
and t(24) =2.42, p=0.02, respectively) (Fig. 3a,
Supplementary Table S6), when neither KYN levels nor the
KYN/KynA ratio had yet changed signicantly from
baseline.
In addition, higher baseline anti-inammatory cytokine
component scores were associated with higher baseline
KYN levels (t(24) =2.94, p=0.007) and higher baseline
KYN/KynA ratio (t(24) =3.47, p=0.002), but not with
KynA levels or the QA/KynA ratio (Supplementary
Table S5). Baseline anti-inammatory component values
predicted change in IDO levels at 230 min post-ketamine,
such that higher anti-inammatory levels were associated
with less of a decrease in IDO levels (t(24) =2.74, p=
0.01) (Fig. 3a, Supplementary Table S6). Higher baseline
anti-inammatory levels were also associated with less of
an increase in KYN levels (t(24) =2.85, p=0.009) and
in the KYN/KynA ratio (t(24) =2.40, p=0.02) at day 1
(Fig. 3a, Supplementary Table S6).
The proinammatory component did not change over
time (Supplementary Table S7), but the anti-inammatory
component was increased at day 3 (t(24) =2.87, p=0.008).
Change in the anti-inammatory component was moderately
and positively correlated with change in KynA levels at day
3(r=0.39, p=0.45) (see Supplementary Table S8).
Discussion
This study is the rst to explore the impact of ketamine on
KYN pathway metabolites in individuals with treatment-
Table 2 Results of mixed models predicting post-ketamine change from baseline in the KYN pathway
Dependent variable 230 min Day 1 Day 3
IDO (ln) Change from baseline (95% CI) 0.16 (0.27 to 0.06) 0.24 (0.35 to 0.12) 0.2 (0.31 to 0.1)
Statistical test t(34) =3.05, p=0.004 t(34) =4.12, p=0.0002 t(34) =3.91, p=0.0004
KYN (ln) Change from baseline (95% CI) 0.07 (0.110.26) 0.5 (0.230.76) 0.39 (0.080.7)
Statistical test t(34) =0.77, p=0.45 t(34) =3.69, p=0.0008 t(34) =2.44, p=0.02
KynA (ln) Change from baseline (95% CI) 0.03 (0.040.1) 0.12 (0.040.2) 0.15 (0.060.23)
Statistical test t(34) =0.82, p=0.42 t(34) =2.85, p=0.007 t(34) =3.36, p=0.002
QA (ln) Change from baseline (95% CI) 0.01 (0.010.04) 0.01 (0.010.04) 0.03 (0.020.08)
Statistical test t(34) =1.28, p=0.21 t(34) =0.94, p=0.35 t(34) =1.26, p=0.22
KYN/KynA Change from baseline (95% CI) 0.01 (0.030.06) 0.09 (0.030.15) 0.07 (0.010.14)
Statistical test t(34) =0.59, p=0.56 t(34) =2.91, p=0.006 t(34) =1.76, p=0.087
QA/KYN Change from baseline (95% CI) 0 (0.01 to 0.01) 0.02 (0.03 to 0.01) 0.01 (0.02 to 0.01)
Statistical test t(34) =0.24, p=0.81 t(34) =3.31, p=0.002 t(34) =0.91, p=0.37
KynA/QA Change from baseline (95% CI) 0.09 (0.320.13) 0.03 (0.180.25) 0.12 (0.420.19)
Statistical test t(34) =0.82, p=0.42 t(34) =0.29, p=0.78 t(34) =0.75, p=0.46
Results of a mixed model with empirical sandwich variance estimator and repeated effect of time. Type 3 tests are shown in Supplementary
Table S2. Table shows specic contrast between estimated values at baseline and follow-up (illustrated in Fig. 2). Age, body mass index (BMI),
and type of mood stabilizer were included as covariates in all analyses
Bold values indicate statistical signicance p< 0.05
IDO indoleamine 2,3-dioxygenase, KYN kynurenine, KynA kynurenic acid, QA quinolinic acid, ln natural logarithm
The kynurenine pathway and bipolar disorder: intersection of the monoaminergic and glutamatergic. . .
Content courtesy of Springer Nature, terms of use apply. Rights reserved
resistant BD. A number of salient ndings emerged. First,
ketamine signicantly lowered IDO levels at 230 min, day
1, and day 3 post-ketamine infusion. Surprisingly,
participants who had higher baseline levels of anti-
inammatory cytokines experienced less of an immediate
(230 min post-infusion) decrease in IDO levels. Second, and
Fig. 2 Results of mixed models: post-ketamine change in the kynur-
enine (KYN) pathway. Least square mean estimated scores (with
standard error) are plotted by time point. Signicance refers to change
from baseline (60 min). IDO indoleamine 2,3-dioxygenase, KYN
kynurenine, KynA kynurenic acid, QA quinolinic acid. Results of the
full analysis can be found in Supplementary Table S2
B. Kadriu et al.
Content courtesy of Springer Nature, terms of use apply. Rights reserved
in contrast, plasma levels of KYN and KynA were sig-
nicantly increased on days 1 and 3 relative to baseline.
Third, the relatively neuroprotective index ratio KYN/
KynA was upregulated in response to ketamine at day 1.
Interestingly, increased baseline levels of anti-inammatory
cytokines were associated with less change at day 1 for
KYN and the KYN/KynA ratio, but not for KynA itself.
Finally, a signicantly lower QA/KYN index ratio was
observed, but only at day 1 post-ketamine infusion, which
coincides with ketamines peak antidepressant effects [34];
given that this ratio is broadly accepted as an index of high
inammation and excitotoxicity, any decrease reects a
desirable outcome.
IDO is an intracellular enzyme mainly produced by
immune cells and the brain and appears to be critical to
some forms of immunologically mediated depression
[11,21,23,29,48]. Preclinical studies also found that IDO
is induced in both brain and periphery after a systemic
inammatory challenge [14,48,49]. During depressive
episodes, stress and immune activation enhance the con-
version of tryptophan to KYN, mostly via IDO induction. In
addition, numerous studies have shown that proin-
ammatory cytokines such as IFN-γ, IFN-α, IL-6, and TNF-
αrobustly induce IDO production via immune cells [50
52], which subsequently shifts tryptophan metabolism away
from the liver [53]. Furthermore, IDO levels are also
affected by oxidative stress and LPS injection [5]. During
LPS-induced brain inammation, most KYN and QA pro-
duction is mediated via IDO induction [54]. In humans,
vulnerability to cytokine-induced depression was found to
be enhanced by a polymorphism in the IDO gene, providing
a possible explanation for the clinical heterogeneity sur-
rounding the occurrence of depressive symptoms following
immune system activation [55]. In this context, genetic
manipulation or pharmacological inhibition of IDO was
found to abolish the LPS-induced depressive phenotype in
mouse models independently of cytokine induction, sug-
gesting that IDO itself is sufcient to trigger depressive-like
behaviors in mice [56]. Studies assessing indirect IDO
activity (measured via the KYN/tryptophan ratio) found that
increased plasma IDO levels mediated the link between
inammation and depressive symptomatology [57]. The
present data support the evidence that IDO is critical for
shunting KYN towards the microglial pathway, but also
suggest that ketamines robust ability to signicantly lower
IDO levels at 230 min, day 1, and day 3 may protect against
this eventuality. Interestingly, we found that higher baseline
IDO levels were associated with less severe depressive
symptoms at baseline but were unrelated to degree of
antidepressant response to ketamine.
In addition, our nding that KYN and KynA levels were
signicantly upregulated in individuals with BD compared
Fig. 3 Baseline inammatory cytokine component scores as mod-
erators of change in the kynurenine (KYN) pathway (a), and baseline
KYN levels as moderators of change in depressive symptom ratings
(b). (a) Results of a mixed model with the component scores of
proinammatory (top) or anti-inammatory (bottom) cytokines entered
as moderators of change in KYN pathway analytes (Y-axis) at three
time points post-ketamine infusion (X-axis). T-values (all df =24) for
the simple slope of the moderator for change at each time point are
plotted; positive values indicate that higher cytokine component scores
were associated with increases in respective KYN pathway analytes,
and negative values indicate that higher cytokine component scores
were associated with decreases in respective KYN pathway analytes.
(b) Results of a mixed model with baseline KYN pathway member
entered as a moderator of change in depressive symptom ratings
(Y-axis) post-ketamine infusion. T-values (all df =33) for the simple
slope of the moderator for change at each time point are plotted;
positive values indicate that higher baseline KYN pathway con-
centrations were associated with less improvement in depressive
symptoms, and negative values indicate that higher baseline KYN
pathway concentrations were associated with more improvement in
depressive symptoms. In both models, age, body mass index (BMI),
and type of mood stabilizer were included as covariates. Results of the
full analysis can be found in Supplementary Table S5 and Supple-
mentary Table S6. IDO indoleamine 2,3-dioxygenase, KYN kynur-
enine, KynA kynurenic acid, QA quinolinic acid
The kynurenine pathway and bipolar disorder: intersection of the monoaminergic and glutamatergic. . .
Content courtesy of Springer Nature, terms of use apply. Rights reserved
with baseline in response to a single ketamine infusion at
both days 1 and 3 (Fig. 2) broadly supports a number of
recent results linking the KYN pathway and depression. For
instance, a large, recent meta-analysis demonstrated that,
compared with healthy volunteers, individuals with
depression had lower overall plasma levels of KYN and
KynA and higher levels of QA [58]. Other studies found
lower levels of KYN metabolites in depressed individuals
[22] as well as in adolescents with melancholic depression
[59]. The severity of depressive symptoms in suicidal
individuals has also been linked to increased plasma levels
of QA and decreased plasma levels of KynA [20,31].
Another study found lower KYN levels and a higher QA/
KYN ratio in participants with major depressive disorder
(MDD) compared with healthy controls; notably, in that
study, higher baseline KYN levels predicted remission in
response to adjunctive treatment with celecoxib, suggesting
that these metabolites were associated with better anti-
inammatory outcomes [60].
In this context, the present study found that higher baseline
levels of KynA were associated with greater improvement in
Depressed Mood score at 230 min and day 3 and with
Negative Cognition score at day 3. In contrast, lower baseline
KYN levels were linked to improved Anhedonia score at day
3. Interestingly, higher baseline QA levels were associated
with greater improvement in Depressed Mood score post-
ketamine infusion at 230 min and day 3, and with greater
improvement in Negative Cognition score at day 3.
With regard to QA in particular, selective serotonin
reuptake inhibitor treatment was found to reduce QA levels
in the rodent brain [61]; in contrast, this study found that
ketamine had no direct impact on QA levels in humans,
thoughas noted aboveit did signicantly affect other
KYN byproduct metabolites, including IDO, KYN, and
KynA. Interestingly, previous postmortem studies found
elevated QA levels in the brains of severely depressed or
suicidal individuals [33], suggesting an excess of QAs
precursor, KYN, which would thus be available for con-
version to QA. There are two potential reasons why, under
pathological conditions, the QA pathway appears to pre-
dominate over the neuroprotective KynA pathway [62].
First, while microglia produce QA, astrocytes produce
KynA; studies have shown that, in response to increased
microglial activity, MDD participants typically have sig-
nicant astrocytic dysfunction in critical areas such as the
subgenual prefrontal cortex [63,64]. Second, QA binds
with tenfold greater afnity to NR2B NMDA glutamate
receptor subunits, which are preferentially localized in the
prefrontal cortex, amygdala, hippocampus, and ventral
striatum [65]. These regions thus bear a greater excitotoxic
burden, which often reects activation of the KYN pathway
in the brain. KynA has no such preferential localization,
though it should be noted that KynA inhibition can lead to
inammation and excitotoxicity. Previous studies found that
a higher plasma KynA/QA ratioa putative neuroprotec-
tive markerwas positively correlated with hippocampal
and amygdalar volumes [66].
KYN readily crosses the blood-brain barrier. Overall,
40% of KYN in the CNS is produced in the brain, and 60%
is supplied by TDO, which is present only in the liver [67].
For this reason, it is almost certain that the increased plasma
levels of KYN and KynA observed in the present study after
ketamine administration derive from the liver. Interestingly,
peripheral KYN possesses pronounced anti-inammatory
properties; it is known to block T-cell proliferation, induce
T-cell death, and suppress the activity of natural killer cells
and antigen-presenting cells (e.g., dendritic cells, mono-
cytes, and macrophages) [29,68]. Thus, ketamine-induced
increases in hepatic KYN production would be expected to
counter the substantial peripheral proinammatory state
associated with depression. However, increased hepatic
KYN production would also present a greater KYN load to
the brain. While this might ordinarily increase production of
toxic metabolites, the ketamine-induced suppression of IDO
could protect against this potential eventuality.
Given the chronicity and long-lasting changes underlying
BD, considerable evidence suggests that epigenetic mod-
ications are involved in the pathophysiology of both
depression [69] and glutamatergic dysregulation [70]. Epi-
genetic mechanisms also help regulate KYN 3-
monooxygenase (KMO), a critical enzyme in the KYN
pathway that inuences bioactive KYN pathway metabo-
lites [71]. Recent genetic, epigenetic, and pharmacological
studies are targeting KMO as a way of impacting the
bioactive byproducts of the KYN pathway, including KynA
[72,73]. Interestingly, preclinical studies found that keta-
mines rapid-acting antidepressant actions are in part
mediated epigenetically [74,75]. In this context, it is highly
likely that ketamines effects on the KYN pathway may be
mediated by epigenetic mechanisms.
One key strength of this study is that our participants
were well-characterized and hospitalized for several weeks
before and after ketamine administration. The study was
also associated with several limitations, including the
absence of a control group. In addition, ten participants
were missing inammatory cytokine data. Because CSF
measures were lacking, another potential limitation is that
only peripheral measures of KYN pathway mediators were
assessed. Finally, the lack of ethnic diversity in the cohort
may limit the generalizability of the results.
Despite these limitations, this study is the rst to
demonstrate that ketamine, in addition to exerting rapid and
sustained antidepressant effects, also impacts key compo-
nents of the KYN pathway. These rate-limiting enzymes and
analytes are highly impacted by inammation and stress,
underscoring ketamines acute anti-inammatory effects.
B. Kadriu et al.
Content courtesy of Springer Nature, terms of use apply. Rights reserved
Acknowledgements Funding for this work was supported by the
Intramural Research Program at the National Institute of Mental
Health, National Institutes of Health (IRP-NIMH-NIH) (ZIA-
MH002857; NCT00088699; 04-M-0222); by a NARSAD Independent
Investigator to CAZ; by a Brain & Behavior Mood Disorders Research
Award to CAZ; and by the Intramural Research Program at the
National Institute of Aging (RM). The authors thank the 7SE research
unit and staff for their support.
Compliance with ethical standards
Conict of interest CAZ is listed as a co-inventor on a patent for the
use of ketamine in major depression and suicidal ideation. CAZ and
RM are listed as co-inventors on a patent for the use of (2R,6R)-
hydroxynorketamine, (S)-dehydronorketamine, and other stereo-
isomeric dehydro and hydroxylated metabolites of (R,S)-ketamine
metabolites in the treatment of depression and neuropathic pain; and as
co-inventors on a patent application for the use of (2R,6R)-hydro-
xynorketamine and (2S,6S)-hydroxynorketamine in the treatment of
depression, anxiety, anhedonia, suicidal ideation, and posttraumatic
stress disorders. They have assigned their patent rights to the US
government but will share a percentage of any royalties that may be
received by the government. The remaining authors declare that they
have no conict of interest.
Publishers note Springer Nature remains neutral with regard to
jurisdictional claims in published maps and institutional afliations.
References
1. Manji HK, Quiroz JA, Payne JL, Singh J, Lopes BP, Viegas JS,
et al. The underlying neurobiology of bipolar disorder. World
Psychiatry. 2003;2:13646.
2. Haroon E, Fleischer CC, Felger JC, Chen X, Woolwine BJ, Patel
T, et al. Conceptual convergence: increased inammation is
associated with increased basal ganglia glutamate in patients with
major depression. Mol Psychiatry. 2016;21:13517.
3. Birner A, Platzer M, Bengesser SA, Dalkner N, Fellendorf FT,
Queissner R, et al. Increased breakdown of kynurenine towards its
neurotoxic branch in bipolar disorder. PLoS ONE. 2017;12:
e0172699.
4. Savitz J, Drevets WC. Bipolar and major depressive disorder:
neuroimaging the developmental-degenerative divide. Neurosci
Biobehav Rev. 2009;33:699771.
5. Leonard BE. Inammation and depression: a causal or coin-
cidental link to the pathophysiology? Acta Neuropsychiatr. 2018;
30:116.
6. Strasser B, Becker K, Fuchs D, Gostner JM. Kynurenine pathway
metabolism and immune activation: peripheral measurements in
psychiatric and co-morbid conditions. Neuropharmacology.
2017;112:28696.
7. Marazziti D, Baroni S, Picchetti M, Piccinni A, Silvestri S, Del-
lOsso L. [New developments on the serotonin hypothesis of
depression: shunt of tryptophan]. Riv Psichiatr. 2013;48:2334.
8. Miller AH. Conceptual conuence: the kynurenine pathway as a
common target for ketamine and the convergence of the inam-
mation and glutamate hypotheses of depression. Neuropsycho-
pharmacology. 2013;38:16078.
9. Schwarcz R, Stone TW. The kynurenine pathway and the brain:
challenges, controversies and promises. Neuropharmacology.
2017;112:23747.
10. Dantzer R, OConnor JC, Freund GG, Johnson RW, Kelley KW.
From inammation to sickness and depression: when the immune
system subjugates the brain. Nat Rev Neurosci. 2008;9:4656.
11. Andre C, OConnor JC, Kelley KW, Lestage J, Dantzer R, Cas-
tanon N. Spatio-temporal differences in the prole of murine brain
expression of proinammatory cytokines and indoleamine 2,3-
dioxygenase in response to peripheral lipopolysaccharide admin-
istration. J Neuroimmunol. 2008;200:909.
12. Reus GZ, Jansen K, Titus S, Carvalho AF, Gabbay V, Quevedo J.
Kynurenine pathway dysfunction in the pathophysiology and
treatment of depression: evidences from animal and human stu-
dies. J Psychiatry Res. 2015;68:31628.
13. Gibney SM, Fagan EM, Waldron AM, OByrne J, Connor TJ,
Harkin A. Inhibition of stress-induced hepatic tryptophan 2,3-
dioxygenase exhibits antidepressant activity in an animal model of
depressive behaviour. Int J Neuropsychopharmacol. 2014;17:
91728.
14. Heisler JM, OConnor JC. Indoleamine 2,3-dioxygenase-depen-
dent neurotoxic kynurenine metabolism mediates inammation-
induced decit in recognition memory. Brain Behav Immun.
2015;50:11524.
15. Guillemin GJ, Kerr SJ, Smythe GA, Smith DG, Kapoor V,
Armati PJ, et al. Kynurenine pathway metabolism in human
astrocytes: a paradox for neuronal protection. J Neurochem.
2001;78:84253.
16. Stone TW. Neuropharmacology of quinolinic and kynurenic
acids. Pharm Rev. 1993;45:30979.
17. Vecsei L, Szalardy L, Fulop F, Toldi J. Kynurenines in the CNS:
recent advances and new questions. Nat Rev Drug Discov.
2013;12:6482.
18. Potter MC, Elmer GI, Bergeron R, Albuquerque EX, Guidetti P,
Wu HQ, et al. Reduction of endogenous kynurenic acid formation
enhances extracellular glutamate, hippocampal plasticity, and
cognitive behavior. Neuropsychopharmacology. 2010;35:173442.
19. Ganong AH, Cotman CW. Kynurenic acid and quinolinic acid act
at N-methyl-D-aspartate receptors in the rat hippocampus. J
Pharm Exp Ther. 1986;236:2939.
20. Heyes MP, Saito K, Crowley JS, Davis LE, Demitrack MA, Der
M, et al. Quinolinic acid and kynurenine pathway metabolism in
inammatory and non-inammatory neurological disease. Brain.
1992;115:124973.
21. Dantzer R, OConnor JC, Lawson MA, Kelley KW.
Inammation-associated depression: from serotonin to kynur-
enine. Psychoneuroendocrinology. 2011;36:42636.
22. Myint AM, Kim YK, Verkerk R, Scharpe S, Steinbusch H,
Leonard B. Kynurenine pathway in major depression: evidence of
impaired neuroprotection. J Affect Disord. 2007;98:14351.
23. Anderson G, Maes M. Bipolar disorder: role of immune-
inammatory cytokines, oxidative and nitrosative stress and
tryptophan catabolites. Curr Psychiatry Rep. 2015;17:8.
24. Parrott JM, OConnor JC. Kynurenine 3-monooxygenase: an inu-
ential mediator of neuropathology. Front Psychiatry. 2015;6:116.
25. Harrison NA, Brydon L, Walker C, Gray MA, Steptoe A,
Critchley HD. Inammation causes mood changes through
alterations in subgenual cingulate activity and mesolimbic con-
nectivity. Biol Psychiatry. 2009;66:40714.
26. Savitz J. The kynurenine pathway: a nger in every pie. Mol
Psychiatry. 2019. https://doi.org/10.1038/s41380-019-0414-4.
[Epub ahead of print].
27. Prinz M, Priller J. The role of peripheral immune cells in the CNS
in steady state and disease. Nat Neurosci. 2017;20:13644.
28. Rao JS, Harry GJ, Rapoport SI, Kim HW. Increased excitotoxicity
and neuroinammatory markers in postmortem frontal cortex from
bipolar disorder patients. Mol Psychiatry. 2010;15:38492.
29. Bay-Richter C, Linderholm KR, Lim CK, Samuelsson M,
Traskman-Bendz L, Guillemin GJ, et al. A role for inammatory
metabolites as modulators of the glutamate N-methyl-D-aspartate
receptor in depression and suicidality. Brain Behav Immun.
2015;43:1107.
The kynurenine pathway and bipolar disorder: intersection of the monoaminergic and glutamatergic. . .
Content courtesy of Springer Nature, terms of use apply. Rights reserved
30. Bryleva EY, Brundin L. Kynurenine pathway metabolites and
suicidality. Neuropharmacology. 2017;112:32430.
31. Erhardt S, Lim CK, Linderholm KR, Janelidze S, Lindqvist D,
Samuelsson M, et al. Connecting inammation with glutamate
agonism in suicidality. Neuropsychopharmacology. 2013;38:74352.
32. Busse M, Busse S, Myint AM, Gos T, Dobrowolny H, Muller UJ,
et al. Decreased quinolinic acid in the hippocampus of depressive
patients: evidence for local anti-inammatory and neuroprotective
responses? Eur Arch Psychiatry Clin Neurosci. 2015;265:3219.
33. Steiner J, Walter M, Gos T, Guillemin GJ, Bernstein HG, Sarnyai
Z, et al. Severe depression is associated with increased microglial
quinolinic acid in subregions of the anterior cingulate gyrus:
evidence for an immune-modulated glutamatergic neuro-
transmission? J Neuroinammation. 2011;8:94.
34. Zarate CA Jr., Brutsche NE, Ibrahim L, Franco-Chaves J, Dia-
zgranados N, Cravchik A, et al. Replication of ketamines anti-
depressant efcacy in bipolar depression: a randomized controlled
add-on trial. Biol Psychiatry. 2012;71:93946.
35. Diazgranados N, Ibrahim L, Brutsche NE, Newberg A, Kronstein
P, Khalife S, et al. A randomized add-on trial of an N-methyl-D-
aspartate antagonist in treatment-resistant bipolar depression. Arch
Gen Psychiatry. 2010;67:793802.
36. Autry AE, Adachi M, Nosyreva E, Na ES, Los MF, Cheng PF,
et al. NMDA receptor blockade at rest triggers rapid behavioural
antidepressant responses. Nature. 2011;475:915.
37. Duman RS, Aghajanian GK, Sanacora G, Krystal JH. Synaptic
plasticity and depression: new insights from stress and rapid-
acting antidepressants. Nat Med. 2016;22:23849.
38. Kiraly DD, Horn SR, Van Dam NT, Costi S, Schwartz J, Kim-
Schulze S, et al. Altered peripheral immune proles in treatment-
resistant depression: response to ketamine and prediction of
treatment outcome. Transl Psychiatry. 2017;7:e1065.
39. Kadriu B, Gold PW, Luckenbaugh DA, Lener MS, Ballard ED,
Niciu MJ, et al. Acute ketamine administration corrects abnormal
inammatory bone markers in major depressive disorder. Mol
Psychiatry. 2018;23:162631.
40. Machado-Vieira R, Gold PW, Luckenbaugh DA, Ballard ED,
Richards EM, Henter ID, et al. The role of adipokines in the rapid
antidepressant effects of ketamine. Mol Psychiatry. 2017;22:12733.
41. Walker AK, Budac DP, Bisulco S, Lee AW, Smith RA, Beenders
B, et al. NMDA receptor blockade by ketamine abrogates
lipopolysaccharide-induced depressive-like behavior in C57BL/6J
mice. Neuropsychopharmacology. 2013;38:160916.
42. Moaddel R, Shardell M, Khadeer M, Lovett J, Kadriu B, Ravi-
chandran S, et al. Plasma metabolomic proling of a ketamine and
placebo crossover trial of major depressive disorder and healthy
control subjects. Psychopharmacology. 2018;235:301730.
43. Beck AT, Ward CH, Mendelson M, Mock J, Erbaugh J. An
inventory for measuring depression. Arch Gen Psychiatry. 1961;4:
56171.
44. Hamilton M. A rating scale for depression. J Neurol Neurosurg
Psychiatry. 1960;23:5662.
45. Montgomery SA, Asberg M. A new depression scale designed to
be sensitive to change. Br J Psychiatry. 1979;134:3829.
46. Snaith RP, Hamilton M, Morley S, Humayan A, Hargreaves D,
Trigwell P. A scale for the assessment of hedonic tone the Snaith-
Hamilton pleasure scale. Br J Psychiatry. 1995;167:99103.
47. Ballard ED, Yarrington JS, Farmer CA, Lener MS, Kadriu B,
Lally N, et al. Parsing the heterogeneity of depression: an
exploratory factor analysis across commonly used depression
rating scales. J Affect Disord. 2018;231:517.
48. Dobos N, de Vries EF, Kema IP, Patas K, Prins M, Nijholt IM,
et al. The role of indoleamine 2,3-dioxygenase in a mouse model
of neuroinammation-induced depression. J Alzheimers Dis.
2012;28:90515.
49. OConnor JC, Lawson MA, Andre C, Briley EM, Szegedi SS,
Lestage J, et al. Induction of IDO by bacille Calmette-Guerin is
responsible for development of murine depressive-like behavior. J
Immunol. 2009;182:320212.
50. Fatokun AA, Hunt NH, Ball HJ. Indoleamine 2,3-dioxygenase 2
(IDO2) and the kynurenine pathway: characteristics and potential
roles in health and disease. Amino Acids. 2013;45:131929.
51. Hughes MM, Connor TJ, Harkin A. Stress-related immune mar-
kers in depression: implications for treatment. Int J Neu-
ropsychopharmacol. 2016;19:pyw001.
52. Wigner P, Czarny P, Galecki P, Su KP, Sliwinski T. The mole-
cular aspects of oxidative & nitrosative stress and the tryptophan
catabolites pathway (TRYCATs) as potential causes of depres-
sion. Psychiatry Res. 2018;262:56674.
53. Moffett JR, Blinder KL, Venkateshan CN, Namboodiri MA.
Differential effects of kynurenine and tryptophan treatment on
quinolinate immunoreactivity in rat lymphoid and non-lymphoid
organs. Cell Tissue Res. 1998;293:52534.
54. Kita T, Morrison PF, Heyes MP, Markey SP. Effects of systemic
and central nervous system localized inammation on the con-
tributions of metabolic precursors to the L-kynurenine and qui-
nolinic acid pools in brain. J Neurochem. 2002;82:25868.
55. Smith AK, Simon JS, Gustafson EL, Noviello S, Cubells JF,
Epstein MP, et al. Association of a polymorphism in the indo-
leamine- 2,3-dioxygenase gene and interferon-alpha-induced
depression in patients with chronic hepatitis C. Mol Psychiatry.
2012;17:7819.
56. Lawson MA, Parrott JM, McCusker RH, Dantzer R, Kelley
KW, OConnor JC. Intracerebroventricular administration of
lipopolysaccharide induces indoleamine-2,3-dioxygenase-depen-
dent depression-like behaviors. J Neuroinammation. 2013;10:87.
57. Quak J, Doornbos B, Roest AM, Duivis HE, Vogelzangs N,
Nolen WA, et al. Does tryptophan degradation along the kynur-
enine pathway mediate the association between pro-inammatory
immune activity and depressive symptoms? Psychoneur-
oendocrinology. 2014;45:20210.
58. Ogyu K, Kubo K, Noda Y, Iwata Y, Tsugawa S, Omura Y, et al.
Kynurenine pathway in depression: a systematic review and meta-
analysis. Neurosci Biobehav Rev. 2018;90:1625.
59. Gabbay V, Klein RG, Katz Y, Mendoza S, Guttman LE, Alonso
CM, et al. The possible role of the kynurenine pathway in ado-
lescent depression with melancholic features. J Child Psychol
Psychiatry. 2010;51:93543.
60. Krause D, Myint AM, Schuett C, Musil R, Dehning S, Cerovecki
A, et al. High kynurenine (a tryptophan metabolite) predicts
remission in patients with major depression to add-on treatment
with celecoxib. Front Psychiatry. 2017;8:16.
61. Eskelund A, Li Y, Budac DP, Muller HK, Gulinello M, Sanchez
C, et al. Drugs with antidepressant properties affect tryptophan
metabolites differently in rodent models with depression-like
behavior. J Neurochem. 2017;142:11831.
62. Muller N, Myint AM, Schwarz MJ. The impact of neuroimmune
dysregulation on neuroprotection and neurotoxicity in psychiatric
disordersrelation to drug treatment. Dialogues Clin Neurosci.
2009;11:31932.
63. Ongur D, Drevets WC, Price JL. Glial reduction in the subgenual
prefrontal cortex in mood disorders. Proc Natl Acad Sci USA.
1998;95:132905.
64. Rajkowska G, Stockmeier CA. Astrocyte pathology in major
depressive disorder: insights from human postmortem brain tissue.
Curr Drug Targets. 2013;14:122536.
65. de Carvalho LP, Bochet P, Rossier J. The endogenous agonist
quinolinic acid and the non endogenous homoquinolinic acid
discriminate between NMDAR2 receptor subunits. Neurochem
Int. 1996;28:44552.
B. Kadriu et al.
Content courtesy of Springer Nature, terms of use apply. Rights reserved
66. Savitz J, Drevets WC, Smith CM, Victor TA, Wurfel BE, Bell-
gowan PS, et al. Putative neuroprotective and neurotoxic kynur-
enine pathway metabolites are associated with hippocampal and
amygdalar volumes in subjects with major depressive disorder.
Neuropsychopharmacology. 2015;40:46371.
67. Maes M, Leonard BE, Myint AM, Kubera M, Verkerk R. The new
5-HThypothesis of depression: cell-mediated immune activation
induces indoleamine 2,3-dioxygenase, which leads to lower
plasma tryptophan and an increased synthesis of detrimental
tryptophan catabolites (TRYCATs), both of which contribute to
the onset of depression. Prog Neuropsychopharmacol Biol Psy-
chiatry. 2011;35:70221.
68. Della Chiesa M, Carlomagno S, Frumento G, Balsamo M, Cantoni
C, Conte R, et al. The tryptophan catabolite L-kynurenine inhibits
the surface expression of NKp46- and NKG2D-activating recep-
tors and regulates NK-cell function. Blood. 2006;108:411825.
69. Krishnan V, Nestler EJ. The molecular neurobiology of depres-
sion. Nature. 2008;455:894902.
70. Veldic M, Millischer V, Port JD, Ho AM, Jia YF, Geske JR, et al.
Genetic variant in SLC1A2 is associated with elevated anterior
cingulate cortex glutamate and lifetime history of rapid cycling.
Transl Psychiatry. 2019;23:149.
71. Giorgini F, Huang SY, Sathyasaikumar KV, Notarangelo FM,
Thomas MA, Tararina M, et al. Targeted deletion of kynurenine 3-
monooxygenase in mice: a new tool for studying kynurenine
pathway metabolism in periphery and brain. J Biol Chem.
2013;288:3655466.
72. Amaral M, Levy C, Heyes DJ, Late P, Outeiro TF, Giorgini F,
et al. Structural basis of kynurenine 3-monooxygenase inhibition.
Nature. 2013;496:3825.
73. Smith JR, Jamie JF, Guillemin GJ. Kynurenine-3-mono-
oxygenase: a review of structure, mechanism, and inhibitors. Drug
Discov Today. 2016;21:31524.
74. Choi M, Lee SH, Wang SE, Ko SY, Song M, Choi JS, et al.
Ketamine produces antidepressant-like effects through
phosphorylation-dependent nuclear export of histone deacety-
lase 5 (HDAC5) in rats. Proc Natl Acad Sci USA. 2015;112:
1575560.
75. Duman RS. Pathophysiology of depression: the concept of
synaptic plasticity. Eur Psychiatry. 2002;17:30610.
The kynurenine pathway and bipolar disorder: intersection of the monoaminergic and glutamatergic. . .
Content courtesy of Springer Nature, terms of use apply. Rights reserved
1.
2.
3.
4.
5.
6.
Terms and Conditions
Springer Nature journal content, brought to you courtesy of Springer Nature Customer Service Center GmbH (“Springer Nature”).
Springer Nature supports a reasonable amount of sharing of research papers by authors, subscribers and authorised users (“Users”), for small-
scale personal, non-commercial use provided that all copyright, trade and service marks and other proprietary notices are maintained. By
accessing, sharing, receiving or otherwise using the Springer Nature journal content you agree to these terms of use (“Terms”). For these
purposes, Springer Nature considers academic use (by researchers and students) to be non-commercial.
These Terms are supplementary and will apply in addition to any applicable website terms and conditions, a relevant site licence or a personal
subscription. These Terms will prevail over any conflict or ambiguity with regards to the relevant terms, a site licence or a personal subscription
(to the extent of the conflict or ambiguity only). For Creative Commons-licensed articles, the terms of the Creative Commons license used will
apply.
We collect and use personal data to provide access to the Springer Nature journal content. We may also use these personal data internally within
ResearchGate and Springer Nature and as agreed share it, in an anonymised way, for purposes of tracking, analysis and reporting. We will not
otherwise disclose your personal data outside the ResearchGate or the Springer Nature group of companies unless we have your permission as
detailed in the Privacy Policy.
While Users may use the Springer Nature journal content for small scale, personal non-commercial use, it is important to note that Users may
not:
use such content for the purpose of providing other users with access on a regular or large scale basis or as a means to circumvent access
control;
use such content where to do so would be considered a criminal or statutory offence in any jurisdiction, or gives rise to civil liability, or is
otherwise unlawful;
falsely or misleadingly imply or suggest endorsement, approval , sponsorship, or association unless explicitly agreed to by Springer Nature in
writing;
use bots or other automated methods to access the content or redirect messages
override any security feature or exclusionary protocol; or
share the content in order to create substitute for Springer Nature products or services or a systematic database of Springer Nature journal
content.
In line with the restriction against commercial use, Springer Nature does not permit the creation of a product or service that creates revenue,
royalties, rent or income from our content or its inclusion as part of a paid for service or for other commercial gain. Springer Nature journal
content cannot be used for inter-library loans and librarians may not upload Springer Nature journal content on a large scale into their, or any
other, institutional repository.
These terms of use are reviewed regularly and may be amended at any time. Springer Nature is not obligated to publish any information or
content on this website and may remove it or features or functionality at our sole discretion, at any time with or without notice. Springer Nature
may revoke this licence to you at any time and remove access to any copies of the Springer Nature journal content which have been saved.
To the fullest extent permitted by law, Springer Nature makes no warranties, representations or guarantees to Users, either express or implied
with respect to the Springer nature journal content and all parties disclaim and waive any implied warranties or warranties imposed by law,
including merchantability or fitness for any particular purpose.
Please note that these rights do not automatically extend to content, data or other material published by Springer Nature that may be licensed
from third parties.
If you would like to use or distribute our Springer Nature journal content to a wider audience or on a regular basis or in any other manner not
expressly permitted by these Terms, please contact Springer Nature at
onlineservice@springernature.com
ResearchGate has not been able to resolve any citations for this publication.
Article
Full-text available
Glutamatergic dysregulation is implicated in the neurobiology of mood disorders. This study investigated the relationship between the anterior cingulate cortex (AC) glutamate, as measured by proton magnetic resonance spectroscopy (1H-MRS), and single-nucleotide polymorphisms (SNPs) from four genes (GLUL, SLC1A3, SLC1A2, and SLC1A7) that regulate the extracellular glutamate in 26 depressed patients with major depressive disorder (MDD; n = 15) and bipolar disorder (BD; n = 11). Two SNPs (rs3812778 and rs3829280), in perfect linkage disequilibrium, in the 3′ untranslated region of the EAAT2 gene SLC1A2, were associated with AC glutamate, with minor allele carriers having significantly higher glutamate levels (p < 0.001) in comparison with common allele homozygotes. In silico analysis revealed an association of minor allele carriers of rs3812778/rs382920 with an upregulation of the astrocytic marker CD44 localized downstream of SLC1A2 on chromosome 11. Finally, we tested the disease relevance of these SNPs in a large group of depressed patients [MDD (n = 458); BD (n = 1473)] and found that minor allele carriers had a significantly higher risk for rapid cycling (p = 0.006). Further work is encouraged to delineate the functional impact of excitatory amino acid transporter genetic variation on CD44 associated physiology and glutamatergic neurotransmission, specifically glutamate–glutamine cycling, and its contribution to subphenotypes of mood disorders.
Article
Full-text available
(R,S)-Ketamine produces rapid, robust, and sustained antidepressant effects in major depressive disorder. Specifically, its pharmacological efficacy in treatment refractory depression is considered a major breakthrough in the field. However, the mechanism of action of ketamine’s rapid effect remains to be determined. In order to identify pathways that are responsible for ketamine’s effect, a targeted metabolomic approach was carried out using a double-blind, placebo-controlled crossover design, with infusion order randomized with medication-free patients with treatment-resistant major depressive disorder (29 subjects) and healthy controls (25 subjects). The metabolomic profile of these subjects was characterized at multiple time points, and a comprehensive analysis was investigated between the following: MDD and healthy controls, treatment and placebo in both groups and the corresponding response to ketamine treatment. Ketamine treatment resulted in a general increase in circulating sphingomyelins, levels which were not correlated with response. Ketamine response resulted in more pronounced effects in the kynurenine pathway and the arginine pathway at 4 h post-infusion, where a larger decrease in circulating kynurenine levels and a larger increase in the bioavailability of arginine were observed in responders to ketamine treatment, suggesting possible mechanisms for response to ketamine treatment.
Article
Full-text available
Background: Due to the heterogeneity of depressive symptoms-which can include depressed mood, anhedonia, negative cognitive biases, and altered activity levels-researchers often use a combination of depression rating scales to assess symptoms. This study sought to identify unidimensional constructs measured across rating scales for depression and to evaluate these constructs across clinical trials of a rapid-acting antidepressant (ketamine). Methods: Exploratory factor analysis (EFA) was conducted on baseline ratings from the Beck Depression Inventory (BDI), the Hamilton Depression Rating Scale (HAM-D), the Montgomery-Asberg Depression Rating Scale (MADRS), and the Snaith-Hamilton Pleasure Rating Scale (SHAPS). Inpatients with major depressive disorder (n = 76) or bipolar depression (n = 43) were participating in clinical ketamine trials. The trajectories of the resulting unidimensional scores were evaluated in 41 subjects with bipolar depression who participated in clinical ketamine trials. Results: The best solution, which exhibited excellent fit to the data, comprised eight factors: Depressed Mood, Tension, Negative Cognition, Impaired Sleep, Suicidal Thoughts, Reduced Appetite, Anhedonia, and Amotivation. Various response patterns were observed across the clinical trial data, both in treatment effect (ketamine versus placebo) and in degree of placebo response, suggesting that use of these unidimensional constructs may reveal patterns not observed with traditional scoring of individual instruments. Limitations: Limitations include: 1) small sample (and related inability to confirm measurement invariance); 2) absence of an independent sample for confirmation of factor structure; and 3) the treatment-resistant nature of the population, which may limit generalizability. Conclusions: The empirical identification of unidimensional constructs creates more refined scores that may elucidate the connection between specific symptoms and underlying pathophysiology.
Article
Full-text available
A subset of patients with depression have elevated levels of inflammatory cytokines, and some studies demonstrate interaction between inflammatory factors and treatment outcome. However, most studies focus on only a narrow subset of factors in a patient sample. In the current study, we analyzed broad immune profiles in blood from patients with treatment-resistant depression (TRD) at baseline and following treatment with the glutamate modulator ketamine. Serum was analyzed from 26 healthy control and 33 actively depressed TRD patients free of antidepressant medication, and matched for age, sex and body mass index. All subjects provided baseline blood samples, and TRD subjects had additional blood draw at 4 and 24 h following intravenous infusion of ketamine (0.5 mg kg⁻¹). Samples underwent multiplex analysis of 41 cytokines, chemokines and growth factors using quantitative immunoassay technology. Our a priori hypothesis was that TRD patients would show elevations in canonical pro-inflammatory cytokines; analyses demonstrated significant elevation of the pro-inflammatory cytokine interleukin-6. Further exploratory analyses revealed significant regulation of four additional soluble factors in patients with TRD. Several cytokines showed transient changes in level after ketamine, but none correlated with treatment response. Low pretreatment levels of fibroblast growth factor 2 were associated with ketamine treatment response. In sum, we found that patients with TRD demonstrate a unique pattern of increased inflammatory mediators, chemokines and colony-stimulating factors, providing support for the immune hypothesis of TRD. These patterns suggest novel treatment targets for the subset of patients with TRD who evidence dysregulated immune functioning.
Article
Full-text available
Introduction Bipolar disorder (BD) is a chronic psychiatric disease which can take most different and unpredictable courses. It is accompanied by unspecific brainstructural changes and cognitive decline. The neurobiological underpinnings of these processes are still unclear. Emerging evidence suggests that tryptophan catabolites (TRYCATs), which involve all metabolites of tryptophan towards the kynurenine (KYN) branch, are involved in the etiology as well as in the course of BD. They are proposed to be mediators of immune-inflammation and neurodegeneration. In this study we measured the levels of KYN and its main catabolites consisting of the neurotoxic hydroxykynurenine (3-HK), the more neuroprotective kynurenic acid (KYNA) and anthranilic acid (AA) and evaluated the ratios between end-products and substrates as proxies for the specific enzymatic activity (3-HK/KYN, KYNA/KYN, AA/KYN) as well as 3-HK/KYNA as a proxy for neurotoxic vs. neuroprotective end-product relation in individuals with BD compared to healthy controls (HC). Methods We took peripheral TRYCAT blood levels of 143 euthymic to mild depressive BD patients and 101 HC. For statistical analyses MANCOVA’s controlled for age, sex, body mass index, cardiovascular disease and smoking were performed. Results The levels of KYNA (F = 5,579; p <.05) were reduced in BD compared to HC. The enzymatic activity of the kynurenine-3-monooxygenase (KMO) reflected by the 3-HK/KYN ratio was increased in BD individuals compared to HC (F = 5,394; p <.05). Additionally the ratio of 3-HK/KYNA was increased in individuals with BD compared to healthy controls (F = 11,357; p <.01). Discussion In conclusion our findings subserve the concept of KYN -pathway alterations in the pathophysiology of BD. We present evidence of increased breakdown towards the neurotoxic branch in KYN metabolism even in a euthymic to mild depressive state in BD. From literature we know that depression and mania are accompanied by inflammatory states which should be capable to produce an even greater imbalance due to activation of key enzymes in the neurotoxic direction of KYN -conversion. These processes could finally be involved in the development of unspecific brain structural changes and cognitive deficits which are prevalent in BD. Further research should focus on state dependent changes in TRYCATs and its relation to cognition, brain structure and staging parameters.
Article
The kynurenine pathway (KP) plays a critical role in generating cellular energy in the form of nicotinamide adenine dinucleotide (NAD+). Because energy requirements are substantially increased during an immune response, the KP is a key regulator of the immune system. Perhaps more importantly in the context of psychiatry, many kynurenines are neuroactive, modulating neuroplasticity and/or exerting neurotoxic effects in part through their effects on NMDA receptor signaling and glutamatergic neurotransmission. As such, it is not surprising that the kynurenines have been implicated in psychiatric illness in the context of inflammation. However, because of their neuromodulatory properties, the kynurenines are not just additional members of a list of inflammatory mediators linked with psychiatric illness, but in preclinical studies have been shown to be necessary components of the behavioral analogs of depression and schizophrenia-like cognitive deficits. Further, as the title suggests, the KP is regulated by, and in turn regulates multiple other physiological systems that are commonly disrupted in psychiatric disorders, including endocrine, metabolic, and hormonal systems. This review provides a broad overview of the mechanistic pathways through which the kynurenines interact with these systems, thus impacting emotion, cognition, pain, metabolic function, and aging, and in so doing potentially increasing the risk of developing psychiatric disorders. Novel therapeutic approaches targeting the KP are discussed. Moreover, electroconvulsive therapy, ketamine, physical exercise, and certain non-steroidal anti-inflammatories have been shown to alter kynurenine metabolism, raising the possibility that kynurenine metabolites may have utility as treatment response or therapeutic monitoring biomarkers.
Article
Abnormalities of the kynurenine (KYN) pathway may be implicated in the pathophysiology of depression. However, the relationships between depression and each metabolite of the KYN pathway remain uncertain. Therefore, we conducted a meta-analysis about the levels of the metabolites of KYN pathway between patients with depression and controls. Out of 899 initial records, we identified 22 articles to form the empirical basis. Seventeen, 10, and 18 studies examined levels of kynurenic acid (KYNA), quinolinic acid (QUIN), and KYN, respectively. KYNA and KYN levels were lower in patients with depression in comparison to controls, while QUIN levels did not differ between the two groups. Antidepressant-free patients showed decreased KYNA levels and increased QUIN levels compared with controls. Male ratios of the samples were negatively associated with study SMDs for KYNA. In conclusion, this meta-analysis revealed that patients with depression had decreased level of KYNA and KYN, whereas antidepressant-free patients showed increased level of QUIN. Nevertheless, given the heterogeneity among their sample characteristics, further research is clearly needed.
Article
Depression is the most common mental disorder in the world. It is estimated that 350 million people suffer from depression worldwide. Depressive disorders will have become the second most frequent health problem globally by the year 2020, just behind ischemic heart disease. The causes of depressive disorders are not fully known. Previous studies showed that impaired tryptophan catabolites pathway, oxidative and nitrosative stress may play an important role in the pathogenesis of depression. Patients with depression have lower plasma levels of superoxide dismutase and glutathione peroxidise in comparison to controls. Moreover, depressed patients are characterized by decreased plasma levels of zinc, coenzyme Q10, albumin, uric acid, vitamin E and glutathione. Abnormal nitric oxidative production and nitric oxide synthase activity are also associated with depression. A dysfunction of the tryptophan catabolites pathway, indicated by increased levels of tryptophan 2,3-dioxygenase and indoleamine 2,3-dioxygenase, is also involved in the development of depression. Furthermore, increased levels of kynurenine and quinolinic acid might cause depression. Moreover, studies to date indicate that 8-oxyguanine, malondialdehyde, and 8-iso-prostaglandin F2α may serve as possible biomarkers. Additionally, regulation of defective mechanisms may provide a promising direction for the development of new and effective therapies.
Article
Patients with major depressive disorder (MDD) have clinically relevant, significant decreases in bone mineral density (BMD). We sought to determine if predictive markers of bone inflammation-the osteoprotegerin (OPG)-RANK-RANKL system or osteopontin (OPN)-play a role in the bone abnormalities associated with MDD and, if so, whether ketamine treatment corrected the abnormalities. The OPG-RANK-RANKL system plays the principal role in determining the balance between bone resorption and bone formation. RANKL is the osteoclast differentiating factor and diminishes BMD. OPG is a decoy receptor for RANKL, thereby increasing BMD. OPN is the bone glue that acts as a scaffold between bone tissues matrix composition to bind them together and is an important component of bone strength and fracture resistance. Twenty-eight medication-free inpatients with treatment-resistant MDD and 16 healthy controls (HCs) participated in the study. Peripheral bone marker levels and their responses to IV ketamine infusion in MDD patients and HCs were measured at four time points: at baseline, and post-infusion at 230 min, Day 1, and Day 3. Patients with MDD had significant decreases in baseline OPG/RANKL ratio and in plasma OPN levels. Ketamine significantly increased both the OPG/RANKL ratio and plasma OPN levels, and significantly decreased RANKL levels. Bone marker levels in HCs remained unaltered. We conclude that the OPG-RANK-RANKL system and the OPN system play important roles in the serious bone abnormalities associated with MDD. These data suggest that, in addition to its antidepressant effects, ketamine also has a salutary effect on a major medical complication of depressive illness.Molecular Psychiatry advance online publication, 30 May 2017; doi:10.1038/mp.2017.109.
Article
The metabolism of tryptophan through kynurenine and serotonin pathways is linked to depression. Here, effects of different drugs with antidepressant properties (vortioxetine, fluoxetine, and ketamine) on various tryptophan metabolites in different brain regions and plasma were examined using tandem mass spectrometry ( LC ‐ MS / MS ), in Flinders Sensitive Line rats, a genetic rat model of depression, and its controls: Flinders Sensitive Line and Sprague–Dawley rats. Protein levels of kynurenine pathway enzymes were measured in the brains and livers of these rat strains. Furthermore, effects of vortioxetine on tryptophan metabolites were assessed in the cortical regions of lupus mice ( MRL /MpJ‐Fas Ipr ), a murine model of increased depression‐like behavior associated with inflammation. Sustained vortioxetine or fluoxetine (at doses aimed to fully occupy serotonin transporter via food or drinking water for at least 14 days) reduced levels of the excitotoxin quinolinic acid ( QUIN ) in various brain regions in all rats. Furthermore, chronic vortioxetine reduced levels of QUIN in MRL /MpJ‐Fas Ipr mice. Acute i.p. administration of fluoxetine (10 mg/kg) or vortioxetine (10 mg/kg) led to reduced brain 5‐hydroxyindoleacetic acid in Sprague–Dawley rats (2, 4, 6, and 8 h) and a similar trend was evident in Flinders Sensitive Line and Flinders Sensitive Line rats after 4 h. In contrast, single or repeated administration of ketamine (15 mg/kg i.p.) did not induce significant changes in metabolite levels. In conclusion, sustained vortioxetine and fluoxetine administration decreased QUIN independent of species, while ketamine was ineffective. These results support the hypothesis that modulating tryptophan metabolism may be part of the mechanism of action for some antidepressants. image