Ky Nure Nine Addiction

You might also like

Download as pdf or txt
Download as pdf or txt
You are on page 1of 17

Pharmacology & Therapeutics 223 (2021) 107807

Contents lists available at ScienceDirect

Pharmacology & Therapeutics

journal homepage: www.elsevier.com/locate/pharmthera

Addiction and the kynurenine pathway: A new dancing couple?


Nuria Morales-Puerto a,b,c,d,1, Pablo Giménez-Gómez a,b,c,d,1, Mercedes Pérez-Hernández a,b,c,d,
Cristina Abuin-Martínez a,b,c,d, Leticia Gil de Biedma-Elduayen a,b,c,d, Rebeca Vidal a,b,c,d,
María Dolores Gutiérrez-López a,b,c,d, Esther O'Shea a,b,c,d,⁎, María Isabel Colado a,b,c,d,⁎
a
Departamento de Farmacología y Toxicología, Facultad de Medicina, Universidad Complutense, Madrid, Spain
b
Instituto de Investigación Sanitaria Hospital 12 de Octubre, Madrid, Spain
c
Red de Trastornos Adictivos del Instituto de Salud Carlos III, Madrid, Spain
d
Instituto Universitario de Investigación Neuroquímica (IUIN), Facultad de Medicina, Universidad Complutense, Madrid, Spain

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

Available online 19 January 2021 Drug use poses a serious threat to health systems throughout the world and the number of consumers rises re-
lentlessly every year. The kynurenine pathway, main pathway of tryptophan degradation, has drawn interest in
this field due to its relationship with addictive behaviour. Recently it has been confirmed that modulation of
Keywords: kynurenine metabolism at certain stages of the pathway can reduce, prevent or abolish drug seeking-like behav-
Kynurenine pathway iours in studies with several different drugs. In this review, we present an up-to-date summary of the evidences
Addiction of a relationship between drug use and the kynurenine pathway, both the alterations of the pathway due to drug
Ethanol use as well as modulation of the pathway as a potential approach to treat drug addiction. The review discusses
Nicotine ethanol, nicotine, cannabis, amphetamines, cocaine and opioids and new prospects in the drug research field
Cannabis
are proposed.
Ro 61–8048
© 2021 Published by Elsevier Inc.

Contents

1. Introduction . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 2
2. Neurobiology of addiction . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 2
3. Tryptophan metabolism and the kynurenine pathway . . . . . . . . . . . . . . . . . . . . . . . . . . . . 2
4. Neuromodulation of brain circuits associated with addiction by kynurenine and its metabolites. . . . . . . . . . 4
5. Alcohol . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 6
6. Nicotine . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 7
7. Cannabis . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 10
8. Amphetamine and its derivatives . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 10
9. Cocaine . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 11
10. Opioids: a special case . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 12
11. Future prospects and new trends . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 13
Declaration of Competing Interest . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 14
Acknowledgements . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 14
References. . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 14

Abbreviations: AhR, Aryl hydrocarbon receptor; BBB, Blood-brain barrier; CA, Cinnabarinic acid; CB1/2, Cannabinoid receptor 1/2; CBD, Cannabidiol; CNS, Central nervous system; DA,
Dopamine; EtOH, Ethanol; GABA, γ-aminobutyric acid; GPR35, G protein-coupled receptor 35; 3-HK, 3-hydroxykynurenine; 5-HT, Serotonin; IDO, Indolamine 2,3-dioxygenase; KAT,
Kynurenine aminotransferase; KMO, Kynurenine 3-monooxygenase; KYN, Kynurenine; KYNA, Kynurenic acid; mGluR, Metabotropic glutamate receptor; NAc, Nucleus accumbens;
nAChR, Nicotinic acetylcholine receptor; NMDA, N-methyl-D-aspartate; PFC, Prefrontal cortex; QA, Quinolinic acid; TDO, Tryptophan 2,3-dioxygenase; THC, Tetrahydrocannabinol; TRP,
Tryptophan; VTA, Ventral tegmental area; XA, Xanthurenic acid.
⁎ Corresponding authors at: Departamento de Farmacología y Toxicología, Facultad de Medicina, Universidad Complutense, Pza. Ramón y Cajal s/n, 28040 Madrid, Spain.
E-mail addresses: estheros@med.ucm.es (E. O'Shea), colado@med.ucm.es (M.I. Colado).
1
Contributed equally.

https://doi.org/10.1016/j.pharmthera.2021.107807
0163-7258/© 2021 Published by Elsevier Inc.
N. Morales-Puerto, P. Giménez-Gómez, M. Pérez-Hernández et al. Pharmacology & Therapeutics 223 (2021) 107807

1. Introduction by the activation of the extended amygdala. This brain area is composed
of the central nucleus of the amygdala, bed nucleus of the stria
Drug abuse and addiction are a major global health problem, as is re- terminalis and the shell of the NAc and plays a key role in the integration
vealed in the World Drug Report of 2019 published by the United Na- of stress systems with hedonic processing systems, in order to produce
tions Office on Drugs and Crime. According to the report, in 2017 the negative emotional state that will subsequently establish the nega-
about 271 million people declared having used drugs in the previous tive reinforcement associated with the development of the addiction
year, which represents an increase of 30% compared with 2009. More- (Koob & Volkow, 2010). The reward obtained after drug consumption
over, currently there are 35 million people diagnosed with drug use dis- is substituted by a reduction in DA release that decreases motivation
orders all over the world. These data together indicate a high burden of for non-drug stimuli (Melis, Spiga, & Diana, 2005). In addition, in the
disease with over half a million deaths and 42 million years of “healthy” withdrawal phase, the activation of the release of corticotropin-
life lost due to drug use (World Health Organization, 2019a). Despite releasing factor and the increase in corticosterone and adrenocortico-
the devastating impact of addiction on society, pharmacological treat- tropic hormone levels in the amygdala promote the engagement of
ments and behavioral therapies are far from being effective for most users in drug consumption in order to avoid this aversive mood
people. Hence, new therapeutic strategies are necessary to combat (Koob, 2008).
these disorders. The last stage of this process is the craving stage. In this last step of
The kynurenine pathway, the main route of tryptophan (TRP) degra- the addiction cycle, an individual has a persistently down-regulated
dation, is implicated in several physiological processes, such as neuronal state and may seek their substance of choice to ameliorate the anhedo-
and immunological functions or ageing processes (Stone, Stoy, & nia and other unpleasant and undesirable consequences of that state
Darlington, 2013). Furthermore, the kynurenine pathway has been im- (Fogaça et al., 2016a). Evidence from animal models show that there
plicated in a wide range of diseases of the central nervous system (CNS), are three possible mechanisms involving different brain areas and,
including neurodegenerative and neuropsychiatric disorders (Platten, mainly, the neurotransmitter glutamate by which an individual can re-
Nollen, Röhrig, Fallarino, & Opitz, 2019). In line with this, during the instate drug-seeking behaviour. The first one is drug-induced reinstate-
last decades it has been proposed that the kynurenine pathway can con- ment, by a dose that can act as a priming and in which the medial PFC/
tribute to or modulate addictive behaviour. These findings open the NAc/ventral pallidum circuit play a key role (McFarland & Kalivas,
door to the study of the involvement of the kynurenine pathway in ad- 2001). The second one involves cue-reinstatement and relies on struc-
dictive disorders and offers a new opportunity to establish the tures such as the basolateral amygdala and the PFC (Everitt & Wolf,
kynurenine pathway as a new pharmacological target in the treatment 2002; Knackstedt & Kalivas, 2009). The last one involves the extended
of addictive disorders. amygdala and, in addition to glutamate, corticotropin-releasing factor
in a stress-induced reinstatement mechanism (Shaham, Shalev, Lu, De
2. Neurobiology of addiction Wit, & Stewart, 2003).

The key brain area associated with addiction is the limbic system, 3. Tryptophan metabolism and the kynurenine pathway
which comprises the cingulate gyrus, amygdala, hippocampus, prefron-
tal cortex (PFC), ventral tegmental area (VTA) and the nucleus accum- TRP, as an essential amino acid, is acquired from the diet, after which
bens (NAc). All these areas are related to reward, emotion and is either destined to the formation of proteins or undergoes metabolic
punishment, thus boding critical importance in addiction (Fogaça, degradation. The main metabolic route of TRP degradation is the
Campos, & Guimarães, 2016a). Reward is defined as any event that in- kynurenine pathway, which eventually leads to the synthesis of redox
creases the probability of a response with a positive hedonic compo- cofactor nicotinamide adenine dinucleotide (Fig. 2). Up to 95% of TRP
nent. All known drugs of abuse act stimulating the brain reward is metabolized to kynurenine (KYN), while the remaining 5% is trans-
system and changes in this system are key to understanding the devel- formed into serotonin (5-HT) (Ikeda et al., 1965; Leklem, 1971). KYN
opment of addiction (Koob & Volkow, 2016). Limbic structures give he- is formed by the action of the enzymes indolamine 2,3-dioxygenase
donic value to stimuli and in this effect dopamine (DA), a (IDO), present in most tissues (Yamazaki, Kuroiwa, Takikawa, & Kido,
monoaminergic neurotransmitter produced in the substantia nigra 1985), and tryptophan 2,3-dioxygenase 2 (TDO2, here TDO), found
and in the VTA, plays a key role. The dopaminergic connection between mainly in the liver (Rongvaux, Andris, Van Gool, & Leo, 2003). IDO exists
the VTA and the NAc is usurped by drugs of abuse (Taber, Black, Porrino, as two different proteins coded for on adjacent genes in humans and
& Hurley, 2012). In this sense, all drugs produce an increase in DA re- mice (IDO1 and IDO2; Badawy, 2017). Whereas IDO1 is widely
lease in the NAc, although the exact mechanism by which drugs of expressed, exhibits high affinity for TRP and has been shown to have
abuse can promote the mentioned DA liberation varies among the dif- an important role in systemic TRP catabolism in particular when in-
ferent classes of substances of abuse (Fig. 1). duced by inflammatory stimuli (Larkin et al., 2016), IDO2 shows a
The most accepted theory about the addiction process proposes a more restricted pattern of expression, a lower affinity for TRP and re-
disorder that includes three phases in which users of drugs of abuse duced catalytic activity compared with IDO1 (Metz et al., 2007). Al-
pass from positive reinforcement and voluntary use to compulsive con- though studies indicate a non-redundant role for this form of the
sumption with negative reinforcement in the absence of drug (Koob & enzyme in the control of inflammation and adaptive immunity, its dele-
Volkow, 2010). The first stage of these three-stage cycle is the binge tion does not alter plasma concentrations of KYN (Metz et al., 2014),
or intoxication stage. This stage is influenced by substance pharmacoki- thus, its contribution to systemic TRP catabolism has yet to be defined.
netics and administration methods (Fogaça et al., 2016a; Fogaça, In the mammalian brain, KYN can be synthesised by IDO1, though
Campos, & Guimarães, 2016b) but relies on the release of DA in the more than half of brain KYN is taken up from the periphery (60%), indi-
NAc, crucial for the acute reinforcing effects of drugs, and the perception cating that it can be transported across the blood-brain barrier (BBB)
of pleasure and enjoyment (Koob & Volkow, 2010). The exact mecha- (Fukui, Schwarcz, Rapoport, Takada, & Smith, 1991). From KYN, the
nism by which a particular drug produces the release of DA will be de- pathway splits in two directions: the neurotoxic branch, towards the
scribed in the section corresponding to each drug of abuse. formation of quinolinic acid (QA) by the action of the enzyme
During the second stage, the withdrawal phase, the brain cannot kynurenine 3-monooxygenase (KMO) (Guillemin, 2012); and the neu-
regulate its experience of pleasure and reward from the drugs of roprotective branch, by the action of the enzyme kynurenine amino-
abuse independently due to their chronic effects, that include a down- transferase (KAT) leading to the formation of kynurenic acid (KYNA)
regulation of reward-related neurotransmitter levels (Koob & Le Moal, (Schwarcz, Bruno, Muchowski, & Wu, 2012a, 2012b). The KYNA/QA
2001). In this phase, a negative emotional state predominates, caused ratio has been proposed recently as a putative neuroprotection index,

2
N. Morales-Puerto, P. Giménez-Gómez, M. Pérez-Hernández et al. Pharmacology & Therapeutics 223 (2021) 107807

Fig. 1. Sites of action of different drugs in the VTA-NAc connection. Although converging in the release of DA in the NAc, each drug of abuse can act through different mechanisms in order to
promote this circuitry signalling. With the exception of amphetamines, all drugs discussed in this review can alter the activity of glutamatergic afferences to VTA, as well as the activity of
local GABAergic interneurons, causing an increment of the former's and an inhibition in the latter's input. EtOH would act on GABAergic neurons by blockade of glutamatergic receptors, i.e.
AMPA and NMDA, while nicotine, cannabis and opioids would activate nAChR, CB1 and MOR receptors, respectively. EtOH and nicotine have the ability to directly activate dopaminergic
neurons by interacting with G protein-coupled inwardly rectifying potassium channels, in the case of EtOH, or nAChR, in the case of nicotine. In the NAc, cocaine and amphetamines exert
their effects by increasing the presence of DA on the synapse. On the one hand, both drugs diminish DA reuptake by blockade of the dopamine transporter. Additionally, amphetamines
induce reverse transport of DA. On the other hand, cocaine increases the concentration of DA in the synapse by desensibilization of the D2 autoreceptor. AMPA: α-amino-3-hydroxy-5-
methyl-4-isoxazolepropionic acid receptor; CB1: cannabinoid receptor 1; D2R: dopamine autoreceptor 2; DA: dopamine; DAT: dopamine transporter; EtOH: ethanol; GIRK: G protein-coupled
inwardly rectifying potassium channel; MOR: μ opioid receptor; nAChR: nicotinic acetylcholine receptor; NMDA: N-methyl-D-aspartate receptor; VTA: ventral tegmental area.

by which decreases in the ratio can signal inflammation-related pathol- nicotinic acetylcholine receptor (nAChR), blocking glutamate release,
ogy (Savitz et al., 2015). and regulated the expression of α4β2nAChR (Hilmas et al., 2001)
QA is one of the most studied metabolites of KYN since its interaction (Fig. 3). However, further experiments in order to replicate this event
with the N-methyl-D-aspartate (NMDA) receptor complex, an ionotropic have had mixed outcomes: while some authors could not reproduce the
glutamate receptor, can cause excitotoxic neuronal cell loss and convul- results, even when using high concentrations of KYNA (Arnaiz-Cot et al.,
sions (Stone & Perkins, 1981). In contrast, KYNA is an endogenous 2008; Dobelis, Staley, & Cooper, 2012; Mok, Fricker, Weil, & Kew, 2009;
NMDA receptor antagonist, which acts mainly at the glycine site on the Stone, 2020), the same group in subsequent assays and also other re-
receptor complex (Birch, Grossman, & Hayes, 1988; Kessler, Terramani, searchers did so, both in vitro and in vivo (Alkondon et al., 2011; Grilli
Lynch, & Baudry, 1989; Perkins & Stone, 1982). It is well-known that in et al., 2006; Lopes et al., 2007). To this day this interaction continues to ac-
many neurodegenerative disorders, for instance Parkinson's disease and cumulate supporting evidence, although it remains to be unequivocally
Alzheimer's disease, and in stroke and epilepsy, the activation of excit- confirmed and is thus regarded as dubious by some experts. In the next
atory amino acid receptors, such as the NMDA receptor, plays a pivotal section, we will elaborate on the interaction between KYNA and its
role (Gagliardi, 2000; Greenamyre & Young, 1989; Shaw, 1993). Numer- brain targets in more detail.
ous studies in the literature have attempted to clarify whether KYNA, as Since KYNA has a very limited ability to cross the BBB (Fukui et al.,
an excitatory amino acid receptor antagonist, can exert a therapeutic ef- 1991), efforts have been made to raise its brain levels in order to pro-
fect in these neurological disorders (Hertelendy, Toldi, Fülöp, & Vécsei, mote its neuroprotective influence. To this end, KYNA derivatives have
2018; Mangas et al., 2018; Tanaka, Bohár, Martos, Telegdy, & Vécsei, been synthesised (e.g. glucosamine-KYNA, 4-chloro-KYNA and 7-
2020). Furthermore, KYNA can interact with several other receptors. chloro-KYNA), in order to produce compounds similar to KYNA in
Sites of action for this metabolite have been described in the astrocytic their ability to act on the glutamate receptor but better transported
G-protein coupled receptor 35 (GPR35) (Wang et al., 2006). Also, KYNA across the BBB (Deora et al., 2017; Gellért et al., 2012). Moreover, at
is an agonist at the aryl hydrocarbon receptor (AhR) (DiNatale et al., least three more strategies have been established for the purpose of pro-
2010). Finally, there is one last receptor with which KYNA appears to in- ducing increases in the cerebral concentration of KYNA. The use of pro-
teract. In the early 2000's, a study using cultured hippocampal neurons benecid, an inhibitor of the transport of organic acids such as KYN and
showed that KYNA non-competitively inhibited the presynaptic α7 KYNA, blocks the exit of these compounds out of the brain, thus

3
N. Morales-Puerto, P. Giménez-Gómez, M. Pérez-Hernández et al. Pharmacology & Therapeutics 223 (2021) 107807

Fig. 2. Kynurenine pathway of TRP degradation and effect of drugs on KYN metabolites. While a small portion of circulating TRP is destined to the synthesis of proteins or serotonin, 95% of it
is metabolized through the kynurenine pathway, which ultimately generates two main products: the neuroprotective KYNA and the neurotoxic QA. Several drugs of abuse have been
shown to affect the levels of metabolites of this pathway. Cocaine can decrease KYNA and anthranilic acid concentration, while leaving KYN and QA unaltered. EtOH exposure causes
an increase in KYN levels, but it does not affect KYNA. Nicotine raises KYNA levels. Amphetamine reduces both KYN and KYNA. 3-HAO: 3-hydroxyanthranilic acid oxygenase; AADC:
aromatic-L-amino acid decarboxylase; EtOH: ethanol; IDO: indoleamine-2,3-dioxygenase; KAT: kynurenine amino transferase; KMO: kynurenine 3-monooxygenase; KYN: kynurenine; KYNA:
kynurenic acid; KYNU: kynureninase; QA: quinolinic acid; QPRT: quinolinic acid phosphoribosyl transferase; TDO: tryptophan-2,3-dioxygenase; TPH: tryptophan hydroxylase; TRP: tryptophan.

increasing their cerebral concentration (Nilsson, Linderholm, & Erhardt, 4. Neuromodulation of brain circuits associated with addiction by
2006). The second strategy consists in the administration of exogenous kynurenine and its metabolites
L-kynurenine to increase both peripheral and central levels of KYN and
KYNA. These studies have been performed pre- and postnatally and In spite of acting through different mechanisms of action involving
showed that the administration of KYN can cause cognitive deficits several neurotransmitter systems and eliciting varying effects outside
(Alexander et al., 2013; Pocivavsek, Wu, Elmer, Bruno, & Schwarcz, the brain reward circuitry, all drugs of abuse ultimately activate the
2012), learning deficits (Pocivavsek, Thomas, Elmer, Bruno, & mesolimbic pathway after acute administration: i.e. by acting at different
Schwarcz, 2014) and a schizophrenic-like phenotype (Goeden et al., levels, they directly or indirectly increase dopaminergic transmission
2017). The third strategy involves the use of enzyme inhibitors which from the VTA to the NAc in the ventral striatum (Nestler, 2005). This do-
block the synthesis of the neurotoxic QA and redirect KYN metabolism paminergic transmission mediates the hedonic consequences of a rein-
towards KYNA. In this sense, Ro 61–8048, a KMO inhibitor (Röver, forcing stimulus, promoting associative learning about the stimulus or
Cesura, Huguenin, Kettler, & Szente, 1997), is one of the most promising anticipating its rewarding effects (Di Chiara, 1999). Hence, the
strategies for the treatment of neurodegenerative diseases or other pa- mesolimbic circuit is considered the neuroanatomical substrate for drug
thologies such as drug abuse (Zwilling et al., 2011). The administration dependence. It is widely modulated by several neurotransmitter systems,
of Ro 61–8048 produces a marked increase in both KYN and KYNA in especially by glutamatergic inputs from PFC, hippocampus, amygdala and
plasma and in various brain areas of mice and rats. Since Ro 61–8048 ex- other areas, which, together, are named “brain reward regions” (Russo &
hibits low blood-brain penetration the increases observed are derived Nestler, 2013). Drug-induced modifications in NAc glutamate neurotrans-
from the increases in the periphery. Moreover, since KYNA does not mission have been identified as an underlying factor driving drug-taking,
cross the BBB, the increase in KYNA in the brain is due to the conversion relapse, withdrawal and other drug-associated behaviours for several
of peripherally-derived KYN. A low brain penetrability could be advan- drugs (Kalivas, LaLumiere, Knackstedt, & Shen, 2009).
tageous by maximizing clinical benefit and managing toxicity in combi- The kynurenine metabolite that has received most attention due to
nation therapies (Reinhart & Kelly, 2011). Although there is concern its neuromodulatory role is KYNA. The mechanism by which KYNA
that the increase in brain KYN, in the absence of brain KMO inhibition, can counteract the addictive effects of drugs of abuse seems to involve
might lead to an increased conversion to QA (Muneer, 2020) this does the regulation of glutamatergic transmission through interactions with
not appear to be the case since QA levels in the brains of mice were un- several of its target receptors. Firstly, KYNA shows a great affinity to
altered following treatment with Ro 61–8048 (Chiarugi & Moroni, NMDA receptors, where it functions as an antagonist by interacting at
1999) or JM6 (a prodrug of Ro 61–8048; Zwilling et al., 2011). Thus, pe- the glycine binding site (Birch et al., 1988; Kessler et al., 1989; Stone &
ripheral inhibition of KMO appears to be a valuable mechanism for in- Perkins, 1981). This relationship is well-documented, given that since
creasing levels of KYN and KYNA in the CNS. its discovery, hundreds of scientists have verified this antagonism

4
N. Morales-Puerto, P. Giménez-Gómez, M. Pérez-Hernández et al. Pharmacology & Therapeutics 223 (2021) 107807

Fig. 3. Targets of the kynurenine pathway metabolites in the CNS. Several compounds derived from kynurenine metabolism exert effects on different receptors on the brain. KYNA
neuroprotective properties may come from its ability to antagonise both the NMDA and the AMPA glutamate receptor, as well as, potentially, the α7nAChR. On the contrary, QA
neurotoxicity is due to stimulation of both the NMDA and AMPA receptors. Moreover, KYNA is an agonist of astrocytic GPR35 and transcription factor AhR, which is also a target for
KYN and CA. XA participates on glutamatergic transmission in several stages: on the one hand, it prevents glutamate reuptake via inhibition of vesicular transporters; on the other
hand, alongside with CA, it can activate metabotropic glutamate receptors. Green arrows represent activation, red lines represent inhibition. 3-HK: 3-hydroxykynurenic acid; α7nAChR:
alpha-7 nicotinic acetylcholine receptor; AhR: aryl-hydrocarbon receptor; AMPAR: α-amino-3-hydroxy-5-methyl-4-isoxazolepropionic acid receptor; BBB: blood-brain barrier; CA: cinnabarinic
acid; CNS: central nervous system; GPR35: G protein-coupled receptor 35; KYN: kynurenine; KYNA: kynurenic acid; mGluR: metabotropic glutamate receptor; NMDAR: N-methyl-D-aspartate
receptor; QA: quinolinic acid; TRP: tryptophan; vGLUT: vesicular glutamate transporter; XA: xanturenic acid.

(Moroni, Cozzi, Sili, & Mannaioni, 2012; Schwarcz et al., 2012a, 2012b; attention in the field of drug addiction, since electrophysiological stud-
Stone & Darlington, 2002). The relevance of the KYNA-NMDA receptor ies have demonstrated that physiologically relevant concentrations of
interaction to the drug addiction field relies especially on the involve- KYNA block the activity of this receptor in a non-competitive and
ment of NMDA receptors in two critical aspects. On the one hand, cogni- voltage-independent manner (Hilmas et al., 2001). Within the
tive processes, which are overall negatively affected by drug mesolimbic pathway, α7nAChRs are mainly located at the terminals of
consumption, can undergo alterations due to imbalances in the glutamatergic afferences that modulate the activity of medium-sized
kynurenine pathway (Akagbosu, Evans, Gulick, Suckow, & Bucci, 2012; spiny neurons of the NAc (Zappettini et al., 2014) and dopaminergic
Chess, Simoni, Alling, & Bucci, 2007; Pocivavsek et al., 2012; Ramirez neurons of the VTA (Jones & Wonnacott, 2004). The activation of these
Ortega et al., 2020). For instance, the increased concentrations of receptors induces the release of glutamate that subsequently enhances
KYNA found in the brains of schizophrenic patients (Erhardt, Oberg, & the release of DA in NAc from VTA dopaminergic neurons. Therefore,
Engberg, 2001; Linderholm et al., 2012; Nilsson et al., 2005), which negative modulation of KYNA on α7nAChR reduces mesolimbic activity.
would cause a glutamatergic hypofunction via NMDA receptor antago- Moreover, KYNA has been described to act as a ligand of the orphan
nism, could be the explanation for the negative symptoms of schizo- GPR35 receptor (Wang et al., 2006), whose activation may contribute to
phrenia, in particular those involving cognitive impairment (Chess the decrease in extracellular glutamate levels and, therefore, to a reduc-
et al., 2007; Shepard, Joy, Clerkin, & Schwarcz, 2003). On the other tion in excitatory transmission (Albuquerque & Schwarcz, 2013). All
hand, neural plasticity, a key factor in addictive behaviour, depends on these evidences indicate that KYNA has a role modulating glutamatergic
events of long-term potentiation or long-term depression involving neurotransmission and therefore potentially altering several aspects of
NMDA receptors on dopaminergic neurons which ultimately generate addictive processes.
modifications in the neural circuitry that lead to compulsive consump- In addition to acting at receptors involved in glutamatergic signal-
tion of substances of abuse (Lüscher & Malenka, 2011; Zweifel, Argilli, ling, KYNA, along with KYN, acts as an agonist of the AhR receptor
Bonci, & Palmiter, 2008). (DiNatale et al., 2010), a cytoplasmic receptor of pollutants or xenobi-
Additionally, despite its need of definitive validation, the role of otics which impacts on neuronal proliferation, differentiation, and sur-
KYNA as a negative allosteric modulator of α7nAChRs is getting vival (Suzuki, Hidaka, Kumagai, & Yamamoto, 2020) and could

5
N. Morales-Puerto, P. Giménez-Gómez, M. Pérez-Hernández et al. Pharmacology & Therapeutics 223 (2021) 107807

therefore also have an impact on the development of drug addiction. rectifying potassium channels located in VTA dopaminergic neurons,
AhR has been proposed as a cellular sensor of changes in the cellular mi- producing an increase in VTA cell firing. Moreover, glutamatergic and
lieu and as such its participates in several biological processes highly rel- GABAergic synapses are also altered: EtOH acts reducing the activity
evant to tissue homeostasis, but it has also been implicated in the of γ-aminobutyric acid (GABA) interneurons and increasing the activity
development of a broad of pathological conditions (Bock, 2020). Addi- of glutamate inputs on VTA dopaminergic neurons (Fig. 1) (Juarez &
tionally, gut microbiota produces a broad spectrum of metabolic prod- Han, 2016).
ucts that interact with the host organism, among them TRP
metabolites (Rooks & Garrett, 2016), which can act as low-affinity 5.1. Changes in kynurenine pathway associated with ethanol
AhR ligands (Dvořák, Sokol, & Mani, 2020). Finally, dietary products administration
such as 3,3′-diindolylmethane, a natural product from the Brassica
genus with therapeutic potential, acts as a positive modulator of AhR Exposure to EtOH has been repeatedly studied in the context of TRP
(Yin, Chen, Mao, Wang, & Chen, 2012). Taken together all these data in- metabolism and, although most of the studies in humans focused on 5-
dicates the promiscuity of this receptor and the complexity of its study. HT (LeMarquand, Pihl, & Benkelfat, 1994), preclinical studies in animals
Although we have thoroughly reviewed the effects of KYNA, since it have established that EtOH administration can modify TDO activity
is a neuromodulator with a large number of targets, we cannot forget leading to KYN accumulation (Badawy, 2002). These studies have also
the role that QA might play in addiction due to its neurotoxic capacity, been validated in humans, demonstrating that various doses of EtOH
nor should other metabolites be overlooked, such as xanthurenic acid produce an elevation in plasmatic KYN levels via liver TDO activation
(XA), and cinnabarinic acid (CA) (Fig. 3) (Lowe et al., 2014). Similar to (Badawy, 2002; Badawy, Doughrty, Marsh-Richard, & Steptoe, 2009).
KYNA, CA also activates the AhR receptor and thus may similarly mod- This effect on the kynurenine pathway has been observed even after
ulate the development of drug addiction. In addition, it is a weak prenatal alcohol exposure in human embryonic stem cell-derived neu-
orthosteric agonist of metabotropic glutamate receptor 4 (mGluR) ral lineages, providing evidence to support the study of the relationship
(Fazio et al., 2012), receptors that are located presynaptically and inhibit between foetal alcohol spectrum disorders and the kynurenine pathway
neurotransmitter release (Nicoletti et al., 2011). Although physiological (Palmer et al., 2012).
brain levels of CA are below those required to activate these receptors, The activation of the kynurenine pathway observed under experi-
levels become detectable under inflammatory conditions (Fazio et al., mental conditions after alcohol exposure can also be found in the few
2012). Thus, under inflammatory conditions produced by several studies conducted in abstinent patients diagnosed with alcohol use dis-
drugs of abuse, CA may activate mGluR4 and restore the homeostasis order, reinforcing the idea that there is a relationship between EtOH
of extracellular glutamate levels which is altered following drug use consumption and this metabolic pathway of TRP. In this sense, in a
(Kalivas et al., 2009). Recently, LSP2-9166, a mGluR4/mGluR7 agonist, study carried out with 169 patients, the results revealed no changes in
has been shown to selectively reduce ethanol (EtOH) consumption, re- plasmatic TRP but KYN levels were augmented. The changes in KYN
inforcing efficacy of EtOH and reacquisition of EtOH self-administration levels were independent of the presence of depression; however an as-
after abstinence (Lebourgeois et al., 2018) suggesting that modulation sociation with duration of abstinence and alcohol use disorder severity
of the metabolite might produce reductions in consumption of drugs. was found (Neupane, Lien, Martinez, Hestad, & Bramness, 2015). An-
XA appears to interact indirectly with group II inhibitory mGluR2 and other study examined TRP metabolism in alcohol-dependent patients
mGluR3 possibly acting as an allosteric agonist. Activation of these re- assessing short-term withdrawal (4 weeks abstinence) and long-term
ceptors, as with mGluR4, could lead to a reduction of extrasynaptic withdrawal (12 weeks abstinence). In line with previous results, levels
levels of glutamate and, thus, negatively regulate brain reward function of TRP remained unchanged, while an elevation of KYN was found in
(Kenny, Boutrel, Gasparini, Koob, & Markou, 2005). In support of this, week 4 which was even greater at week 12 (Gleissenthall et al., 2014).
activation of the receptors elevates intracranial self-stimulation thresh- In the same direction, preclinical studies show that EtOH withdrawal in-
olds and decreases relapse to cocaine-seeking behaviour (Weiss & creases IDO1 activity in the PFC of mice (Dos Santos, Junqueira Ayres, de
Baptista, 2003). In addition, XA may also inhibit glutamatergic transmis- Sousa Pinto, Silveira, et al., 2021). Altogether, these evidences suggest a
sion by blocking transport of L-glutamate into synaptic vesicles thereby possible role of the kynurenine pathway in the mediation of the in-
ultimately reducing the synaptic release of L-glutamate (Bartlett, creased stress sensibility in post-withdrawal alcohol-dependent pa-
Esslinger, Thompson, & Bridges, 1998) and restoring glutamate tients. This imbalance in TRP metabolism can last even after
homeostasis. protracted abstinence as revealed by a recent study in which plasmatic
concentration of KYN was increased while that of KYNA was decreased
5. Alcohol (Vidal et al., 2020). Nonetheless, the relationship between the
kynurenine pathway and EtOH consumption may well be more com-
Alcohol, or EtOH, is the most widely consumed drug in the world. It plex. Recent research has shown that KYNA can be found in the order
has been estimated that harmful use of alcohol produces 3 million of micromolar concentrations in several commercial alcoholic bever-
deaths every year, which represents 1 out of 20 of all deaths in the ages including red wine (Turska et al., 2019), which may also contain
world (5.3%). More than three quarters of these were among men. picolinic acid and QA (Yılmaz & Gökmen, 2020), suggesting that EtOH
These deaths occur relatively early in life; in the 20–39 years age consumption may affect the kynurenine pathway not only through its
group 13.5% of the total deaths are alcohol attributable. Overall, the alteration, but also by affecting substrate availability.
harmful use of alcohol causes more than 5% of the global disease burden. In animal models, our laboratory has studied the effect of voluntary
Its consumption is related to more than 200 diseases and injury condi- consumption of EtOH with the Drinking in the Dark paradigm (a para-
tions. Moreover, the consequences are not only limited to the health digm that has been established as a model of binge drinking) on TRP,
sphere: alcohol abuse brings economic and social losses to the society KYN and 5-HT levels in plasma and in limbic forebrain, a brain area, an-
at large (World Health Organization, 2019a). Pharmacological treat- terior to the optic chiasm excluding the olfactory bulbs, which contains
ments for alcohol use disorder can be sorted into several groups de- the reward system area (Wang, Liu, Harvey-White, Zimmer, & Kunos,
pending on the strategy of action: aversive drugs, such as disulfiram; 2003). Our results showed that there were no changes in TRP, KYN or
anticraving drugs, including naltrexone and nalmefene (which is only 5-HT in either plasma or limbic forebrain, which suggests that this
authorised for EtOH addiction in Europe); and other drugs of unknown model cannot modify TRP metabolism or the kynurenine pathway
mechanism, such as acamprosate (Kranzler & Soyka, 2018; Mutschler, (Giménez-Gómez et al., 2018). In order to further explore the role of
Grosshans, Soyka, & Rösner, 2016). EtOH produces DA release by several EtOH consumption in the kynurenine pathway, we used the Chronic In-
mechanisms. Firstly, EtOH directly binds to G protein-coupled inwardly termittent paradigm, a model validated to produce changes in the

6
N. Morales-Puerto, P. Giménez-Gómez, M. Pérez-Hernández et al. Pharmacology & Therapeutics 223 (2021) 107807

neuroimmune component of chronic EtOH abuse. Our data demonstrate connexions to VTA, in order to reduce their firing and for the latter, fol-
that this model can alter the kynurenine pathway producing an increase lowing withdrawal, antagonism of NMDA receptors. These results to-
in KYN both in plasma and brain. It is the first study showing that EtOH gether highlight the usefulness of manipulating the kynurenine
consumption can modulate the kynurenine pathway in the brain of ro- pathway as a pharmacological tool to modify EtOH consumption and
dents (Giménez-Gómez et al., 2019). In our study, EtOH consumption point to a possible modulator of alcohol drinking behaviour.
produces an alteration in the permeability of the gut and the extravasa- Finally, in the last year a relationship between the emotional deficits
tion of LPS, a proinflammatory compound that induces the activation of caused by ethanol consumption and the kynurenine pathway has been
IDO1 (Wang et al., 2010) and consequently increases the levels of KYN established. EtOH consumption produces mood (Boden & Fergusson,
in brain. Taken together all this evidence indicates that the mechanisms 2011) or anxiety alterations (Kliethermes, 2005). Our group has con-
implicated in EtOH-induced alterations of kynurenine pathway are firmed that the Chronic Intermittent paradigm of EtOH consumption
complex, and at least three ways exist. The first is the direct contribution produces depressive symptomatology in mice via elevation of KYN
of KYN metabolites from alcoholic beverages (Turska et al., 2019). The levels. Moreover, treatment with antibiotics counteracts these effects.
second implicates the regulation of IDO1 by LPS, and this mechanism re- Our data reveal the involvement of the microbiota in the consequences
quires more attention in the future since it is supported by the observa- on mood of EtOH consumption via the alteration of kynurenine path-
tion that EtOH consumption produces extravasation of LPS in both way (Giménez-Gómez et al., 2019). In the same line, it was recently ev-
humans and rodents (Giménez-Gómez et al., 2019; Orio, Alen, Pavón, idenced that an IDO1 inhibitor can reduce emotional deficits caused by
Serrano, & García-Bueno, 2019). Finally, EtOH produces an increase in EtOH addiction (Jiang et al., 2020). Taken together, these data indicate
glucocorticoids (Lu & Richardson, 2014) and it is well-established that that the kynurenine pathway plays a role in the development of mood
glucocorticoids can activate TDO (Badawy, 2017). disorders due to EtOH consumption and open a new door for the treat-
ment of these alcohol-related pathologies.
5.2. Pharmacological modulation of kynurenine pathway and effects on
ethanol use 6. Nicotine

The first evidence of the relationship between the manipulation of According to World Health Organization, more than 1.1 billion peo-
the kynurenine pathway and EtOH was obtained when studying the ple over the age of 15 years smoked tobacco in 2016, which accounts for
link between the consumption of this drug and the hole reflex in ro- more than a fifth of the global population. Tobacco use is a major risk
dents. Intraperitoneal administration of KYNA, XA and picolinic acid at- factor for cardiovascular and respiratory diseases, for over 20 different
tenuated the alterations in the hole reflex produced by low dose EtOH types of cancer and for other pathologies. It is estimated that more
while the intracerebroventricular administration completely abolished than 8 million people suffer a tobacco-related death yearly, most of
them (Table 1) (Lapin, Mizaev, Prakh'e, & Ryzhkov, 1991). them taking place in low or middle income countries (World Health
Furthermore, KYN and KYNA may modulate mechanisms specifi- Organization, 2019b).
cally related to EtOH addiction. Regarding KYN, our laboratory showed Although most of the harm of tobacco smoking comes from the inha-
recently that the exogenous administration of L-kynurenine can reduce lation of smoke and other chemicals present in tobacco preparations,
EtOH consumption in mice under the Drinking in the Dark paradigm the main addictive and psychoactive compound of tobacco is nicotine.
(Giménez-Gómez et al., 2018). Moreover, both metabolites act as strong Nicotine is an agonist of nAChR, which is highly expressed in VTA dopa-
inhibitors of the mammalian liver mitochondrial low Km aldehyde de- minergic neurons. Thus, nicotine can produce an increase in DA release
hydrogenase, a key enzyme in the metabolism of EtOH (Fogaça et al., by their direct effect in VTA dopaminergic neurons. Complementary to
2016a), both in vitro (Badawy & Morgan, 2007) and in vivo (Badawy, this effect, nicotine also acts on GABA interneurons and glutamatergic
Bano, & Steptoe, 2011). In this line, there are evidences that indicate afferents via the nAChR located in these neurons, with the final conse-
that KYNA, 3-hydroxykynurenine (3−HK) and 3-hydroxyanthranilic quence of an increase in DA release via desensitization of GABA neurons
acid administered by intraperitoneal injection to rodents block the ac- and an increased glutamate potentiation (Fig. 1) (Juarez & Han, 2016).
tivity of aldehyde dehydrogenase and this inhibition could induce aver- Pharmacological treatments for smoking cessation include nicotine re-
sion towards EtOH (Table 1). These results imply that it is possible to placement therapy (patch, gums, …) (Wadgave & Nagesh, 2016),
manipulate the kynurenine pathway to reduce EtOH consumption via bupropion (antidepressant) (Tran, Ho, & Varghese Gupta, 2019) and
an aversive mechanism (Badawy et al., 2011), similar to what occurs varenicline (partial nicotinic agonist) (Jordan & Xi, 2018).
with disulfiram (Hald, Jacobsen, & Larsen, 2009). .
Recently, the use of Ro 61–8048, a selective KMO inhibitor, as a phar-
macological tool in order to produce a strong elevation in KYNA has 6.1. Changes in kynurenine pathway associated with nicotine
demonstrated effectiveness in manipulating EtOH consumption or the administration
reinstatement of EtOH. Cue-induced reinstatement can be abolished
by administration of Ro 61–8048 (Table 1) (Vengeliene, Cannella, In the early 2000's, several studies analysed the effect of nicotine ex-
Takahashi, & Spanagel, 2016). In the same line, our group has demon- posure on KYNA concentration in the brain of experimental animals.
strated that Ro 61–8048 can reduce EtOH consumption in the Drinking Prolonged nicotine administration caused significant changes in KYNA
in the Dark model. Ro 61–8048 prevented EtOH-induced DA release in levels. These nicotine-induced changes occurred as period-of-expo-
the NAc shell thanks to its ability to increase KYNA concentration in sure-dependent fluctuations of KYNA concentration, where KYNA levels
brain, acting as a negative allosteric modulator of α7nAChRs. The use decreased when the time of exposure was 4, 5 or 6 days, whereas longer
of PNU120596, a selective positive allosteric modulator of α7nAChRs, treatments (a 10- or 15-day treatment) caused an increment in KYNA.
allowed us to verify the involvement of the named receptors, since its The effect of nicotine on KYNA concentration was also dose-
administration prevented the effects of Ro 61–8048 (Table 1) (Fig. 4) dependent and non-region-specific, since similar effects were observed
(Giménez-Gómez et al., 2018), an outcome consistent with those ob- in different brain areas, i.e. hippocampus, striatum and frontal cortex.
served in studies with other abuse substances by other groups However, these changes were not accompanied by alterations on
(Justinova et al., 2013; Secci et al., 2017; Vengeliene et al., 2016). The KYNA serum levels nor KYN brain and serum levels (Hilmas et al.,
exact mechanism by which an elevation in KYNA can reduce EtOH 2001; Rassoulpour, Wu, Ferre, & Schwarcz, 2005). These results were
self-administration and its reinstatement needs more investigation to corroborated in in vitro experiments and, using several nAChR inhibi-
be clearly elucidated, but the most promising hypotheses appear to be tors, ruled out the involvement of the direct activation of nAChR by nic-
for the former, modulation of the α7nAChRs in glutamatergic otine administration in the KYNA increase. Moreover, they suggested an

7
N. Morales-Puerto, P. Giménez-Gómez, M. Pérez-Hernández et al. Pharmacology & Therapeutics 223 (2021) 107807

Table 1
Modulation of the kynurenine pathway and effects associated with drug abuse. ALDH: aldehyde dehydrogenase; EtOH: ethanol; KMO: kynurenine 3-monooxygenase..

Compound Structure Function Use related to drug abuse

Kynurenine Metabolite of the kynurenine pathway • Reduction of nicotine-evoked glutamatergic


signalling (Koshy Cherian et al., 2014)
• Reduction of EtOH consumption
(Giménez-Gómez et al., 2018)

Kynurenic acid Metabolite of the kynurenine pathway • Prevention of the effect of EtOH in the
hole reflex (Lapin et al., 1991)
• Blockade of the activity of ALDH
(aversion to EtOH) (Badawy et al., 2011)

3-Hydroxykynurenine Metabolite of the kynurenine pathway • Blockade of the activity of ALDH


(Aversion to EtOH) (Badawy et al., 2011)

3-Hydroxyanthranilic acid Metabolite of the kynurenine pathway • Blockade of the activity of ALDH
(Aversion to EtOH) (Badawy et al., 2011)

Picolinic acid Metabolite of the kynurenine pathway • Prevention of the effect of EtOH in the
hole reflex (Lapin et al., 1991)

Xanthurenic acid Metabolite of the kynurenine pathway • Prevention of the effect of EtOH in the
hole reflex (Lapin et al., 1991)

Ro 61–8048 KMO inhibitor • Prevention of cannabinoid abuse


(Justinova et al., 2013)
• Abolition of EtOH cue-induced reinstatement
(Vengeliene et al., 2016)
• Reduction of EtOH consumption
(Giménez-Gómez et al., 2018)

enhancement in KAT I and KAT II activities as responsible for the change PNU120596 to modulate the activity of α7nAChRs, the effects of Ro
in KYNA production, but the exact mechanism is unknown (Zielińska, 61–8048 on reinforcement and relapse were not observed, although
Kuc, Zgrajka, Turski, & Dekundy, 2009). these findings did not agree across different animal species (Fig. 4)
Not much has been studied regarding nicotine addiction and (Secci et al., 2017). The mechanism through which an elevation in
kynurenine pathway alterations in humans. In 2006, it was determined KYNA levels can reduce nicotine self-administration involves gluta-
that serum IDO activity was lower in smokers than in non-smokers. This matergic connections from the cortex to VTA and NAc. When nicotine
effect was strong and immediate but of short duration, given that the ac- produces an increase in glutamatergic firing in these connections,
tivities were similar in non-smokers and smokers that had not smoked KYNA may act as an allosteric negative modulator of the α7nAChR in
in the two days previous to the study (Pertovaara et al., 2006). these glutamatergic neurons, therefore blocking the effect of nicotine
on the reward system. We should not, however, overlook the participa-
6.2. Pharmacological modulation of kynurenine pathway and effects on tion of NMDA receptor antagonism by KYNA in VTA dopaminergic neu-
nicotine use rons, which would make these neurons unresponsive to the
glutamatergic firing induced by nicotine.
KMO has been the target of several studies that aim to reduce nico- A recent study proposed modulation of KMO gene expression as a
tine consumption. Pharmacological inhibition of KMO diminishes the therapeutic target for smoking initiation and dependence. Bioinformat-
reinforcing effects of nicotine in animal models. Ro 61–8048 succeeded ics analysis identified a non-coding RNA region which could be used as a
in reducing nicotine self-administration at different doses, as well as genetic tool to modulate KMO expression. This non-coding RNA region
relapse-like effects due to reexposure to nicotine. nAChR seemed to be encodes for two microRNA, has-miR-5096 and has-miR-1285-3p, both
involved in the mechanism of action of Ro 61–8048. When using of which bind to a regulatory region of the KMO mRNA. Downregulation

8
N. Morales-Puerto, P. Giménez-Gómez, M. Pérez-Hernández et al. Pharmacology & Therapeutics 223 (2021) 107807

Fig. 4. Mechanism of DA release inhibition by Ro 61–8048. The risk of addiction of a given drug of abuse depends on the release of DA in the NAc from projection neurons located in the VTA.
Several studies using different drugs, such as EtOH, nicotine, cannabis or cocaine, have aimed to elucidate the mechanism by which Ro 61–8048 is able to prevent or reduce some of the
drug-related behaviours (self-administration, relapse-like effects, …). The most popular explanation involves the inhibition of α7nAChR by KYNA. Ro 61–8048 binds in a non-competitive
manner to KMO enzyme, causing an allosteric change that blocks the metabolism of KYN towards 3-HK. In this situation, KYN is mainly converted by KATs to KYNA, increasing significantly
its concentration. The antagonism of α7nAChR by KYNA causes an interruption of glutamatergic signalling from cortex to VTA. As a result, dopaminergic neurons in VTA stop firing and DA
is not released in the NAc, thus ceasing the activation of the reward circuits induced by drug exposure. 3-HK: 3-hydroxykynurenine; α7nAChR: α7 nicotinic acetylcholine receptor; KAT:
kynurenine aminotransferase; KMO: kynurenine monooxygenase; KYN: kynurenine; KYNA: kynurenic acid; NAc: nucleus accumbens; VTA: ventral tegmental area.

of KMO via microRNA would result in an increase of KYNA levels, thus upregulation of KYNA occurs and would be an alternative approach
promoting α7nAChR inhibition (Aziz, Abdel-Salam, Al-Obaide, Alobydi, to restoring NMDA receptor function in glutamatergic neurons. In sup-
& Al-Humaish, 2018). port of this, a study with Wistar male rats showed that decreasing
The link between nicotine and the kynurenine pathway, in particular KYNA levels via inhibition of KAT II enzyme with PF-04859989, a po-
KYNA, is especially interesting in the context of schizophrenia. Schizo- tent KAT II inhibitor, restored KYN or nicotine administration-evoked
phrenia is a complex neuropsychiatric disorder with patients often glutamate release events in the PFC. Although these experiments did
exhibiting a high degree of cognitive impairment whose underlying not aim to determine the targeted receptors for the observed effects,
cause is thought to involve glutamatergic dysfunction. Schizophrenic the authors, based on previous work in which they observed that
patients also have elevated KYNA brain concentration compared nicotine-evoked glutamatergic transients were unaffected by knock-
with control subjects (Erhardt et al., 2001; Linderholm et al., 2012) out of this nAChR, argue that, while representing a believable explana-
and increased KYNA levels have been linked to cognitive deficits in tion, inhibition of α7nAChR may not be the main system involved in
animal models (Akagbosu et al., 2012). Thus, it has been proposed the presented results, but rather interference of KYNA on the activity
that the elevated KYNA concentration produces aberrant glutamater- of ionotropic glutamate receptors appears a more likely target
gic signalling. Interestingly, schizophrenic patients exhibit a high prev- (Koshy Cherian et al., 2014; Parikh, Ji, Decker, & Sarter, 2010). This hy-
alence of nicotine abuse (Hughes, Hatsukami, Mitchell, & Dahlgren, pothesis is consistent with evidence obtained a decade before, where
1986). A plausible explanation for nicotine use by schizophrenic pa- electrophysiological experiments on Sprague-Dawley rats demon-
tients might be that, given the antagonism of KYNA on α7nAChR, nic- strated that the inhibitory effect of increased KYNA levels on
otine consumption would enhance the activity of these receptors in nicotine-evoked VTA dopaminergic neuron increase in firing was
glutamatergic neurons, thereby alleviating the glutamatergic dysfunc- prevented by the partial agonist at the glycine site of the NMDA re-
tion. Thus, KYNA and the pro-cognitive effects of nicotine can be pro- ceptor, D-cycloserine (Erhardt, Schwieler, & Engberg, 2002).
posed as a link between these two pathologies. Decreasing KYNA Finally, a recent study has shown results that may speak in favour of
levels could be a therapeutic target in pathologies where an the synergy between the activity of these two receptors (Phenis et al.,

9
N. Morales-Puerto, P. Giménez-Gómez, M. Pérez-Hernández et al. Pharmacology & Therapeutics 223 (2021) 107807

2020). Administration of an acute injection of KYN reduced the On the effects of THC on the CNS, experiments with Wistar rats
performance of adult rats in a working memory task, while the showed that KYNA levels in the PFC of adult offspring of dams treated
co-administration of positive allosteric modulators of nAChR succeeded with THC during the last week of pregnancy were elevated, and an
in maintaining normal levels of performance in this test while a NMDA increase in KAT I mRNA levels was also found. The same work de-
agonist did not. However, when exposed to NMDA antagonist 7-Chloro- monstrated that administration of KYN during adulthood after prenatal
KYNA, deficits in the working memory task were also observed. Hence, exposure to THC exacerbated the increase in KYNA concentration in the
these results highlight the involvement of both nAChR and NMDA re- PFC compared with control animals. The observed changes occurred
ceptors in cognitive deficits induced by increases in KYNA levels. specifically in the adult animal, given that fetal and maternal plasma
levels of KYNA and KAT I and KAT II mRNA remained unaltered after
7. Cannabis prenatal administration of THC (Beggiato, Ieraci, Tomasini, Schwarcz,
& Ferraro, 2020).
In several countries, cannabis is the most abused illicit drug,
both among the general and young population. In 2016, 192 million 7.2. Pharmacological modulation of kynurenine pathway and effects on
people used it at least once in the previous year. Perception of low cannabis or THC use
risk of harm in addition to easy access makes cannabis the most
common substance initiated in adolescent years. In fact, from the Pharmacological modulation of brain KYNA levels by using KMO
aforementioned 192 million cannabis users, 13.8 million were inhibitors may become an effective treatment for marijuana depen-
aged 15–16. Cannabis use is usually concomitant with and also dence. A study in Sprague-Dawley rats showed that systemic admin-
the gateway to other drugs of abuse (World Health Organization, istration of Ro 61–8048 caused an elevation of KYNA levels in VTA
2019b). and NAc, resulting in a blockade of THC-induced DA release in
Cannabis, also commonly known as marijuana, is obtained from the these brain regions. In behavioral experiments with rats and mon-
Cannabis plant, endemic to Asia but nowadays cultivated worldwide. keys, self-administration of the synthetic cannabinoid WIN
Tetrahydrocannabinol (THC) is the main psychoactive compound in 55,212–2 decreased significantly after Ro 61–8048 treatment. A sim-
cannabis and is a partial agonist of the cannabinoid receptor 1 (CB1), ilar effect was observed in the rewarding effects of THC. Ro 61–8048
which mediates its psychoactive effects (Fogaça et al., 2016a). Cannabis administration also avoided relapse to THC use after re-exposure to
acts producing an increase in DA release in NAc by at least two mecha- THC or WIN 55,212–2. The actions of Ro 61–8048 were achieved
nisms. The first one involves an increase in DA neural firing by decreas- through negative modulation of α7nAChR via increase of KYNA
ing GABAergic inhibition of dopaminergic neurons, a mechanism in levels, given that the administration of PNU120596 prevented this
which the CB1 receptors are implicated (Oleson & Cheer, 2012). Second- effect (Table 1) (Fig. 4) (Justinova et al., 2013). As we described
ary to that, cannabinoids such as THC produce the activation of excit- above, THC can increase the release of DA via an increase in the firing
atory glutamatergic pyramidal neurons that project from PFC to the of glutamatergic neurons that project to both VTA and NAc. In this
VTA and NAc (Fig. 1) (Pistis, Muntoni, Pillolla, & Gessa, 2002; Pistis, sense, KYNA can block the activity of these glutamatergic neurons
Porcu, Melis, Diana, & Gessa, 2001). via its effect on α7nAChR and finally produce a decrease in DA re-
lease in NAc. Likewise, similar to the case of nicotine, KYNA could
7.1. Changes in kynurenine pathway associated with cannabis or THC also be acting through inhibition of NMDA receptors on dopaminer-
administration gic neurons, blocking its firing.
Glutamate plays a central role in the mechanism by which Ro
Cannabis active metabolites influence several steps of TRP metabo- 61–8048 exerts its effects on marijuana cessation. In experiments with
lism. In 1972, it was demonstrated that cannabis extract, as well as primary astrocyte cultures of rat cerebral cortex, the addition of either
pure THC, could increase the activity of two hepatic enzymes of rat, rimonabant (CB1 receptor antagonist) or nanomolar concentrations of
one of them being TDO (Poddar & Ghosh, 1972). Later in 1974, the KYNA to the culture medium managed to prevent THC-induced in-
same research group pointed out that cannabidiol (CBD, non- creases in extracellular glutamate levels. Furthermore, in in vivo studies
psychoactive cannabis compound) did not increase TDO activity per with rats, Ro 61–8048 also inhibited THC-induced glutamate release in
se, but in combination with THC potentiated the effect of the latter the rat reward-related brain areas, namely VTA, NAc and medial PFC,
(Poddar, Bhattacharyya, & Ghosh, 1974). thus preventing signalling among these brain regions (Secci et al.,
THC and CBD might exert a biphasic stimulation of TRP degradation 2019).
towards KYN by IDO in immune cells (Jenny, Santer, Pirich, Schennach,
& Fuchs, 2009; Jenny, Schröcksnadel, Überall, & Fuchs, 2010). At 8. Amphetamine and its derivatives
nanomolar concentrations of both THC and CBD, activation of CB1- and
CB2-receptor increased IDO activity in peripheral blood mononuclear The group of amphetamines comprises a series of compounds,
cells. On the other hand, micromolar concentrations of these cannabi- including amphetamine itself and substituted amphetamines,
noids down-regulated TRP metabolism, with the effect of CBD being such as methamphetamine and MDMA, among others. This chem-
greater. This effect suggests an implication of cannabinoids in the regu- ically diverse group of drugs acts directly in the CNS and has as pri-
lation of 5-HT levels, due to the increment of circulating TRP (Jenny mary targets the transporters of monoamine neurotransmitters,
et al., 2010). i.e. DA and 5-HT (Sitte & Freissmuth, 2015). According to the
Regarding the nervous system, di Giacomo et al. (2020a) studied the World Health Organization Report, major markets for metham-
effect of CBD and cannabigerol on the hypothalamic cell line Hypo-E22 phetamine continue to grow and it is currently the second greatest
under oxidative stress due to exposure to H2O2. They found that both drug threat in the US, after heroin. Another harmful feature of
CBD and cannabigerol presented neuroprotective effects regarded as a the amphetamines is its dynamic market: each year, hundreds of
reduction of the ratio 3-HK/KYNA, suggesting a blockade of the neuro- new synthetic amphetamine-like substances are synthesised and
toxic branch of the kynurenine pathway. The same authors later dem- added to the traffic (World Health Organization, 2019b). Interest-
onstrated that cultured PFC tissue treated with CBD, after an ingly, some compounds of this group, such as MDMA or amphet-
excitotoxic depolarizing stimulus, was protected against neurotoxicity amine have been, or even still are, used to treat some disorders,
due to increased 3-HK levels, at the same time as the decrease in such as attention deficit and hyperactivity disorder or narcolepsy
KYNA levels was reverted, while cannabigerol did not prevent these ef- (Heal, Smith, Gosden, & Nutt, 2013). In fact, MDMA has recently
fects (di Giacomo et al., 2020b). been proposed as a new pharmacotherapy for refractory post-

10
N. Morales-Puerto, P. Giménez-Gómez, M. Pérez-Hernández et al. Pharmacology & Therapeutics 223 (2021) 107807

traumatic stress disorder (Mithoefer, Grob, & Brewerton, 2016). identifying a relationship between the endogenous KYNA levels in the
Amphetamines alter DA release by two main mechanisms: the striatum and NMDA receptor-mediated excitotoxic events (Poeggeler
first one involves the binding to the dopamine transporter, pro- et al., 2007).
ducing an inhibition of DA reuptake and increasing the extracellu- In another report, the authors demonstrated that subchronic pre-
lar concentration of DA in NAc; the second one is exclusive of treatment with KYN, which induced an increase in KYNA levels, poten-
the amphetamines, where DA release is achieved through dopa- tiated the D-amphetamine-induced DA release in the rat NAc and
mine transporter-mediated reverse transport (Fig. 1) (Dela Peña, reduced its inhibitory effect on firing rate and burst firing activity of
Gevorkiana, & Shi, 2015). VTA dopaminergic neurons (Olsson et al., 2009). This pharmacological
approach was also tested in mouse to study the relationship between
8.1. Changes in kynurenine pathway associated with administration of am- KYNA concentration and the locomotor response induced by D-
phetamine and its derivatives amphetamine, showing that subchronic pretreatment with KYN pro-
duced a potentiation of D-amphetamine-induced locomotor activity,
D-amphetamine is one of the drugs that has been widely used to consistent with the results mentioned above (Olsson, Larsson, &
study the kynurenine pathway functions in the brain. Acute administra- Erhardt, 2012). In line with these observations, Kmo−/− mouse model
tion of D-amphetamine induces a significant dose-dependent short- injected with D-amphetamine showed a potentiation of the horizontal
term decrease of KYNA concentration in the striatum of rat brain. This activity induced by the drug (Erhardt et al., 2017). However, these ef-
effect is noticeable not only in adult rat brain, but also during the post- fects were not observed after acute KYN pretreatment, suggesting that
natal period (Poeggeler, Rassoulpour, Guidetti, Wu, & Schwarcz, 1998; subchronic elevated KYNA levels exposure may induce some adaptive
Rassoulpour, Wu, Poeggeler, & Schwarcz, 1998). The same authors ob- mechanisms such as changes in axonal transport or α7nAChR expres-
served that the D-amphetamine-induced reduction in KYNA levels sion (Olsson et al., 2009, 2012). All together, these works highlight the
was prevented in postnatal and adult rat striatum by using the D2 re- potential implications of the regulation of KYNA concentration on D-
ceptor antagonist haloperidol, suggesting a potential role of D- amphetamine-induced dopaminergic effects in striatum but to this
amphetamine-induced striatal DA changes in the regulation of KYNA day, there is no evidence of the effect of pharmacological modulation
concentration (Poeggeler et al., 1998). Moreover, this decrease in of the kynurenine pathway on amphetamine consumption.
KYNA concentration also occurred in other brain regions such as cortex, Regarding MDMA, we recently demonstrated in rats that enzymatic
hippocampus and cerebellum. However, no changes in other TDO activity in the liver is increased three hours after MDMA adminis-
kynurenine pathway metabolites such as KYN, 3-HK nor QA concentra- tration, resulting in elevated concentrations of plasma KYN and, since
tions were detected in striatum after D-amphetamine injection KYN can cross the BBB, the increase observed in periphery was also
(Rassoulpour et al., 1998). found in several brain regions, such as hippocampus. In addition, we
Regarding the periphery, acute D-amphetamine treatment does not also observed increased concentrations of hippocampal TRP and a ten-
seem to induce any short-term changes in KYN or KYNA in rat liver dency to increase in plasma. We speculate with the possibility that
(Rassoulpour et al., 1998). Yet, a recent study in mice showed that re- MDMA increases the availability of free plasma TRP, which can subse-
peated administration of D-amphetamine over an 11-day period in- quently cross the BBB, which could explain the increase observed in
duced a decrease in alpha-aminoadipic acid, a substrate for KAT-II that rat brain. MDMA acts inhibiting tryptophan hydroxylase (O'Shea et al.,
can dictate the availability of KAT-II for the transamination of KYN to 2006), producing a decrease in 5-HT and an increase in TRP, the sub-
KYNA (Schwarcz et al., 2012a, 2012b), and consequently produces a de- strate for TDO, an enzyme whose activity can be regulated by the pres-
crease in KYN plasma levels (Vanaveski et al., 2018). Moreover, it has ence of substrate (Bender, 1983) producing finally an increase in KYN
been described that amphetamine is a competitive concentration- levels.
dependent inhibitor of the activity of KAT and kynurenine hydrolase We further demonstrated that TDO inhibitor 680C91 potentiates the
liver enzymes (Moussa, El-Ezaby, & Farid, 1978). However, the apparent long-term serotonergic neurotoxicity induced by a neurotoxic dose of
inconsistency of some of the results presented in these reports may be MDMA while, on the contrary, L-kynurenine administered together
explained by the use of different D-amphetamine concentrations, the with probenecid attenuates the brain damage induced by MDMA
disparate drug administration protocols (from acute to chronic admin- (Abuin et al., submitted for publication).
istration) or the intrinsic differences between rat and mouse This study also revealed that modulating AhR activity by using
kynurenine pathway activity (Murakami & Saito, 2013). antagonists (i.e. CH-223191) or positive modulators, such as 3, 3′-
Little is known about the effect of amphetamine derivatives on the diindolylmethane, leads to a potentiation or attenuation of MDMA-
kynurenine pathway. Concerning MDMA, a randomized controlled induced toxicity, respectively. Taking into account that KYN and KYNA
study of healthy subjects showed no changes on KYN and TRP plasma are agonists of AhR (Opitz et al., 2011), these results suggest a role of
concentration after a single dose during the first 24 h after exposure KYN and presumably KYNA in the mechanism of action underlying
(Boxler, Liechti, Schmid, Kraemer, & Steuer, 2017). However, studies MDMA-induced toxicity. Thus, an increase in KYN levels or the adminis-
in rat carried out by our group showed that a neurotoxic dose of tration of 3, 3′-diindolylmethane would produce the translocation of
MDMA induced a short-term increase of TRP in hippocampus while AhR to the nucleus counteracting the MDMA-induced toxicity, but
KYN was elevated both in hippocampus and plasma, an effect mediated more research is needed in order to characterise the exact role of AhR
by activation of the liver enzyme TDO (Abuin et al., submitted for in the MDMA-induced toxicity.
publication). Considering the moderate rise in the recreational use of these drugs
and the renewed interest of some of these drugs as a therapeutic strat-
8.2. Pharmacological modulation of kynurenine pathway and effects on egy to treat different neurological disorders, the pharmacological mod-
amphetamine and its derivatives use ulation of kynurenine pathway may be an interesting approach to
prevent harmful events derived of the administration of D-
Several approaches have been used to determine the role of amphetamine and its derivatives.
kynurenine pathway on D-amphetamine effects in CNS, employing
mainly pharmacological and genetic tools. It has been shown that the 9. Cocaine
administration of Ro 61–8048 prevents the D-amphetamine-induced
reduction in KYNA concentration and the increase of NMDA-induced Cocaine is a highly addictive illegal drug which comes from the coca
neuronal vulnerability in striatal tissue of postnatal rats. These effects plant, known for thousands of years (Biondich & Joslin, 2016). Purified
were independent of the striatal DA release induced by the drug, cocaine, which was first isolated more than 100 years ago, has been

11
N. Morales-Puerto, P. Giménez-Gómez, M. Pérez-Hernández et al. Pharmacology & Therapeutics 223 (2021) 107807

traditionally used in the treatment for a variety of diseases and as anaes- induced DA increases in the NAc are mediated via the inhibition of the
thetic to block local pain. Although it was banned when its harmful ef- dopamine transporter on which KYNA or other kynurenine pathway
fects and addictive power were acknowledged in the beginning of the metabolites have no effects (Fig. 1).
1900's, cocaine became popular in the 80's for recreational purposes
among young people in Europe, North America and the producer coun-
10. Opioids: a special case
tries of South America. After a progressive fall at the beginning of the
21st century, global cocaine manufacture reached its highest level in
Opiates are defined as the derivatives of opium alkaloids, the most
history in 2016, with parallel evidence that cocaine use rose from
relevant being morphine, codeine and heroin. The term opioid com-
2013 to 2016. The routes of administration are flexible: consumers
prises compounds of natural, semisynthetic, synthetic and endogenous
can use it nasally, orally, intravenously or by inhalation. It is because
origin that act like morphine, interact with opioid receptors either as ag-
of this that cocaine addiction is a major health issue, not only for the
onists or antagonists and the effects of which are completely
psychological and medical problems, but also, among other aspects,
antagonised by naloxone (Dinis-Oliveira, 2019).
for the spread of infectious diseases, such as AIDS or hepatitis (World
Opioids are effective analgesics in the treatment of pain. However, in
Health Organization, 2019b).
addition to the side effects of these drugs, one of the most serious ad-
Cocaine produces its effects in the CNS by preventing DA reup-
verse consequences is potential opioid abuse and addiction after re-
take via transporter block, thus prolonging neurotransmitter pres-
peated use (Compton, Jones, & Baldwin, 2016). The opioid crisis
ence in the synapsis of the NAc and amplifying the stimulus in the
continues increasing, in part due to the illicit intake of fentanyl and its
mesolimbic reward areas (Fogaça et al., 2016b). Furthermore, it
potent analogues, which has resulted in a greater propensity to over-
also acts producing desensibilization of the D2-autorreceptor
dose (Spencer, Warner, Bastian, Trinidad, & Hedegaard, 2019).
resulting in increased levels of DA in NAc. Finally cocaine can in-
Opioids are a major concern in many countries because of the severe
crease glutamate transmission and decrease GABAergic transmis-
health consequences associated with their use. In 2017, the use of opi-
sion (Fig. 1) (Juarez & Han, 2016).
oids accounted for 66% of the deaths attributed to drug use disorders.
Approximately 53.4 million people used opioids (both illegal and
9.1. Changes in kynurenine pathway associated with cocaine
prescription opioids for non-medical purposes) in 2017, which corre-
administration
sponds to 1.1% of the global population aged 15–64 (World Health
Organization, 2019b). The mechanism by which opioid consumption
Little is known about the consequences of cocaine consumption
produces reward involves activation of opioid receptors in both VTA
on the kynurenine pathway. The first study in the literature using
and NAc. Opioids bind to μ-opioid receptors and decrease GABAergic
18 cocaine-dependent individuals and 10 drug-free controls
cell firing. This decrease disinhibits VTA dopaminergic neurons, increas-
established that there is a relationship between chronic exposure
ing DA release in NAc. Moreover, opioid consumption can also enhance
to cocaine and alteration in TRP metabolism. Despite this study fo-
glutamatergic signalling in VTA dopaminergic neurons (Fig. 1) (Juarez &
cusing on 5-HT metabolism, it also described a reduction in
Han, 2016).
anthranilic acid without alteration in KYN or 3-HK levels (Patkar
et al., 2009). A recent study from our lab using 110 cocaine users
and 65 controls examined for the first time the kynurenine pathway 10.1. Kynurenine pathway and opioids
including metabolites such as QA and KYNA in plasma. As previously
described, there were no changes in TRP or KYN levels, nor in QA, but To our knowledge, no research on the effect of opioids on the
we found a decrease in KYNA levels (Araos et al., 2019). Both results kynurenine pathway has been conducted, but a few studies have looked
together indicate that alterations in kynurenine pathway metabo- at the effects of some opioids on TRP levels. These studies indicate that
lites can occur without finding changes in KYN or TRP. Cocaine pro- morphine addiction may be associated with uptake of TRP from the
duces a decrease in KYNA without affecting the rest of the blood to the brain (Larson & Takemori, 1977; Zaitsu et al., 2014). More-
kynurenine pathway, which suggests a modulation by cocaine of over, a single injection of heroin produces an increase of TRP urine
KAT-II but which requires further study. levels. Therefore, changes in the levels of these peripheral metabolites
may be considered as possible biomarkers of heroin consumption and
9.2. Pharmacological modulation of kynurenine pathway and effects on withdrawal (Zheng et al., 2013).
cocaine use Regarding the possibilities of pharmacological modulation of the
kynurenine pathway as an approach to opioid treatment, the evi-
Once again, redirecting the kynurenine pathway towards KYNA for- dence suggests that activation of the NMDA receptor, of which
mation by manipulation of KMO enzyme has proven to be a useful phar- KYNA is antagonist, could be involved in the development of mor-
macological tool to block cocaine effects. In male Wistar rats, cocaine phine dependence (Trujillo & Akil, 1991) and in the development
relapse-like behaviour was avoided when KMO was inhibited. As with of analgesic tolerance (Marek, Ben-Eliyahu, Vaccarino, & Liebeskind,
alcohol, reestablishment of cue-induced cocaine-seeking conduct was 1991). In this sense, KYNA could act reducing the symptoms that
prevented by intraperitoneal administration of Ro 61–8048 (Table 1) lead to relapse by blocking NMDA receptors. Moreover, a possible
(Vengeliene et al., 2016). There is evidence in literature of increased glu- modulation of opioid dependence by KYNA could take place at the
tamatergic transmission during cocaine withdrawal. The effect of KYNA level of the opioid receptors themselves. Although it appears that
on NMDA receptors may alleviate withdrawal symptoms due to a KYNA has no affinity for any of the opioid receptors, i.e. μ, κ and δ
decrease in glutamate activity and effectively prevent withdrawal. (Zador et al., 2014), KYNA administration altered opioid receptor
Nonetheless, in contrast to the abolishment of THC and nicotine self- signalling by modifying the activity of G proteins coupled to these
administration by Ro 61–8048 treatment, KMO inhibition did not affect receptors by a mechanism that involves the NMDA receptor, since
cocaine self-administration (Table 1) (Justinova et al., 2013; Secci et al., antagonism of the NMDA receptor with MK-801 prevented the
2017). As in the experiments described above with other drugs, KMO KYNA-induced effect (Samavati et al., 2017).
inhibition by Ro 61–8048 treatment shifted the metabolism of the All this evidence suggests that, although there is not much literature
kynurenine pathway leading to a significant increase in plasma KYNA on the relationship between the kynurenine pathway and opioid addic-
concentration, with the expected consequences of either NMDA recep- tion, there may be an opportunity for treatment of this drug use by
tor or α7nAChR inhibition. However, a possible explanation for the lack harnessing the effect that the kynurenine pathway exerts on the modu-
of effect of Ro 61–8048 on cocaine self-administration is that cocaine- lation of pain.

12
N. Morales-Puerto, P. Giménez-Gómez, M. Pérez-Hernández et al. Pharmacology & Therapeutics 223 (2021) 107807

10.2. Pain modulation: a link between the kynurenine pathway and opioids significant source of inhibitory control (Airavaara et al., 2016; Polter,
Barcomb, Tsuda, & Kauer, 2018; Simmons, Petko, & Paladini, 2017;
The pharmacological manipulation of the kynurenine pathway, en- Smith, Vento, Chao, Good, & Jhou, 2019). Since there are NMDA recep-
hancing the activity of KYNA, has been proposed as a possible target in tors located in these GABA interneurons (Fig. 3) (Nugent, Penick, &
the regulation of neuronal hyperexcitability and glutamatergic neuro- Kauer, 2007) and given its role as NMDA receptors antagonist it is pos-
transmission that are involved in pain generation, and the antinociceptive sible that KYNA can modulate GABA interneurons, opening the door to a
actions of KYNA may be achieved through two of its target receptors. The new promising strategy to modulate DA release.
administration of KYN and probenecid inhibits the NMDA receptor in an- The rest of the receptors through which the metabolites of the
imal models of trigeminal activation and sensitization (Knyihár-Csillik kynurenine pathway can exert their effect, however, should not be
et al., 2007), increases KYNA levels in cerebrospinal fluid and prevents neglected. In the future, the role that modulation of the kynurenine
allodynia in a neuropathic pain model (Pineda-Farias et al., 2013). These pathway may play, after the consumption of substances, on other re-
results are consistent with those from studies using direct blockade of ceptors, such as GPR35, should be characterised. This is so since there
NMDA receptors. For instance, sub-anaesthetic doses of ketamine, a is evidence that this receptor can play a relevant role in analgesia
NMDA antagonist, were found to moderately decrease acute pain and (Larigot, Juricek, Dairou, & Coumoul, 2018; Nourbakhsh, Atabaki, &
hyperalgesia in a recent meta-analysis study of human patients Roohbakhsh, 2018). Moreover, regarding AhR, this receptor impacts
(Thompson et al., 2019) and to reduce the aversive response to noxious on neuronal proliferation, differentiation, and survival (Suzuki et al.,
stimuli in rodent chronic pain models by supressing NMDA hyperactivity 2020), key components of the development of addiction to several
(Zhou et al., 2018). drugs (Koob & Volkow, 2016). In this sense, the manipulation of this re-
On the other hand, KMO inhibition decreases neuropathic pain in- ceptor or their ligands opens a new opportunity in order to protect
tensity after chronic constriction injury to the sciatic nerve and potenti- against neurotoxicity caused by drug abuse and drug addiction.
ates the analgesic properties of morphine (Rojewska, Piotrowska, Even more relevant could be the case of mGluR 2/3 and their possi-
Makuch, Przewlocka, & Mika, 2016). More recently, these authors stud- ble modulation by XA. There is evidence implying that this receptor
ied the role of GPR35 in nociceptive transmission and showed that in- family may be involved in pathologies such as anxiety, depression or
trathecal administration of zaprinast, a GPR35 agonist, and KYNA chronic stress-related disorders (Peterlik, Flor, & P., & Uschold-
reduces thermal and tactile hypersensitivity after chronic constriction Schmidt, N., 2015). Moreover, the modulation of these receptors has an-
injury and enhances the effectiveness of morphine and buprenorphine tipsychotic and anti-stress effects (Swanson & Schoepp, 2003). Many of
(Resta et al., 2016; Rojewska, Ciapała, & Mika, 2019; Rojewska, these pathologies occur in the presence of drug use and, conversely,
Ciapała, Piotrowska, Makuch, & Mika, 2018). they can be caused by drug use or abuse. Further, the kynurenine path-
All in all, modulation of the kynurenine pathway may be effective in way is also related to these pathologies (Platten et al., 2019). In this way,
the treatment of pain. Elevations in KYNA via direct infusion of KYN, the kynurenine pathway could play a relevant role in the neuropsychi-
KYNA or inhibiting KMO seems the most appropriate strategy. Its effects atric disorders caused by drug use. This would constitute a new avenue
on NMDA receptors and as an endogenous agonist of GPR35, a receptor to address the relationship between the kynurenine pathway and sub-
whose activation could cause analgesia (Cosi et al., 2011) provide a new stance use, focusing on the consequences of consumption.
opportunity to possibly substitute opioids for compounds that modulate In the future, not only will it be important to characterise the effect
pain through the kynurenine pathway route, thus reducing their poten- of the receptors on which the kynurenine pathway acts. During recent
tial risk of addiction. years, the microbiota-gut-brain axis has been established as a mecha-
nism that can play a relevant role in various mental illness (Zhu et al.,
11. Future prospects and new trends 2017). It is well known that microbiota can regulate TRP availability
for KYN metabolism, with downstream effects on CNS function
Throughout this review we have unravelled the relationship be- (Kennedy, Cryan, Dinan, & Clarke, 2017). These evidences oblige us to
tween the kynurenine pathway and drug consumption. Scientific litera- pay special attention in the coming years to the effect of drugs of
ture indicates that there is an association between the kynurenine abuse on the kynurenine pathway, but also on the intestinal microbiota.
pathway and EtOH, nicotine, cannabis, amphetamines and cocaine. This opens a totally new opportunity to explain, through these mecha-
But even in these cases the characterisation of the kynurenine pathway nisms, key aspects related to drugs of abuse such as the transition
has not been exhaustive. Most studies available on the literature only from voluntary consumption to addiction or consumption-associated
characterised the effect of the drug on KYN as a part of the TRP metab- pathologies. Another strategy would entail the study of the effect of
olism. Only a few studies have focused on KYNA or QA, and there are no early life administration of KYN in order to determine the importance
evidences in the literature about the effect that drug use can have on of this critical window in the addiction processes. It is known that ad-
other metabolites such as anthranilic acid, CA, XA or picolinic acid. Con- ministration of KYN in early life produces an elevation in KYN levels in
sidering that some of these metabolites have neuroactive properties adulthood (Alexander et al., 2013) and affects contextual memory
(Fazio et al., 2015) it is necessary to study the role that drug consump- (Pocivavsek et al., 2014). Evaluating how a pretreatment with KYN or
tion may have on these compounds. In this way, progress could be other compounds of the kynurenine pathway could affect the consump-
made towards a full understanding of the state of the kynurenine path- tion of drugs or other parameters related to addiction in the long term
way in relation to drug use. Moreover, the mechanisms by which drugs could constitute an interesting approach in the future.
of abuse alter the kynurenine pathway are known only for a few drugs, Finally, the most studied pharmacological compound has been Ro
and so further research is necessary to improve our knowledge in this 61–8048. This compound must be tested on the rest of substances
point. with the aim to elucidate whether it is an effective tool to reduce the
Similarly, there is also no evidence of the effect that drug use has on consumption of those substances in which glutamate, through the
altering the targets of the kynurenine pathway. In the field of drug α7nAChR receptors, plays an essential role or if, in addition, we are
abuse, the alleged role of KYNA in the modulation of α7nAChR is un- faced with a proposal for general treatment for addictions. This com-
doubtedly one of the most promising strategies for the future and has pound should especially be assessed in the case of opioids: it is likely
already been shown to be effective in reducing the consumption of var- that modulating pain through this compound can become an alternative
ious drugs via the modulation of DA release. However, GABA interneu- treatment to opioids, thus preventing its consumption and subsequent
rons also play a key role in the regulation of DA release (Bouarab, abuse. Nonetheless, even when most of the experimental evidence on
Thompson, & Polter, 2019). There is evidence in the literature that the functioning of Ro 61–8048 has been obtained in relation to its mod-
local VTA GABA neurons synapse onto VTA DA neurons and provide a ulation of α7nAChR receptors, specifically in the case of EtOH, nicotine

13
N. Morales-Puerto, P. Giménez-Gómez, M. Pérez-Hernández et al. Pharmacology & Therapeutics 223 (2021) 107807

and cannabis, we should not forget that there is a possibility that Badawy, A., Doughrty, D. M., Marsh-Richard, D. M., & Steptoe, A. (2009). Activation of liver
tryptophan pyrrolase mediates the decrease in tryptophan availability to the brain
another mechanism may be involved: the prevention of drug self- after acute alcohol consumption by normal subjects. Alcohol and Alcoholism (Oxford,
administration observed after Ro 61–8048 treatment may be a conse- Oxfordshire) 44, 267–271.
quence of the blockade of NMDA receptors located on VTA dopaminer- Badawy, A., Bano, S., & Steptoe, A. (2011). Tryptophan in alcoholism treatment I:
Kynurenine metabolites inhibit the rat liver mitochondrial low Km aldehyde dehy-
gic neurons due to the increment in KYNA concentration. Future drogenase activity, elevate blood acetaldehyde concentration and induce aversion
research could benefit from elucidating all targets implicated in Ro to alcohol. Alcohol and Alcoholism (Oxford, Oxfordshire) 46, 651–660.
61–8048 mechanism of action. Bartlett, R. D., Esslinger, C. S., Thompson, C. M., & Bridges, R. J. (1998). Substituted quino-
Moreover, it is necessary to approach other therapeutic strategies. lines as inhibitors of l-glutamate transport into synaptic vesicles. Neuropharmacology
37, 839–846.
There is little evidence of the role that IDO1 inhibition can play in rela- Beggiato, S., Ieraci, A., Tomasini, M. C., Schwarcz, R., & Ferraro, L. (2020). Prenatal THC ex-
tion to drug abuse. There are several clinical trials involving IDO1 inhib- posure raises kynurenic acid levels in the prefrontal cortex of adult rats. Progress in
itors studying the role of the kynurenine pathway in pathologies such as Neuropsychopharmacology and Biological Psychiatry 100.
Bender, D. A. (1983). Biochemistry of tryptophan in health and disease. Molecular Aspects
cancer (Platten et al., 2019). The good results of this approach should of Medicine 6, 101–197.
lead addiction research to evaluate these inhibitors in order to charac- Biondich, A. S., & Joslin, J. D. (2016). Coca: The history and medical significance of an an-
terise the role of IDO1 in aspects such as alteration of the immune sys- cient andean tradition. Emergency Medicine International 2016.
tem produced by the consumption of drugs or aspects related to their Birch, P. J., Grossman, C. J., & Hayes, A. G. (1988). Kynurenate and FG9041 have both com-
petitive and non-competitive antagonist actions at excitatory amino acid receptors.
toxicity. All these perspectives together, on the one hand the correct European Journal of Pharmacology 151, 313–315.
characterisation of the kynurenine pathway and, on the other hand, Bock, K. W. (2020). Aryl hydrocarbon receptor (AHR) functions: Balancing opposing pro-
the use of all the available pharmacological tools, may in the future cesses including inflammatory reactions. Biochemical Pharmacology 178.
Boden, J. M., & Fergusson, D. M. (2011). Alcohol and depression. Addiction 106, 906–914.
allow a full understanding of the relationship that exists between the
Bouarab, C., Thompson, B., & Polter, A. M. (2019). VTA GABA neurons at the Interface of
kynurenine pathway and substance abuse. A relationship that we now stress and reward. Frontiers in Neural Circuits 13, 1–14.
only begin to intuit. Boxler, M. I., Liechti, M. E., Schmid, Y., Kraemer, T., & Steuer, A. E. (2017). First time view
on human metabolome changes after a single intake of 3,4-methylenedioxymetham-
phetamine in healthy placebo-controlled subjects. Journal of Proteome Research 16,
3310–3320.
Declaration of Competing Interest Chess, A. C., Simoni, M. K., Alling, T. E., & Bucci, D. J. (2007). Elevations of endogenous
kynurenic acid produce spatial working memory deficits. Schizophrenia Bulletin 33,
797–804.
The authors declare that there are no conflicts of interest. Chiarugi, A., & Moroni, F. (1999). Quinolinic acid formation in immune-activated mice:
Studies with (m-nitrobenzoyl)-alanine (mNBA) and 3,4-dimethoxy-[-N-4-(−3-ni-
Acknowledgements trophenyl) thiazol-2yl]-benzenesulfonamide (Ro 61-8048), two potent and selective
inhibitors of kynurenine hydroxylase. Neuropharmacology 38, 1225–1233.
Compton, W. M., Jones, C. M., & Baldwin, G. T. (2016). Relationship between nonmedical
This study was supported by Ministerio de Ciencia e Innovación prescription-opioid use and heroin use. New England Journal of Medicine 374,
(Grants SAF2013-40592, SAF2016-78864-R and PID2019-105847RB- 154–163.
I00), Ministerio de Sanidad, Consumo y Bienestar Social [Plan Nacional Cosi, C., Mannaioni, G., Cozzi, A., Carl, V., Sili, M., Cavone, L., et al. (2011). G-protein
coupled receptor 35 (GPR35) activation and inflammatory pain: Studies on the
sobre Drogas (PNSD) Grants 2014I015, 2017I017 and 2019I025], antinociceptive effects of kynurenic acid and zaprinast. Neuropharmacology 60,
Instituto de Salud Carlos III Redes Temáticas de Investigación 1227–1231.
Cooperativa en Salud-Red de Trastornos Adictivos (ISCII RETICS-RTA) Dela Peña, I., Gevorkiana, R., & Shi, W. -X. (2015). Psychostimulants affect dopamine
transmission through both dopamine transporter-dependent and independent
Grants RD12/0028/0002 and RD16/0017/0021, and Universidad mechanisms HHS public access. European Journal of Pharmacology 764, 562–570.
Complutense de Madrid Grant 910258. Deora, G. S., Kantham, S., Chan, S., Dighe, S. N., Veliyath, S. K., McColl, G., et al. (2017). Mul-
tifunctional analogs of Kynurenic acid for the treatment of Alzheimer’s disease: Syn-
References thesis, pharmacology, and molecular modeling studies. ACS Chemical Neuroscience 8,
2667–2675.
Airavaara, M., Lahti, L., Haugas, M., Tikker, L., Voutilainen, M. H., Anttila, J., et al. (2016). Di Chiara, G. (1999). Drug addiction as dopamine-dependent associative learning disor-
Differentiation and molecular heterogeneity of inhibitory and excitatory neurons as- der. European Journal of Pharmacology 375, 13–30.
sociated with midbrain dopaminergic nuclei. Development 143, 516–523. di Giacomo, V., Chiavaroli, A., Orlando, G., Cataldi, A., Rapino, M., di Valerio, V., et al.
Akagbosu, C. O., Evans, G. C., Gulick, D., Suckow, R. F., & Bucci, D. J. (2012). Exposure to (2020a). Neuroprotective and neuromodulatory effects induced by cannabidiol and
kynurenic acid during adolescence produces memory deficits in adulthood. cannabigerol in rat hypo-E22 cells and isolated hypothalamus. Antioxidants 9, 1–14.
Schizophrenia Bulletin 38, 769–778. di Giacomo, V., Chiavaroli, A., Recinella, L., Orlando, G., Cataldi, A., Rapino, M., et al.
Albuquerque, E. X., & Schwarcz, R. (2013). Kynurenic acid as an antagonist of α7 nicotinic (2020b). Antioxidant and neuroprotective effects induced by cannabidiol and
acetylcholine receptors in the brain: Facts and challenges. Biochemical Pharmacology cannabigerol in rat CTX-TNA2 astrocytes and isolated cortexes. International Journal
85, 1027–1032. of Molecular Sciences 21, 3575.
Alexander, K. S., Pocivavsek, A., Wu, H. Q., Pershing, M. L., Schwarcz, R., & Bruno, J. P. DiNatale, B. C., Murray, I. A., Schroeder, J. C., Flaveny, C. A., Lahoti, T. S., Laurenzana, E., et al.
(2013). Early developmental elevations of brain kynurenic acid impair cognitive flex- (2010). Kynurenic acid is a potent endogenous aryl hydrocarbon receptor ligand that
ibility in adults: Reversal with galantamine. Neuroscience 238, 19–28. synergistically induces interleukin-6 in the presence of inflammatory signaling.
Alkondon, M., Pereira, E. F. R., Eisenberg, H. M., Kajii, Y., Schwarcz, R., & Albuquerque, E. X. Toxicological Sciences : An Official Journal of the Society of Toxicology 115, 89–97.
(2011). Age dependency of inhibition of α7 nicotinic receptors and tonically active N- Dinis-Oliveira, R. J. (2019). Metabolism and metabolomics of opiates: A long way of foren-
methyl-D-aspartate receptors by endogenously produced kynurenic acid in the brain. sic implications to unravel. Journal of Forensic and Legal Medicine 61, 128–140.
Journal of Pharmacology and Experimental Therapeutics 337, 572–582. Dobelis, P., Staley, K. J., & Cooper, D. C. (2012). Lack of modulation of nicotinic acetylcho-
Araos, P., Vidal, R., O’Shea, E., Pedraz, M., García-Marchena, N., Serrano, A., et al. (2019). line alpha-7 receptor currents by Kynurenic acid in adult hippocampal interneurons.
Serotonin is the main tryptophan metabolite associated with psychiatric comorbidity PLoS One 7, 1–6.
in abstinent cocaine-addicted patients. Scientific Reports 9, 1–11. Dos Santos, L. C., Junqueira Ayres, D. D., de Sousa Pinto, Í. A., Silveira, M. A., & Albino, M. de
Arnaiz-Cot, J. J., González, J. C., Sobrado, M., Baldelli, P., Carbone, E., Gandía, L., et al. (2008). C., Holanda, V. A. D., et al. (2021). Early and late behavioral consequences of ethanol
Allosteric modulation of α7 nicotinic receptors selectively depolarizes hippocampal withdrawal: Focus on brain indoleamine 2,3 dioxygenase activity. Alcohol 90, 1–9.
interneurons, enhancing spontaneous GABAergic transmission. European Journal of Dvořák, Z., Sokol, H., & Mani, S. (2020). Drug mimicry: Promiscuous receptors PXR
Neuroscience 27, 1097–1110. and AhR, and microbial metabolite interactions in the intestine. Trends in
Aziz, H. A., Abdel-Salam, G., Al-Obaide, I., Alobydi, H. W., & Al-Humaish, S. (2018). Pharmacological Sciences 41, 900–908.
Kynurenine 3-monooxygenase gene associated with nicotine initiation and addic- Erhardt, S., Oberg, H., & Engberg, G. (2001). Pharmacologically elevated levels of endoge-
tion: Analysis of novel regulatory features at 5′ and 3’-regions. Frontiers in Genetics nous kynurenic acid prevent nicotine-induced activation of nigral dopamine neurons.
9, 198. Naunyn-Schmiedeberg’s Archives of Pharmacology 363, 21–27.
Badawy, A. (2002). Tryptophan metabolism in alcoholism. Nutrition Research Reviews 15, Erhardt, S., Schwieler, L., & Engberg, G. (2002). Excitatory and inhibitory responses of do-
123–152. pamine neurons in the ventral tegmental area to nicotine. Synapse 43, 227–237.
Badawy, A. (2017). Kynurenine pathway of tryptophan metabolism: Regulatory and func- Erhardt, S., Pocivavsek, A., Repici, M., Liu, X. C., Imbeault, S., Maddison, D., et al. (2017).
tional aspects. International Journal of Tryptophan Research 10, 1–20. Adaptive and behavioral changes in kynurenine 3-monooxygenase knockout mice:
Badawy, A., & Morgan, C. J. (2007). Tryptophan metabolites as potent inhibitors of alde- Relevance to psychotic disorders. Biological Psychiatry 82, 756–765.
hyde dehydrogenase activity and potential alcoholism-aversion therapeutic agents. Everitt, B. J., & Wolf, M. E. (2002). Psychomotor stimulant addiction: A neural systems
International Congress Series 1304, 344–351. perspective. Journal of Neuroscience 22, 3312–3320.

14
N. Morales-Puerto, P. Giménez-Gómez, M. Pérez-Hernández et al. Pharmacology & Therapeutics 223 (2021) 107807

Fazio, F., Lionetto, L., Molinaro, G., Bertrand, H. O., Acher, F., Ngomba, R. T., et al. (2012). Kliethermes, C. L. (2005). Anxiety-like behaviors following chronic ethanol exposure.
Cinnabarinic acid, an endogenous metabolite of the kynurenine pathway, activates Neuroscience and Biobehavioral Reviews 28, 837–850.
type 4 metabotropic glutamate receptors. Molecular Pharmacology 81, 643–656. Knackstedt, L. A., & Kalivas, P. W. (2009). Glutamate and reinstatement. Current Opinion in
Fazio, F., Lionetto, L., Curto, M., Iacovelli, L., Cavallari, M., Zappulla, C., et al. (2015). Pharmacology 9, 59–64.
Xanthurenic acid activates mGlu2/3 metabotropic glutamate receptors and is a po- Knyihár-Csillik, E., Toldi, J., Mihály, A., Krisztin-Péva, B., Chadaide, Z., Németh, H., et al.
tential trait marker for schizophrenia. Scientific Reports 5. (2007). Kynurenine in combination with probenecid mitigates the stimulation-
Fogaça, M. V., Campos, A. C., & Guimarães, F. S. (2016a). Neuropathology of drug addictions induced increase of c-fos immunoreactivity of the rat caudal trigeminal nucleus in
and substance misuse volume 1. Amsterdam: Elsevier. an experimental migraine model. Journal of Neural Transmission 114, 417–421.
Fogaça, M. V., Campos, A. C., & Guimarães, F. S. (2016b). Neuropathology of drug addictions Koob, G. F. (2008). A role for brain stress systems in addiction. Neuron 59, 11–34.
and substance misuse volume 2. Amsterdam: Elsevier. Koob, G. F., & Le Moal, M. (2001). Drug addiction, dysregulation of reward, and allostasis.
Fukui, S., Schwarcz, R., Rapoport, S. I., Takada, Y., & Smith, Q. R. (1991). Blood-brain barrier Neuropsychopharmacology 24, 97–129.
transport of kynurenines: Implications for brain synthesis and metabolism. Journal of Koob, G. F., & Volkow, N. D. (2010). Neurocircuitry of addiction. Neuropsychopharmacology 35,
Neurochemistry 56, 2007–2017. 217–238.
Gagliardi, R. J. (2000). Neuroprotection, excitotoxicicity and NMDA antagonists. Arquivos Koob, G. F., & Volkow, N. D. (2016). Neurobiology of addiction: A neurocircuitry analysis.
de Neuro-Psiquiatria 58, 583–588. The Lancet Psychiatry 3, 760–773.
Gellért, L., Varga, D., Ruszka, M., Toldi, J., Farkas, T., Szatmári, I., et al. (2012). Behavioural Koshy Cherian, A., Gritton, H., Johnson, D. E., Young, D., Kozak, R., & Sarter, M. (2014). A
studies with a newly developed neuroprotective KYNA-amide. Journal of Neural systemically-available kynurenine aminotransferase II (KAT II) inhibitor restores
Transmission 119, 165–172. nicotine-evoked glutamatergic activity in the cortex of rats. Neuropharmacology 82,
Giménez-Gómez, P., Pérez-Hernández, M., Gutiérrez-López, M. D., Vidal, R., Abuin- 41–48.
Martínez, C., O’Shea, E., et al. (2018). Increasing kynurenine brain levels reduces eth- Kranzler, H. R., & Soyka, M. (2018). Diagnosis and pharmacotherapy of alcohol use disor-
anol consumption in mice by inhibiting dopamine release in nucleus accumbens. der a review. JAMA : The Journal of the American Medical Association 320, 815–824.
Neuropharmacology 135, 581–591.
Lapin, I. P., Mizaev, S., Prakh’e, I. B., & Ryzhkov, I. V. (1991). Kynurenine protection against
Giménez-Gómez, P., Pérez-Hernández, M., O’Shea, E., Caso, J. R., Martín-Hernandez, D., ethanol-induced disorders of the burrowing reflex in mice and rats. Zhurnal Vyssheĭ
Cervera, L. A., et al. (2019). Changes in brain kynurenine levels via gut microbiota Nervnoĭ Deiatelnosti Imeni I P Pavlova 41, 551–557.
and gut-barrier disruption induced by chronic ethanol exposure in mice. The FASEB
Larigot, L., Juricek, L., Dairou, J., & Coumoul, X. (2018). AhR signaling pathways and regu-
Journal 33, 12900–12914.
latory functions. Biochimie Open 7, 1–9.
Gleissenthall, G., Geisler, S., Malik, P., Kemmler, G., Benicke, H., Fuchs, D., et al. (2014).
Larkin, P. B., Sathyasaikumar, K. V., Notarangelo, F. M., Funakoshi, H., Nakamura, T.,
Tryptophan metabolism in post-withdrawal alcohol-dependent patients. Alcohol
Schwarcz, R., et al. (2016). Tryptophan 2,3-dioxygenase and indoleamine 2,3-
and Alcoholism 49, 251–255.
dioxygenase 1 make separate, tissue-specific contributions to basal and
Goeden, N., Notarangelo, F. M., Pocivavsek, A., Beggiato, S., Bonnin, A., & Schwarcz, R.
inflammation-induced kynurenine pathway metabolism in mice. Biochimica et
(2017). Prenatal dynamics of kynurenine pathway metabolism in mice: Focus on
Biophysica Acta - General Subjects 1860.
Kynurenic acid. Developmental Neuroscience 39, 519–528.
Larson, A. A., & Takemori, A. E. (1977). Effect of narcotics on the uptake of serotonin pre-
Greenamyre, J. T., & Young, A. B. (1989). Excitatory amino acids and Alzheimer’s disease.
cursors by the rat brain. Journal of Pharmacology and Experimental Therapeutics 200,
Neurobiology of Aging 10, 593–602.
216–223.
Grilli, M., Raiteri, L., Patti, L., Parodi, M., Robino, F., Raiteri, M., et al. (2006). Modulation of
Lebourgeois, S., Vilpoux, C., Jeanblanc, J., Acher, F., Marie, N., Noble, F., et al. (2018). Phar-
the function of presynaptic α7 and non-α7 nicotinic receptors by the tryptophan me-
macological activation of mGlu4 and mGlu7 receptors, by LSP2-9166, reduces ethanol
tabolites, 5-hydroxyindole and kynurenate in mouse brain. British Journal of
consumption and relapse in rat. Neuropharmacology 133, 163–170.
Pharmacology 149, 724–732.
Leklem, J. E. (1971). Quantitative aspects of tryptophan metabolism in humans and other
Guillemin, G. J. (2012). Quinolinic acid: Neurotoxicity. FEBS Journal 279, 1355.
species: A review. The American Journal of Clinical Nutrition 24, 659–672.
Hald, J., Jacobsen, E., & Larsen, V. (2009). The sensitizing effect of tetraethylthiuramdis-
LeMarquand, D., Pihl, R. O., & Benkelfat, C. (1994). Serotonin and alcohol intake, abuse,
ulphide (antabuse) to ethylalcohol. Acta Pharmacologica et Toxicologica 4, 285–296.
and dependence: Clinical evidence. Biological Psychiatry 36, 326–337.
Heal, D. J., Smith, S. L., Gosden, J., & Nutt, D. J. (2013). Amphetamine, past and present - A
Linderholm, K. R., Skogh, E., Olsson, S. K., Dahl, M. -L., Holtze, M., Engberg, G., et al. (2012).
pharmacological and clinical perspective. Journal of Psychopharmacology 27, 479–496.
Increased levels of kynurenine and Kynurenic acid in the CSF of patients with schizo-
Hertelendy, P., Toldi, J., Fülöp, F., & Vécsei, L. (2018). Ischemic stroke and kynurenines:
phrenia. Schizophrenia Bulletin 38, 426–432.
Medicinal chemistry aspects. Current Medicinal Chemistry 25, 5945–5957.
Lopes, C., Pereira, E. F. R., Wu, H. -Q., Purushottamachar, P., Njar, V., Schwarcz, R., et al.
Hilmas, C., Pereira, E. F. R., Alkondon, M., Rassoulpour, A., Schwarcz, R., & Albuquerque, E.
(2007). Competitive antagonism between the nicotinic allosteric potentiating ligand
X. (2001). The brain metabolite Kynurenic acid inhibits α7 nicotinic receptor activity
galantamine and kynurenic acid at alpha7* nicotinic receptors. The Journal of
and increases non-α7 nicotinic receptor expression: Physiopathological implications.
Pharmacology and Experimental Therapeutics 322, 48–58.
The Journal of Neuroscience 21, 7463–7473.
Hughes, J. R., Hatsukami, D. K., Mitchell, J. E., & Dahlgren, L. A. (1986). Prevalence of Lowe, M. M., Mold, J. E., Kanwar, B., Huang, Y., Louie, A., Pollastri, M. P., et al. (2014). Iden-
smoking among psychiatric outpatients. American Journal of Psychiatry 143, 993–997. tification of cinnabarinic acid as a novel endogenous aryl hydrocarbon receptor li-
gand that drives IL-22 production. PLoS One 9.
Ikeda, M., Tsuji, H., Nakamura, S., Ichiyama, A., Nishizuka, Y., & Hayaishi, O. (1965). Studies
on the biosynthesis of nicotinamide adenine dinucleotide. ii. A role of picolinic car- Lu, Y. L., & Richardson, H. N. (2014). Alcohol, stress hormones, and the prefrontal cortex: A
boxylase in the biosynthesis of nicotinamide adenine dinucleotide from tryptophan proposed pathway to the dark side of addiction. Neuroscience 277, 139–151.
in mammals. The Journal of Biological Chemistry 240, 1395–1401. Lüscher, C., & Malenka, R. C. (2011). Drug-evoked synaptic plasticity in addiction: From
Jenny, M., Santer, E., Pirich, E., Schennach, H., & Fuchs, D. (2009). Δ9-tetrahydrocan- molecular changes to circuit remodeling. Neuron 69, 650–663.
nabinol and cannabidiol modulate mitogen-induced tryptophan degradation and Mangas, A., Heredia, M., Riolobos, A., de la Fuente, A., Criado, J. M., Yajeya, J., et al. (2018).
neopterin formation in peripheral blood mononuclear cells in vitro. Journal of Overexpression of kynurenic acid and 3-hydroxyanthranilic acid after rat traumatic
Neuroimmunology 207, 75–82. brain injury. European Journal of Histochemistry 62, 278–284.
Jenny, M., Schröcksnadel, S., Überall, F., & Fuchs, D. (2010). The potential role of cannabi- Marek, P., Ben-Eliyahu, S., Vaccarino, A., & Liebeskind, J. C. (1991). Delayed application of
noids in modulating serotonergic signaling by their influence on tryptophan metab- MK-801 attenuates development of morphine tolerance in rats. Brain Research 558,
olism. Pharmaceuticals 3, 2647–2660. 163–165.
Jiang, X., Lin, Q., Xu, L., Chen, Z., Yan, Q., Chen, L., et al. (2020). Indoleamine-2,3- McFarland, K., & Kalivas, P. W. (2001). The circuitry mediating cocaine-induced reinstate-
dioxygenase mediates emotional deficits by the kynurenine/tryptophan pathway in ment of drug-seeking behavior. Journal of Neuroscience 21, 8655–8663.
the ethanol addiction/withdrawal mouse model. Frontiers in Cellular Neuroscience 14. Melis, M., Spiga, S., & Diana, M. (2005). The dopamine hypothesis of drug addiction:
Jones, I. W., & Wonnacott, S. (2004). Cellular/molecular precise localization of α7 nicotinic hypodopaminergic state. International Review of Neurobiology 63, 101–154.
acetylcholine receptors on glutamatergic axon terminals in the rat ventral tegmental Metz, R., DuHadaway, J. B., Kamasani, U., Laury-Kleintop, L., Muller, A. J., & Prendergast, G.
area. The Journal of Neuroscience 24, 11244–11252. C. (2007). Novel tryptophan catabolic enzyme IDO2 is the preferred biochemical tar-
Jordan, C. J., & Xi, Z. X. (2018). Discovery and development of varenicline for smoking ces- get of the antitumor indoleamine 2,3-dioxygenase inhibitory compound D-1-methyl-
sation. Expert Opinion on Drug Discovery 13, 671–683. tryptophan. Cancer Research 67, 7082–7087.
Juarez, B., & Han, M. H. (2016). Diversity of dopaminergic neural circuits in response to Metz, R., Smith, C., DuHadaway, J. B., Chandler, P., Baban, B., Merlo, L. M. F., et al. (2014).
drug exposure. Neuropsychopharmacology 41, 2424–2446. IDO2 is critical for IDO1-mediated T-cell regulation and exerts a non-redundant func-
Justinova, Z., Mascia, P., Wu, H. -Q., Secci, M. E., Redhi, G. H., Panlilio, L. V., et al. (2013). tion in inflammation. International Immunology 26, 357–367.
Reducing cannabinoid abuse and preventing relapse by enhancing endogenous Mithoefer, M. C., Grob, C. S., & Brewerton, T. D. (2016). Novel psychopharmacological
brain levels of kynurenic acid. Nature Neuroscience 16, 1652–1661. therapies for psychiatric disorders: Psilocybin and MDMA. The Lancet Psychiatry 3,
Kalivas, P. W., LaLumiere, R. T., Knackstedt, L., & Shen, H. (2009). Glutamate transmission 481–488.
in addiction. Neuropharmacology 56, 169–173. Mok, M. H. S., Fricker, A. C., Weil, A., & Kew, J. N. C. (2009). Electrophysiological character-
Kennedy, P. J., Cryan, J. F., Dinan, T. G., & Clarke, G. (2017). Kynurenine pathway metabo- isation of the actions of kynurenic acid at ligand-gated ion channels.
lism and the microbiota-gut-brain axis. Neuropharmacology 112, 399–412. Neuropharmacology 57, 242–249.
Kenny, P. J., Boutrel, B., Gasparini, F., Koob, G. F., & Markou, A. (2005). Metabotropic glu- Moroni, F., Cozzi, A., Sili, M., & Mannaioni, G. (2012). Kynurenic acid: A metabolite with
tamate 5 receptor blockade may attenuate cocaine self-administration by decreasing multiple actions and multiple targets in brain and periphery. Journal of Neural
brain reward function in rats. Psychopharmacology 179, 247–254. Transmission 119, 133–139.
Kessler, M., Terramani, T., Lynch, G., & Baudry, M. (1989). A Glycine site associated with Moussa, N. M., El-Ezaby, M. S., & Farid, S. (1978). Effect of amphetamine on kyunrenine
N-methyl‑d-aspartic acid receptors: Characterization and identification of a new aminotransferase and kynurenine hydrolase activities in normal mouse liver.
class of antagonists. Journal of Neurochemistry 52, 1319–1328. Chemical & Pharmaceutical Bulletin 26, 853–856.

15
N. Morales-Puerto, P. Giménez-Gómez, M. Pérez-Hernández et al. Pharmacology & Therapeutics 223 (2021) 107807

Muneer, A. (2020). Kynurenine pathway of tryptophan metabolism in neuropsy- Poddar, M. K., & Ghosh, J. J. (1972). Effect of cannabis extract, Δ9-tetrahydrocannabinol
chiatric disorders: Pathophysiologic and therapeutic considerations. Clinical and lysergic acid diethylamide on rat liver enzymes. Biochemical Pharmacology 21,
Psychopharmacology and Neuroscience 18, 507–526. 3301–3303.
Murakami, Y., & Saito, K. (2013). Species and cell types difference in tryptophan metabo- Poddar, M. K., Bhattacharyya, K. C., & Ghosh, J. J. (1974). Potentiating effect of cannabidiol
lism. International Journal of Tryptophan Research 6, 47–54. on Δ9-tetrahydrocannabinol-induced changes in hepatic enzymes. Biochemical
Mutschler, J., Grosshans, M., Soyka, M., & Rösner, S. (2016). Current findings and mecha- Pharmacology 23, 758–759.
nisms of action of disulfiram in the treatment of alcohol dependence. Poeggeler, B., Rassoulpour, A., Guidetti, P., Wu, H. Q., & Schwarcz, R. (1998). Dopaminergic
Pharmacopsychiatry 49, 137–141. control of kynurenate levels and N-methyl-D-aspartate toxicity in the developing rat
Nestler, E. J. (2005). The neurobiology of cocaine addiction. Science & Practice Perspectives striatum. Developmental Neuroscience 20, 146–153.
3, 4–10. Poeggeler, B., Rassoulpour, A., Wu, H. Q., Guidetti, P., Roberts, R. C., & Schwarcz, R. (2007).
Neupane, S. P., Lien, L., Martinez, P., Hestad, K., & Bramness, J. G. (2015). The relationship Dopamine receptor activation reveals a novel, kynurenate-sensitive component of
of alcohol use disorders and depressive symptoms to tryptophan metabolism: Cross- striatal N-methyl-d-aspartate neurotoxicity. Neuroscience 148, 188–197.
sectional data from a Nepalese alcohol treatment sample. Alcoholism, Clinical and Polter, A. M., Barcomb, K., Tsuda, A. C., & Kauer, J. A. (2018). Synaptic function and plastic-
Experimental Research 39, 514–521. ity in identified inhibitory inputs onto VTA dopamine neurons. European Journal of
Nicoletti, F., Bockaert, J., Collingridge, G. L., Conn, P. J., Ferraguti, F., Schoepp, D. D., et al. Neuroscience 47, 1208–1218.
(2011). Metabotropic glutamate receptors: From the workbench to the bedside. Ramirez Ortega, D., Ovalle Rodríguez, P., Pineda, B., González Esquivel, D. F., Ramos
Neuropharmacology 60, 1017–1041. Chávez, L. A., Vázquez Cervantes, G. I., et al. (2020). Kynurenine pathway as a new
Nilsson, L. K., Linderholm, K. R., Engberg, G., Paulson, L., Blennow, K., Lindström, L. H., et al. target of cognitive impairment induced by lead toxicity during the lactation.
(2005). Elevated levels of kynurenic acid in the cerebrospinal fluid of male patients Scientific Reports, 10.
with schizophrenia. Schizophrenia Research 80, 315–322. Rassoulpour, A., Wu, H. -Q., Poeggeler, B., & Schwarcz, R. (1998). Systemic d-
Nilsson, L. K., Linderholm, K. R., & Erhardt, S. (2006). Subchronic treatment with amphetamine administration causes a reduction of kynurenic acid levels in rat
kynurenine and probenecid: Effects on prepulse inhibition and firing of midbrain do- brain. Brain Research 802, 111–118.
pamine neurons. Journal of Neural Transmission 113, 557–571. Rassoulpour, A., Wu, H. -Q., Ferre, S., & Schwarcz, R. (2005). Nanomolar concentrations of
Nourbakhsh, F., Atabaki, R., & Roohbakhsh, A. (2018). The role of orphan G protein- kynurenic acid reduce extracellular dopamine levels in the striatum. Journal of
coupled receptors in the modulation of pain: A review. Life Sciences 212, 59–69. Neurochemistry 93, 762–765.
Nugent, F. S., Penick, E. C., & Kauer, J. A. (2007). Opioids block long-term potentiation of Reinhart, P. H., & Kelly, J. W. (2011). Treating the periphery to ameliorate neurodegener-
inhibitory synapses. Nature 446, 1086–1090. ative diseases. Cell 145, 813–814.
Oleson, E. B., & Cheer, J. F. (2012). A brain on cannabinoids: The role of dopamine release Resta, F., Masi, A., Sili, M., Laurino, A., Moroni, F., & Mannaioni, G. (2016). Kynurenic acid
in reward seeking. Cold Spring Harbor Perspectives in Medicine 2. and zaprinast induce analgesia by modulating HCN channels through GPR35 activa-
Olsson, S. K., Andersson, A. S., Linderholm, K. R., Holtze, M., Nilsson-Todd, L. K., & tion. Neuropharmacology 108, 136–143.
Schwieler, et al. (2009). Elevated levels of kynurenic acid change the dopaminergic Rojewska, E., Piotrowska, A., Makuch, W., Przewlocka, B., & Mika, J. (2016). Pharmacolog-
response to amphetamine: Implications for schizophrenia. International Journal of ical kynurenine 3-monooxygenase enzyme inhibition significantly reduces neuro-
Neuropsychopharmacology 12, 501–512. pathic pain in a rat model. Neuropharmacology 102, 80–91.
Olsson, S. K., Larsson, M. K., & Erhardt, S. (2012). Subchronic elevation of brain kynurenic Rojewska, E., Ciapała, K., Piotrowska, A., Makuch, W., & Mika, J. (2018). Pharmacological
acid augments amphetamine-induced locomotor response in mice. Journal of Neural inhibition of indoleamine 2,3-dioxygenase-2 and kynurenine 3-monooxygenase, en-
Transmission 119, 155–163. zymes of the kynurenine pathway, Significantly diminishes neuropathic pain in a rat
Opitz, C. A., Litzenburger, U. M., Sahm, F., Ott, M., Tritschler, I., Trump, S., et al. (2011). An model. Frontiers in Pharmacology 9.
endogenous tumour-promoting ligand of the human aryl hydrocarbon receptor.
Rojewska, E., Ciapała, K., & Mika, J. (2019). Kynurenic acid and zaprinast diminished
Nature 478, 197–203.
CXCL17-evoked pain-related behaviour and enhanced morphine analgesia in a
Orio, L., Alen, F., Pavón, F. J., Serrano, A., & García-Bueno, B. (2019). Oleoylethanolamide,
mouse neuropathic pain model. Pharmacological Reports 71, 139–148.
neuroinflammation, and alcohol abuse. Frontiers in Molecular Neuroscience 11.
Rongvaux, A., Andris, F., Van Gool, F., & Leo, O. (2003). Reconstructing eukaryotic NAD
O’Shea, E., Orio, L., Escobedo, I., Sanchez, V., Camarero, J., Green, A. R., et al. (2006).
metabolism. BioEssays 25, 683–690.
MDMA-induced neurotoxicity: Long-term effects on 5-HT biosynthesis and the influ-
Rooks, M. G., & Garrett, W. S. (2016). Gut microbiota, metabolites and host immunity.
ence of ambient temperature. British Journal of Pharmacology 148, 778–785.
Nature Reviews Immunology 16, 341–352.
Palmer, J. A., Poenitzsch, A. M., Smith, S. M., Conard, K. R., West, P. R., & Cezar, G. G. (2012).
Röver, S., Cesura, A. M., Huguenin, P., Kettler, R., & Szente, A. (1997). Synthesis and bio-
Metabolic biomarkers of prenatal alcohol exposure in human embryonic stem cell-
chemical evaluation of N-(4-phenylthiazol-2-yl)benzenesulfonamides as high-
derived neural lineages. Alcoholism: Clinical and Experimental Research 36,
affinity inhibitors of kynurenine 3-hydroxylase. Journal of Medicinal Chemistry 40,
1314–1324.
4378–4385.
Parikh, V., Ji, J., Decker, M. W., & Sarter, M. (2010). Prefrontal β2 subunit-containing and
Russo, S. J., & Nestler, E. J. (2013). The brain reward circuitry in mood disorders. Nature
α7 nicotinic acetylcholine receptors differentially control glutamatergic and choliner-
Reviews Neuroscience 14, 609–625.
gic signaling. The Journal of Neuroscience 30, 3518–3530.
Patkar, A. A., Rozen, S., Mannelli, P., Matson, W., Pae, C. U., Krishnan, K. R., et al. (2009). Samavati, R., Zádor, F., Szűcs, E., Tuka, B., Martos, D., Veres, G., et al. (2017). Kynurenic acid
Alterations in tryptophan and purine metabolism in cocaine addiction: A and its analogue can alter the opioid receptor G-protein signaling after acute treat-
metabolomic study. Psychopharmacology 206, 479–489. ment via NMDA receptor in rat cortex and striatum. Journal of the Neurological
Perkins, M. N., & Stone, T. W. (1982). An iontophoretic investigation of the actions of con- Sciences 376, 63–70.
vulsant kynurenines and their interaction with the endogenous excitant quinolinic Savitz, J., Drevets, W. C., Wurfel, B. E., Ford, B. N., Bellgowan, P. S. F., Victor, T. A., et al.
acid. Brain Research 247, 184–187. (2015). Reduction of kynurenic acid to quinolinic acid ratio in both the depressed
Pertovaara, M., Heliövaara, M., Raitala, A., Oja, S. S., Knekt, P., & Hurme, M. (2006). The ac- and remitted phases of major depressive disorder. Brain, Behavior, and Immunity 46,
tivity of the immunoregulatory enzyme indoleamine 2,3-dioxygenase is decreased in 55–59.
smokers. Clinical and Experimental Immunology 145, 469–473. Schwarcz, R., Bruno, J. P., Muchowski, P. J., & Wu, H. -Q. (2012a). Kynurenines in the mam-
Peterlik, D., Flor, J., & P., & Uschold-Schmidt, N. (2015). The emerging role of metabotropic malian brain: When physiology meets pathology. Nature Reviews. Neuroscience 13,
glutamate receptors in the pathophysiology of chronic stress-related disorders. 465–477.
Current Neuropharmacology 14, 514–539. Schwarcz, R., Bruno, J. P., Muchowski, P. J., & Wu, H. -Q. (2012b). Kynurenines in the mam-
Phenis, D., Vunck, S. A., Valentini, V., Arias, H., Schwarcz, R., & Bruno, J. P. (2020). Activa- malian brain: When physiology meets pathology. Nature Reviews. Neuroscience 13,
tion of alpha7 nicotinic and NMDA receptors is necessary for performance in a work- 465–477.
ing memory task. Psychopharmacology 237, 1723–1735. Secci, M. E., Auber, A., Panlilio, L. V., Redhi, G. H., Thorndike, E. B., Schindler, C. W., et al.
Pineda-Farias, J. B., Pérez-Severiano, F., González-Esquivel, D. F., Barragán-Iglesias, P., (2017). Attenuating nicotine reinforcement and relapse by enhancing endog-
Bravo-Hernández, M., Cervantes-Durán, C., et al. (2013). The L-kynurenine-probene- enous brain levels of kynurenic acid in rats and squirrel monkeys.
cid combination reduces neuropathic pain in rats. European Journal of Pain 17, Neuropsychopharmacology 42, 1619–1629.
1365–1373. Secci, M. E., Mascia, P., Sagheddu, C., Beggiato, S., Melis, M., Borelli, A. C., et al. (2019). As-
Pistis, M., Porcu, G., Melis, M., Diana, M., & Gessa, G. L. (2001). Effects of cannabinoids on trocytic mechanisms involving kynurenic acid control Δ 9 -tetrahydrocannabinol-in-
prefrontal neuronal responses to ventral tegmental area stimulation. European Jour- duced increases in glutamate release in brain reward-processing areas. Molecular
nal of Neuroscience 14, 96–102. Neurobiology 56, 3563–3575.
Pistis, M., Muntoni, A. L., Pillolla, G., & Gessa, G. L. (2002). Cannabinoids inhibit excitatory Shaham, Y., Shalev, U., Lu, L., De Wit, H., & Stewart, J. (2003). The reinstatement model of
inputs to neurons in the shell of the nucleus accumbens: An in vivo electrophysiolog- drug relapse: History, methodology and major findings. Psychopharmacology 168,
ical study. European Journal of Neuroscience 15, 1795–1802. 3–20.
Platten, M., Nollen, E. A. A., Röhrig, U. F., Fallarino, F., & Opitz, C. A. (2019). Tryptophan me- Shaw, P. J. (1993). Excitatory amino acid receptors, excitotoxicity, and the human nervous
tabolism as a common therapeutic target in cancer, neurodegeneration and beyond. system. Current Opinion in Neurology and Neurosurgery 6, 414–422.
Nature Reviews Drug Discovery 18, 379–401. Shepard, P. D., Joy, B., Clerkin, L., & Schwarcz, R. (2003). Micromolar brain levels of
Pocivavsek, A., Wu, H. Q., Elmer, G. I., Bruno, J. P., & Schwarcz, R. (2012). Pre- and postnatal kynurenic acid are associated with a disruption of auditory sensory gating in the
exposure to kynurenine causes cognitive deficits in adulthood. European Journal of rat. Neuropsychopharmacology 28, 1454–1462.
Neuroscience 35, 1605–1612. Simmons, D. N. V., Petko, A. K., & Paladini, C. A. (2017). Differential expression of long-
Pocivavsek, A., Thomas, M. A. R., Elmer, G. I., Bruno, J. P., & Schwarcz, R. (2014). Continu- term potentiation among identified inhibitory inputs to dopamine neurons. Journal
ous kynurenine administration during the prenatal period, but not during adoles- of Neurophysiology 118, 1998–2008.
cence, causes learning and memory deficits in adult rats. Psychopharmacology 231, Sitte, H. H., & Freissmuth, M. (2015). Amphetamines, new psychoactive drugs and the
2799–2809. monoamine transporter cycle. Trends in Pharmacological Sciences 36, 41–50.

16
N. Morales-Puerto, P. Giménez-Gómez, M. Pérez-Hernández et al. Pharmacology & Therapeutics 223 (2021) 107807

Smith, R. J., Vento, P. J., Chao, Y. S., Good, C. H., & Jhou, T. C. (2019). Gene expression and Wang, J., Simonavicius, N., Wu, X., Swaminath, G., Reagan, J., Tian, H., et al. (2006).
neurochemical characterization of the rostromedial tegmental nucleus (RMTg) in Kynurenic acid as a ligand for orphan G protein-coupled receptor GPR35. Journal of
rats and mice. Brain Structure and Function 224, 219–238. Biological Chemistry 281, 22021–22028.
Spencer, M. R., Warner, M., Bastian, B. A., Trinidad, J. P., & Hedegaard, H. (2019). Drug Wang, Y., Devkota, S., Musch, M. W., Jabri, B., Nagler, C., Antonopoulos, D. A., et al. (2010).
overdose deaths involving fentanyl, 2011-2016. National Vital Statistics Reports 68, Regional mucosa-associated microbiota determine physiological expression of TLR2
1–19. and TLR4 in murine colon. PLoS One 5.
Stone, T. W. (2020). Does kynurenic acid act on nicotinic receptors? An assessment of the Weiss, F., & Baptista, M. (2003). mGlu2/3 agonist (LY379268) selectively reverses condi-
evidence. Journal of Neurochemistry 152, 627–649. tioned reinstatement of cocaine seeking behavior. Behavioural Pharmacology 14.
Stone, T. W., & Darlington, L. G. (2002). Endogenous kynurenines as targets for drug dis- World Health Organization (2019a). Global status report on alcohol and health 2018. Vi-
covery and development. Nature Reviews Drug Discovery 1, 609–620. enna: United Nations.
Stone, T. W., & Perkins, M. N. (1981). Quinolinic acid: A potent endogenous excitant at World Health Organization (2019b). World drug report 2019 – Global overview of drug de-
amino acid receptors in CNS. European Journal of Pharmacology 72, 411–412. mand and supply. Vienna: United Nations.
Stone, T. W., Stoy, N., & Darlington, L. G. (2013). An expanding range of targets for Yamazaki, F., Kuroiwa, T., Takikawa, O., & Kido, R. (1985). Human indolylamine 2,3-
kynurenine metabolites of tryptophan. Trends in Pharmacological Sciences 34, dioxygenase. Its tissue distribution, and characterization of the placental enzyme.
136–143. Biochemical Journal 230, 635–638.
Suzuki, T., Hidaka, T., Kumagai, Y., & Yamamoto, M. (2020). Environmental pollutants and Yılmaz, C., & Gökmen, V. (2020). Formation of amino acid derivatives in white and red
the immune response. Nature Immunology 21, 1486–1495. wines during fermentation: Effects of non-Saccharomyces yeasts and Oenococcus
Swanson, C. J., & Schoepp, D. D. (2003). A role for noradrenergic transmission in the ac- oeni. Food Chemistry, 128415.
tions of phencyclidine and the antipsychotic and antistress effects of mGlu2/3 recep- Yin, X. F., Chen, J., Mao, W., Wang, Y. H., & Chen, M. H. (2012). A selective aryl hydrocarbon
tor agonists. Annals of the New York Academy of Sciences 1003, 309–317. receptor modulator 3,3’-Diindolylmethane inhibits gastric cancer cell growth. Journal
Taber, K. H., Black, D. N., Porrino, L. J., & Hurley, R. A. (2012). Neuroanatomy of dopamine: of Experimental & Clinical Cancer Research 31.
Reward and addiction. Journal of Neuropsychiatry and Clinical Neurosciences 24, 1–4. Zador, F., Samavati, R., Szlavicz, E., Tuka, B., Bojnik, E., Fulop, F., et al. (2014). Inhibition of
Tanaka, M., Bohár, Z., Martos, D., Telegdy, G., & Vécsei, L. (2020). Antidepressant-like ef- opioid receptor mediated G-protein activity after chronic administration of kynurenic
fects of kynurenic acid in a modified forced swim test. Pharmacological Reports 72, acid and its derivative without direct binding to opioid receptors. CNS & Neurological
449–455. Disorders Drug Targets 13, 1520–1529.
Thompson, T., Whiter, F., Gallop, K., Veronese, N., Solmi, M., Newton, P., et al. (2019). Zaitsu, K., Miyawaki, I., Bando, K., Horie, H., Shima, N., Katagi, M., et al. (2014). Metabolic
NMDA receptor antagonists and pain relief: A meta-analysis of experimental trials. profiling of urine and blood plasma in rat models of drug addiction on the basis of
Neurology 92, 1652–1662. morphine, methamphetamine, and cocaine-induced conditioned place preference.
Tran, A. X., Ho, T. T., & Varghese Gupta, S. (2019). Role of CYP2B6 pharmacogenomics in Analytical and Bioanalytical Chemistry 406, 1339–1354.
bupropion-mediated smoking cessation. Journal of Clinical Pharmacy and Zappettini, S., Grilli, M., Olivero, G., Chen, J., Padolecchia, C., Pittaluga, A., et al. (2014). Nic-
Therapeutics 44, 174–179. otinic α7 receptor activation selectively potentiates the function of NMDA receptors
Trujillo, K., & Akil, H. (1991). Inhibition of morphine tolerance and dependence by the in glutamatergic terminals of the nucleus accumbens. Frontiers in Cellular
NMDA receptor antagonist MK-801. Science 251, 85–87. Neuroscience 8.
Turska, M., Rutyna, R., Paluszkiewicz, M., Terlecka, P., Dobrowolski, A., Pelak, J., et al. Zheng, T., Liu, L., Aa, J., Wang, G., Cao, B., Li, M., et al. (2013). Metabolic phenotype of rats
(2019). Presence of kynurenic acid in alcoholic beverages – Is this good news, or exposed to heroin and potential markers of heroin abuse. Drug and Alcohol
bad news? Medical Hypotheses 122, 200–205. Dependence 127, 177–186.
Vanaveski, T., Narvik, J., Innos, J., Philips, M. A., Ottas, A., Plaas, M., et al. (2018). Repeated Zhou, H., Zhang, Q., Martinez, E., Dale, J., Hu, S., Zhang, E., Liu, K., et al. (2018). Ketamine
administration of D-amphetamine induces distinct alterations in behavior and me- reduces aversion in rodent pain models by suppressing hyperactivity of the anterior
tabolite levels in 129Sv and Bl6 mouse strains. Frontiers in Neuroscience 12. cingulate cortex. Nature Communications 9.
Vengeliene, V., Cannella, N., Takahashi, T., & Spanagel, R. (2016). Metabolic shift of the Zhu, X., Han, Y., Du, J., Liu, R., Jin, K., & Yi, W. (2017). Microbiota-gut-brain axis and the
kynurenine pathway impairs alcohol and cocaine seeking and relapse. central nervous system. Oncotarget 8, 53829–53838.
Psychopharmacology 10, 1–11. Zielińska, E., Kuc, D., Zgrajka, W., Turski, W. A., & Dekundy, A. (2009). Long-term exposure
Vidal, R., García-Marchena, N., O’Shea, E., Requena-Ocaña, N., Flores-López, M., Araos, P., to nicotine markedly reduces kynurenic acid in rat brain — In vitro and ex vivo evi-
et al. (2020). Plasma tryptophan and kynurenine pathway metabolites in abstinent dence. Toxicology and Applied Pharmacology 240, 174–179.
patients with alcohol use disorder and high prevalence of psychiatric comorbidity. Zweifel, L. S., Argilli, E., Bonci, A., & Palmiter, R. D. (2008). Role of NMDA receptors in do-
Progress in Neuro-Psychopharmacology and Biological Psychiatry 100. pamine neurons for plasticity and addictive behaviors. Neuron 59(3), 486–496.
Wadgave, U., & Nagesh, L. (2016). Nicotine replacement therapy: An overview. Zwilling, D., Huang, S. Y., Sathyasaikumar, K. V., Notarangelo, F. M., Guidetti, P., Wu, H. Q.,
International Journal of Health Sciences 10, 425–435. et al. (2011). Kynurenine 3-monooxygenase inhibition in blood ameliorates neurode-
Wang, L., Liu, J., Harvey-White, J., Zimmer, A., & Kunos, G. (2003). Endocannabinoid sig- generation. Cell 145, 863–874.
naling via cannabinoid receptor 1 is involved in ethanol preference and its age-
dependent decline in mice. Proceedings of the National Academy of Sciences of the
United States of America 100, 1393–1398.

17

You might also like