You are on page 1of 30

672342

research-article2016
JOP0010.1177/0269881116672342Journal of PsychopharmacologyMillan et al.

Commentary

The frontal cortex as a network


hub controlling mood and cognition:
Probing its neurochemical substrates Journal of Psychopharmacology

for improved therapy of psychiatric and


1­–30
© The Author(s) 2016
Reprints and permissions:
neurological disorders sagepub.co.uk/journalsPermissions.nav
DOI: 10.1177/0269881116672342
jop.sagepub.com

Mark J Millan, Jean-Michel Rivet and Alain Gobert

Abstract
The highly-interconnected and neurochemically-rich frontal cortex plays a crucial role in the regulation of mood and cognition, domains disrupted
in depression and other central nervous system disorders, and it is an important site of action for their therapeutic control. For improving our
understanding of the function and dysfunction of the frontal cortex, and for identifying improved treatments, quantification of extracellular pools of
neuromodulators by microdialysis in freely-moving rodents has proven indispensable. This approach has revealed a complex mesh of autoreceptor and
heteroceptor interactions amongst monoaminergic pathways, and led from selective 5-HT reuptake inhibitors to novel classes of multi-target drugs
for treating depression like the mixed α2-adrenoceptor/5-HT reuptake inhibitor, S35966, and the clinically-launched vortioxetine and vilazodone.
Moreover, integration of non-monoaminergic actions resulted in the discovery and development of the innovative melatonin receptor agonist/5-
HT2C receptor antagonist, Agomelatine. Melatonin levels, like those of corticosterone and the “social hormone”, oxytocin, can now be quantified by
microdialysis over the full 24 h daily cycle. Further, the introduction of procedures for measuring extracellular histamine and acetylcholine has provided
insights into strategies for improving cognition by, for example, blockade of 5-HT6 and/or dopamine D3 receptors. The challenge of concurrently
determining extracellular levels of GABA, glutamate, d-serine, glycine, kynurenate and other amino acids, and of clarifying their interactions with
monoamines, has also been resolved. This has proven important for characterizing the actions of glycine reuptake inhibitors that indirectly augment
transmission at N-methyl-d-aspartate receptors, and of “glutamatergic antidepressants” like ketamine, mGluR5 antagonists and positive modulators
of AMPA receptors (including S47445). Most recently, quantification of the neurotoxic proteins Aβ42 and Tau has extended microdialysis studies to
the pathogenesis of neurodegenerative disorders, and another frontier currently being broached is microRNAs. The present article discusses the above
themes, focusses on recent advances, highlights opportunities for clinical “translation”, and suggests avenues for further progress.

Keywords
Microdialysis, frontal cortex, depression, Alzheimer’s, schizophrenia, antidepressant, antipsychotic, Tau, β-amyloid, monoamine, glutamate, GABA,
mGluR, AMPA, NMDA, ketamine

The frontal cortex as a


therapeutically-important centre
controlling mood and cognition
To improve our understanding of the role of the FCX, and to
The frontal cortex (FCX) is heavily innervated by ascending mono- acquire insights into potentially improved therapies, it is critical
aminergic, cholinergic and histaminergic pathways, contains a rich to characterize interactions amongst the many neuromodulators
population of modulatory GABAergic interneurons, and sends underpinning its complex operations. This enterprise has most
major projections (mainly glutamatergic) to other cortical regions effectively been led by preclinical work in rodents. One fun-
and sub-cortical centres (Adolphs, 2009; Alexander et al., 1986; damental approach consists in the sampling of extracellular
Lewis, 2012; Millan et al., 2012a; Robbins and Arnsten, 2009; Zanto levels of specific neuromodulators by microdialysis coupled to
et al., 2011). It is a crucial network hub in the control of core func- various methods of detection and quantification, where possible
tions like mood, cognition and social behaviour. Accordingly, dis-
ruption of the FCX is implicated in the pathogenesis of depression,
schizophrenia and many other central nervous system (CNS) disor- Pole for Therapeutic Innovation in CNS disorders, IDR Servier,
Croissy-sur-Seine, France
ders, and the FCX is a pivotal region underlying the therapeutic
actions of pharmacotherapeutic, psychosocial-cognitive-behavioural Corresponding author:
and stimulation-based treatments for their control (Anticevic et al., Mark J Millan, Pole for Therapeutic Innovation in CNS disorders, IDR
2015; Barr et al., 2009; Cerqueira et al., 2007; Holmes and Wellman, Servier, 125 chemin de ronde, 78290 Croissy-sur-Seine, France.
2009; Millan et al., 2012a, 2015, 2016; Navailles et al., 2010). Email: mark.millan@fr.netgrs.com

Downloaded from jop.sagepub.com at CORNELL UNIV on October 18, 2016


2 Journal of Psychopharmacology

accompanied by electrophysiological monitoring of the activity •• Third, recently-launched multi-target drugs for treating
of the neurons in question (Di Chiara, 1990; Di Chiara et al., depression, like vortioxetine, that were developed around
1996; Flik et al., 2015; Lejeune et al., 1994, 1997; Millan et al., the notion of more effectively exploiting monoaminergic
2000a,2001b; 2007a: Sharp et al., 1986; Ungerstedt, 1991). interactions in the FCX and other structures controlling
Extracellular levels of neurotransmitters and other neuromodula- mood and cognition.
tors generally reflect a balance of tightly-regulated processes of •• Fourth, Agomelatine (clinically-available) and other anti-
synthesis and release versus clearance by reuptake and enzymatic depressants that target not only monoamines for mood-
degradation: microdialysis offers a means for probing all these relief but also other, complementary, non-monoaminergic
mechanisms, together with their modulation by potential thera- mechanisms.
peutic agents. •• Fifth, modulation of the release of acetylcholine (ACh)
In this regard, an early focus of microdialysis studies was and histamine in the FCX in relation to their control of
monoamines, in particular the modulation of their extracellular cognitive processes – bearing in mind that cognitive dys-
levels in the FCX by drugs for treating anxiety, depression and function in depression has been, in comparison with
psychosis – including tricyclic-structured reuptake blockers like schizophrenia, neglected.
clomipramine, monoamine oxidase inhibitors and selective 5-HT •• Sixth, measurements of extracellular levels of hormones
reuptake inhibitors (SSRIs). Indeed, towards the end of the (melatonin, corticosterone and oxytocin) that play impor-
1990s, microdialysis was being intensively exploited to answer tant roles in the pathogenesis and potentially control of
two core and interrelated questions: (1) what is the influence of psychiatric disorders.
drugs for treating depression upon extracellular levels of 5-HT, •• Seventh, the complex challenge of quantifying glutamate,
noradrenaline (NA) and dopamine (DA) in the FCX and other GABA and other amino acids in FCX dialysates, and
key regions like the hippocampus and (2) what is the nature of linking their release, reuptake and actions both to mono-
interactions amongst monoaminergic projections in the FCX. It aminergic transmission and to novel strategies for treat-
was thought – correctly, as it turned out – that clarifying these ing depression and schizophrenia.
issues would help in the conception and evaluation of novel •• Eighth, the equally formidable hurdle of quantifying
classes of antidepressant acting via multiple and complementary extracellular levels of proteins incriminated in neurologi-
mechanisms of action. cal disorders, such as β-amyloid (Aβ) and Tau.
A comprehensive review article dedicated to the above themes •• Ninth, the translational bridging of preclinical studies of
was published in this journal by the present authors at the turn of FCX neurochemistry to work that can be undertaken in
the Millennium, together with a survey of perspectives for future human subjects, notably in the framework of clinical
progress (Millan et al., 2000a). trials.
•• Finally, some new domains of “R and D” which are likely
to evolve over the coming years.
Aims and organization of the review
The purpose of the present review is to pick up on those perspec- While this Review covers, then, a substantial amount of terri-
tives and to examine the progress that has been made over the past tory, in view of its “Anniversary” nature, the citation list is illus-
15 years or so as regards our knowledge of, ability to probe, and trative rather than comprehensive. Further, it has somewhat a
therapeutic exploitation of the FCX in the control of depression historical flavour in focussing on key articles that have appeared
and other CNS disorders. In addition, we consider what might be over the last 15–20 years, and in incorporating a representative
achieved over the next decade of progress, and it is emphasized selection of our own work in this field.
that the pursuit of microdialysis studies of the FCX and other CNS
regions remains fundamental, not least since such work can
increasingly be “translated” into clinical investigations. Ipso facto, Technological progress: from
it would be unwise indeed to discontinue this line of neurochemi- monoamines to neuromodulators to
cal research. Rather than an exhaustive review, the paper high-
lights and visually illustrates some major advances and themes hormones to proteins
of the last decade and a half, including a palette of hitherto- Dialysis of the FCX in freely-moving animals:
unpublished yet noteworthy observations from the author’s lab.
The paper focusses upon the following (“Top Ten”) facets of
the basic paradigm
research which have rapidly progressed over the past 15 years or The core dialysis paradigm permitting quantification of extracel-
so and – in most cases – will continue to do so. lular levels of neuromodulators in rats or mice is depicted in
Figure 1. As indicated in the Figure and legend, the system is
•• First, methodological progress that now permit quantifi- highly flexible, with one to two probes introduced and measure-
cation of a vast range of neuromodulators in dialysates ments undertaken in freely-moving animals fully recovered (for
from the FCX in comparison to earlier studies that at least a week) following implantation. Obviously, for all neuro-
focussed primarily on monoamines. modulators determined (Table 1), measurements need to be taken
•• Second, recent developments in our understanding of in “normal” subjects. In addition, studies undertaken during
how monoaminergic pathways interact in the FCX, and behavioural tasks, in “knockout and overexpressing” mice, in
incorporating some newer mechanisms – like roles for response to therapeutic agents, and employing genetic, environ-
5-HT6 receptors and trace amines – that were poorly char- mental and pharmacological models for depression and other
acterized at the time of the original review. CNS disorders, are of interest.

Downloaded from jop.sagepub.com at CORNELL UNIV on October 18, 2016


Millan et al. 3

Figure 1.  Schema illustrating typical dialysis probe placement in brain, the diversity of structures and neuromodulators that can be dialysed, and
methods used for detection.
Dialysis can be undertaken with one, two or, albeit rarely, more probes and is performed in freely-moving animals, equipped with a swivel, tether and collection tube
connecting the dialysis probe the sample collector. “CMA 11” is a standard microdialysis probe with a membrane cutoff of 6 kiloDaltons (kDa) sufficient to allow for the
passage of neurotransmitters. However, other probes can be used with cutoffs as high as 300 kDa, notably when proteins are to be measured. As a rule, and for studies of
drug actions, following stabilization of baseline values after probe introduction (an hour or so), samples are taken for a few hours. However, they can be taken round the
clock for 24 hours to cover an entire diurnal cycle (Figure 3). See text and Table 1 for additional information on methodology for quantifying specific neuromodulators in
dialysates.

Simultaneous quantification of 5-HT, DA and Profiting from the refinement of detection methods, including
NA in single dialysate samples high performance liquid chromatography (HPLC) coupled to
coulometric (electrochemical) detection, in the mid-1990s we
Reflecting limited technical means and the comparatively low elaborated an exceptionally sensitive and reliable method for
sensitivity of detection systems, most of the first wave of dial- simultaneously and rapidly (minutes) quantifying all three mono-
ysis studies was oriented around measures of DA and monoam- amines in a single and small volume (20 μL) dialysate sample of
inemetabolites in the striatum (Bonhomme et al., 1995; Di the FCX (Figure 2). Moreover, these studies could be undertaken
Chiara, 1990; Leviel et al., 1989; Nisell et al., 1994; Rivet in freely-moving rats (Gobert et al., 1998) – and, subsequently,
et al., 1994; Sharp et al., 1986; Ungerstedt, 1991; Zetterström mice (Di Cara et al., 2011). This proved a decisive advance allow-
et al., 1983). This work was subsequently extended to 5-HT ing for the refined characterization of: (1) the control of monoam-
and NA as well as the FCX and other cerebral regions. Further, ine release, breakdown and reuptake in the FCX; (2) the influence
there was increasing interest in drugs for treating depression of drugs on these parameters and (3) reciprocal interactions
and in dopamine/serotonin antagonists for treating psychosis amongst dopaminergic, serotonergic and adrenergic pathways to
(“atypical antipsychotics”). With some exceptions, experiments the FCX. This work was summarized in a tome which appeared in
were still performed in anaesthetized animals (Celada et al., this journal (Millan et al., 2000a) and has since been further devel-
2004; Gardier et al., 1996; Hertel et al., 1999; Hjorth and oped both by us and by others employing similar techniques (e.g.
Auerbach, 1996; Jordan et al., 1994; Millan et al., 2007b; Flik et al., 2015; Kehr et al., 2001; Yoshitake et al., 2004).
Nomikos et al., 1992; Schoemaker et al., 1997; Westerink More recently, mass spectrometry (MS) systems have been
et al., 1987; Zhang and Beyer, 2006). introduced with a similar ability to detect multiple monoamines

Downloaded from jop.sagepub.com at CORNELL UNIV on October 18, 2016


4 Journal of Psychopharmacology

Table 1.  Summary of neuromodulators quantified in microdialysate samples of the frontal cortex, and of the technologies used for their
determination.

Extracellular neuromodulator Technique employed Limit of detection Basal level in dialysates Key citation

5-HT, dopamine, noradrenaline HPLC-ECD 30–60 pM 400 pM Gobert et al., 1998


5-HT, dopamine, noradrenaline HPLC-MS 70–700 pM 400 pM Gobert, unpub. obs.
Acetylcholine HPLC-ECD 30 pM 1.3 nM Gobert et al., 2003
Acetylcholine HPLC-MS 100 pM 1.3 nM Gobert, unpub. obs.
Glutamate, aspartate, d-serine, PEA HPLC-fluorimetry 2–4 nM 0.33–0.5 µM Gobert et al., 2011
Glycine, l-serine, taurine HPLC-fluorimetry 2–4 nM 2.6–4.5 µM Gobert et al., 2011
Glutamine HPLC-fluorimetry 2 nM 2.3 µM Gobert et al., 2011
GABA HPLC-fluorimetry 2 nM 10 nM Gobert et al., 2011
Histamine HPLC-fluorimetry 0.5 nM 320 nM Rivet, unpub. obs.
Kynurenate HPLC-fluorimetry 0.2 nM 0.8 nM Gobert et al., 2011
Melatonin HPLC-fluorimetry 1 nM 9 nM Rivet, unpub. obs.
Corticosterone Enzymatic immunoAssay 30 pM 700 pM Bouchez et al., 2012
Aβ1-42 & Aβ1-40 ELISA 0.1 & 0.6 pM 0.5 & 2.5 pM Gobert et al., 2014
Tau protein ELISA 1.5 pM 250 pM Gobert et al., 2014
Tau protein HPLC-MS 20 pM 250 pM Gobert, unpub. obs.

The neuromodulators depicted, and the methods indicated, are those which we have studied in our lab. As outlined in the text, other biologically-active molecules can be
determined in extracellular samples, and additional technologies used for detection and quantification. Note the marked differences in basal levels of the neuromodula-
tors in microdialysates and the need to attain appropriate sensitivity for reliable detection. All basal values are from frontal cortex microdialysates except melatonin
(pineal gland) and corticosterone (basolateral amygdala). See also Figures 2 and 3 for visual illustration. Abbreviations: ECD, electrochemical detection; ELISA, enzyme-
linked immunosorbent assay; HPLC, high performance liquid chromatography; MS, mass spectrometry; and PEA, phosphoethanolamine.

in parallel (Bergh et al., 2016; Hows et al., 2004; Kim et al., especially opportune in view of: (1) increasing recognition that
2016). They have the advantage of improved resolution for dis- the cognitive deficits of schizophrenia are poorly relieved and
tinguishing the individual monoamines, but at the price of lesser even exacerbated by existing drugs for treating psychosis – in
sensitivity (Table1), a potential problem in structures where lev- certain cases, like clozapine, reflecting blockade of muscarinic
els are low – for example, NA in the striatum (Gobert et al., receptors; (2) the ongoing design of novel classes of agent for
2004). In any event, as outlined below, the exploitation of a vari- relieving cognitive dysfunction in schizophrenia and other disor-
ety of procedures has led to further insights into interactions ders by boosting ACh release in FCX and hippocampus and (3)
amongst monoamines and other transmitters in the FCX and sub- the functional relevance of ACh to cognitive and motor dysfunc-
cortical territories, and has facilitated the discovery and charac- tion in neurological disorders (Graef et al., 2011; Kendall et al.,
terization of novel classes of psychotropic agent. 2011; Millan et al., 2012a). More recently, we and others have set
up a rather more robust mass spectrometry (MS) technique to
detect and measure ACh (Figure 2) (Hows et al., 2002).
Quantifying acetylcholine without the need Some examples of work along these lines with potential new
for cholinesterase inhibitors drugs for treating psychosis are given later.
The remarkable efficacy of the ACh-cleaving enzyme, cholinest-
erase, means that extracellular levels of its metabolite, choline, Quantification of histamine compared to the
are vastly (thousand-fold) higher than those of ACh itself. Hence,
metabolite 1-methyl-histamine
a major challenge during the early 2000s remained the need to
reproducibly and sensitively quantify basal levels of ACh in Another frontocortical neurotransmitter involved in cognitive
dialysates without resorting to the addition of cholinesterase processes, in particular attention and vigilance, is histamine
inhibitors to the perfusion medium – or their administration to (Raddatz et al., 2010), but it has also proven something of a head-
animals – in order to artificially boost basal levels of ACh vis-a- ache to reliably quantify. Indeed, the vast majority of studies of
vis choline. Use of these inhibitors has the major disadvantages histaminergic transmission have indirectly estimated activity on
of: modifying the sensitivity of muscarinic (M2/M4) autorecep- the basis of concentrations of its principle metabolite, 1-methyl-
tors; perturbing the physiological status of cholinergic neurones; histamine – usually quantified in tissue. This strategy has obvious
and interfering with their control by pharmacological agents (see disadvantages. Not least, for pharmacological agents, it is difficult
Gobert et al., 2003; Ichikawa et al., 2000; Kehr et al., 2001). to know if any elevation in 1-methyl-histamine levels genuinely
Initially pioneered by Ichikawa et al. (2000), we further reflects a shift in histamine release as opposed to, say, an altera-
developed a novel technique, depicted in Figure 2, that oxida- tion in its catabolism and elimination. (Incidentally, it should be
tively eliminated choline in a pre-column, thereby increasing noted that specific reuptake sites for histamine have not been
assay sensitivity by 10-fold and allowing reliable quantification found, but organic cation transporters appear to be harnessed for
of baseline and stimulated levels of ACh in the FCX and other its clearance (Yoshikawa et al., 2013)). Lack of a means to rapidly,
brain regions (Gobert et al., 2003; Millan et al., 2004). This was reliably and directly quantify extracellular histamine had been a

Downloaded from jop.sagepub.com at CORNELL UNIV on October 18, 2016


Millan et al. 5

Figure 2.  Evolution of the use of dialysis for the quantification of extracellular levels of neuromodulators: from monoamines to amino acids and
acetylcholine (ACh).
Studies of the response of extracellular levels of monoamines in the FCX to antidepressants, and analysis of interactions amongst frontocortical monoaminergic pathways,
necessitated the setting up of a highly-sensitive method for quantifying 5-HT, NA and DA in single dialysate samples of freely-moving rats. With increasing interest in the
role of the FCX in the control of cognition, a technique for determining levels of ACh in the absence of cholinesterase inhibitors (w/o AChE) became important, initially
by electrochemistry, later by use of mass spectrometry. Another challenge was to establish a procedure for concurrently quantifying a whole range of amino acids and
their “false” relatives, taurine and phosphoethanolamine (PEA), in dialysates, together with both d- and l- forms of serine of which the d isomer gates NMDA receptors by
acting at glycine B sites. Differentiation of d- from l- forms of serine required a separate approach, as outlined in the text. Together these strategies facilitate evaluation
of the interrelationship amongst frontocortical populations of glutamatergic and GABAergic neurons, and between neurons and astrocytes, a key issue for many classes of
psychotropic agent (Figure 4). Note that the amino acid chromotagram shown does not include all standards. Amino acids in order of elution: Asp, apartate; Glu, gluta-
mate; Asn, aparagine; His, histidine; Gln, glutamine; Ser, serine (dl); PEA, phosphoethanolamine; Cit, citrulline; Arg, arginine; Gly, glycine; Thr, threonine; Ala, alanine;
Tau, taurine; Tyr, tyrosine and GABA, γ-aminobutyric acid. AAA is aminoadipic acid that was used as an internal standard (8 ng).

serious and persistent hassle in the screening for H3 autoreceptor antagonists per se are unlikely to be effective drugs for treating
antagonists – which disinhibit histamine release – for the treat- depression, association of H3 antagonist properties with inhibi-
ment of Alzheimer’s disease and schizophrenia (Millan et al., tion of 5-HT reuptake has attracted interest (Barbier et al., 2007):
2012a; Raddatz et al., 2010). In view of this issue, as illustrated in such drugs are best characterized neurochemically using micro-
Figure 3, we set up a dialysis procedure for sensitive detection of dialysis measures of extracellular levels of histamine.
histamine and its separation from 1-methyl-histamine. Other
experts have also elaborated approaches for studying the impact
of histamine H3 antagonists and additional drug classes upon Differentiating and quantifying a broad
extracellular levels of histamine in the brain, and for deciphering
palette of d- and l-amino acids
the interrelationships between histaminergic and monoaminergic
transmission (Flik et al., 2011, 2015). Glutamatergic pyramidal neurons interact locally with monoam-
Adrenergic and serotonergic input to the FCX modulates the inergic, cholinergic and other classes of frontocortical neurons,
release of histamine, so elevations in its levels may contribute to they project to other cortical lobes, and they also connect the
the influence of drugs for treating depression upon cognition (see FCX with subcortical structures like the basal ganglia, hippocam-
below) (Flik et al., 2015; Sanchez et al., 2015). Further, while H3 pus and thalamus (Alexander et al., 1986; Millan et al., 2012a;

Downloaded from jop.sagepub.com at CORNELL UNIV on October 18, 2016


6 Journal of Psychopharmacology

Figure 3.  Evolution of the use of dialysis for the quantification of extracellular levels of neuromodulators: from histamine and kynurenate to all-day
sampling of hormones.
Rather than estimating the activity of histaminergic pathways by dosing 1-methyl-histamine in tissue, it was desired to establish a procedure for differentiation of this
metabolite from histamine in dialysates by fluorimetry. Kynurenate is an endogenous antagonist at NMDA, AMPA and α7-nicotonic) receptors but it does not need to
be derivatized like amino acids, so it was necessary to devise a separate method for its independent quantification, as shown in Figure 2. For both corticosterone and
melatonin, continuous 24 h dialysis permitted pursuit of their circadian rhythms in the brain throughout the entire day: corticosterone peaks just around lights off, while
melatonin peaks in the middle of the dark cycle. Note the parallel shifts in its precursor, N-acetyl-melatonin, and the anti-correlated levels of 5-HT from which melatonin
is generated in the pineal gland. Exceptionally, these assays were initially established in the basolateral amygdala (high density of receptors for corticosterone) and
pineal gland (site of melatonin synthesis), respectively.

Musazzi et al., 2015; Sanacora et al., 2008). Equally pivotal to and Javitt, 2010; Millan, 2005a; Park et al., 2015; Wolosker,
the operation of the FCX, GABAergic interneurons exert a pro- 2011; Yang and Svensson, 2008). Further, there is considerable
nounced (usually inhibitory) tone on monoamines and other fron- interest in glutamatergic agents as potential new drugs for treat-
tocortical neurotransmitters (Lewis, 2012; Millan et al., 2012a, ing depression (see below).
2016). Moreover, they play an important role in the synchroniza- These observations emphasize the crucial importance of
tion of glutamatergic networks to orchestrate the top-down con- understanding the patterns and regulation of glutamate and
trol of many functions like mood, cognition, social behaviour, GABA release in the FCX, of the respective significance of neu-
reward, motor performance – as well as impulsive and compul- rons and astrocytes in this regard, and or integrating measures of
sive behaviours (Lewis, 2012; Millan et al., 2012a, 2016; Park other – usually ignored – amino acids like aspartate (Figure 4)
et al., 2015). A disruption of glutamatergic and GABAergic neu- (Errico et al., 2015; Eulenburg and Gomeza, 2010; Fossat et al.,
rones in the FCX is strongly implicated in the pathophysiology of 2012; Gobert et al., 2011; Millan, 2005a; Pehrson and Sanchez,
disorders like depression and schizophrenia. For example, a 2014; Tsai and Lin, 2010; Wolosker, 2011).
hypoactivity of FCX-localized NMDA receptors is implicated in One approach to address all this is microdialysis. Though it
the induction of psychosis, so drugs that increase activity at the lacks the rapid time resolution of voltammetry and electrophysi-
co-agonist glycine B site – of which the endogenous ligands are ology, and questions persist concerning the significance of gluta-
glial-derived glycine and d-serine – have been sought as potential mate levels in dialysates, it is indispensable for evaluating the
new drugs for treating psychosis (Danysz and Parsons, 1998; actions of pharmacological agents which should inherently have
Fossat et al., 2012; Heresco-Levy and Javitt, 2004; Kantrowitz substantial residence times and sustained functional effects

Downloaded from jop.sagepub.com at CORNELL UNIV on October 18, 2016


Millan et al. 7

Figure 4.  Schematic depiction of the interrelationships amongst glutamatergic and GABAergic neurons, and amongst neurons and astrocytes, in the
frontal cortex.
The figure incorporates amino acids, “false” amino acids and kynurenate as quantified in dialysates of FCX (Figures 2 and 3). Note the complex patterns of interplay
between astrocytes and neurons as regards the generation, reuptake and release of neuromodulators: microdialysis has proven important in their characterization.
In addition, there are many interactions mediated via distinct classes of receptor, not all of which can be shown: for example, there are inhibitory GABAB receptors
on glutamatergic terminals. The postsynaptic neurone could be another glutamatergic (pyramidal) cell, a GABAergic interneurone or a monoaminergic terminal etc.
For example, GABAergic neurons are under the excitatatory control of NMDA receptors (see text). Similarly complex configurations occur in other structures, like the
ventrotegmental area where clusters of dopaminergic cell bodies projecting to the FCX are located. Kynurenate also behaves as an antagonist at AMPA receptors and
α7-nicotinic receptors. Abbreviations as follows: Asc-T: alanine, serine, cysteine transporter; Asp, aspartate; D-AAO, d-amino acid oxidase; EAAT, excitatory amino acid
transporter; IDO, indolamine 2,3-dioxygenase; GABAT, GABA transaminase; GAD, 1-glutamic acid decarboxylase; GAT, GABA transporter; Glu, glutamate; Gln, glutamine;
Gln-ase, glutaminase; Gly; glycine; GlyT1, glycine transporter; K-3-OH, kynurenine 3-hydroxylase; KAT, kynurenine aminotransferase; Kyn, kynurenine; NR, NMDA receptor;
Ser-HMT, serine hydroxymethyltransferase; SNTA, specific neutral amino acid transporter; TCA, tricarboxylic acid cycle; TDO, tryptophan 2,3-dioxygenase; Xc-, cysteine/
glutamate antiporter; and Trp, trytophan.

(Gobert et al., 2011; Van der Zeyden et al., 2008). To this end, (a catabolic product of membrane-enriched phosphotidylethan-
then, we developed a robust and sensitive system capable of con- olamine) provides a readout for a loss of cellular integrity due to
currently quantifying more than a dozen amino acids in dialysates excitotoxicity (Vance, 2008) – as provoked by, for example, a
of FCX (Figure 2). This was based on naphthalene-2,3-dicarboal- high dose of NMDA (Table 2) (Gobert et al., 2011). Finally, in a
dehyde/cyanide derivitization coupled to HPLC binary gradient separate run using contrasting conditions (mobile phase, sodium
reverse phase separation and fluorescence detection. In addition, acetate (20 mM), zinc acetate (200 mM) and methanol (10%)),
we introduced a technical innovation (derivatization with ortho- fluorimetric measurement of the endogenous NMDA, AMPA and
phthaldehyde and Boc-l-cysteine for chiral separation) to allow α7-nicotonic receptor antagonist, kynurenate (kynurenic acid),
for complementary differentiation of “d” (NMDA co-agonist) was achieved (Erhardt et al., 2009; Gobert et al., 2011; Millan,
versus “l” (precursor) forms of serine (Gobert et al., 2011). As 2005a).
can be seen from Figure 2, it was also possible to determine lev- As described elsewhere, all these procedures and measures
els of taurine in parallel. This osmoregulatory and neuroprotec- were subjected to rigorous pharmacological characterization
tive compound behaves as a weak GABAergic agonist and is (Table 2) (Gobert et al., 2011). The following key examples may
under the excitatory control of NMDA receptors (Gobert et al., be briefly mentioned (see Table 2). First, the GABAB autorecep-
2011; Wu et al., 2005). While the underlying mechanisms are not tor and heteroceptor agonist, Baclofen, reduced levels of GABA
entirely clear, taurine appears to positively affect cognition, itself and also those of the excitatory and co-localized transmit-
mood and motivation, especially when combined with other pro- ters, glutamate and asparate. Second, NMDA elevated levels of
cognitive and energy-promoting agents (Curry and Stasio, 2009). both taurine and PEA, and reduced levels of d-serine, actions
The simultaneous quantification of phosphoethanolamine (PEA) abolished by the competitive NMDA receptor antagonist, CPP.

Downloaded from jop.sagepub.com at CORNELL UNIV on October 18, 2016


8 Journal of Psychopharmacology

Third, levels of kynurenate were increased by administration of

50.1 ± 3.9a
15.8 ± 0.4

17.5 ± 0.1
16.7 ± 0.4
14.3 ± 0.9

15.6 ± 1.0
13.6 ± 2.0

17.2 ± 1.1
14.5 ± 1.1

vehicle. As previously (Gobert et al., 2011), values shown represent “Area under the curve” analysis: that is, the global drug influence expressed across the 3 h dialysis period as % × minutes/1000. Doses are in mg/kg, but note that
15.6 ±0.9

the dose of ketamine was 80.0 for its influence on amino acids. Data are means ± SEMs with N values between 5 and 8. aP < 0.05 to corresponding vehicles in Newman–Keuls test following ANOVA. Abbreviations: ago, agonist at the
its precursor, kynurenine, an effect blocked by inhibition of the

The findings shown, which incorporate some documented in Gobert et al. (2011), are based upon dialysis studies performed over a period of 3 h following injection of the agent shown, with effects determined in comparison to
converting enzyme, amino-oxyacetate (Gobert et al., 2011).
PEA

ND
This microdialysis procedure is highly instructive for exam-
25.6 ± 2.2a ining the actions of drugs for treating depression and psychosis

18.4 ± 0.9a

19.8 ± 1.0a

21.2 ± 1.4a
15.8 ± 0.6

17.6 ± 0.5

15.1 ± 1.1
15.3 ± 1.3
17.7 ± 1.6

16.9 ± 1.2
as well as pro-cognitive agents (see below). Nonetheless, a word

Glycine B site on NDMA receptors; ant, antagonist; Ch Bl, Channel Blocker; co-ago, co-agonist; DA, dopamine; Gly-RI, glycine reuptake inhibitor; NA, noradrenaline; ND, not determined; PEA, phosphoethanolamine.
15.1 ±0.5
Taurine

of caution, especially for glutamate. With the exception of


blockers of glutamate reuptake, its extracellular levels are quite
resistant to modification, even for mechanisms that are known to
influence glutamatergic transmission (Gobert et al., 2011; Van
12.0 ± 0.9a
14.1 ± 0.9a
13.8 ± 1.1a
17.0 ± 1.0
17.1 ± 1.0
15.1 ± 2.6

19.7 ± 1.3
18.6 ± 1.0

18.5 ± 1.3
16.8 ± 1.4
15.7 ±0.4
der Zeyden et al., 2008). Thus, the sensitivity of this and other
GABA

dialysis systems to small and rapid alterations in glutamate


release “felt” by postsynaptic glutamatergic receptors may be
limited and insufficient for monitoring dynamic shifts in signal-
162.5 ± 16.7a

ling. Also, the architecture of various classes of glutamatergic


14.6 ± 1.1a

28.9 ± 2.3a
24.4 ± 0.4a
17.1 ± 0.5

15.5 ± 0.4
16.8 ± 1.1
19.8 ± 1.1

16.5 ± 1.2

15.9 ± 0.8

17.1 ± 0.6

receptor at synapses is complex with both intra- and extra-


Glycine

synaptic distributions: localized differences in concentration


differentially seen by these populations cannot be registered by
microdialysis (Danysz and Parsons, 1998; Papouin and Oliet,
Table 2.  Influence of glutamatergic agents upon dialysate levels of monoamines and amino acids in the frontal cortex of freely-moving rats.

2014). Thus, for glutamate in particular, the importance of other


571.1 ± 194.6a

measures of its activity, especially those with rapid time resolu-


14.9 ± 0.8a

13.3 ± 1.0a
17.5 ± 0.4

15.4 ± 0.5
17.7 ± 0.6
18.4 ± 0.7
16.0 ± 0.8

17.9 ± 1.3
18.6 ± 0.5
18.7 ± 0.9

tion should be underlined (Alvarsson et al., 2015).


d-Serine

Twenty four hour monitoring of centrally-


active hormones implicated in psychiatric
13.3 ± 1.3a
19.0 ± 0.7a

12.7 ± 1.5a

11.6 ± 1.0a
16.7 ± 0.7

16.1 ± 1.5

15.8 ± 0.7
15.4 ± 0.5
16.3 ± 1.3

13.7 ± 0.7
15.0 ± 1.2
Aspartate

disorders: melatonin, corticosterone and


oxytocin
Kynurenate provides a convenient link to a further set of proce-
14.6 ± 1.4a
20.6 ± 0.2a

14.4 ± 1.0a
17.6 ± 0.5

17.9 ± 1.7
16.1 ± 0.4

16.5 ± 0.9
15.9 ± 0.7
22.4 ± 4.1

16.0 ± 1.0
Glutamate

dures that were established for dosing central levels of hormones


16.7 ±1.0

implicated in the pathogenesis and management of depression


and other psychiatric disorders. Like kynurenate, melatonin is
ultimately derived from the precursor, tryptophan, albeit follow-
ing an alternative biosynthetic route in which 5-HT is trans-
27.5 ± 2.2a
25.5 ± 2.8a

26.3 ± 2.8a

22.2 ± 1.7a
26.1 ± 3.6a
31.0 ± 5.2a
18.0 ± 0.6

19.5 ± 1.9

16.4 ± 0.6
20.5 ± 1.1

20.3 ± 0.5

formed (by 5-HT-N-acetyltransferase) into its acetylated


derivative, N-acetyl-5-HT (nor-melatonin), which is then con-
DA

verted into melatonin by hydroxy-indole-O-methyl transferase


(Catena-Dell’Osso et al., 2012). The synthesis of melatonin
25.8 ± 1.8a
29.4 ± 4.4a

24.7 ± 2.6a

26.0 ± 3.3a
20.6 ± 1.4

20.5 ± 1.1

18.8 ± 1.5
20.8 ± 1.2
21.8 ± 0.9
20.4 ± 1.1

21.1 ± 1.2

occurs in the pineal gland which was the logical place, rather than
the FCX, to validate a fluorometric method that we devised for its
measurement. Moreover, in view of the pronounced diurnal
NA

rhythm in secretion of melatonin, and its crucial role in the circa-


dian scheduling of other rhythms and behaviours (Catena-
16.7 ± 0.5

16.7 ± 0.9
18.0 ± 1.1
17.1 ± 1.2
15.1 ± 0.2
17.5 ± 2.2
14.4 ± 0.9

17.4 ± 2.2
18.3 ± 0.6
18.1 ± 2.3
17.4 ±1.0

Dell’Osso et al., 2012), this was an opportunity to set up a system


allowing for monitoring of the daily fluctuations of extracellular
5-HT

levels of neuromodulators over 24 h. As shown in Figure 3, the


fluorometric strategy chosen permitted not only demonstration of
NMDA co-ago
NMDA co-ago

the night-time peak in melatonin levels but also, and rather ele-
NMDA Ch Bl
mGluR5 ant
GABAB ago

AMPA PAM
NMDA ago
NMDA ant

gantly, a concomitant elevation in N-acetyl-5-HT coupled to an


Activity

Gly-RI
Gly-RI

anti-correlated reduction in their precursor, 5-HT. The sensitivity


of this procedure was sufficient to allow deployment in the FCX


and other brain structures, and it is obviously informative as a
direct measure of how pharmacological agents and animal mod-
ORG 24598 (10.0, s.c.)
LY392,098 (10.0, s.c.)
Ketamine (10.0, s.c.)

Sarcosine (800, i.p.)


Baclofen (10.0, s.c.)

els for depression influence the diurnal organization of biological


d-Serine (160, s.c.)
Glycine (800, i.p.)
Vehicle (s.c., i.p.)
NMDA (40.0, s.c.)
Drug (dose, route)

MPEP (10.0, i.p.)

rhythms.
CPP (20.0, s.c.)

Another hormone which likewise displays a striking circadian


rhythm is corticosterone (CS), secreted by the adrenal gland
downstream of the hypothalamo-hypophyseal axis. CS secretion
is a typical response to “stress” and closely linked to multiple – if

Downloaded from jop.sagepub.com at CORNELL UNIV on October 18, 2016


Millan et al. 9

not all – CNS disorders (Cerqueira et al., 2007; Lupien et al., Wotjak et al., 2008; Zoicas et al., 2014). It would be particularly
2009). Indeed, protracted and uncontrolled increases in CS secre- interesting to evaluate the influence of novel (pharmacotherapeu-
tion are incriminated in the etiology of anxio-depressive states, tic and other) strategies for treating autism and schizophrenia
disruption of mood and cognition, and structural and functional upon the release of oxytocin by dialysis procedures. More gener-
changes in corticolimbic structures like the FCX, hippocampus ally, this remains a fruitful line of investigation, and numerous
and amygdala (Cerqueira et al., 2007; De Quervain et al., 2009; other hormones – peptidergic, steroid and other – interrelated to
Holmes and Wellmann, 2009; Lupien et al., 2009; Millan, 2003, depressive and other psychiatric states, warrant analysis by dialy-
2006). The central actions of CS reflect its recruitment of nuclear sis procedures (Clough, 2005; Wotjak et al., 2008).
mineralocorticoid receptors (high affinity) and glucocorticoid
receptors (low affinity, mainly activated during stress and peak
circadian levels) (De Quervain et al., 2009; Lupien et al., 2009). Procedures for detecting and measuring
Antagonists of glucocorticoid receptors have attracted interest extracellular levels of neurotoxic proteins in
for the control of psychotic depression, but issues of safety ham- dialysates
pered their development and they no longer appear to be in active
research (Millan, 2006). Notwithstanding the importance of cen- Specialist teams have been studying dialysis levels of the neuro-
tral actions of CS, its levels have almost invariably been moni- toxic proteins, Aβ42 and Tau, for several years in humans, mostly
tored in the periphery, so it was useful to set up a sensitive under conditions of traumatic brain injury (Marklund et al., 2009;
enzymatic immunoassay for reliably quantifying the far lower 2014; Ungerstedt and Rostami, 2004). Further, CSF levels of
levels of CS in the brain. This was undertaken in the basolateral Aβ42, total Tau and phosphorylated Tau are considered reliable
amygdala in view of its well-documented role in the cerebral biomarkers of early Alzheimer’s disease (Hulstaert et al., 1999;
response to CS under conditions of stress, and to complement Olsson et al., 2016). Ironically, there have been very few reports
studies performed elsewhere of hippocampal levels of CS in on extracellular levels of Aβ42 and Tau in rodents – despite the
mice and rats (Béracochéa et al., 2011; Droste et al., 2009). frantic levels of research incriminating this infamous double act
Pharmacological, psychological and mild physical stressors all in the pathogenesis of Alzheimer’s disease (Iqbal et al., 2016).
provoked robust and time-constrained elevations in basolateral One simple reason is that it is very difficult to detect and quantify
amygdala levels of CS which were abolished by cojoint blockade Aβ42 and Tau in the extracellular environment – in comparison
of central vasopressin1B and corticotrophin releasing factor1 to the fairly straightforward task of measuring these neurotoxic
receptors upstream of the hypothalamo-pituitary axis (Bouchez proteins in tissue. Nonetheless, employing a microdialysis tech-
et al., 2012; Murat et al., 2012). Furthermore, the circadian nique coupled to enzyme-linked immunosorbent assay (ELISA)
rhythm of CS secretion, coordinated by the suprachiasmatic quantification, Cirrito et al. (2003) showed that soluble Aβ42 is
nucleus (Nader et al., 2010), could be fully reproduced by dialy- released into brain interstitial fluid in mice overexpressing amy-
sis quantification in the basolateral amygdala, with the character- loid precursor protein.
istic increase commencing in anticipation of the wake (dark for We have recently established our own microdialysis proto-
rats) period, peak levels attained and maintained over several cols to address this issue (Gobert et al., 2014). Using the P301L
hours during this active phase of the cycle, and a return to base- mouse (Lewis et al., 2000), it has proven possible to simultane-
line occurring just before the onset of the inactive (lights on) ously quantify levels of Aβ1–40 and Aβ1–42 (ca. 4000 Daltons
period (Figure 3) (Mohawk et al., 2007). Quantification of CS each in size) and total Tau protein (ca. 50,000 Daltons) in dialy-
levels in the FCX is also feasible and of especial interest since it sis samples of the FCX using an ELISA assay coupled to elec-
is there that CS exerts several actions, such as modulation of trochemiluminescence detection (Figure 5) (Gobert et al.,
glutamatergic transmission, related to the pathophysiology of 2014). Inasmuch as the dialysis membrane used for this work
depression (Dwivedi et al., 2015; Musazzi et al., 2015; permits passage of molecules of up to about 300,000 Daltons
Palamarchouk et al., 2009). (Figure 1), all these peptides can easily penetrate into the probe.
Another hormone to evoke is oxytocin in view of its impor- While it is likely for Tau that microdialysis detects a mixture of
tant role, expressed across the FCX and several cerebral struc- monomers, dimers and small oligomers (up to hexamers),
tures, in the promotion of social behaviour and social cognition Aβ1–40 and Aβ1–42 may be present as even larger conglomerates
(Bukovskaya and Shmukler, 2016; Goodson and Thomson, – though in both cases proteins must be soluble for passage,
2010). Disruption of these social functions is a fundamental detection and quantification. By use of a single site ELISA, it
feature not only of autism-spectrum disorders but also of may soon be possible to specifically distinguish Tau oligomers
schizophrenia, depression and even neurological disorders like from monomers (Takeda et al., 2016). Another complication is,
Alzheimer’s disease (Adolphs, 2009; Goodson and Thomson, however, the vast array of different forms of Tau created by
2010; Henry et al., 2016; Millan et al., 2012a). Thus, modulation post-translational modifications like phosphorylation and acet-
of extracellular levels of oxytocin in discrete brain regions under ylation (Iqbal et al., 2016). For their detection and differentia-
conditions of rewarding or conflicting social interaction, and in tion, proteomics and phosphoproteomics plus ELISA may
response to depression or pharmacological agents, is of consider- prove helpful, but this remains difficult in view of the very low
able interest, yet there is surprisingly little information available. quantities of individual variants of Tau in the extracellular
While we have regrettably not had the opportunity to measure space (Brinkmalm et al., 2015; Fu et al., 2015; Takeda et al.,
this social hormone ourselves, several procedures have been pro- 2016). Thus, rigorous microdialysis detection and quantifica-
posed in order to do so (Mabrouk and Kennedy, 2012). Their util- tion of neurotoxic proteins in rodents, parallelling work under-
ity and promise has been unveiled in behavioural paradigms like taken in human CSF (Brinkmalm et al., 2015; Olsson et al.,
fear-based learning and social interaction (Ross et al., 2009; 2016; Takeda et al., 2016), remains a work in progress.

Downloaded from jop.sagepub.com at CORNELL UNIV on October 18, 2016


10 Journal of Psychopharmacology

Figure 5.  Evolution of the use of dialysis for the quantification of extracellular levels of neuromodulators: quantification of Tau protein and
ß-amyloid in dialysates of the FCX.
The proteins were all determined with a highly sensitive and selective ELISA (enzyme-linked immunosorbent assay) technique, as outlined in the text and Table 1. Their
levels were determined in “P301L” mice bearing a Tau mutation associated with Tau over-production as compared to their wild-type counterparts with the same genetic
background. In comparison to wild-type animals, Tau levels were significantly higher in P301L mice at 16 months when behavioural symptoms, cognitive deficits and
neuropathological changes appear. Conversely, there was no difference to wild-type mice for levels of ß-amyloid 1–40 or 1–42. Data are means ± SEMs. *P < 0.01 to wild-
type animals in Newman–Keuls test following ANOVA with time as a dependent variable. N was 6–8 per value.

Extracellular levels of other biologically- (3) measures of interactions with endothelial cell-enriched
important molecules P-glycoproteins – that eject many compounds from the brain
(Cremers et al., 2012, 2016; Ejsing et al., 2005; Josserand et al.,
The above discussion evoked a quite wide spectrum of biologi- 2006).
cally-active substances found in the extracellular space and rele- Ideally, one can simultaneously dose the drug under investi-
vant to the genesis and control of depression and other CNS gation in the FCX as well as the levels of the neurotransmitter
disorders, but the list is not exhaustive. Second messengers which it is modifying: for example, extracellular levels of DA
(cAMP and cGMP) and growth factors like brain derived neuro- together with its precursor, l-DOPA, following administration
trophic factor (BDNF), for example, merit a brief comment, as of the latter to rats (De Deurwaerdère et al., 2016). This
well as other neuropeptides like substance P, somatostatin and approach has been less enacted than one might imagine, yet it
neurotensin (Cunha et al., 2010; Pepicelli et al., 2004; Vaka et al., remains a translationally-relevant option for providing direct
2011; Wojtak et al., 2008; Yoshitake et al., 2013). answers to the question of just how well an agent actually gets
into the brain, and whether it reaches concentrations sufficient
to exert actions of interest – beneficial or undesirable (Cremers
Dialysis quantification of pharmacological
et al., 2012, 2016; Weber et al., 2013). Another use is for moni-
agents toring long-term studies where it is important to be sure of the
Finally, at least in the academic environment, the major focus of cerebral concentration of a drug throughout treatment. For
dialysis is on quantitation of extracellular levels of endogenous example, chronic administration of fluoxetine over three weeks
neuromodulators, yet monitoring of pharmacological agents leads to a large (more than two-fold) elevation in basal extracel-
themselves is likewise feasible, providing an indication of entry lular levels of 5-HT in FCX not owing to fluoxetine-induced
into the brain, distribution and (with appropriate calculations) 5-HT1A autoreceptor desensitization per se, but mainly due to
concentrations attained cerebrally (Cremers et al., 2012, 2016; the long half-life of its bio-active metabolite, nor-fluoxetine,
Deguchi et al., 2002; De Lange et al., 2005). This strategy is which was found at very high levels in microdialysates. In con-
complementary to (and may confirm or refute) observations trast, this was not the case for the SNRI, S33005, which has a
made to forecast passage in the brain using: (1) in silico pre- shorter half-life and no equivalent bioactive metabolite (Gobert
diction; (2) cellular models of blood–brain-barrier passage and et al., unpublished observations; Millan et al., 2001a).

Downloaded from jop.sagepub.com at CORNELL UNIV on October 18, 2016


Millan et al. 11

Monoaminergic interplay in the FCX: functionally active and, inasmuch as they are more prevalent in
human dopaminergic cell bodies, they may be of greater clinical
overview of core interactions and significance than usually thought – also as a mechanism of
recent developments neuroprotection (Collo et al., 2012; Joyce and Millan, 2007;
Lejeune et al., 1997; Pich and Collo, 2015).
Regulation of frontocortical monoaminergic
While dopaminergic neurons do not exert any marked influ-
transmission as a basis for therapeutic ence on their serotonergic and adrenergic counterparts, the latter
progress restrain dopaminergic and serotonergic pathways via  α2-AR
Millan et al. (2000a) was principally devoted to the interrelation- heteroceptors localized on their terminals: mirroring autorecep-
ships amongst monoaminergic pathways innervating the FCX, tors, α2A-ARs appear to predominate as heteroceptors over their
how they regulate their own and each other’s activity via reup- α2C counterparts (Gobert et al., 1999, 2003; Hertel et al., 1999;
take sites, auto- and heteroceptors, and how improved knowledge Millan et al., 2000a, 2000b, 2000c; Milan, 2009). Importantly
of these interactions might be used as a basis for developing α2-AR heteroceptors on dopaminergic terminals are tonically
novel classes of antidepressant. The following paragraphs sum- active, whereas those on serotonergic terminals are not, at least as
marize this core paradigm and some recent – though surprisingly judged by the consequences of their blockade. Beta-ARs may
minor – refinements in our understanding of the interplay also have some minor excitatory influence on dopaminergic pro-
amongst frontocortical monoaminergic projections. It is worth jections (Celada et al., 2004; Gobert and Millan 1999a; Hjorth
noting in this context that widely-heralded studies in genetically- and Sharp, 1993) which are subject to a stabilizing and globally
modified mice largely converged with findings acquired using inhibitory influence of α1A/1B-ARs at the level of their cell bodies
boring-old – yet selective and well-characterized – pharmaco- (Lejeune et al., 1994).
logical agents. As for serotonergic fibres, their influence on neighbouring
In the subsequent sections, key examples are given of mono- dopaminergic and adrenergic projections is pronounced, com-
aminergic antidepressants that were conceived, discovered, plex and – even today – not fully understood (Blier and El
developed and – in two cases – launched based on an improved Mansari, 2013; Millan et al., 2000a; Milan, 2009; Sanchez et al.,
understanding of interactions amongst monoaminergic pathways 2015). The best documented control is expressed via 5-HT2C
in the FCX (Millan, 2006, 2009, 2014b; Sahli et al., 2016; receptors which exert a robust and inhibitory influence on DA
Sanchez et al., 2015). and NA release in FCX: irrespective of whether these sites show
At a somewhat later date, this purely monoaminergic frame- constitutive activity in native tissue, their inhibitory control is
work of FCX function was expanded to embrace complementary exerted tonically (Dekeyne et al., 2008; Di Giovanni and De
non-monoaminergic mechanisms. This led to the elaboration of Deurwaerdère, 2016; Millan et al., 2000a, 2003; Millan, 2005b).
further multi-target concepts for improving the treatment of Reconciling the excitatory actions of 5-HT2C receptors with
depression (Millan, 2006), including the therapeutically- inhibition of adrenergic and dopaminergic pathways has neces-
employed agomelatine (De Bodinat et al., 2010; Millan et al., sitated the presence of intervening GABAergic interneurons,
2003; 2005), which is likewise evoked below. located both in the ventrotegmental area and locus coereleus
and at the terminal level in the FCX (Di Giovanni and De
Deurwaerdère, 2016; Millan, 2005b; Santana and Artigas, 2016).
How monoaminergic terminals interact in the A modest excitatory impact of 5-HT2A receptors has also been
FCX: updating the paradigm seen on DA and NA liberation, possibly direct and/or heterosyn-
aptic and passing by excitatory glutamatergic input (Celada
Serotonergic, adrenergic and dopaminergic projections to the et al., 2013; Gobert and Millan, 1999b). Despite some intriguing
FCX are all equipped with reuptake sites for 5-HT (SERT), NA (or odd to the skeptic) observations in knockout mice and their
(NAT) and DA (DAT), respectively, though the latter are scarce staunch defence by a dwindling handful of devotees, 5-HT2B
and DA is also cleared by the more numerous NATs on adrener- receptors are the poor relatives of 5-HT2A and 5-HT2C receptors
gic terminals. SERTs and NATs are, of course, the targets of and merely lurk in the background. In any event, in addition to
SSRIs and NARIs, respectively, and they are both concomitantly postsynaptic sites, there may be a small population of 5-HT2B
blocked by 5-HT/NA reuptake blockers (SNRIs) like venlafaxine receptors on 5-HT neurons that exert a variable, excitatory influ-
and S33005 (Barnes and Sharp, 1999; Blier and De Montigny, ence on 5-HT release, and 5-HT2B sites were speculated to be
1994; Celada et al., 2004; Millan et al., 2000a, 2001a). somehow involved in the actions of SSRIs (Diaz et al., 2016;
Serotonergic, adrenergic and dopaminergic neurons also possess Launay et al., 2006; McDevitt and Neumaier, 2011). While they
inhibitory autoreceptors: 5-HT1A and 5-HT1D (cell body)/5-HT1B should not be ignored, compared to their big brother, 5-HT2C
(terminal), α2-AR (cell bodies and terminals) and D2/D3(cell bod- receptors, 5-HT2B receptors do not seem to have a prominent
ies and terminals) (Barnes and Sharp, 1999; Gardier et al., 1996; role in the control of monoaminergic transmission in the FCX.
Hjorth and Auerbach, 1996; Hjorth et al., 2000; Lejeune et al., Recently, a role for 5-HT3 receptors in the control of mono-
1997; Lejeune and Millan, 1998; McDevitt and Neumaier, 2011; aminergic transmission was revived, possibly by excitation of
Millan et al., 1995, 2000a, 2000b, 2000c; Rivet et al., 1994). It is GABAergic neurons, but the robustness and therapeutic rele-
now clear that the α2A-AR subtype predominates as an autore- vance of this control is uncertain (Celada et al., 2013; Mørk et al.,
ceptor but α2C-ARs fulfill a complementary and subsidiary role 2012; Riga et al., 2016; Sanchez et al 2015). 5-HT4 receptors
(Gobert et al., 1998, 1999, 2003; Hertel et al., 1999; Millan have been increasingly implicated in the mediation of antidepres-
et al., 2000a, 2000b; Milan, 2009). D2 autoreceptors are quanti- sant properties, even leading to the suggestion that they can (indi-
tatively predominant in rodents, but D3 autoreceptors are also rectly) desensitize 5-HT1A autoreceptors (Samuels et al., 2016;

Downloaded from jop.sagepub.com at CORNELL UNIV on October 18, 2016


12 Journal of Psychopharmacology

Vidal et al., 2014). 5-HT5 receptors appear to be present in the depression and psychosis but clinical feedback is awaited (Lam
raphe, but there is little or no information on control of seroton- et al., 2015; Revel et al., 2012).
ergic (or catecholaminergic) transmission, perhaps due to a lack To summarize, the monoaminergic framework articulated by
of pharmacological tools (Millan, 2006; Thomas, 2006). As for Millan et al. (2000a) has not, in the past 15 years, been radically
5-HT6 receptors, there is evidence for a facilitatory influence of corrected or modified, although the roles of multiple 5-HT recep-
their blockade on adrenergic and dopaminergic pathways, but tor subtypes are now better characterized. Further, a modulatory
results have been variable and sometimes contradictory (for ago- influence for trace amines has been established, and interactions
nists and antagonists), possibly due to opposing actions of 5-HT6 between monoaminergic autoreceptors and transporters have
receptors expressed via GABAergic versus glutamatergic neu- become important. The monoaminergic framework of 2000 has
rons (Kendall et al., 2011; King et al., 2008; Schechter et al., then continued to provide a coherent schema for devising new
2008). Finally, 5-HT7 receptor antagonism has been evoked as a classes of drugs for treating depression, and especially those pos-
potential antidepressant strategy (alone and coupled to SERT sessing actions at novel combinations of monoaminergic receptor
blockade) and this may reflect enhanced 5-HT release due to and reuptake site. Nonetheless, things have evidently moved on
actions expressed in raphe nuclei, directly and in interaction with and a major emphasis today is the identification of drugs for
GABAergic interneurons, but data remain limited (Bonaventure treating depression and psychosis that act primarily via mecha-
et al., 2007; Kusek et al., 2015; Mnie-Filali et al., 2011). nisms that are not solely – or at all – monoaminergic. Some
Needless to say, one should not forget the inhibitory autore- insights into the above strategies and their interrelationships with
ceptor roles of 5-HT1B and 5-HT1D receptors (De Groote et al., monoaminergic transmission are given below.
2003; Gardier et al., 1996; Gobert et al., 1998; Gobert and Millan,
1999a; McDevitt and Neumaier, 2011), and there must be a final
word on the 5-HT1A receptors which have been studied in breadth From monoamine reuptake inhibitors
and depth for decades. While blockade of 5-HT1A autoreceptors to novel multi-target classes of
enhances the influence of SSRIs on dialysis levels of 5-HT,
5-HT1A receptor agonists and partial agonists, alone and co-
antidepressant
administered with SSRIs/SNRIs, enhance extracellular levels of Coupling 5-HT1A receptor activity to
DA and NA in FCX, actions linked to antidepressant properties inhibition of 5-HT reuptake: vilazodone and
(Blier and El Mansari, 2013; Gobert et al., 1998, 1999; Millan
et al., 2000a; Millan, 2006). A role for postsynaptic 5-HT1A
vortioxetine
receptors (presumably located on GABAergic interneurons) has One of the earliest ideas of how to accelerate antidepressant onset
been evoked in the influence of 5-HT1A receptor agonists/partial of action was based on the somatodendritic localization of the
agonists on DA/NA release. Nonetheless, on balance, it appears 5-HT1A subtype and its inhibitory influence on serotonergic neu-
that recruitment of 5-HT1A autoreceptors is mainly involved rons. It was reasoned that 5-HT1A autoreceptors must brake and
leading to downstream removal of 5-HT2C receptor-mediated slow the actions of SSRIs by a negative feedback mechanism,
excitation of GABAergic interneurons (Gobert et al., 1998; leading to efforts to develop 5-HT1A receptor antagonists and to
Lejeune and Millan, 1998; Santana and Artigas, 2016; Sanchez associate SERT inhibition and 5-HT1A autoreceptor blockade
et al., 2015). An important point relevant to drug design is that either by drug combinations or by their integration into a single
postsynaptic 5-HT1A receptors are, in addition to 5-HT1A autore- chemical entity (Blier and De Montigny, 1994; Celada et al.,
ceptors, involved in the mediation of antidepressant properties, 2004; Gardier et al., 1996; Hjorth et al 2000; McDevitt and
including those triggered by inhibition of 5-HT reuptake (see Neumaier, 2011; Millan, 2014a; Millan et al., 1995). Later, as
below) (McDevitt and Neumaier, 2011; Millan, 2006; Samuels remarked above, it was realized that postsynaptic 5-HT1A recep-
et al., 2016; Sanchez et al., 2015). As regards these postsynaptic tors contribute to the antidepressant actions of SSRIs (Millan,
sites, it has been suggested that they influence the activity of 2006; Samuels et al., 2016), so this appeared to be a case of gain-
ascending monoaminergic pathways by virtue of feedback loops ing on the roundabout yet losing on the swings. Indeed, 5-HT1A
running to their cell bodies (Hajós et al., 2003). antagonist/SSRI association studies ultimately failed to improve
Finally, trace amine associated receptors 1 (recruited by trace on the efficacy of SSRIs and, despite considerable efforts, no
amines like tyramine and phenylethylamine) have emerged to combined 5-HT1A autoreceptor antagonist/SERT blocker has
modulate the activity of monoaminergic neurones and the libera- proven clinical efficacy and reached the market (Blier and El
tion of monoamines in the FCX and sub-cortical regions. They Mansari, 2013; Celada et al., 2004; Millan, 2006, 2009, 2014a;
influence, in particular, dopaminergic and serotonergic transmis- Sanchez et al., 2015).
sion, including the response to drugs suppressing reuptake and/or A subtle shift in emphasis led to the notion of coupling 5-HT1A
impacting mechanisms of release (Di Cara et al., 2011; Ledonne receptor partial agonist properties to 5-HT reuptake inhibition: this
et al., 2010). In addition, like dopamine D3 and other classes of should permit some postsynaptic efficacy yet also – it is claimed
autoreceptor, trace amine associated receptors 1 physically and – realize the more rapid 5-HT1A autoreceptor desensitization com-
functionally interact with monoamine transporters, thereby pared to a SSRI alone (Celada et al., 2004, 2013; McDevitt and
unveiling a novel dimension to the fine-control of extracellular Neumaier, 2011; Millan et al., 2000a). In fact, it is not so easy to
levels of monoamines in frontocortical and subcortical structures see why partial agonist properties (drug) should more effectively
(Castro-Hernández et al., 2015; Miller, 2011). Trace amine desensitize 5-HT1A autoreceptors than SSRI-elevated levels of
associated receptors 1 are also localized postsynaptically to 5-HT (a full agonist) itself. Nonetheless, 5-HT1A autoreceptor par-
monoaminergic projections (Pei et al., 2016). They are intrigu- tial agonists/SERT blockers are easier to find (often by accident)
ing candidates targets for novel classes of drugs for treating than pure 5-HT1A antagonists/SERT blockers so this was all rather

Downloaded from jop.sagepub.com at CORNELL UNIV on October 18, 2016


Millan et al. 13

convenient. Moreover, 5-HT1A partial agonism enhances fronto- Dialling in DA as well as 5-HT and NA
cortical release of NA and DA alone and in association with SSRIs transporters
(Gobert et al., 1998, 1999) and, in contrast to a pure 5-HT1A antag-
onist, one would expect anxiolytic properties from partial agonist Compromised dopaminergic transmission in both the FCX and
properties in the absence of disruptive side-effects (Millan, 2003; mesolimbic regions is implicated in the mood, reward, cognitive
Millan et al., 1995). To cut a long story short, this manner of and even sexual deficits of depression, and all clinically-effective
exploiting monoaminergic networks was eventually vindicated antidepressants either elevate FCX levels of DA and/or reinforce
with the launching of vilazodone, a partial 5-HT1A autoreceptor postsynaptic DA signalling in limbic regions (reviewed in Millan,
partial agonist/SERT blocker for depression (Ashby et al., 2013; 2006). Hence, there has been interest in adding DA reuptake inhi-
Hughes et al., 2005; Sahli et al., 2016). Vilazodone robustly ele- bition to blockade of SERTs and NATs. This is chemically quite
vates FCX levels of 5-HT and possesses antidepressant and anxio- straightforward – indeed, it not infrequently happens without any
lytic actions in preclinical models. Further, upon chronic conscious effort at all (Lane, 2015; Millan 2009, 2014a).
administration, it results in dorsal raphe 5-HT1A autoreceptor Accordingly, a whole range of “triple inhibitors” have been
desensitization yet an enhancement in the activity of postsynaptic described (Lane, 2015; Millan, 2009). However, rigorous clinical
5-HT1A receptors on CA3 neurones in the hippocampus – mimick- demonstration of antidepressant properties at well-tolerated
ing other classes of antidepressant (El Mansari et al., 2015; Sahli doses is awaited and several – like tesofensine – have been re-
et al., 2016). For reasons that remain unclear, it does not seem to routed to other indications like the treatment of addiction and
increase FCX release of NA and DA: how this unexpected (and neurological disorders. Furthermore, direct elevation of extracel-
presumable) drawback influences therapeutic efficacy for treating lular levels of DA in limbic structures incurs the risk of abuse,
depression (and symptoms of anxiety) compared with SSRIs dependence and psychosis, while “triple inhibitors” may also
remains to be clarified (El Mansari et al., 2015; Hughes et al., have autonomic/cardiac-cardiovascular side-effects and cause
2005; Sahli et al., 2016; Thase et al., 2014). insomnia (Millan, 2009; Lane, 2015).
Another recently-launched antidepressant, vortioxetine, like-
wise emerged from studies of monoaminergic neurochemistry in Blockade of α2-adrenoceptors to disinhibit
the FCX, and the question as to how a SSRI profile might be
upgraded by concomitantly manipulating discrete classes of
NA and DA release in FCX
5-HT receptor. Initially, this project was again oriented around Another way of reinforcing the influence of antidepressants on
SERTs and antagonist actions at 5-HT1A autoreceptors (Sanchez DA – as well as NA – levels in FCX would be to block tonically-
et al., 2015). However, it was redirected (or redirected itself to active α2-ARs on the terminals of dopaminergic and adrenergic
the gratification of astute pharmacologists and chemists) towards fibres innervating the FXC (vide supra): since α2-ARs do not in
a broader and multi-target profile that embraced not only 5-HT1A this way control mesolimbic dopaminergic pathways, there is – in
receptor agonist properties, but also partial agonism at 5-HT1B contrast to triple inhibitors – no risk of excess DA release in sub-
receptors as well as antagonist properties at 5-HT1D, 5-HT3 and cortical regions (Gobert et al., 1999; Hertel et al., 1999; Millan
5-HT7 receptors (Bang-Andersen et al., 2011; Mørk et al., 2012; et al., 2000a, 2000b). Mirroring the blockade/desensitization of
Pehrson et al., 2013; Riga et al., 2016; Sanchez et al., 2015). 5-HT1A autoreceptors, pharmacological co-antagonism of α2-ARs
5-HT1D and 5-HT7 receptor blockade may help promote seroton- should rapidly reproduce the progressive desensitization of pre-
ergic transmission and participate in antidepressant properties synaptic α2-ARs seen upon long-term exposure to antidepressants
(see above) (Mnie-Falili et al., 2011). Further, the most potent that elevate extracellular levels of NA (Gobert et al., 1998, 1999;
action of vortioxetine, 5-HT3 blockade, apart from opposing Millan et al., 2000a, 2000b; Sanacora et al., 2004; Serres et al.,
nausea during treatment, may offer some modest benefit in treat- 2012). Further, α2-ARs blockade should counter sexual dysfunc-
ing symptoms of anxiety (Baldwin et al., 2016; Millan, 2003). tion and, via enhanced ACh release, favour cognitive performance
Further, 5-HT3 blockade was suggested to participate in the facil- (Coupland et al., 1994; Millan, 2006). This reasoning led several
itation by vortioxetine of FCX levels of monoamines, possibly by groups to develop mixed α2-ARs/inhibitors of 5-HT and/or NA
blocking excitatory 5-HT3 receptors on GABAergic interneurons reuptake (Andrés et al., 2007; Cordi et al., 2001; Gobert et al.,
(Pehrson and Sanchez, 2014; Riga et al., 2016). This GABAergic 2002; Serres et al., 2012). One such agent is the spiroimidazoline
action may also lead to the disinhibiton of glutamatergic trans- S35966, a member of a small family of compounds that have high
missionand hence favour synaptic plasticity and cognitive func- affinity for both SERTs and NATs, yet negligible affinity for
tion (Pehrson and Sanchez, 2014; Riga et al., 2016). However, an DATs, together with high affinity for α2-ARs. The only other
induction of frontocortical ACh and histamine release by vortiox- activity of significance is, interestingly enough – and resembling
etine – triggered by its distinctive serotonergic properties – offers vortioxetine – antagonist properties at 5-HT3 receptors (pKi 7.5
a complementary substrate for pro-cognitive properties in pre- for S35966) (Cordi et al., 2001; Gobert et al., 2002). As shown in
clinical models (Pehrson et al., 2013; Sanchez et al., 2015). In Figure 6, this receptor profile is distinctive as compared with a
line with these observations, there are encouraging findings with broad range of antidepressants. This includes mianserin that
vortioxetine and cognitive performance both in preclinical mod- blocks α2-ARs (and 5-HT3) receptors but not SERT and which, in
els and in patients, notably elderly subjects (Katona et al., 2012; contrast to S35966, potently blocks α1-ARs and histamine H1
McIntyre et al., 2016). However, it remains to be seen in further receptors risking autonomic and sedative side-effects (Millan,
direct and rigorous comparative trials just how the influence of 2006). The accompanying Figure 7 shows that venlafaxine
vortioxetine on depressed mood, anxious symptoms and impaired inhibits NA neurons due to release of NA onto α2-ARs (Millan
cognition compares with other classes of antidepressant (Baldwin et al., 2001a),whereas S35966 is auto-protected from feedback-
et al., 2016; Thase et al., 2016). inhibition and preserves locus coeruleus firing by virtue of its

Downloaded from jop.sagepub.com at CORNELL UNIV on October 18, 2016


14 Journal of Psychopharmacology

Figure 6.  Overall binding profile of a novel class of mixed α2-AR antagonists/SNRIs in comparison to other classes of antidepressant agent.
S35966 – and highly-related “S” ligands – behave as inhibitors of 5-HT and NA reuptake (pInhibitory Concentration50 values shown for rat synaptosomes) and, in contrast
to venlafaxine, as antagonists at α2-ARs (pKb value shown for blockade of Gαi/o-protein coupling at hα2A-ARs). The comparable pKi values for S35966 at these sites and
for a dozen other sites underlying beneficial and deleterious actions of antidepressants (Gobert et al., 2002; Millan, 2006) were used to construct (by principal com-
ponent analysis) a dendrogramme: this formally revealed the distinctive overall receptor-binding profile of S35966 and its relatives compared to venlafaxine and other
“categories” of antidepressant. Note that, like venlafaxine and other SNRIs and in contrast to the other classes of antidepressant, S35966 had low affinity (pKi, < 5.5) for
histamine H1 receptors, α1-ARs and muscarinic M1 receptors eliciting autonomic/cardiovascular side-effects (Gobert et al., 2002; Millan, 2006).

antagonist properties at α2-AR autoreceptors. Correspondingly, unacceptable cardiovascular side-effects, in particular hyperten-
S35966 induces a profound increase in NA release in FCX (and sion due to blockade of α2-ARs on sympathetic terminals. Thus,
hippocampus) even greater than co-administration of venlafaxine despite this preclinical proof of concept, α2-AR autoreceptor
and a α2-AR antagonist. Likewise, S35966 is more effective than blockade for reinforcing the efficacy of SERT-NAT blockers has
venlafaxine in elevating DA levels in FCX (Gobert et al., 2002). not (yet) been translated into the clinic.
Conversely, the efficacy of S35966 and venlafaxine for increasing
5-HT levels in FCX is similar, possibly since α2-ARs on 5-HT
neurones are not tonically active – S35966 does not block 5-HT1A Blockade of 5-HT2C receptors for improving
or 5-HT1B/1D autoreceptors. However, this remains to be clarified. antidepressant efficacy
Compared to venlafaxine, the robust neurochemical profile of
S35966 translates into marked antidepressant properties, for As mentioned above, blockade of excitatory 5-HT2C receptors
example, in a forced swim test. It is also associated with the more on GABAergic interneurons inhibitory to dopaminergic and
rapid alteration of biomarkers of onset of antidepressant efficacy adrenergic pathways elevates release of DA and NA in the
upon chronic administration to rats: down-regulation of 5-HT2A FCX. Accordingly, selective antagonists and inverse agonists at
receptors, up-regulation of BDNF, and modulation of neuroplasti- 5-HT2C receptors, despite not changing 5-HT release in FCX,
city genes like Arc and Shank (Serres et al., 2012). Moreover, possess antidepressant properties and – probably reflecting
S35966 can be distinguished from venlafaxine by its induction of blockade of 5-HT2C sites in limbic regions like the hippocampus
ACh release in FXC and precognitive properties in preclinical and amygdala – they also display anxiolytic actions (Dekeyne
models (Gobert et al., 2002). et al., 2008; Di Giovianni and De Deurwaerdère, 2016; Millan,
In view of the rather striking profile of S35966 (Figure 7), it 2005b, 2006). However, no selective antagonist or inverse agonist
might not unreasonably be asked why it has not become available has ever been clinically developed. One factor that hampered
to patients, notably for those resistant to SSRIs and SNRIs. In clinical progress was the persistent lack of a positron emission
fact, it was not even evaluated in humans owing to concerns of tomography (PET) ligand to verify 5-HT2C receptor engagement

Downloaded from jop.sagepub.com at CORNELL UNIV on October 18, 2016


Millan et al. 15

Figure 7.  Overview of the influence of S35966 compared to venlafaxine on monoaminergic transmission in the FCX, and antidepressant properties in
the forced-swim test in rats.
Panel A, Activity of adrenergic cell bodies in the locus coeruleus: The α2-AR antagonist, atipamezole, blocks the inhibitory influence of venlafaxine which releases NA
onto α2-AR autoreceptors – intrinsically blocked by S35966 itself. Panel B, Extracellular levels of NA in FCX: atipamezole blocks α2-AR feedback inhibition by venlafaxine
to potentiate increases in levels of NA which are even more pronounced for S35966 alone – active dose-dependently from 0.16–10.0 mg/kg, s.c. (not shown). Panel C,
Extracellular levels of DA in FCX: S35966 has a more pronounced action than venlafaxine which via NA indirectly activates α2-ARs on dopaminergic terminals. Panel D,
Extracellular levels of 5-HT in FCX: no difference between the drugs, possibly since α2-ARs on serotonergic terminals are not tonically active. Panel E, Extracellular levels
of ACh in the FCX: S35966 elevates ACh by blocking inhibitory α2-ARs on their terminals. Panel F, Reduction of immobility in a forced-swim test of antidepressant proper-
ties: S35966 at equivalent doses to those increasing FCX levels of monoamines is more potent than venlafaxine and has a greater maximal effect. Drugs given i.v. for
Panel A, 3 min pre-measure; for Panels B to E, drugs were given s.c. as shown by arrows; and for Panel F, drugs were given s.c. three times, 24, 1 h and 30 min pre-test.
All data are means ± SEMs.*P < 0.01 for drug to vehicle, and # for S35966 versus Venlafaxine, in Newman–Keuls test following ANOVA. N = 6–8 per value.

in human subjects. In the case of S32006, its non-development (1-(1-phenylethyl)-spiro[indole-3,4′-piperidin]-2(1H)-one)


was also partly due to the pre-existence of agomelatine and the which was designed to retain the mixed 5-HT2C/SNRI-like prop-
decision to explore other avenues for exploiting 5-HT2C recep- erties of older agents like amitriptyline yet ditch their unfavour-
tors. For example, the union of α2-AR and 5-HT2C antagonist able actions at Na+/Ca++ channels, histamine H1 and muscarinic
properties into a single compound. By doing so, it was hoped to receptors (Millan, 2006). The appropriate binding profile was
mimic the beneficial effects of mianserin and mirtazapine, achieved without undue difficulty (pKi values for SERT/NAT/5-
which block both these sites, while avoiding their detrimental HT2C receptors 8.2/7.9/7.4 respectively versus less than 6.0 for
actions due to off-target effects at, as mentioned above, α1-ARs histamine H1 receptors, muscarinic M1 receptors and α1-ARs.
and histamine H1 receptors as well as, albeit less markedly, Further, S42141 robustly induced frontocortical release of NA,
muscarinic M1 receptors. DA and 5-HT, was active in tests of antidepressant activity in
This was realized with the discovery of S32212, which rodents to the same degree as amitriptyline and – in contrast to
revealed a promising preclinical profile in terms of its induction the latter – displayed anxiolytic properties (Millan et al., unpub-
of FCX levels of NA and DA and robust actions in tests of anti- lished observations). While S42141 escaped the fate of S32212
depressant, anxiolytic and pro-cognitive properties, yet it had to at ion channels, it was discarded for what is euphemistically ref-
be abandoned owing to unanticipated actions at ion channels ered to as “strategic” considerations.
that may have triggered cardiac side-effects (Dekeyne et al., Thus, it proved possible to design and validate new 5-HT2C
2012; Millan et al., 2012b). Another potential, dual-acting receptor-based agents for manipulating monoaminergic trans-
antidepressant based on 5-HT2C antagonism was S42141 mission in FCX, and to demonstrate using microdialysis their

Downloaded from jop.sagepub.com at CORNELL UNIV on October 18, 2016


16 Journal of Psychopharmacology

Figure 8.  A conceptual framework for the design of antidepressant agents acting either selectively or at pairs of monoaminergic and/or non-
monoaminergic target.
The figure highlights a selection of mechanisms that have been directly examined in the lab of the authors. Drugs in violet are selective and those in pink are dual-
acting. The latter include agomelatine which has been launched for the clinical treatment of depression. S32212 and S41744 have been extensively described in the
literature, S47445 has also recently been documented, while S35966 and S42141 are both outlined herein. All the actions shown would be expected to improve mood in
depression, though they may not be sufficient alone. In green are indicated some additional benefits of the non-monoaminergic property in question though there may
be others, and the actions indicated are not necessarily exclusive. For example, melatoninergic mechanisms may also reduce anxiety, while positive allosteric modulation
(PAM) of AMPA receptors may influence circadian rhythms.

pronounced reinforcement of frontocortical monoaminergic improves symptoms of anxiety as well as depressed mood
transmission. However, as so often in “R and D” for psychiatric (Stein et al., 2013).
disorders, they fell either at the intimidating hurdle of safety Several other novel concepts have been explored for the rein-
pharmacology or failed to gel with current development priori- forcement of monoaminergic transmission in the FCX by graft-
ties. More cheerfully, as noted below, greater luck was had with ing additional actions onto a core and validated monoaminergic
a further 5-HT2C-plus ligand, agomelatine. mechanism. For example, mixed neurokinin (NK1) receptors
antagonists/5-HT reuptake inhibitors where blockade of NK1
receptors moderates (by analogy to 5-HT autoreceptor blockade)
Coupling non-monoaminergic properties to the impact of 5-HT reuptake suppression on extracellular levels
monoaminergic actions of 5-HT in FCX – and in parallel increases the frontocortical
release of NA and DA (Gobert et al., 2009; Guiard et al., 2006;
As discussed elsewhere (Millan 2006, 2009, 2014a), a further
Millan et al., 2010). These and related agents have not yet
and inherently rich strategy for improving the clinically-vali-
reached patients, but this still appears to be a promising avenue
dated profiles of monoaminergic antidepressants, largely –
for progress.
though not only – articulated around blockade of SERTs,
In this light, as recently analyzed elsewhere (Millan, 2014a),
NATs and 5-HT2C receptors, is to integrate actions at non-
an important question is whether to either: (1) employ associa-
monoaminergic classes of receptor (Figure 8). One major exam-
tions of two separate drugs or (2) integrate dual activity into a
ple is provided by the combined melatonin MT1 and MT2 recep-
single compound. On balance, the latter approach is the more
tor agonist/5-HT2C receptor neutral antagonist, agomelatine,
promising, but the former is well adapted to drug reorientation.
launched for the treatment of depression in Europe (De Barardis
The most appropriate multi-target strategy will depend upon drug
et al., 2015; De Bodinat et al., 2010; Kamal et al., 2015; Millan
availability, the mechanism of action sought and the patient pop-
et al., 2003, 2005, 2011; Soumier et al., 2009). This agent has
ulations to be treated. Whatever the strategy, microdialysis evalu-
unfortunately been handicapped by the need to control for the
ation of the impact on frontocortical release of monoamines is
risk of rare hepatic side-effects (Perlemutter et al., 2016) but
likely to be a pivotal step in preclinical characterization.
remarkably it is still the only currently-available antidepressant
with a non-monoaminergic component of action. In addition to
the advantages of 5-HT2C receptor antagonism (see above), Coupling regulation of ACh release to
melatonergic properties were conceived to promote sleep and
resynchronize depressed patients with insomnia and disrupted
strategies for improving cognition
circadian rhythms (Catena-Dell’Osso et al., 2012; De Berardis As mentioned above, several monoaminergic mechanisms
et al., 2015; De Bodinat et al., 2010). Further, reflecting its (directly and indirectly) engaged by clinically-employed antide-
unique and synergistic mechanism of action, agomelatine pressants can promote release of ACh in the FCX, including

Downloaded from jop.sagepub.com at CORNELL UNIV on October 18, 2016


Millan et al. 17

5-HT1A partial agonism, 5-HT4 agonism and blockade of α2-ARs receptors which denigrates cognitive performance, blockade of
(Gobert et al., 2003; Millan, 2006; Millan et al., 1995; King et al., D3 receptors in the FCX is associated with a remarkably broad-
2008; Peñas-Cazorla and Vilaró, 2015; Sanchez et al., 2015). based and robust pattern of pro-cognitive activity in rodent and
This should promote performance in certain cognitive tasks, an primate models of pro-cognitive properties: moreover, pro-cog-
issue of importance since cognitive deficits in depression have nitive actions are at least partly expressed in the FCX and (3) the
been neglected relative to schizophrenia, yet they can sometimes potential antipsychotic properties of D3 receptor blockade would
be quite disabling (Marazitti et al., 2010; Millan et al 2012a). be of utility in the treatment of both schizophrenia and
In fact, α2-AR antagonist properties (possessed by mianserin Alzheimer’s disease, as well as psychotic depression (Gross
for example) may be a double-edged sword since frontocortical et al., 2013; Loiseau and Millan, 2009; Millan and Brocco, 2008;
populations of postsynaptic α2-AR (probably on pyramidal Millan et al., 2007a; Nakajima et al., 2013; Watson et al., 2012).
neurons) promote attention, working memory and certain other It is, thus, of interest that sub-maximal and just effective doses of
components of cognitive function (Coupland et al., 1994; Millan, 5-HT6 and D3 receptor antagonists cooperatively elevated levels
2006; Millan et al., 2015; Ramos and Arnsten, 2007; Robbins of ACh in the FCX (Figure 9). Both 5-HT6 and D3 receptor antag-
and Arnsten, 2009; Sallee, 2010). onists are effective alone in rat models of social and object recog-
Further, it remains difficult to extrapolate from pro-cognitive nition – expressing their actions in the FCX itself (Loiseau and
observations in rodent tests to real-world function in human Millan, 2009; Loiseau et al., 2008; Watson et al., 2012). Mirroring
patients (see below), and no antidepressant has yet been given a their neurochemical interaction, the association at sub-active
market authorization for restoring cognitive performance in doses of 5-HT6 and D3 receptor antagonists “synergistically”
patients (Millan et al., 2012a; Rosenblatt et al., 2015). In this improved performance in a model of social recognition. This
light, a particularly interesting compound to pursue will be vorti- interaction was confirmed by use of two different 5-HT6 antago-
oxetine which, as outlined above, releases ACh (as well as DA, nists (Figure 9).
NA and histamine) via serotonergic mechanisms without block- Although we have failed to find an influence of 5-HT6 antago-
ing muscarinic or other classes of receptor that may compromise nists upon extracellular levels of glutamate (Rivet, unpublished
potential pro-cognitive properties, and which has shown positive observations), this may reflect technical and kinetic limitations
effects on cognition in rodents and patients (Katona et al., 2012; (see above), so an influence on glutamatergic transmission should
McIntyre et al., 2016; Pehrson et al., 2013; Rosenblatt et al., not be excluded. Indeed, pro-cognitive properties of 5-HT6
2015; Sanchez et al., 2015). antagonists were abolished by the NMDA receptor antagonist,
One class of receptor that vortioxetine does not interact with dizocilpine, suggesting dependence on functionally-intact
is the 5-HT6 receptor. Antagonists at this site have been designed NMDA receptors that are known to modulate social cognition,
for the improvement of cognition primarily in Alzheimer’s dis- likely by actions in the FCX (De Bruin and Kruse 2015;
ease and schizophrenia, at least partially due to their enhance- Shimazaki et al., 2010). Interestingly, pro-cognitive properties of
ment of cholinergic transmission, and they are still in active D3 antagonists expressed in the FCX are dependent upon the
development, at least for the former indication (De Bruin and release of d-serine from astrocytes onto the co-agonist site of
Kruse, 2015; Hirst et al., 2006; King et al., 2008; Loiseau et al., NMDA receptors in FCX (Dallerac et al., 2014). This provides a
2008; Meffre et al., 2012). Indeed, they possess strikingly robust fascinating example of (D3/5-HT6 receptor driven) molecular
patterns of pro-cognitive properties in preclinical models (op. convergence in the control of cognition via glutamatergic/d-ser-
cit.) but, as pointed out above, the neurochemistry of 5-HT6 ine signalling at FCX-localized NMDA receptors.
receptors is complex and actions at GABAergic and glutamater- Dual dopamine D3/5-HT6 receptor antagonists are just one
gic as well as catecholaminergic pathways are relevant to the example of an unjustly neglected yet potentially important pal-
influence of 5-HT6 receptor ligands upon cognition (De Bruin ette of designer multi-target drugs for promoting release of ACh
and Kruse, 2015; Kendall et al., 2011; King et al., 2008; Schechter and other transmitters controlling cognition in the FCX, and
et al., 2008). hence countering cognitive dysfunction in depression and other
We have found a facilitatory influence of 5-HT6 antagonists CNS disorders.
upon FCX (and hippocampal) release of DA and NA (Figure 9),
and inhibition of GABAergic interneurons may be at least
partially responsible for this observation (De Bruin and Kruse Influence of potential glutamatergic
2015; Schechter et al., 2008). On the other hand, and rather sur- drugs for treating depression on
prisingly, the influence of 5-HT6 antagonists upon FCX levels of extracellular levels of monoamines
ACh (De Bruin and Kruse 2015, Hirst et al., 2006) has proven to
be significant yet quite modest under the rigorous dialysis condi-
and amino acids in FCX
tions (no cholinesterase inhibitors) that we have developed. Thus, The pervasive contribution of glutamatergic mechanisms to fron-
by analogy to multi-target agents for relieving depressed mood, tocortical networks controlling mood, cognition and other func-
the question arises as to whether 5-HT6 antagonism might best be tions disrupted in depressed states, and evidence for a perturbation
coupled to complementary pharmacological properties for fur- of glutamatergic signalling in depression, has kindled interest in
ther strengthening the induction of ACh release in FCX and, potential “glutamatergic” drugs for treating depression. In this
accordingly, pro-cognitive properties. regard, it is useful to recall the notion of a push/pull (accelerator/
One promising candidate would be blockade of dopamine D3 brake) influence of glutamatergic projections on monoaminergic
receptors (Saavedra et al., 2016) since: (1) this mechanism pathways: that is direct, excitatory actions counterbalanced
consistently reinforces frontocortical (though not hippocampal) and finely-tuned by indirect, GABAergic interneuron-mediated
cholinergic transmission; (2) in contrast to antagonism of D2 inhibition. This can explain some at first sight contradictory

Downloaded from jop.sagepub.com at CORNELL UNIV on October 18, 2016


18 Journal of Psychopharmacology

Figure 9.  Complementary 5-HT6 and D3 receptor antagonist properties for promoting cognitive performance: a neurochemical and behavioural
characterization in rats.
Panel A, Influence of the selective 5-HT6 receptor antagonist, SB271,046, upon extracellular levels of monoamines in the FCX. Panel B, Influence of SB271,046, and of
the selective D3 receptor antagonist, S33084, alone and together upon extracellular levels of ACh in the FCX. Panel C, Influence of combined administration of SB271,046
and S33084 upon social recognition in rats. Both these drugs are dose-dependently (0.16–10.0 and 0.01–0.63 mg/kg respectively) active in this procedure (Loiseau
et al., 2008; Loiseau and Millan, 2009; Millan et al., 2007): they were combined at sub-active doses to test for synergy. Panel D, Influence of combined administration
of another selective 5-HT6 receptor antagonist, SB258,585 plus S33084, upon social recognition. Panel E: Blockade of the pro-cognitive properties of S271,046 in the
social recognition test by the NMDA receptor channel blocker, dizocilpine. Drugs given s.c. For Panels A and B, arrows indicate times of drug administration. For Panels
C to E, T2 (test two) − T1 (test one, 2 h earlier) is the difference in exploration time during a 5 min text by the treated adult rat of the same conspecific. A minus score
indicates good retention in the second test (low exploration rate as familiar). In control experiments when a new rat is presented (not shown), T2 − T1 is close to zero
(see Figure 10 and Loiseau and Millan, 2009). The drugs were given 1 min after T1. All data are means ± SEMs. *P < 0.01 for SB271,046 to vehicle in Panel A. For Panels
B to D, #P < 0.01 for SB271,046/S33084 versus SB271,046 or S33084 alone in Newman–Keuls test following ANOVA. In Panel E, P < 0.01 for Dizocilpine/SB271,046 vs
Vehicle/SB271,016. N = 6–8 per value.

observations such as why both a NMDA receptor antagonist and we have seen no influence of mGluR5 antagonists like “MPEP”
antagonist may activate dopaminergic and adrenergic transmis- upon 5-HT levels in dialysates of FCX (Table 2). We likewise
sion (Table 2) (Celada et al., 2013; Millan, 2005a). observed little influence on extracellular levels of DA and NA in
While potential antidepressant actions of mGluR2 receptor FCX, supporting a mechanism of action distinct from conven-
antagonists and mGluR7 receptor agonists have been evoked tional antidepressants, observations well aligned with the work of
(Campo et al., 2011; Millan, 2014a; Palucha et al., 2007), a larger Lindemann et al (2015) on another mGluR5 antagonist. As
body of evidence has focussed on blockade of mGluR5 receptors shown in Table 2 and consistent with actions downstream of glu-
as a strategy for treating depression (Hughes et al., 2012; tamatergic terminals, MPEP exerted negligible influence upon
Lindemann et al., 2015; Pałucha-Poniewiera et al., 2013; dialysate levels of most amino acids, including taurine and PEA,
Pomierny-Chamioło et al., 2010) This focus partly reflects their underpinning the benign influence of mGluR5 antagonists on
functional coupling to NMDA receptors, but also compound overall cellular function and integrity (Inta et al., 2012). Indeed,
availability since mGluR5 antagonists have been intensively the only effect seen was a modest decrease in GABA levels, pre-
studied in Fragile X syndrome and, more recently, Alzheimer’s sumably reflecting relief on an excitatory mGluR5 tone on
disease (Dölen et al., 2010; Kumar et al., 2015; Millan 2013). GABAergic neurones (Sarihi et al., 2008).
Antidepressant actions of mGluR5 antagonists have been As mentioned above, mGluR5 and NMDA receptors are func-
reported in a variety of rodent models, and it has been suggested tionally coupled, and there is evidence that an interference with
that serotonergic mechanisms may be involved in their effects activity at NMDA receptors is involved in the antidepressant
(Fukumoto and Chaki, 2015). However, not all studies agree and actions of mGluR5 antagonists (Fukumoto and Chaki, 2015;

Downloaded from jop.sagepub.com at CORNELL UNIV on October 18, 2016


Millan et al. 19

Hughes et al., 2012; Pałucha-Poniewiera et al., 2013; Pomierny- is attractive, either as “dual-acting ligands” or as a combinatin of
Chamioło et al., 2010). This renders comparisons with the drugs (Abdallah et al., 2015; Fitzpatrick et al., 2016; Li et al.,
NMDA channel blocker ketamine of interest. Ketamine is cur- 2003; Millan, 2014a).
rently enjoying something of renaissance since it rapidly allevi-
ates depressed mood upon i.v. (and possibly intranasal)
administration to humans, and also has pronounced antidepres- Influence of glycine reuptake
sant actions in rodents (Park et al., 2015). In contrast to the inhibitors, glycine and d-serine, upon
mGluR5 antagonist, MPEP, ketamine enhances frontocortical dialysis levels of amino acids and
release of NA and DA – even at lowish doses. A prompt elevation
in frontocortical DA and NA release may be related to the rapid-
monoamines in FCX
onset antidepressant actions of ketamine, though a role for sero- Inhibitors of glycine reuptake (Gly-RI) elevate extracellular lev-
tonergic transmission should not be excluded, especially at els of glycine and hence indirectly promote glycine-mediated
higher yet clinically-relevant doses (Du Jardin et al., 2016). activation of the co-agonist glycine B site on NMDA receptors
The decrease in extracellular levels of GABA seen with keta- (Millan, 2005a). It has been suggested – based on preclinical
mine (Table 2) – albeit at a high dose – agrees with previous observations – that they may be useful in the treatment of depres-
studies suggesting that it suppresses tonic NMDA receptor sion (Pałucha-Poniewiera et al., 2013; Park et al., 2015). In this
(NR2B subtype) excitation of GABAergic interneurons. As a rule regard, it is of note that the Gly-RI, sarcosine, exerts partial ago-
of thumb, NMDA receptor channel blockers like ketamine have nist actions at NMDA receptors but it is unclear how this action
more robust actions than competitive NMDA receptor antago- relates to potential antidepressant properties (Chen et al., 2015;
nists like CPP which did not affect levels of GABA (Table 2). Park et al., 2015). Conversely, the modest increase by sarcosine
Nonetheless, in this light, it is apposite to mention “off-target” and another Gly-RI, ORG 24598, of extracellular levels of DA in
actions of ketamine independent of NMDA receptors which FCX may more readily be assimiliated into a positive influence
should not be neglected in interpreting its actions (Abdallah upon depressed mood (Table 2) (Alberati et al., 2012; Nagy et al.,
et al., 2015; Gobert et al., 2011; Park et al., 2015; Zhou et al., 2010).
2015). At a high dose, ketamine elevated levels of glutamate and It is worth stressing the selectivity of the actions of sarcosine
aspartate in FCX, confirming previous work with NMDA recep- inasmuch as levels of d-serine and glutamate were unchanged at
tor channel blockers, and presumably reflecting a loss of doses that elevated dialysis levels of glycine (Figure 11 and Table
GABAergic inhibition (Table 2) (Abdallah et al., 2015; Park 2). Though sarcosine provoked an increase in taurine levels, phe-
et al., 2015; Zhou et al., 2015). It is thought that glutamate nylethanolamine was not – in contrast to NMDA – affected,
released by ketamine downstream of GABAergic neurones underscoring the more benign impact of sarcosine on cellular
recruits AMPA receptors that themselves stimulate BDNF and integrity (Table 2). Collectively, these data support the selectivity
mammalian target of rapamycin (mTOR) signalling, leading to of sarcosine for glycine transporters and for increasing glycine
expression of antidepressant properties (Abdallah et al., 2015; levels at doses equivalent to those used clinically as an adjunct
Park et al., 2015). Interestingly, independently of glutamate to antipsychotics – albeit with mitigated success (Chue, 2013;
release, ketamine metabolites also exert antidepressant properties Heresco-Levy and Javitt, 2004; Kantrowitz and Javitt, 2010;
via recruitment of AMPA receptors, though it is not clear exactly Millan, 2005a). These findings are not trivial since, to our knowl-
how they exert their actions (Zanos et al., 2016). edge, they comprise the first comprehensive characterization of
The presumptive role of AMPA receptors in the antidepressant its neurochemical profile – decades after initiation of clincical
actions of ketamine (though it is not certain that they are sufficient trials! (Gobert et al., 2011; Kantrowitz and Javitt, 2010).
rather than just necessary) has rekindled interest in AMPA recep- As regards ORG 24598 (Brown et al., 2001), it likewise
tors themselves as targets for antidepressant agents (Farley et al., exerted little influence on amino acids other than glycine, again
2010; Li et al., 2003; Nations et al., 2012; Park et al., 2015; Zanos supporting the safety and selectivity of Gly-RIs (Figure 11)
et al 2016). For example, the AMPA receptor positive allosteric (Alberati et al., 2012). Findings in schizophrenia are not to our
modulator (PAM), S47445, is under development for the treat- knowledge available for ORG 24598, despite preclinical and
ment of cognitive and mood deficits in Alzheimer’s disease clinical support with bitopertine (as monotherapy) for the hypoth-
(Bretin et al., 2015). In rat procedures of antidepressant proper- esis that Gly-RIs may improve negative and cognitive symptoms
ties, AMPA receptor PAMs like LY392,098 are active at doses (Alberati et al., 2012; Bugarski-Kirola et al., 2014; Heresco-Levy
corresponding to those expressing their pro-cognitive properties and Javitt, 2004; Kantrowitz and Javitt, 2010; Liem-Moolenaar
(Figure 10). As for the underlying neurochemical mechanisms, et al., 2010; Umbricht et al., 2014). Less positively, bitopertine
LY392,098 did not affect NA or 5-HT levels in FCX dialysates, seems to have an inverted dose–response curve and, despite
though it did exert a mild increase in DA levels suggesting that encouraging Phase II observations, Phase III findings were
dopaminergic mechanisms in the FCX might be involved in the ambivalent (Bugarski-Kirola et al., 2014; Umbricht et al., 2014).
antidepressant actions of AMPA receptor PAMs: this awaits con- Nonetheless, neurochemistry was crucial in getting the drug to
firmation. LY392,098 exerted little influence on amino acids patients, and it was elegantly shown by measuring glycine in
(Table 2), in line with the notion that it acts downstream of human CSF that the doses of bitopertine used in clinical trials
the NMDA receptor/GABAergic interneurone/glutamatergic axis were equivalent to those yielding positive neurochemical and
engaged by ketamine. Furthermore, the lack of influence of behavioural effects in animals (Alberati et al., 2012; Bugarski-
LY392,098 upon taurine and PEA supports a lack of excitotoxic- Kirola et al., 2014; Hofmann et al., 2016; Umbricht et al., 2014).
ity. Finally, AMPA receptor PAMs may enhance the actions of By analogy to the Gly-RI, sarcosine, glycine itself has
SSRIs and, as depicted in Figure 8, the notion of their association been extensively evaluated as an adjunct in schizophrenia

Downloaded from jop.sagepub.com at CORNELL UNIV on October 18, 2016


20 Journal of Psychopharmacology

Figure 10.  Influence of AMPA receptor positive allosteric modulators upon monoaminergic transmission in the FCX in comparison to antidepressant
and pro-cognitive properties in rats.
Panel A, Influence of the AMPA receptor PAM, LY392,098, on extracellular levels of 5-HT in FCX in comparison to the SSRI, citalopram. Panel B, Influence of LY392,098
on extracellular levels of NA in FCX in comparison to the NARI, reboxetine. Panel C, Influence of LY392,098 on extracellular levels of DA in FCX in comparison to the DA
reuptake inhibitor, bupropion. Panel D, Facilitation by LY392,098 of the recognition of a familiar conspecific in a social recognition procedure. For a description of the
procedure, see legend to Figure 8 and Loiseau et al. (2008). Panel E, Influence of LY392,098 on immobility in a forced swim test of antidepressant properties. Drugs were
given 24 h, 1 h and 30 min prior to the test. Panel F, Influence of another AMPA PAM, LY404,187, on immobility in a forced swim test employing the same procedure.
Drugs were given i.p., except for Panel F. All data are means ± SEMs. *P < 0.01 for drug versus vehicle in Newman–Keuls test following ANOVA. N = 6–8 per value.

(Heresco-Levy and Javitt, 2004; Kantrowitz and Javitt, 2010), (Gobert et al., 2011). Interestingly, glycine provoked a significant
yet its neurochemical profile has never been thoroughly charac- increase in extracellular levels of DA and NA in FCX. Though
terized. Using our novel dialysis system, it was found that doses we have not formally analyzed the underlying mechanisms, this
comparable with those used clinically elicited a colossal increase reinforcement in frontocortical dopaminergic transmission – which
in dialysis level of glycine, dwarfing the effects of Gly-RIs mirrors the effects of some mixed dopaminergic/serotoninergic
(Figure 11) (Alberati et al., 2012; Gobert et al., 2011; Kantrowitz drugs for treating psychosis (Millan et al., 1998a,b) – might be
and Javitt, 2010). It is unclear whether this observation suggests interpreted as supporting the utility of Gly-RIs and glycine B
that Gly-RIs do not attain levels sufficiently high for robust clini- ligands for control of the cognitive and negative symptoms of
cal effects or, more likely, that the effect of glycine itself on schizophrenia (Bugarski-Kirola et al., 2014; Kantrowitz and
extracellular glycine – assuming that extrapolation to humans is Javitt, 2010; Millan, 2005a; Umbricht et al., 2014; Yang and
valid – is extremely high. Also striking is the influence of glycine Svensson, 2008). However, this remains to be proven and clinical
upon a broad range of amino acids, including a decrease in d- data with glycine in schizophrenia have been (as for the glycine
serine release. This may be compensatory for the elevation in B site partial agonist, d-cyloserine) ambivalent (Heresco-Levy
glycine levels/enhanced NMDA receptor engagement, and a and Javitt, 2004; Kantrowitz and Javitt, 2010).
reduction in d-serine levels was also seen upon administration of Finally, in contrast to this broad pattern of changes seen with
NMDA (Table 2). By analogy to NDMA, the diminution in d-serine glycine, d-serine – which has also yielded chequered results as an
release induced by glycine was blunted by the competitive adjunct in schizophrenia (Heresco-Levy and Javitt, 2004;
NMDA receptor antagonist, CPP, suggesting partial and media- Kantrowitz and Javitt, 2010) – was remarkably selective in exclu-
tion by NMDA receptors (Gobert et al., 2011; Rivet, unpublished sively increasing dialysate levels of d-serine (without affecting
observations). It should also be mentioned that the decline in l-serine) (Gobert et al., 2011). This suggests that these two agents
d-serine levels was specific in that l-serine was not affected should not be considered as interchangeable, in line with their

Downloaded from jop.sagepub.com at CORNELL UNIV on October 18, 2016


Millan et al. 21

Figure 11.  Influence of glycine, d-serine and two glycine reuptake inhibitors upon extracellular levels of DA, NA, glycine and d-serine in FCX in rats.
Panels A and B, Dose-dependent influence of glycine upon dialysis levels of DA and NA. (5-HT was not affected (not shown). Panel C, Dose-dependent influence of
glycine compared to d-serine and the two glycine reuptake inhibitors, sarcosine and ORG 24598, upon dialysis levels of glycine in FCX. Panel D, Dose-dependent influence
of d-serine compared to glycine, sarcosine and ORG 24598, upon dialysis levels of d-serine in FCX. Note that glycine decreases the levels of d-serine whereas d-serine does
not affect levels of glycine. All data are means ± SEMs. *P < 0.01 for drug versus vehicle in Newman–Keuls test following ANOVA. N = 6–8 per value.

contrasting patterns of action at NMDA receptor subtypes and term refers to the use in animals of procedures and readouts,
functional profiles (Danysz and Parsons, 1998; Fossat et al., models and measures that are as close to and as relevant as pos-
2012). Moreover, these data are reassuring as regards the safety of sible to those used in humans in order to optimize the transition
d-serine as an agent for probing glycine B receptor function. On from drug discovery to clinical drug development (Hendriksen
the other hand, d-serine should not be regarded as entirely benign and Groenink, 2015; Kas et al., 2011; Millan, 2008; Pratt et al.,
inasmuch as over-production and over-activation of endogenous 2012). Arguably, this is exactly what pharmacologists and
pools has been implicated in the pathophysiology of Alzheimer’s clinicians have been trying to do since the very advent of drug
disease and may, together with Aβ42 and Tau, serve as a useful discovery – though the trendy buzz-word “translational” was
biomarker for this disorder (Madeira et al., 2015). only invented much later. “Translational research” is not, then, a
Collectively, the above observations underpin the importance conceptual revolution at all, but rather a procedural revision inas-
of microdialysis: (1) for evaluating the influence of glutamater- much as we have, thanks to technological progress, a broader
gic agents upon FCX function and determining their potential range of possibilities available than several decades ago – and
therapeutic pertinence and (2) for measuring extracellular levels indeed in 2000. There is also far greater understanding of the
of amino acids and exploring their relevance to the pathogenesis neural substrates of CNS disorders and of the mechanisms of
and treatment (by other drug classes) of CNS disorders. action of therapeutics - information which helps guide transla-
tional studies. Finally, partly due to many failures in the clinical
development of drugs for treating CNS disorders, the mindset has
Translating insights from FCX changed. There is a much wider perception of the importance of
neurochemistry into humans: a work reciprocally bridging animal and human studies, and of the need
for two-way traffic: human studies driving preclinical designs
in progress and vice versa (Bespalov et al., 2016; Millan et al., 2015).
“Translational” has become something of a catchword in recent This is epitomized by the “MATRICS/CNTRICS” initiative to
years, and it is sometimes used for almost anything and every- harmonize preclinical and clinical measures of cognitive perfor-
thing that is done pre-clinically in terms of its ostensible rele- mance and its modulation by potential medication (Buchanan
vance to – and pursuit by – clinical studies. More precisely, the et al., 2010; Millan and Bales, 2013; Young et al., 2009). Further,

Downloaded from jop.sagepub.com at CORNELL UNIV on October 18, 2016


22 Journal of Psychopharmacology

functional magnetic resonance imaging (fMRI) is being “back- Thus, it is clearly possibly to expand the translation of experi-
translated” (albeit laboriously) into mice and rats; it is relatively mental neurochemistry/microdialysis of the FCX and other cer-
easy to undertake parallel electroencephalographic studies in ebral regions, but there is a need for additional work along these
rodents and humans; “PK/PD studies” of drug exposure are lines.
becoming de rigeur; and PET, imaging and other measures of
target engagement are quasi-mandatory (Millan, 2008; Millan
et al., 2015; Murphy and Mackay, 2011; Sakkalis, 2011; Progress and perspectives: concluding
Whittington et al., 2010; Vyas et al., 2011). comments
It is not unreasonable then to ask how microdialysis and neu-
rochemistry in rodents can be translationally advanced into, and Considerable progress has been made, and continues to be made,
inform, clinical characterization of novel chemical entities. in our understanding of the FCX by the application of microdi-
In fact, this continues to present challenges, not least since one alysis coupled to increasingly refined and broad-based methods
cannot implant probes into the brains of human volunteers or for quantification of biologically-active molecules from neuro-
patients – other than those undergoing neurosurgical procedures transmitters to hormones to metabolites to growth factors to sec-
– participating in trials of drug efficacy. Nonetheless, there are ond messengers to neurotoxic proteins. As outlined here, the
several possibilities for progress. lion’s share of work to date has been devoted, not altogether sur-
First, as pointed out above, in elegant and rigorous studies of prisingly, to neurotransmitters. Nonetheless, neurotoxic proteins
the novel Gly-RI, bitopertine, its influence upon extracellular incriminated in the “spreading” of neurological disorders, such as
levels of glycine in the FCX of rodents was compared with its Aβ42 and tau – as well as other proteins like “homeobox” mes-
influence upon their levels in the CSF of humans and compara- sengers (Prochiantz and Di Nardo, 2015) – may well become a
bility established as a foundation for selecting doses for clinical major focus of future work. Cytokines and other pro and anti-
trials (Bugarski-Kirola et al., 2014; Hofmann et al., 2016; inflammatory mediators will also attract considerable interest in
Umbricht et al., 2014). This general approach, complemented by view of evidence that neuroinflammation is a prominent feature
measures of the drug itself, can be applied to other biologically- of both psychiatric and neurological disorders (Millan et al.,
active molecules impacted by pharmacotherapy. This may well 2016; Yamanaka et al., 2015).
prove possible in the near future as regards neurotoxic Aβ42 and A rather new class of intercellular mediators derived from
Tau which can be reliably measured in human CSF, and by neurons, astrocytes and microglia, are microRNAs: they are
microdialysis in the interstitial fluid of rodents (see above) highly stable in the extracellular environment – since they are
(Brinkmalm et al., 2015; Olsson et al., 2016; Takeda et al., 2016). bound to argonaut proteins and/or protected in exosomes (Bartel,
Second, PET also has a broader role than just in the definition 2009; Danborg et al., 2014; Millan, 2011, 2013, 2014b).
of drug target engagement, notably in the extension of neuro- MicroRNAs are of considerable interest due to their key roles in
chemical work in rodents to studies in humans. One example is coordinating neuronal function at the cell and circuit level, their
provided by PET ligands at D2/D3 receptors: interference with release by neurones and other cell types, and their promise as
their binding potential can be used as a surrogate measure of the potential CSF and plasma biomarkers of depression, Alzheimer’s
availability of endogenous DA and hence its extracellular levels. and other diseases (Bartel, 2009; Danborg et al., 2014; Millan,
This has proven useful in studies of schizophrenia to estimate the 2014b). It is recommended and predicted that miRNAs and other
(increased) occupation of sub-cortical D2 receptors (FCX D2/D3 classes of ncRNA should become important – and translatable –
receptor density is too low to be visualized) by endogenous DA themes of future research into FCX function using microdialysis
and hence deduce a hyperactivity/hypersensitivity of striatal and other approaches. Underpinning this contention, it has now
dopaminergic pathways (Kegeles et al., 2010). The approach can been reported that microRNAs can be recuperated and quantified
also be applied to drugs that release DA such as agents for treat- in cerebral microdialysates in humans (Bache et al., 2005).
ing attention deficit hyperactivity disorder, like methylphenidate, Incorporation of the above-mentioned and other important
which acts via actions in the FCX (Robbins and Arnsten, 2009; molecules into microdialysis studies will be facilitated by ongo-
Millan et al., 2015; Sallee, 2010). ing progress in the sensitivity and precision of methods of detec-
Third, glutamatergic and GABAergic systems can be exam- tion and quantification. In this regard, it would be instructive to
inedby magnetic resonance spectroscopy. While most studies undertake studies across the entire cascade of neurotransmission
have focused on a composite “Glx” (glutamate + glutamine or in time and in space: for example, in determining the influence of
glutamate + glutamine + GABA) signal, technical advances are a novel potential antidepressant not only upon extracellular lev-
helping facilitate their differentiation, and other metabolites can els of 5-HT and glutamate, but also upon second messengers like
be determined concurrently, such as the acetylcholine catabolite, cGMP and cAMP, as well as, in the context of chronic adminis-
choline (Poels et al., 2014; Stzrelecki et al., 2015). One drawback tration, BDNF. Such work could be coupled to complementary
is that results reflect levels in whole tissue, with little or no studies of intracellular events, like signalling cascades, kinase
distinction between intracellular, intra and extrasynaptic com- recruitment and gene transcription. The question of whether keta-
partments: hence it remains unclear to what extent measurements mine (and related compounds) can achieve sustained as well as
genuinely reflect neurotransmission and its modulation, in par- rapid-onset relief of depression is still under discussion (Park
ticular for GABA (Myers et al., 2016; Poels et al., 2014). et al., 2015), accentuating the importance of examining the long-
Finally, to conclude on a simple note, microdialysis quantifi- term as well as short-term effects of novel agents; work which is
cation of central levels of hormones can be reproduced without still insufficiently undertaken today.
any great difficulty in humans by measures of their levels in As for the practicalities of measurements themselves, while
either the CSF or plasma. conventional one to several hours periods of measurement are

Downloaded from jop.sagepub.com at CORNELL UNIV on October 18, 2016


Millan et al. 23

well-adapted to studies of pharmacological agents – and models reciprocal and fruitful exchanges with other experimental (and
of CNS disorders where baseline levels are important – it would clinical) domains. The setting up of a neurochemistry/microdialysis
be interesting to shorten sample times to have greater insights lab requires expertise, commitment, patience and a significant
into the dynamics of events in the extracellular/peri-synaptic financial investment. Nonetheless, in the long-term, the running
space. At the other end of the temporal scale, as shown above for of a focussed yet flexible and polyvalent microdialysis unit is not
hormones, it would also be important to systematically examine only instructive and productive, but less expensive and more effi-
fluctuations across the daily cycle, (Figure 3) both as regards the cient than the piecemeal farming out of specific studies to outside
influence of potential medication (which may be administered organizations.
either in the mornings and/or evenings (De Bodinat et al., 2010)) As amply illustrated above, enormous progress has been
and the characteristics of genetically-modified mice and rodent made since the turn of the Millenium (see Millan et al., 2000a,
models for CNS disorders. Two simple examples extracted from this journal) in the application of microdialysis to our under-
the above discussion are: first, AMPA receptor PAMs, since standing of frontocortical networks, their disruption in CNS dis-
AMPA receptors provide a major optic drive to the master pace- orders, and the characterization of novel therapeutic strategies for
maker in the suprachiasmatic nucleus (Mizoro et al., 2010) and, their treatment. However, there is still plenty of progress that can
second, animal models for depression, bearing in mind the and should be made. A microdialysis approach to studying the
problem of desynchronization encountered by many patients neurochemistry of the FCX and other cerebral structures remains
(Catena-Dell’Osso et al., 2012). a translationally-relevant and crucial pillar of modern neurosci-
Another important domain of future progress would be to ence and drug-discovery for both psychiatric and neurological
better interlink the neurochemistry of the FCX with studies of its disorders.
global network function as revealed by electroencephalography
and fMRI (Millan et al., 2012a). As pointed out above, microdi- Acknowledgements
alysis is complementary to these strategies which provide little
direct information on the precise neurochemical substrates We thank all our many colleagues and collaborators for their excellent
underpinning circuit operation, disruption and restitution work over the years. In particular, L Cistarelli and R Billiras for their
(Murphy and Mackay, 2011). devotion and their virtuoso performance and analysis of microdialysis
Moving in the opposite direction in terms of scale – though studies in rats and mice. G Bouchez, R Boulanger, B Di Cara, L De
sticking to networks – a further domain of progress would be to Groote, F Panayi and G Rollin-Jego are also thanked for very fruitful
systematically study neuromodulators in the extracellular envi- participation with microdialysis; A Dekeyne, M Brocco, F Lejeune, F
Loiseau, C Mannoury la Cour and A Newman-Tancredi are thanked for
ronment of primary cultures of FCX and especially of cortical
important contributions to the characterization of drugs discussed herein,
neurons, networks and organoids derived from human (and and A Cordi, G Lavielle, G de Nanteuil and JL Peglion and colleagues are
murine) induced pluripotent cells. Studies could be undertaken thanked for long-term support with excellent medicinal chemistry.
both in cells from normal subjects and in patients with CNS dis- Note this paper has been written in accordance with the new NbN
orders impacting the FCX; under baseline conditions and upon nomenclature guidelines that Journal of Psychopharmacology now
exposure to known and potentially novel classes of medication endorses: http://nbnomenclature.org/_static/docs/NbN_Glossary.pdf
(Brennand et al., 2015).
Though frontocortical organoids derived from human induced Declaration of conflicting interests
pluripotent cells are still somewhat esoteric, the more common
The authors declared the following potential conflicts of interest with
and critical use of microdialysis would be in the thorough charac-
respect to the research, authorship, and/or publication of this article: All
terization of animal models for CNS disorders, especially under the authors are full-time employees of the Institut de Recherche Servier
the influence of both symptomatic and disease-modifying drugs. and have no other interests to declare.
Such studies are reasonably well-established in, say, schizophre-
nia research (Millan et al., 2016), yet they are still very thin on
the ground and urgently need intensification with animal models
Funding
forAlzheimer’s disease and other neurological disorders. And not The authors received no financial support for the research, authorship,
just Aβ42 and Tau: there is a need to maintain a broad perspec- and/or publication of this article.
tive. For example, it would be naïve in the extreme to imagine
that one should only be interested in dosing neurotoxic proteins References
when developing a potential Tau or β-amyloid disease-modifying Abdallah CG, Sanacora G, Duman RS, et al. (2015) Ketamine and rapid-
therapy for Alzheimer’s disease: just as for symptomatic therapy, acting antidepressants: a window into a new neurobiology for mood
it is critical to look at relevant neurotransmitters from monoam- disorder therapeutics. Annu Rev Med 66: 509–523.
ines to ACh to glutamate. This assertion is underpinned by the Adolphs R (2009) The social brain: neural basis of social knowledge.
crucial insights provided by microdialysis studies of the influ- Annu Rev Psychol 60: 693–716
ence of vortioxetine on neurotransmitters other than 5-HT Alberati D, Moreau JL, Lengyel J, et al. (2012) Glycine reuptake inhibi-
(Sanchez et al., 2015), and of l-DOPA upon neurotransmitters tor RG1678: a pharmacologic characterization of an investigational
other than DA (De Deurwaerdère et al., 2016). agent for the treatment of schizophrenia. Neuropharmacology 62:
1152–1161.
Finally, from a pragmatic and economic point of view, where
Alexander GE, DeLong MR and Strick PL (1986) Parallel organization
feasible, the most effective and informative way to pursue micro- of functionally segregated circuits linking basal ganglia and cortex.
dialysis and neurochemistry is hand in hand with complementary Annu Rev Neurosci 9: 357–381.
strategies for model and drug characterization. Feedback is Alvarsson A, Zhang X, Stan TL, et al. (2015) Modulation by Trace
instantaneous and dynamic when in-house, profiting from Amine-Associated Receptor 1 of Experimental Parkinsonism,

Downloaded from jop.sagepub.com at CORNELL UNIV on October 18, 2016


24 Journal of Psychopharmacology

L-DOPA Responsivity, and Glutamatergic Neurotransmission. allosteric modulator of AMPA type glutamate receptors. Eur Neuro-
J Neurosci 35: 14057–14069. psychopharmacol 25(Suppl. 2): S248–S249.
Andrés JI, Alcázar J, Alonso JM, et al. (2007) Tricyclic isoxazolines: Brinkmalm A, Portelius E, Öhrfelt A, et al. (2015) Explorative and tar-
identification of R226161 as a potential new antidepressant that com- geted neuroproteomics in Alzheimer’s disease Biochim Biophys Acta
bines potent serotonin reuptake inhibition and alpha2-adrenoceptor 1854: 769–778.
antagonism. Bioorg Med Chem 15: 3649–3660. Brown A, Carlyle I, Clark J, et al. (2001) Discovery and SAR of ORG
Anticevic A, Corlett PR, Cole MW, et al. (2015) N-methyl-D-aspartate 24598 - a selective glycine uptake inhibitor. Bioorg Med Chem Lett
5-HT1A receptor antagonist effects on prefrontal cortical connectiv- 11: 2007–2009.
ity better model early than chronic schizophrenia. Biol Psychiatry Buchanan RW, Keefe RS, Umbricht D, et al. (2010) The FDA-NIMH-
77: 569–580. MATRICS guidelines for clinical trial design of cognitive-enhancing
Ashby CR Jr, Kehne JH, Bartoszyk GD, et al. (2013) Electrophysiologi- drugs: what do we know 5 years later? Schizophr Bull 37: 1209–
cal evidence for rapid 5-HT1A autoreceptor inhibition byvilazodone, 1217.
a 5-HT1A receptor partial agonist and 5-HT reuptake inhibitor. Eur J Bugarski-Kirola D, Wang A, Abi-Saab D, et al. (2014) A phase II/III trial
Pharmacol 714: 359–365. of bitopertin monotherapy compared with placebo in patients with
Bache S, Rasmussen R, Rossing M, et al. (2015) Detection and quantifica- an acute exacerbation of schizophrenia - results from the CandleLyte
tion of microRNA in cerebral microdialysate. J Transl Med 13: 149. study. Eur Neuropsychopharmacol 24: 1024–1036.
Baldwin DS, Florea I, Jacobsen PL, et al. 2016 A meta-analysis of the Bukovskaya O and Shmukler A (2016) Oxytocin and social cognition
efficacy of vortioxetine in patients with major depressive disorder and schizophrenia: a systematic review. Psychiatr Q 87: 521–543.
(MDD) and high levels of anxiety symptoms. J Affective Disord 206: Campo B, Kalinichev M, Lambeng N, et al. (2011) Characterization of an
140–150. mGluR2/3 negative allosteric modulator in rodent models of depres-
Bang-Andersen B, Ruhland T, Jørgensen M, et al. (2011) Discovery of sion. J Neurogenet 25: 152–166.
1-[2-(2,4-dimethylphenylsulfanyl)phenyl]piperazine (Lu AA21004): Castro-Hernández J, Afonso-Oramas D, Cruz-Muros, et al. (2015) Pro-
a novel multimodal compound for the treatment of major depressive longed treatment with pramipexole promotes physical interaction of
disorder. J Med Chem 54: 3206–3222. striatal dopamine D3 autoreceptors with dopamine transporters to
Barbier AJ, Aluisio L, Lord B, et al. (2007) Pharmacological character- reduce dopamine uptake Neurobiol Dis 74: 325–335.
ization of JNJ-28583867, a histamine H3 receptor antagonist and Catena-Dell’Osso M, Marazziti D, Rotella F, et al. (2012) Emerging tar-
serotonin reuptake inhibitor. Eur J Pharmacol 576: 43–45. gets for the pharmacological treatment of depression: focus on mela-
Barnes NM and Sharp T (1999) A review of central5-HT receptors and tonergic system. Curr Med Chem 19: 428–437.
their function. Neuropharmacology 38: 1083–1152. Celada P, Puig M, Amargós-Bosch M, et al. (2004) The therapeutic role
Barr MS, Farzan F, Rusjan PM, et al. (2009) Potentiation of gamma oscil-
of 5-HT1A and 5-HT2A receptors in depression. J Psychiatry Neu-
latory activity through repetitive transcranial magnetic stimulation
rosci 29: 252–265.
of the dorsolateral prefrontal cortex. Neuropsychopharmacology 34:
Celada P, Puig MV and Artigas F (2013) Serotonin modulation of corti-
2359–2367.
cal neurons and networks. Front Integr Neurosci 2013 Apr 19; 7: 25.
Bartel DP (2009) MicroRNAs: target recognition and regulatory func-
doi: 10.3389/fnint.2013.00025.
tions. Cell 136: 215–233.
Cerqueira JJ, Maillet F, Almeida OF, et al. (2007) The prefrontal cortex
Béracochéa D, Tronche C, Coutan M, et al. (2011) Interaction between
as a key target of the maladaptive response to stress J Neurosci 27:
diazepam and hippocampal corticosterone after acute stress: impact
2781–2787.
on memory in middle-aged mice. Front Behav Neurosci 5: 14.
Chen KT, Tsai MH, Wu CH, et al. (2015) AMPA Receptor-mTOR acti-
Bergh MS, Bogen IL, Lundanes E, et al. (2016) Validated methods for
vation is required for the antidepressant-like effects of Sarcosine dur-
determination of neurotransmitters and metabolites in rodent brain
ing the forced swim test in rats: Insertion of AMPA Receptor may
tissue and extracellular fluid by reversed phase UHPLC-MS/MS.
play a role. Front Behav Neurosci 2015; 9: 162.
J Chromatogr B: Anal Technol Biomed Life Sci 1028: 120–129.
Bespalov A, Steckler T, Altevogt B, et al. (2016) Failed trials for central Chue P (2013) Glycine reuptake inhibition as a new therapeutic approach
nervous system disorders do not necessarily invalidate preclinical in schizophrenia: focus on the glycine transporter 1 (GlyT1). Curr
models and drug targets. Nat Rev Drug Discovery 15: 516–519. Pharm Des 19: 1311–1320.
Blier P and de Montigny C (1994) Current advances and trends in the Cirrito JR, May PC, O’Dell MA, et al. (2003) In vivo assessment of
treatment of depression Trends Pharmacol Sci 15: 220–226. brain interstitial fluid with microdialysis reveals plaque-associated
Blier P and El Mansari M. (2013) Serotonin and beyond: therapeutics changes in amyloid-beta metabolism and half-life. J Neurosci 23:
for major depression. Philos Trans R Soc London B: Biol Sci 368: 8844-8853.
20120536. Clough GF (2005). Microdialysis of large molecules. AAPS J 7: E686–E692.
Bonaventure P, Kelly L, Aluisio L, et al. (2007) Selective blockade of Collo G, Bono F, Cavalleri L, et al. (2012) Pre-synaptic dopamine D(3)
5-hydroxytryptamine (5-HT)7 receptors enhances 5-HT transmission, receptor mediates cocaine-induced structural plasticity in mesence-
antidepressant-like behavior, and rapid eye movement sleep suppres- phalic dopaminergic neurons via ERK and Akt pathways. J Neuro-
sion induced by citalopram in rodents. J Pharmacol Exp Ther 321: chem 120: 765–778.
690–698. Coupland NJ, Bailey JE, Wilson SJ, et al. (1994) A pharmacodynamic
Bonhomme N, De Deurwaèrdere P, Le Moal M, et al. (1995). Evidence study of the alpha2-adrenergic receptor antagonist ethoxyidazoxan
for 5-HT4 receptor subtype involvement in the enhancement of stria- in healthy volunteers. Clin Pharmacol Ther 56: 420–429.
tal dopamine release induced by serotonin: a microdialysis study in Cordi AA, Berque-Bestel I, Persigand T, et al. (2001) Potential antide-
the halothane-anesthetized rat. Neuropharmacology. 34: 269–279. pressants display combined α2-adrenoceptor antagonist and mono-
Bouchez G, Millan MJ, Rivet JM, et al. (2012) Quantification of extra- amine uptake inhibitor properties. J Med Chem 44: 787–805.
cellular levels of corticosterone in the basolateral amygdaloid com- Cremers TI, Flik G, Folgering JH, et al. (2016) Development of a rat
plex of freely-moving rats: a dialysis study of circadian variation and plasma and brain extracellular fluid pharmacokinetic model for
stress-induced modulation. Brain Res 1452: 47–46. bupropion and hydroxybupropion based on microdialysis sampling,
Brennand KJ, Marchetto MC, Benvenisty N, et al. (2015) Creating and application to predict human brain concentrations. Drug Metab
patient-specific neural cells for the in vitro study of brain disorders. Dispos 44: 624–633.
Stem Cell Rep 5: 933–945. Cremers TI, Flik G, Hofland C, et al. (2012) Microdialysis evaluation of
Bretin S, Waller D, Martinez Diaz Guerra G, et al. (2015) Antidepres- clozapine and N-desmethylclozapine pharmacokinetics in rat brain
sant- and anxiolytic-like properties of S47445, a novel positive Drug Metab Dispos 40: 1909–1916.

Downloaded from jop.sagepub.com at CORNELL UNIV on October 18, 2016


Millan et al. 25

Cunha C, Brambilla R and Thomas KL (2010) A simple role for BDNF Droste SK, Collins A, Lightman SL, et al. (2009) Distinct, time-depen-
in learning and memory? Front Mol Neurosci 3: 1. dent effects of voluntary exercise on circadian and ultradian rhythms
Curry K and Stasio MJ (2009) The effects of energy drinks alone or with and stress responses of free corticosterone in the rat hippocampus.
alcohol on neuropsychological functioning. Hum Psychopharmcol Endocrinology 150: 4170–4179.
Clin Exp 24: 473–481. Du Jardin KG, Müller HK, Elfving B, et al. (2016) Potential involvement
Dallerac G, Turpin F, Morisot N, et al. (2014) Absence of D-serine of serotonergic signaling in ketamine’s antidepressant actions: A criti-
results in abnormal dopamine–glutamate dialogue in the prefrontal cal review. Prog Neuropsychopharmacol Biol Psychiatry 71: 27–38.
cortex Eur Neuropsychopharmacol 24: S126. Dwivedi Y, Roy B, Lugli G, et al. (2015) Chronic corticosterone-medi-
Danborg PB, Simonsen AH, Waldemar G, et al. (2014) The potential of ated dysregulation of microRNA network in prefrontal cortex of rats:
microRNAs as biofluid markers of neurodegenerative diseases—a relevance to depression pathophysiology. Transl Psychiatry 5: e682.
systematic review. Biomarkers 19: 259–268. Ejsing TB, Pedersen AD and Linnet K (2005) P-glycoprotein interaction
Danysz W and Parsons CG (1998) Glycine and N-methyl-D-aspartate with risperidone and 9-OH-risperidone studied in vitro, in knock-out
receptors: physiological significance and possible therapeutic appli- mice and in drug-drug interaction experiments. Hum Psychophar-
cations. Pharmacol Rev 50: 597–666. macol 20: 493–500.
De Berardis D, Fornaro M, Serroni N, et al. (2015) Agomelatine beyond El Mansari M, Crnic A, Oosterhof C, et al. (2015) Long-term administra-
borders: current evidences of its efficacy in disorders other than tion of the antidepressant vilazodone modulates rat brain monoami-
major depression. Int J Mol Sci 16: 1111–1130. nergic systems. Neuropharmacology 99: 696–704.
De Bodinat C, Guardiola-Lemaitre B, Mocaer E, et al. (2010) Agomela- Erhardt S, Olsson SK and Engberg G (2009) Pharmacological manipula-
tine, the first melatonergic antidepressant: discovery, characteriza- tion of kynurenic acid: potential in the treatment of psychiatric disor-
tion and development Nat Rev Drug Discovery 9: 628–642. ders. CNS Drugs 23: 91–101.
De Bruin NM and Kruse CG (2015) 5-HT6 receptor antagonists: poten- Errico F, D’Argenio V, Sforazzini F, et al. (2015) A role for D-aspartate
tial efficacy for the treatment of cognitive impairment in schizophre- oxidase in schizophrenia and in schizophrenia-related symptoms
nia. Curr Pharm Des 21: 3739–3759. induced by phencyclidine in mice. Transl Psychiatry 5: e512. DOI:
De Deurwaerdère P, Di Giovanni G and Millan MJ (2016) Expanding 10.1038/tp.2015.2.
the repertoire of L-DOPA’s actions: A comprehensive review of its Eulenburg V and Gomeza J (2010) Neurotransmitter transporters
functional neurochemistry. Prog Neurobiol. Epub ahead of print 4 expressed in glial cells as regulators of synapse function. Brain Res
July 2016. DOI: 10.1016/j.pneurobio.2016.07.00. Rev 63: 103–112.
Deguchi Y (2002) Application of in vivo brain microdialysis to the study Farley S, Apazoglou K, Witkin JM, et al. (2010) Antidepressant-like
of blood-brain barrier transport of drugs. Drug Metab Pharmacokin effects of an AMPA receptor potentiator under a chronic mild stress
17: 395–407. paradigm. Int J Neuropsychopharmacol 13: 1207–1218.
De Groote L, Klompmakers AA, Olivier B, et al. (2003) Role of extra- Fitzpatrick CM, Larsen M, Madsen LH, et al. (2016) Positive allosteric
cellular serotonin levels in the effect of 5-HT1B receptor blockade. modulation of α-amino-3-hydroxy-5-methyl-4-isoxazolepropionic
Psychopharmacology (Berlin) 167: 153–158. acid glutamate receptors differentially modulates the behavioural
Dekeyne A, Brocco M, Loiseau F, et al. (2012) S32212, a novel sero- effects of citalopram in mouse models of antidepressant and anxio-
tonin type 2C receptor inverse agonist/α2-adrenoceptor antagonist lytic action. Behav Pharmacol 27: 549–555.
and potential antidepressant: II. A behavioral, neurochemical, and Flik G, Dremencov E, Cremers TI, et al. (2011) The role of cortical and
electrophysiological characterization. J Pharmacol Exp Ther 340: hypothalamic histamine-3 receptors in the modulation of central his-
765–780. tamine neurotransmission: an in vivo electrophysiology and micro-
Dekeyne A, Mannoury la Cour C, Gobert A, et al. (2008) S32006, a novel dialysis study. Eur J Neurosci 34: 1747–1755.
5-HT2C receptor antagonist displaying broad-base antidepressant and Flik G, Folgering JH, Cremers TI, et al. (2015) Interaction between brain
anxiolytic properties in rodent models. Psychopharmacology 199: histamine and serotonin, norepinephrine, and dopamine systems: in
549–568. vivo microdialysis and electrophysiology study. J Mol Neurosci 56:
De Lange EC, Ravenstijn PG, Groenendaal D, et al. (2005) Toward the 320–328.
prediction of CNS drug-effect profiles in physiological and patho- Fossat P, Turpin FR, Sacchi S, et al. (2012) Glial D-serine gates NMDA
logical conditions using microdialysis and mechanism-based phar- receptors at excitatory synapses in prefrontal cortex. Cereb Cortex
macokinetic-pharmacodynamic modelling.AAPS J 7: E532–E543. 22: 595–606.
De Quervain DJ, Aerni A, Schelling G, et al. (2009) Glucocorticoids and Fu Y, Zhao D, Pan B, et al. (2015) Proteomic analysis of protein expres-
the regulation of memory in health and disease. Front Neuroendo- sion throughout disease progression in a mouse model of Alzheim-
crinol 30: 358–370. er’s disease. J Alzheimer’s Dis 47: 915–926.
Diaz SL, Narboux-Nême N, Boutourlinsky K, et al. (2016) Mice lacking Fukumoto K and Chaki S (2015) Involvement of serotonergic system in
the serotonin 5-HT2B receptor as an animal model of resistance to the effect of a metabotropic glutamate 5 receptorantagonist in the
selective serotonin reuptake inhibitors antidepressants. Eur Neuro- novelty-suppressed feeding test. J Pharmacol Sci 127: 57–61.
psychopharmacol 26: 265–279. Gardier AM, Malagie I, Trillat AC, et al. (1996) Role of 5-HT1A autore-
Di Cara B, Maggio R, Aloisi G, et al. (2011) Genetic deletion of trace ceptors in the mechanism of action of serotoninergic antidepressant
amine 1 receptors reveals their role in auto-inhibiting the actions of drugs: recent findings from in vivo microdialysis studies. Fundam
ecstasy (MDMA) J Neurosci 31: 16928–16940 Clin Pharmacol 10: 16–27.
Di Chiara G (1990) In vivo brain dialysis of neurotransmitters. Trends Gobert A, Billiras R, Cistarelli L, et al. (2004) Quantification and phar-
Pharmacol Sci 11: 116–121. macological characterization of dialysate levels of noradrenaline in
Di Chiara G, Tanda G and Carboni E (1996). Estimation of in-vivo neu- the striatum of freely-moving rats: release from adrenergic terminals
rotransmitter release by brain microdialysis: the issue of validity. and modulation by alpha2-autoreceptors. J Neurosci Methods 140:
Behav Pharmacol 7, 640–657. 141–152.
Di Giovanni G and De Deurwaerdère P (2016). New therapeutic oppor- Gobert A, Brocco M, Dekeyne A, et al. (2009) Neurokinin1 antagonists
tunities for 5-HT2C receptor ligands in neuropsychiatric disorders. potentiate antidepressant properties of serotonin reuptake inhibitors,
Pharmacol Ther 57: 125–162. yet blunt their anxiogenic actions: a neurochemical, electrophysio-
Dölen G, Carpenter RL, Ocain TD, et al. (2010) Mechanism-based logical, and behavioral characterization. Neuropsychopharmacology
approaches to treating fragile X. Pharmacol Ther 127: 78–93 34: 1039–1056.

Downloaded from jop.sagepub.com at CORNELL UNIV on October 18, 2016


26 Journal of Psychopharmacology

Gobert A, Cussac D, Lejeune F, et al. (2002) The novel antidepressant, Hirst WD, Stean TO, Rogers DC, et al. (2006) SB-399885 is a potent,
S35966, is a mixed serotonin and noradrenaline reuptake inhibitor selective 5-HT6 receptor antagonist with cognitive enhancing prop-
and an antagonist at α2-adrenoceptors. Eur Neuropsychopharmacol erties in aged rat water maze and novel object recognition models.
12(Suppl 3), 248. Eur J Pharmacol 553: 109–119.
Gobert A, Di Cara B, Cistarelli L, et al. (2003) Piribedil enhances fron- Hjorth S and Auerbach SB (1996) 5-HT1A autoreceptors and the mode
tocortical and hippocampal release of acetylcholine in freely moving of action of selective serotonin reuptake inhibitors (SSRI). Behav
rats by blockade of alpha 2A-adrenoceptors: a dialysis comparison Brain Res 73: 281–283.
to talipexole and quinelorane in the absence of acetylcholinesterase Hjorth S, Bengtsson HJ, Kullberg A, et al. (2000) Serotonin autorecep-
inhibitors. J Pharmacol Exp Ther 305: 338–346. tor function and antidepressant drug action. J Psychopharmacol 14:
Gobert A and Millan MJ (1999a) Modulation of dialysate levels of 177–185.
dopamine, noradrenaline, and serotonin (5-HT) in the frontal cor- Hjorth S and Sharp T (1993) In vivo microdialysis evidence of central
tex of freely-moving rats by (−)-pindolol alone and in association serotonin1A and serotonin1B autoreceptor blocking properties of the
with 5-HT reuptake inhibitors: comparative roles of beta-adrenergic, beta adrenoceptor antagonist, (−)-penbutolol J Pharmacol Exp Ther
5-HT1A, and 5-HT1B receptors. Neuropsychopharmacology 21: 265: 707–712.
268–284. Hofmann C, Pizzagalli F, Boetsch C, et al. (2016) Effects of the glycine
Gobert A and Millan MJ (1999b) Serotonin (5-HT)2A receptor activa- reuptake inhibitors bitopertin and RG7118 on glycine in cerebrospi-
tion enhances dialysate levels of dopamine and noradrenaline, but nal fluid: results of two proofs of mechanism studies in healthy vol-
not 5-HT, in the frontal cortex of freely-moving rats. Neuropharma- unteers. Psychopharmacology (Berlin) 233: 2429–2439.
cology 38: 315–317. Holmes A and Wellman CL (2009) Stress-induced prefrontal reorganiza-
Gobert A, Rivet JM, Audinot V, et al. (1998) Simultaneous quantifica- tion and executive dysfunction in rodents. Neurosci Biobehav Rev
tion of serotonin, dopamine and noradrenaline levels in single frontal 33: 773–783.
cortex dialysates of freely-moving rats reveals a complex pattern of Hows ME, Lacroix L, Heidbreder C, et al. (2004) High-performance
reciprocal auto- and heteroreceptor-mediated control of release. Neu- liquid chromatography/tandem mass spectrometric assay for the
roscience 84: 413–429. simultaneous measurement of dopamine, norepinephrine, 5-hydroxy-
Gobert A, Rivet JM, Billiras R, et al. (2011) Simultaneous quantifica- tryptamine and cocaine in biological samples. J Neurosci Methods
tion of D- vs. L-serine, taurine, kynurenate, phosphoethanolamine 138: 123–132.
and diverse amino acids in frontocortical dialysates of freely-moving Hows ME, Organ AJ, Murray S, et al. (2002) High-performance liquid
rats: differential modulation by N-methyl-D-aspartate (NMDA) and chromatography/tandem mass spectrometric assay for the rapid high
other pharmacological agents. J Neurosci Methods 202: 143–157. sensitivity measurement of basal acetylcholine from microdialy-
Gobert A, Rivet JM, Cistarelli L, et al. (1999) Buspirone modulates basal sates. J Neurosci Methods 121: 33–39.
and fluoxetine-stimulated dialysate levels of dopamine, noradrena- Hughes ZA, Neal SJ, Smith DL, et al. (2012) Negative allosteric modu-
line and serotonin in the frontal cortex of freely moving rats: activa- lation of metabolic glutamate receptor 5 results in broad spectrum
tion of serotonin1A receptors and blockade of α2-adrenergic receptors activity relevant to treatment resistant depression. Neuropharmacol-
underlie its actions. Neuroscience 93: 1251–1262. ogy 66: 202–214.
Gobert A, Rollin-Jego G, Iop F, et al. (2014) Age-dependent changes Hughes ZA, Starr KR, Langmead CJ, et al. (2005) Neurochemical evalu-
of γ-amyloid and tau protein in two mouse models of Alzheimer’s ation of the novel 5-HT1A receptor partial agonist/serotonin reup-
Disease. Soc Neurosci Abstr 40: 788.21. take inhibitor, vilazodone. Eur J Pharmacol 510: 49–57.
Goodson JL and Thompson RR (2010) Nonapeptide mechanisms of Hulstaert F, Blennow K, Ivanoiu A, et al. (1999) Improved discrimina-
social cognition, behavior and species-specific social systems Curr tion of AD patients using beta-amyloid(1-42) and tau levels in CSF.
Opin Neurobiol 20: 784–794. Neurology 52: 1555–1562.
Graef S, Schönknecht P, Sabri O, et al. (2011) Cholinergic receptor sub- Ichikawa J, Dai J and Meltzer HY (2000) Acetylcholinesterase inhibitors
types and their role in cognition, emotion, and vigilance control: An are neither necessary nor desirable for microdialysis studies of brain
overview of preclinical and clinical findings. Psychopharmacology acetylcholine. Curr Sep 19: 37–44.
215: 205–229. Inta D, Filipovic D, Lima-Ojeda JM, et al. (2012) The mGlu5 receptor
Gross G, Wicke K and Drescher KU (2013) Dopamine D3 receptor antag- antagonist MPEP activates specific stress-related brain regions and
onism – still a therapeutic option for the treatment of schizophrenia. lacks neurotoxic effects of the NMDA receptor antagonist MK-801:
Naunyn-Schmiedeberg’s Arch Pharmacol 386: 155–166. significance for the use as anxiolytic/antidepressant drug. Neuro-
Guiard BP, Lanfumey L and Gardier AM (2006) Microdialysis approach pharmacology 62: 2034–2039.
to study serotonin outflow in mice following selective serotonin Iqbal K, Liu F and Gong CX (2016) Tau and neurodegenerative disease:
reuptake inhibitors and substance P (neurokinin 1) receptor antago- the story so far. Nat Rev Neurol 12: 15–27.
nist administration: a review. Curr Drug Targets 7: 187–201. Jordan S, Kramer GL, Zukas PK, et al. (1994) In vivo biogenic amine
Hajós M, Gartside SE, Varga V, et al. (2003) In vivo inhibition of neuronal efflux in medial prefrontal cortex with imipramine, fluoxetine, and
activity in the rat ventromedial prefrontal cortex by midbrain-raphe fluvoxamine. Synapse 18: 294–297.
nuclei: role of 5-HT1A receptors. Neuropharmacology 45: 72–81. Josserand V, Pélerin H, de Bruin B, et al. (2006). Evaluation of drug
Hendriksen H and Groenink L (2015) Back to the future of psychophar- penetration into the brain: A double study by in vivo imaging with
macology: A perspective on animal models in drug discovery. Eur J positron emission tomography and using an in vitro model of the
Pharmacol 1759: 30–41. human blood-brain barrier. J Pharmacol Exp Ther 316: 79–86.
Henry JD, von Hippel W, Molenberghs P, et al. (2016) Clinical assess- Joyce JN and Millan MJ (2007) Dopamine D3 receptor agonists for pro-
ment of social cognitive function in neurological disorders. Nat Rev tection and repair in Parkinson’s disease. Curr Opin Pharmacol 7:
Neurol 12: 28–39. 100–105.
Heresco-Levy U and Javitt DC (2004) Comparative effects of glycine Kamal M, Gbahou F, Guillaume JL, et al. (2015) Convergence of mela-
and D-cycloserine on persistent negative symptoms in schizophre- tonin and serotonin (5-HT) signaling at MT2/5-HT2C receptor het-
nia: a retrospective analysis. Schizophr Res 66: 89–96. eromers J Biol Chem 290: 11537–11534.
Hertel P, Nomikos GG and Svensson TH (1999) Idazoxan preferentially Kantrowitz JT and Javitt DC (2010) N-methyl-D-aspartate (NMDA)
increases dopamine output in the rat medial prefrontal cortex at the receptor dysfunction or dysregulation: the final common pathway on
nerve terminal level. Eur J Pharmacol 371: 153–158. the road to schizophrenia? Brain Res Bull 83: 108–121.

Downloaded from jop.sagepub.com at CORNELL UNIV on October 18, 2016


Millan et al. 27

Kas MJ, Krishnan V, Gould TD, et al. (2011) Advances in multidisci- Li X, Witkin JM, Need AB, et al. (2003) Enhancement of antidepressant
plinary and cross-species approaches to examine the neurobiology potency by a potentiator of AMPA receptors. Cell Mol Neurobiol
of psychiatric disorders. Eur Neuropsychopharmacol 21: 532–544. 23: 419–430.
Katona C, Hansen T and Olsen CK (2012) A randomized, double-blind, Liem-Moolenaar M, Zoethout RW, de Boer P, et al. (2010) The effects
placebo-controlled, duloxetine-referenced, fixed-dose study compar- of the glycine reuptake inhibitor R213129 on the central nervous
ing the efficacy and safety of Lu AA21004 in elderly patients with system and on scopolamine-induced impairments in psychomotor
major depressive disorder. Int Clin Psychopharmacol 27: 215–223. and cognitive function in healthy subjects. J Psychopharmacol 24:
Kegeles LS, Abi-Dargham A, Frankle WG, et al. (2010) Increased syn- 1671–1679.
aptic dopamine function in associative regions of the striatum in Lindemann L, Porter RH, Scharf SH, et al. (2015) Pharmacology of
schizophrenia. Arch Gen Psychiatry 67: 231–239. basimglurant (RO4917523, RG7090), a unique metabotropic glu-
Kehr J, Yoshitake T, Wang FH, et al. (2001) Microdialysis in freely- tamate receptor 5 negative allosteric modulator in clinical develop-
moving mice: determination of acetylcholine, serotonin and nor- ment for depression. J Pharmacol Exp Ther 353: 213–233.
adrenaline release in galanin transgenic mice. J Neurosci Methods Loiseau F and Millan MJ (2009) Blockade of dopamine D3 receptors in
109: 71–80. frontal cortex, but not in sub-cortical structures, enhances social rec-
Kendall I, Slotten HA, Codony X, et al. (2011) E-6801, a 5-HT6 receptor ognition in rats: similar actions of D1 receptor agonists, but not of D2
agonist, improves recognition memory by combined modulation of antagonists. Eur Neuropsychopharmacol 19: 23–33.
cholinergic and glutamatergic neurotransmission in the rat. Psycho- Loiseau F, Dekeyne A and Millan MJ (2008) Pro-cognitive effects of
pharmacology 213, 413–430. 5-HT6 receptor antagonists in the social recognition procedure in
Kim M, Lee JG, Yang CH, et al. (2016) Silica stationary phase-based rats: implication of the frontal cortex. Psychopharmacology (Berlin)
on-line sample enrichment coupled with LC-MS/MS for the quan- 196: 93–104.
tification of dopamine, serotonin and their metabolites in rat brain Lupien SJ, McEwen BS and Gunnar MR (2009) Effects of stress through-
microdialysates. Anal Chim Acta 923: 55–65. out the lifespan on the brain, behaviour and cognition. Nat Rev Neu-
King MV, Marsden CA and Fone KC (2008) A role for the 5-HT(1A), rosci 10: 434–445.
5-HT4 and 5-HT6 receptors in learning and memory. Trends Phar- Mabrouk OS and Kennedy RT (2012) Simultaneous oxytocin and arg-
macol Sci 29: 482–492. vasopressin measurements in microdialysates using capillary liq-
Kumar A, Dhull DK and Mishra PS (2015) Therapeutic potential of uid chromatography-mass spectrometry. J Neurosci Methods 209:
mGluR5 targeting in Alzheimer’s disease. Front Neurosci 9: 215. 127–133.
DOI: 10.3389/fnins.2015.00215. McDevitt RA and Neumaier JF (2011) Regulation of dorsal raphe
Kusek M, Sowa J, Kamińska K, et al. (2015) 5-HT7 receptor modulates nucleus function by serotonin autoreceptors: a behavioral perspec-
GABAergic transmission in the rat dorsal raphe nucleus and controls tive. J Chem Neuroanat 41: 234–246.
cortical release of serotonin. Front Cell Neurosci 9: 324. McIntyre RS, Harrison J, Loft H, et al. (2016) The effects of Vortioxetine
Labrie V and Roder JC (2010) The involvement of the NMDA receptor on cognitive function in patients with major depressive disorder: a
D-serine/glycine site in the pathophysiology and treatment of schizo- meta-analysis of three randomized controlled trials. Int J Neuropsy-
phrenia. Neurosci Biobehav Rev 34: 351–372. chopharmacol. Epub ahead of print 24 August 2016. DOI: 10.1093/
Lam VM, Espinoza S, Gerasimov AS, et al. (2015) In vivo pharmacol- ijnp/pyw055.
ogy of Trace-Amine Associated Receptor 1. Eur J Pharmacol 763: Madeira C, Lourenco MV, Vargas-Lopes C, et al. (2015) D-serine levels
136–142. in Alzheimer’s disease: implications for novel biomarker develop-
Lane RM (2015) Antidepressant drug development: Focus on triple ment. Transl Psychiatry; 5: e561. DOI: 10.1038/tp.2015.52.
monoamine reuptake inhibition. J Psychopharmacol 29: 526–544. Marazziti D, Consoli G, Picchetti M, et al. (2010) Cognitive impairment
Launay JM, Schneider B, Loric S, et al. (2006) Serotonin transport and in major depression. Eur J Pharmacol 626: 83–86.
serotonin transporter-mediated antidepressant recognition are con- Marklund N, Blennow K, Zetterberg H, et al. (2009) Monitoring of
trolled by 5-HT2B receptor signaling in serotonergic neuronal cells brain interstitial total tau and beta amyloid proteins by microdi-
FASEB J 20: 1843–1854. alysis in patients with traumatic brain injury. J Neurosurg 110:
Ledonne A, Federici M, Giustizieri M, et al. (2010) Trace amines depress 1227–1237.
D(2)-autoreceptor-mediated responses on midbrain dopaminergic Marklund N, Farrokhnia N, Hånell A, et al. (2014) Monitoring of
cells. Br J Pharmacol 160: 1509–1520. β-amyloid dynamics after human traumatic brain injury. J Neu-
Lejeune F, Audinot V, Gobert A, et al. (1994) Clozapine inhibits sero- rotrauma 31: 42–55.
toninergic transmission by an action at α1-adrenoceptors not at Meffre J, Chaumont-Dubel S, Mannoury la Cour C, et al. (2012) 5-HT(6)
5-HT1A receptors. Eur J Pharmacol 260: 79–88. receptor recruitment of mTOR as a mechanism for perturbed cogni-
Lejeune F and Millan MJ (1998) Induction of burst firing in ventral tion in schizophrenia. EMBO Mol Med 4: 1043–1056.
tegmental area dopaminergic neurons by activation of serotonin Millan MJ (2003) The neurobiology and control of anxious states. Prog
(5-HT)1A receptors: WAY 100,635-reversible actions of the highly Neurobiol 70: 83–244.
selective ligands, flesinoxan and S 15535. Synapse 30: 172–180. Millan MJ (2005a) N-Methyl-D-aspartate receptors as a target for
Lejeune F, Newman-Tancredi A, Audinot V, et al. (1997) Interactions improved antipsychotic agents: novel insights and clinical perspec-
of (+)- and (−)-8- and 7-hydroxy-2-(di-n-propylamino)tetralin at tives. Psychopharmacology (Berlin) 179: 30–53.
human (h)D3, hD2 and h serotonin1A receptors and their modula- Millan MJ (2005b) Serotonin 5-HT2C receptors as a target for the treat-
tion of the activity of serotoninergic and dopaminergic neurones in ment of depressive and anxious states: focus on novel therapeutic
rats. J Pharmacol Exp Ther 280: 1241–1249. strategies. Therapie 60: 441–460.
Leviel V, Gobert A and Guibert B (1989) Direct observation of dopamine Millan MJ (2006) Multi-target strategies for the improved treatment of
compartmentation in striatal nerve terminal by ‘in vivo’ measurement depressive states: Conceptual foundations and neuronal substrates,
of the specific activity of released dopamine. Brain Res 499: 205–213. drug discovery and therapeutic application. Pharmacol Ther 110:
Lewis DA (2012) Cortical circuit dysfunction and cognitive deficits in 135–370.
schizophrenia - implications for preemptive interventions. Eur J Millan MJ (2008) The discovery and development of pharmacotherapy
Neurosci 35, 1871–1878. for psychiatric disorders: a critical survey of animal and translational
Lewis J, McGowan E, Rockwood J, et al. (2000) Neurofibrillary tangles, models and perspectives for their improvement. In: McArthur RA
amyotrophy and progressive motor disturbance in mice expressing and Borsini F (eds) Animal and Translational Models for CNS Drug
mutant (P301L) tau protein. Nat Genet 25: 402–405. Discovery. London: Academic Press, vol. 1, pp. 1–57.

Downloaded from jop.sagepub.com at CORNELL UNIV on October 18, 2016


28 Journal of Psychopharmacology

Millan MJ (2009) Dual- and triple-acting agents for treating core and co- improving the treatment of psychiatric disorders. Eur Neuropsycho-
morbid symptoms of major depression: novel concepts, new drugs. pharmacol 25: 599–656.
Neurotherapeutics 6: 53–77. Millan MJ, Lejeune F, De Nanteuil G, et al. (2001b) Selective blockade
Millan MJ (2011) MicroRNA in the regulation and expression of seroto- of NK1 receptors facilitates the activity of adrenergic pathways pro-
nergic transmission in the brain and other tissues. Curr Opin Phar- jecting to frontal cortex and dorsal hippocampus in rats. J Neuro-
macol 11: 11–22. chem 76: 1949–1954.
Millan MJ (2013) An epigenetic framework for neurodevelopmental dis- Millan MJ, Lejeune F and Gobert A (2000a) Reciprocal autorecep-
orders: from pathogenesis to therapy. Neuropharmacology 62: 2–82. tor and heteroreceptor control of serotonergic, dopaminergic and
Millan MJ (2014a) On ‘polypharmacy’ and multi-target agents, comple- noradrenergic transmission in the frontal cortex: relevance to
mentary strategies for improving the treatment of depression: a com- the actions of antidepressant agents. J Psychopharmacol 14:
parative appraisal. Int J Neuropsychopharmacol 17: 1009–1037. 114–138.
Millan MJ (2014b) The epigenetic dimension of Alzheimer’s disease: Millan MJ, Lejeune F, Gobert A, et al. (2000b) S18616, a highly potent
causal, consequence, or curiosity? Dialogues Clin Neurosci 16: spiroimidazoline agonist at alpha(2)-adrenoceptors: II. Influence on
373–379. monoaminergic transmission, motor function, and anxiety in com-
Millan MJ and Bales KL (2013) Towards improved animal models for parison with dexmedetomidine and clonidine. J Pharmacol Exp Ther
evaluating social cognition and its disruption in schizophrenia: the 295: 1206–1222.
CNTRICS initiative Neurosci Biobehav Rev 37: 2166–2180. Millan MJ, Mannoury la Cour C, Chanrion B, et al. (2012b) S32212,
Millan MJ and Brocco M (2008) Cognitive impairment in schizophrenia: a novel serotonin type 2C receptor inverse agonist/α2-adrenoceptor
a review of developmental and genetic models, and pro-cognitive antagonist and potential antidepressant: I. A mechanistic character-
profile of the optimised D(3) > D(2) antagonist, S33138. Therapie ization. J Pharmacol Exp Ther 340: 750–764.
63: 187–229. Millan MJ, Marin P, Kamal M, et al. (2011) The melatonergic ago-
Millan MJ, Agid Y, Brüne M, et al. (2012a) Cognitive dysfunction in psy- nist and clinically active antidepressant, agomelatine, is a neutral
chiatric disorders: characteristics, causes and the quest for improved antagonist at 5-HT(2C) receptors. Int J Neuropsychopharmacol 14:
therapy. Nat Rev Drug Discovery 11: 141–168. 768–768.
Millan MJ, Andrieux A, Bartzokis G, et al. (2016) Altering the course of Millan MJ, Newman-Tancredi A, Audinot V, et al. (2000c) Agonist and
schizophrenia: progress and perspectives. Nat Rev Drug Discovery antagonist actions of yohimbine as compared to fluparoxan at alpha2-
15: 485–515. adrenergic receptors (AR)s, serotonin (5-HT)1A, 5-HT1B, 5-HT1D and
Millan MJ, Brocco M, Gobert A, et al. (2005) Anxiolytic properties of dopamine D2 and D3 receptors. Significance for the modulation of
agomelatine, an antidepressant with melatoninergic and serotonergic frontocortical monoaminergic transmission and depressive states.
properties: role of 5-HT2C receptor blockade. Psychopharmacology Synapse 35: 79–95.
(Berlin) 177: 448–458. Millan MJ, Panayi F, Rivet JM, et al. (2007b) The role of microdialysis
Millan MJ, Canton H, Gobert A, et al. (1995) Novel benzodioxopipera- in drug discovery: focus on antipsychotic agents. In: Westerink BHC
zines acting as antagonists at postsynaptic 5-HT1A receptors and as and Cremers TIFH (eds) Handbook of Microdialysis. Amsterdam:
agonists at 5-HT1A autoreceptors: a comparative pharmacological Academic Press, vol. 16, pp. 485–511.
characterization with proposed 5-HT1A antagonists. J Pharmacol Millan MJ, Schreiber R, Dekeyne A, et al. (1998b) S 16924 ((R)-2-[1-
Exp Ther 268: 337–352. [2-(2,3-dihydro-benzo[1,4] dioxin-5-yloxy)-ethyl]-pyrrolidin-3yl]-
Millan MJ, Dekeyne A, Gobert A, et al. (2010) S41744, a dual neurokinin 1-(4-fluoro-phenyl)-ethanone), a novel, potential antipsychotic with
(NK)1 receptor antagonist and serotonin (5-HT) reuptake inhibitor marked serotonin (5-HT)1A agonist properties: II. Functional profile
with potential antidepressant properties: a comparison to aprepitant in comparison to clozapine and haloperidol. J Pharmacol Exp Ther
(MK869) and paroxetine. Eur Neuropsychopharmacol 20: 599–621. 286; 1356–1373.
Millan MJ, Di Cara B, Dekeyne A, et al. (2007a) Selective blockade of Miller GM (2011) The emerging role of trace amine-associated receptor
dopamine D3 vs D2 receptors enhances frontocortical cholinergic 1 in the functional regulation of monoamine transporters and dopa-
transmission and social memory in rats: a parallel neurochemical and minergic activity. J Neurochem 116: 164–176.
behavioural analysis. J Neurochem 100: 1047–1061. Mizoro Y, Yamaguchi Y, Kitazawa R, et al. (2010) Activation of AMPA
Millan MJ, Gobert A, Lejeune F, et al. (2001a) S33005, a novel ligand at receptors in the suprachiasmatic nucleus phase-shifts the mouse cir-
both serotonin and norepinephrine transporters: I. Receptor binding, cadian clock in vivo and in vitro. PLoS One; 5:e10951.
electrophysiological, and neurochemical profile in comparison with Mnie-Filali O, Faure C, Lambas-Senas L, et al. (2011) Pharmacological
venlafaxine, reboxetine, citalopram, and clomipramine. J Pharmacol blockade of 5-HT7 receptors as a putative fast acting antidepressant
Exp Ther 298: 565–580. strategy. Neuropsychopharmacology 36: 1275–1288.
Millan MJ, Gobert A, Lejeune F, et al. (2003) The novel melatonin Mohawk JA, Pargament JM and Lee TM (2007) Circadian dependence
agonist agomelatine (S20098) is an antagonist at 5-hydroxytrypta- of corticosterone release to light exposure in the rat. Physiol Behav
mine2C receptors, blockade of which enhances the activity of fron- 92: 800–806.
tocortical dopaminergic and adrenergic pathways. J Pharmacol Exp Mørk A, Pehrson A, Brennum LT, et al. (2012) Pharmacological
Ther 306: 954–964. effects of Lu AA21004: a novel multimodal compound for the
Millan MJ, Gobert A, Newman-Tancredi A, et al. (1998a) S 16924 ((R)- treatment of major depressive disorder. J Pharmacol Exp Ther
2-[1-[2-(2,3-dihydro-benzo[1,4] dioxin-5-yloxy)-ethyl]-pyrrolidin- 340: 666–675.
3yl]-1-(4-fluoro-phenyl)-ethanone), a novel, potential antipsychotic Murat B, Devost D, Andrés M, et al. (2012) V1b and CRHR1 receptor
with marked serotonin (5-HT)1A agonist properties: I. Receptorial heterodimerization mediates synergistic biological actions of vaso-
and neurochemical profile in comparison with clozapine and halo- pressin and CRH. Mol Endocrinol 26: 502–520.
peridol. J Pharmacol Exp Ther 286: 1341–1355. Murphy SE and Mackay CE (2011) Using MRI to measure drug action:
Millan MJ, Gobert A, Roux S, et al. (2004) The serotonin1A receptor par- caveats and new directions. J Psychopharmacol 25: 1168–1174.
tial agonist S15535 [4-(benzodioxan-5-yl)1-(indan-2-yl)piperazine] Musazzi L, Treccani G and Popoli M (2015) Functional and structural
enhances cholinergic transmission and cognitive function in rodents: remodeling of synapses in prefrontal and frontal cortex induced by
a combined neurochemical and behavioral analysis. J Pharmacol behavioral stress. Front Psychiatry 6: 60.
Exp Ther 311: 190–203. Myers JF, Nutt DJ and Lingford-Hughes AR (2016) γ-Aminobutyric
Millan MJ, Goodwin GM, Meyer-Lindenberg A, et al. (2015) Learning acid as a metabolite: Interpreting magnetic resonance spectroscopy
from the past and looking to the future: Emerging perspectives for experiments. J Psychopharmacol 30: 422–427.

Downloaded from jop.sagepub.com at CORNELL UNIV on October 18, 2016


Millan et al. 29

Nader N, Chrousos GP and Kino T (2010) Interactions of the circadian Poels EM, Kegeles LS, Kantrowitz JT, et al. (2014) Glutamatergic
CLOCK system and the HPA axis. Trends Endocrinol Metab 21: abnormalities in schizophrenia: a review of proton MRS findings.
277–286. Schizophr Res 152: 325–332.
Nakajima S, Gerretsen P, Takeuchi H, et al. (2013) The potential role of Pomierny-Chamioło L, Poleszak E, Pilc A, et al. (2010) NMDA but not
dopamine D3 receptor neurotransmission incognition. Eur Neuropsy- AMPA glutamatergic receptors are involved in the antidepressant-
chopharmacol 23: 799–813. like activity of MTEP during the forced swim test in mice. Pharma-
Nagy K, Marko B and Zsilla G (2010). Alterations in brain extracellular col Rep 62: 1186–1190.
dopamine and glycine levels following combined administration of Pratt J, Winchester C, Dawson N, et al. (2012) Advancing schizophrenia
the glycine transporter type-1 inhibitor Org-24461 and risperidone. drug discovery: optimizing rodent models to bridge the translational
Neurochem Res 35: 2096–2106. gap. Nat Rev Drug Discovery 11: 560–579.
Nations KR, Dogterom P, Bursi R, et al. (2012) Examination of Org 26576, Prochiantz A and Di Nardo AA (2015) Homeoprotein signaling in the
an AMPA receptor positive allosteric modulator, in patients diagnosed developing and adult nervous system. Neuron 85: 911–925.
with major depressive disorder: an exploratory, randomized, double- Raddatz R, Tao M and Hudkins RL (2010) Histamine H3 antagonists for
blind, placebo-controlled trial. J Psychopharmacol 26: 1525–1539. treatment of cognitive deficits in CNS diseases. Curr Top Med Chem
Navailles S, Benazzouz A, Bioulac B, et al. (2010) High-frequency 10: 153–169.
stimulation of the subthalamic nucleus and L-3,4-dihydroxyphenyl- Ramos BP and Arnsten AF (2007) Adrenergic pharmacology and cogni-
alanine inhibit in vivo serotonin release in the prefrontal cortex and tion: focus on the prefrontal cortex. Pharmacol Ther 113: 523–536.
hippocampus in a rat model of Parkinson’s disease. J Neurosci 30: Revel FG, Moreau JL, Gainetdinov RR, et al. (2012) Trace amine-associ-
2356–2364. ated receptor 1 partial agonism reveals novel paradigm for neuropsy-
Nisell M, Nomikos GG and Svensson TH (1994) Systemic nicotine- chiatric therapeutics. Biol Psychiatry 74: 934–942.
induced dopamine release in the rat nucleus accumbens is regulated Riga MS, Sánchez C, Celada P, et al. (2016) Involvement of 5-HT3
by nicotinic receptors in the ventral tegmental area. Synapse 16: 36–44. receptors in the action of vortioxetine in rat brain: Focus on gluta-
Nomikos GG, Arborelius L, Svensson TH, et al. (1992). The novel matergic and GABAergic neurotransmission. Neuropharmacology
5-HT1A receptor antagonist (S)-UH-301 prevents (R)-8-OH-DPAT- 108: 73–81.
induced decrease in interstitial concentrations of serotonin in the rat Rivet JM, Audinot V, Gobert A, et al. (1994) Modulation of mesolimbic
hippocampus. Eur J Pharmacol 216: 373–378. dopamine release by the selective dopamine D3 receptor antagonist,
Olsson B, Lautner R, Andreasson U, et al. (2016) CSF and blood bio- (+)-S 14297. Eur J Pharmacol 265: 175–177.
markers for the diagnosis of Alzheimer’s disease: a systematic Robbins TW and Arnsten AFT (2009) The neuropsychopharmacology
review and meta-analysis. Lancet Neurol 15: 673–684. of fronto-executive function: monoaminergic modulation. Annu Rev
Palamarchouk V, Smagin G and Goeders NE (2009) Self-administered Neurosci 32: 267–287.
and passive cocaine infusions produce different effects on corticos- Rosenblat JD, Kakar R and McIntyre RS (2015) The cognitive effects
terone concentrations in the medial prefrontal cortex (MPC) of rats. of antidepressants in major depressive disorder: a systematic review
Pharmacol Biochem Behav 94: 163–168. and meta-analysis of randomized clinical trials. Int J Neuropsycho-
Palucha A, Klak K, Banski P, et al. (2007) Activation of the mGlu7 pharmacol 19. DOI: 10.1093/ijnp/pyv082.
receptor elicits antidepressant-like effects in mice. Psychopharma- Ross HE, Cole CD, Smith Y, et al. (2009) Characterization of the oxy-
cology 194: 555–562. tocin system regulating affiliative behavior in female prairie voles.
Pałucha-Poniewiera A, Wierońska JM, Brański P, et al. (2013) Is the Neuroscience 162: 892–903.
mGlu5 receptor a possible target for new antidepressant drugs? Saavedra OM, Karila D, Brossard D, et al. (2016) Design and synthesis
Pharmacol Rep 65: 1506–1511. of novel N-sulfonyl-2-indoles that behave as 5-HT6 antagonists with
Papouin T and Oliet SH (2014) Organization, control and function of significant selectivity for D3 over D2 receptors. Biochem Biophys
extrasynaptic NMDA receptors. Philos Trans R Soc London B: Biol Res Commun, in press.
Sci 369: 20130601. Sahli ZT, Banerjee P and Tarazi FI (2016) The preclinical and clinical
Park M, Niciu MJ and Zarate CA, Jr (2015) Novel glutamatergic treatments effects of vilazodone for the treatment of major depressive disorder.
for severe mood disorders. Curr Behav Neurosci Rep 2: 198–208. Expert Opin Drug Discovery 11: 515–523.
Pehrson AL, Cremers T, Bétry C, et al. (2013) Lu AA21004, a novel Sakkalis V (2011) Applied strategies towards EEG/MEG biomarker
multimodal antidepressant, produces regionally selective increases identification in clinical and cognitive research. Biomarkers Med 5:
of multiple neurotransmitters - a rat microdialysis and electrophysi- 93–105.
ology study. Eur Neuropsychopharmacol 23: 133–145. Sallee FR (2010) The role of alpha2-adrenergic agonists in attention-def-
Pehrson AL and Sanchez C (2014) Serotonergic modulation of glutamate icit/hyperactivity disorder. Postgrad Med 122: 78–87.
neurotransmission as a strategy for treating depression and cognitive Samuels BA, Mendez-David I, Faye C, et al. (2016) Serotonin 1A and
dysfunction. CNS Spectr 19: 121–133. serotonin 4 receptors: essential mediators of the neurogenic and
Pei Y, Asif-Malik A and Canales JJ (2016) Trace amines and the trace behavioral actions of antidepressants. Neuroscientist 22: 26–45.
amine-associated receptor 1: pharmacology, neurochemistry, and Sanacora G, Berman RM, Cappiello A, et al. (2004) Addition of the α2-
clinical implications. Front Neurosci 10: 148. antagonist yohimbine to fluoxetine: effects on rate of antidepressant
Peñas-Cazorla R and Vilaró MT (2015) Serotonin 5-HT4 receptors and response. Neuropsychopharmacology 29: 1166–1171.
forebrain cholinergic system: receptor expression in identified cell Sanacora G, Zarate CA, Krystal JH, et al. (2008) Targeting the gluta-
populations. Brain Struct Funct 220: 3413–3434. matergic system to develop novel, improved therapeutics for mood
Pepicelli O, Raiteri M and Fedele E (2004) The NOS/sGC pathway in the disorders. Nat Rev Drug Discovery 7: 426–437.
rat central nervous system: a microdialysis overview. Neurochem Int Sanchez C, Asin KE and Artigas F (2015) Vortioxetine, a novel antide-
45: 787–797. pressant with multimodal activity: review of preclinical and clinical
Perlemuter G, Cacoub P, Valla D, et al. (2016) Characterisation of data. Pharmacol Ther 145:–57.
agomelatine-induced increase in liver enzymes: frequency and risk Santana N and Artigas F (2016) Expression of serotonin2C receptors
factors determined from a pooled analysis of 7605 treated patients. in pyramidal and GABAergic neurons of rat prefrontal cortex: a
CNS Drugs 30: 877–888. comparison with striatum. Cereb Cortex Epub ahead of print 1 June
Pich EM and Collo G (2015) Pharmacological targeting of dopamine D3 2016. DOI: 10.1093/cercor/bhw148.
receptors: Possible clinical applications of selective drugs. Eur Neu- Sarihi A, Jiang B, Komaki A, et al. (2008) Metabotropic glutamate recep-
ropsychopharmacol 25: 1437–1447. tor type 5-dependent long-term potentiation of excitatory synapses

Downloaded from jop.sagepub.com at CORNELL UNIV on October 18, 2016


30 Journal of Psychopharmacology

on fast-spiking GABAergic neurons in mouse visual cortex. J Neu- Vidal R, Castro E, Pilar-Cuéllar F, et al. (2014) Serotonin 5-HT4
rosci 28: 1224–1235. receptors: A new strategy for developing fast acting antidepres-
Schechter LE, Lin Q, Smith DL, et al. (2008) Neuropharmacological pro- sants? Curr Pharm Des 20: 3751–3762.
file of novel and selective 5-HT6 receptor agonists: WAY-181187 Vyas NS, Patel NH and Nijran KS (2011) The use of PET imaging in
and WAY-208466. Neuropsychopharmacology 33: 1323–1335. studying cognition, genetics and pharmacotherapeutic interventions
Schoemaker H, Claustre Y, Fage D, et al. (1997) Neurochemical char- in schizophrenia. Expert Rev Neurother 11: 37–51.
acteristics of amisulpride, and atypical dopamine D2/D3 receptor Watson DJ, Loiseau F, Ingallinesi M, et al. (2012) Selective blockade
antagonist with both presynaptic and limbic selectivity. J Pharmacol of dopamine D3 receptors enhances while D2 receptor antagonism
Exp Ther 280: 83–97. impairs social novelty discrimination and novel object recognition in
Serres F, Millan MJ and Sharp T (2012) Molecular adaptation to chronic rats: a key role for the prefrontal cortex. Neuropsychopharmacology
antidepressant treatment: evidence for a more rapid response to 37: 770–786.
the novel α2-adrenoceptor antagonist/5-HT-noradrenaline reuptake Weber ML, Hofland CM, Shaffer CL, et al. (2013) Therapeutic doses
inhibitor (SNRI), S35966, compared to the SNRI, venlafaxine. Int J of antidepressants are projected not to inhibit human α4β2 nicotinic
Neuropsychopharmacol 15: 617–629. acetylcholine receptors. Neuropharmacology 72: 88–95.
Sharp T, Zetterstrom T and Ungerstedt U (1986) An in vivo study of Westerink BH, Damsma G, Rollema H, et al. (1987) Scope and limita-
dopamine release and metabolism in rat brain regions using intrace- tions of in vivo brain dialysis: a comparison of its application to vari-
rebral dialysis. J Neurochem 47, 113–122. ous neurotransmitter systems. Life Sci 41, 1763–1776.
Shimazaki T, Kaku A and Chaki S (2010) D-Serine and a glycine trans- Whittington MA, Cunningham MO, LeBeau FEN, et al. (2010)
porter-1 inhibitor enhance social memory in rats. Psychopharmacol- Multiple origins of the cortical gamma rhythm. Dev Neurobiol
ogy 20: 263–270. 71: 92–106.
Soumier A, Banasr M, Lortet S, et al. (2009) Mechanisms contributing to Wolosker H (2011) Serine racemase and the serine shuttle between neu-
the phase-dependent regulation of neurogenesis by the novel antide- rons and astrocytes. Biochim Biophys Acta 41: 1546–1560.
pressant, agomelatine, in the adult rat hippocampus. Neuropsycho- Wotjak CT, Landgraf R and Engelmann M (2008) Listening to neuropep-
pharmacology 34: 2390–2403. tides by microdialysis: echoes and new sounds? Pharmacol Biochem
Stein DJ, Picarel-Blanchot F and Kennedy SH (2013) Efficacy of the Behav 90: 125–134.
novel antidepressant agomelatine for anxiety symptoms in major Wu H, Jin Y, Wei J, et al. (2005) Mode of action of taurine as a neuropro-
depression. Hum Psychopharmacol 28: 151–159. tector. Brain Res 1038: 123–131.
Strzelecki D, Podgórski M, Kałużyńska O, et al. (2015) Adding sarco- Yamanaka K, Koistinaho J, Latz E, et al. (2015) Neuroinflammation in
sine to antipsychotic treatment in patients with stable schizophrenia Alzheimer’s disease. Lancet Neurol 14: 388–405.
changes the concentrations of neuronal and glial metabolites in the Yang CR and Svensson KA (2008) Allosteric modulation of
left Dorsolateral Prefrontal Cortex. Int J Mol Sci 16: 24475–24489. NMDA receptor via elevation of brain glycine and D-serine: the
Takeda S, Commins C and DeVos SL (2016) Seed-competent high- therapeutic potential for schizophrenia. Pharmacol Ther 120:
molecular-weight tau species accumulates in the cerebrospinal fluid 317–332.
of Alzheimer’s disease mouse model and human patients. Ann Neu- Yoshikawa T, Naganuma F, Iida T, et al. (2013) Molecular mecha-
rol 80: 355–367. nism of histamine clearance by primary human astrocytes. Glia 61:
Thase ME, Chen D, Edwards J, et al. (2014) Efficacy of vilazodone on 905–916.
anxiety symptoms in patients with major depressive disorder. Int Yoshitake S, Ijiri S, Kehr J, et al. (2013) Concurrent modulation of extra-
Clin Psychopharmacol 29: 351–356. cellular levels of noradrenaline and cAMP during stress and by anx-
Thase ME, Mahableshwarkar AR and Dragheim M (2016) A meta-anal- iogenic- or anxiolytic-like neuropeptides in the prefrontal cortex of
ysis of randomized, placebo-controlled trials of vortioxetine for the awake rats. Neurochem Int 62: 314–323.
treatment of major depressive disorder in adults. Eur Neuropsycho- Yoshitake T, Yoshitake S, Fujino K, et al. (2004) High-sensitive
pharmacol 26: 979–993. liquid chromatographic method for determination of neuronal
Thomas DR (2006) 5-HT5A receptors as a therapeutic target. Pharmacol release of serotonin, noradrenaline and dopamine monitored by
Ther 111: 707–714. microdialysis in the rat prefrontal cortex. J Neurosci Methods 140:
Tran P, Skolnick P, Czobor P, et al. (2012) Efficacy and tolerability of 163–168.
the novel triple reuptake inhibitor amitifadine in the treatment of Young JW, Powell SB and Risbrough V (2009) Using the MATRICS to
patients with major depressive disorder: a randomized, double-blind, guide development of a preclinical cognitive test battery for research
placebo-controlled trial. J Psychiatr Res 46: 64–71. in schizophrenia. Pharmacol Ther 122: 150–202.
Tsai GE and Lin PY (2010) Strategies to enhance N-methyl-D-aspartate Zanos P, Moaddel R, Morris PJ, et al. (2016) NMDAR inhibition-
receptor-mediated neurotransmission in schizophrenia, a critical independent antidepressant actions of ketamine metabolites.
review and meta-analysis. Curr Pharm Des 16: 522–537. Nature 533: 481–486.
Umbricht D, Alberati D, Martin-Facklam M, et al. (2014) Effect of bitop- Zanto FP, Rubens MT, Thangavel A, et al. (2011) Causal role of the
ertin, a glycine reuptake inhibitor, on negative symptoms of schizo- prefrontal cortex in top-down modulation of visual processing and
phrenia: a randomized, double-blind, proof-of-concept study. JAMA working memory Nat Neurosci 14: 656–662.
Psychiatry 71, 637–646. Zetterström T, Sharp T, Marsden CA, et al. (1983) In vivo measurement
Ungerstedt U (1991) Microdialysis - principles and applications for stud- of dopamine and its metabolites by intracerebral dialysis: changes
ies in animals and man. J Intern Med 230: 365–373. after D-amphetamine. J Neurochem 41: 1769–1773.
Ungerstedt U and Rostami E (2004) Microdialysis in neurointensive care. Zhang MY and Beyer CE (2006) Measurement of neurotransmitters
Curr Pharm Des 10: 2145–2152. from extracellular fluid in brain by in vivo microdialysis and
Vaka SR, Murthy SN, Repka MA, et al. (2011) Upregulation of endog- chromatography-mass spectrometry. J Pharm Biomed Anal 40:
enous neurotrophin levels in the brain by intranasal administration of 492–499.
carnosic acid. J Pharm Sci 100: 3139–3345. Zhou Z, Zhang G, Li X, et al. (2015) Loss of phenotype of parvalbumin
Van der Zeyden M, Oldenziel WH, Rea K, et al. (2008) Microdialysis of interneurons in rat prefrontal cortex is involved in antidepressant-
GABA and glutamate: analysis, interpretation and comparison with and propsychotic-like behaviors following acute and repeated ket-
microsensors. Pharmacol Biochem Behav 90: 135–147. amine administration. Mol Neurobiol 51: 808–819.
Vance JE (2008) Phosphatidylserine and phosphatidylethanolamine in Zoicas I, Slattery DA and Neumann ID (2014) Brain oxytocin in social
mammalian cells: two metabolically related aminophospholipids. J fear conditioning and its extinction: involvement of the lateral sep-
Lipid Res 49: 1377–1387. tum. Neuropsychopharmacology 39: 3027–3035.

Downloaded from jop.sagepub.com at CORNELL UNIV on October 18, 2016

You might also like