Hostname: page-component-7c8c6479df-5xszh Total loading time: 0 Render date: 2024-03-27T18:13:40.843Z Has data issue: false hasContentIssue false

Effects of serotonin in the hippocampus: how SSRIs and multimodal antidepressants might regulate pyramidal cell function

Published online by Cambridge University Press:  08 September 2015

Elena Dale*
Affiliation:
Lundbeck Research USA, Paramus, New Jersey, USA
Alan L. Pehrson
Affiliation:
Lundbeck Research USA, Paramus, New Jersey, USA
Theepica Jeyarajah
Affiliation:
Lundbeck Research USA, Paramus, New Jersey, USA
Yan Li
Affiliation:
Lundbeck Research USA, Paramus, New Jersey, USA
Steven C. Leiser
Affiliation:
Lundbeck Research USA, Paramus, New Jersey, USA
Gennady Smagin
Affiliation:
Lundbeck Research USA, Paramus, New Jersey, USA
Christina K. Olsen
Affiliation:
Lundbeck DK, Copenhagen-Valby, Denmark
Connie Sanchez
Affiliation:
Lundbeck Research USA, Paramus, New Jersey, USA
*
*Address for correspondence: Elena Dale, PhD, Lundbeck Research USA, 215 College Rd., Paramus, NJ 07652, USA. (Email: EDAL@lundbeck.com)
Rights & Permissions [Opens in a new window]

Abstract

The hippocampus plays an important role in emotional and cognitive processing, and both of these domains are affected in patients with major depressive disorder (MDD). Extensive preclinical research and the notion that modulation of serotonin (5-HT) neurotransmission plays a key role in the therapeutic efficacy of selective serotonin reuptake inhibitors (SSRIs) support the view that 5-HT is important for hippocampal function in normal and disease-like conditions. The hippocampus is densely innervated by serotonergic fibers, and the majority of 5-HT receptor subtypes are expressed there. Furthermore, hippocampal cells often co-express multiple 5-HT receptor subtypes that can have either complementary or opposing effects on cell function, adding to the complexity of 5-HT neurotransmission. Here we review the current knowledge of how 5-HT, through its various receptor subtypes, modulates hippocampal output and the activity of hippocampal pyramidal cells in rodents. In addition, we discuss the relevance of 5-HT modulation for cognitive processing in rodents and possible clinical implications of these results in patients with MDD. Finally, we review the data on how SSRIs and vortioxetine, an antidepressant with multimodal activity, affect hippocampal function, including cognitive processing, from both a preclinical and clinical perspective.

Type
Review Articles
Creative Commons
Creative Common License - CCCreative Common License - BYCreative Common License - NCCreative Common License - SA
The online version of this article is published within an Open Access environment subject to the conditions of the Creative Commons Attribution-NonCommercial-ShareAlike licence . The written permission of Cambridge University Press must be obtained for commercial re-use.
Copyright
© Cambridge University Press 2015

Clinical Implications

  • The hippocampus is involved in the regulation of emotional and cognitive processing, both of which are compromised in patients with major depressive disorder (MDD).

  • Serotonin (5-HT) plays an important role in hippocampal function.

  • Multiple 5-HT receptors are often co-expressed on the same cell types in the hippocampus with functions that can either be complementary or opposing. Overall, 5-HT appears to inhibit pyramidal cells in the hippocampal circuit in rodents.

  • While selective serotonin reuptake inhibitors (SSRIs) have the potential to normalize hippocampal output under stress conditions and to treat mood symptoms in MDD, their effects on cognitive function are less clear.

  • The multimodal antidepressant vortioxetine has shown clinical efficacy on mood as well as cognitive symptoms in patients with MDD. 5-HT3 receptor inhibition of GABAergic interneurons is thought to play an important role in mediating these effects.

Introduction

There is extensive evidence that depression and other stress-related conditions are associated with hippocampal dysfunction.Reference Femenia, Gomez-Galan, Lindskog and Magara1, Reference Small, Schobel, Buxton, Witter and Barnes2 In several magnetic resonance studies, patients with major depressive disorder (MDD) have reduced hippocampal volumes compared with matched control subjects.Reference Bell-McGinty, Butters, Meltzer, Greer, Reynolds and Becker3Reference Frodl, Koutsouleris and Bottlender5 Furthermore, Sheline et al Reference Sheline, Gado and Kraemer6 have shown that there is an inverse correlation between hippocampal volume and the duration of untreated depression. A meta-analysis of 12 clinical studies indicated that the number of depressive episodes may be correlated with a reduction of hippocampal volume in the right hemisphere.Reference Videbech and Ravnkilde7 Reduced hippocampal volume in MDD patients has also been associated with impaired memory (eg, MacQueen et al Reference MacQueen, Campbell and McEwen8). Memory function related to hippocampal integrity decreases with increasing numbers of depressive episodes.Reference Gorwood, Corruble, Falissard and Goodwin9 In addition, functional magnetic resonance imaging (fMRI) studies of depressed patients have consistently shown overactivity in the frontolimbic circuitry, including the dorsolateral prefrontal cortex and hippocampus during working memory performance.Reference Harvey, Fossati and Pochon10, Reference Walsh, Williams and Brammer11

In addition to disturbances in mood and emotional processing, MDD is associated with deficits in several cognitive domains, including executive function, processing speed, and attention, as well as learning and memory.Reference Hammar and Ardal12Reference Porter, Bourke and Gallagher14 There is evidence that cognitive impairment varies independently of mood state and does not necessarily resolve when the patient is considered to be in clinical remission.Reference Hasselbalch, Knorr and Kessing15 This may imply that cognitive control and the regulation of emotion have distinct neuronal bases in depression.Reference Campbell and MacQueen16 While the literature suggests that antidepressants may potentially treat cognitive dysfunction in some patients with MDD, these studies were not designed to distinguish between the direct effects on cognitive domains versus indirect effects on cognition via improvements in mood. Overall, small sample sizes, methodological constraints, and the absence of replication make it difficult to draw firm conclusions from the majority of these studies.Reference Biringer, Rongve and Lund17, Reference McIntyre, Cha and Soczynska18

Since the selective serotonin (5-HT) reuptake inhibitors (SSRIs) and serotonin norepinephrine (NE) reuptake inhibitors (SNRIs) are the predominant pharmacotherapies used for the treatment of MDD, modulation of serotonergic neurotransmission is assumed to play a pivotal role in achieving their antidepressant efficacy. Many 5-HT receptor subtypes are extensively expressed in the hippocampus. However, even though a large number of preclinical studies in rodents are strongly supportive of antidepressant treatments restoring hippocampal function, their mechanisms of action have not been fully elucidated. Furthermore, it is not well understood how the clinical efficacy of currently used antidepressants might be related to changes in hippocampal function in patients with MDD.Reference Vythilingam, Vermetten and Anderson19 Therefore, a thorough understanding of how 5-HT receptor modulation affects hippocampal functions is essential to the understanding of how antidepressants might work. Here we review the current knowledge of how 5-HT, through its various receptor subtypes, might modulate hippocampal activity in rodents. In addition, we discuss its relevance for cognitive processing and the possible clinical implications for patients with MDD. Finally, we review available data on how SSRIs and vortioxetine, an antidepressant that, in addition to inhibition of 5-HT reuptake, also modulates a number of 5-HT receptor subtypes, affect hippocampus function from a preclinical and clinical perspective.

Anatomy of the Hippocampus

To understand how 5-HT modulates hippocampal function at a molecular level, it is necessary to gain insights into how 5-HT modulates the different cell types and subregions that comprise the hippocampal microcircuits. Along the longitudinal axis, the hippocampus is segregated into dorsal, intermediate, and ventral regions in rodents (reviewed in Fanselow and DongReference Fanselow and Dong20 and Moser and MoserReference Moser and Moser21), and analogous posterior and anterior regions in primates and humansReference Poppenk, Evensmoen, Moscovitch and Nadel22 that project to distinct brain areas.Reference O’Leary and Cryan23 Lesion and electrophysiology studies in rodents have shown that the dorsal hippocampus is primarily involved in the cognitive functions, including spatial learning and memory,Reference Moser, Moser and Andersen24, Reference Pothuizen, Zhang, Jongen-Relo, Feldon and Yee25 whereas the ventral hippocampus is primarily involved in regulating stress, emotion, and anxiety.Reference Bannerman, Grubb, Deacon, Yee, Feldon and Rawlins26Reference Kjelstrup, Tuvnes, Steffenach, Murison, Moser and Moser28 However, this division of functions is somewhat ambiguous, since parts of the ventral hippocampus have been also shown to be involved in memory tasks.Reference Rudy and Matus-Amat29

The hippocampus is subdivided into several distinct zones: the dentate gyrus (DG), CA3, CA2, CA1, and the subiculum regions that were first described by Ramon y Cajal in 1911Reference Ramon y30 and Lorente de Nó in 1934 Reference Lorente de Nó31 (Figure 1). The CA3, CA2, and CA1 regions are sometimes called the hippocampal gyrus or Ammon’s horn. Granule cells in the DG receive projections from the surrounding entorhinal cortex and send their axons, called mossy fibers, to the CA3 area. Pyramidal cells in the CA3 area project axons, known as Schaffer collaterals, to the CA2 and CA1 areas. Pyramidal cells in the CA1 area send their axons to the surrounding deep cortical layers of the entorhinal cortex and to the subiculum which is the final processing stage of the hippocampal microcircuitry (Figure 1). In addition to this main “trisynaptic circuit,” there are direct connections from the superficial layers of the entorhinal cortex to the CA3 and CA1 areas, and synaptic connections from inhibitory gamma-butyric acid (GABA)ergic interneurons to excitatory glutamatergic pyramidal and granule cells within the hippocampus.Reference Lavenex32

Figure 1 Schematic illustration of the rat hippocampal circuit with 5-HT receptor localization. The main areas of the hippocampus, including the dentate gyrus (DG), CA3, CA2, CA1, and the subiculum regions, and synaptic connections between them are indicated. Principal (granule and pyramidal) cells are shown in blue, and interneurons are shown in green. Expression of 5-HT receptor subtypes on hippocampal CA1 and CA3 pyramidal cells, granule cells, and interneurons are shown. References for 5-HT receptor localization are listed Table 1. At least 16 subtypes of interneurons have been identified in the hippocampus; one representative interneuron is shown for illustrative purposes. Note that the 5-HT1A heteroreceptor is expressed at high levels throughout the hippocampus. The 5-HT1B receptor is found at highest levels in the subiculum. Based on histology data, the 5-HT3 receptor is only expressed on the interneurons, and the 5-HT4 receptor is only expressed on pyramidal cells. Other 5-HT receptors subtypes are found on both principal cells and interneurons.

There are 2 types of principal cells in the hippocampal circuit: glutamatergic pyramidal cells in the Ammon’s horn and subiculum regions, and glutamatergic granule cells in the DG (Figure 1). They generally have excitatory effects on the neurons to which they send axon terminals including other glutamatergic and GABAergic, as well monoaminergic [5-HT, norepinephrine (NE), dopamine (DA)], cholinergic, and histaminergic (HA) cells. There are also 3 major populations of GABAergic inhibitory interneurons that can be identified by the expression of the calcium-binding proteins parvalbumin, calbindin, and calretinin. These interneurons can be further subdivided based on their morphology and presence of receptors for neuropeptides and other neurotransmitters. In total at least 16 different subtypes of interneurons have been identified in the hippocampus with different firing properties and functions.Reference Freund and Buzsáki33, Reference Parra, Gulyas and Miles34 Each of these interneuron subpopulations has a distinct placement within the network, as well as a distinct role in modulating the behavior of pyramidal neurons.

Parvalbumin-immunoreactive interneurons are present within the pyramidal cell body layer (divided into stratum oriens and pyramidale), and synapse onto the soma and/or axons of pyramidal neurons.Reference Ribak, Nitsch and Seress35 In contrast, calbindin immunoreactive interneurons are present in the dendritic layers of the hippocampus (divided into stratum radiatum and moleculare)Reference Sloviter36 and are thought to synapse onto pyramidal neuron dendrites.Reference Aznar, Qian, Shah, Rahbek and Knudsen37 Finally, calretinin immunoreactive interneurons frequently form local synaptic connections onto other interneurons.Reference Gulyas, Hajos and Freund38 Thus, hippocampal interneurons can modulate the activity of both pyramidal cells and other interneurons.Reference Gulyas, Hajos and Freund38

The processing of information within the hippocampus is complex and is influenced by multiple neurotransmitters and neuromodulators, including glutamate, GABA, DA, NE, HA, acetylcholine (ACh), and 5-HT. Serotonergic receptors are found on both excitatory cells and inhibitory interneurons. As will be discussed in the following sections, 5-HT neurotransmission can have a direct effect on pyramidal neuron firing by modulating its membrane potential and indirect effects via modulating GABA neurotransmission.

5-HT Receptors in the Rodent Hippocampus

Nearly all of the identified 5-HT receptor subtypes are expressed in the hippocampal circuit in rodents.Reference Berumen, Rodriguez, Miledi and Garcia-Alcocer39 Interestingly, 5-HT fibers often lack direct synaptic contacts, and in many cases 5-HT receptors have been detected on neurons that do not receive serotonergic innervation.Reference Oleskevich, Descarries, Watkins, Seguela and Daszuta40Reference Vizi and Kiss42 This suggests that in the hippocampus, as in other brain areas, 5-HT is released diffusely by volume transmission and acts more as a neuromodulator whose function might be to maintain homeostasis in the brain.

The specificity and diversity of 5-HT signaling arises from at least 14 different receptor subtypes grouped in 7 receptor families with distinct characteristics and expression patterns (Table 1, Figures 1 and 2). The 5-HT1 receptor family is inhibitory; it is coupled to G-protein-coupled inwardly rectifying potassium (GIRK) channels and Gi/o proteins. Activation of GIRK channels exerts hyperpolarizing effects on cell function. Activation of Gi/o proteins inhibits adenylyl cyclase and decreases cyclic adenosine monophosphate (cAMP) concentration. The 5-HT2 receptor family is stimulatory and signals via activation of Gq proteins that are coupled to phospholipase C. Phospholipase C hydrolyses membrane phosphoinositides into inositol triphosphate (IP3) and diacylglycerol (DAG), which elevate intracellular calcium. One of the downstream targets of 5-HT2 receptors are potassium leak channels. Activation of 5-HT2 receptors closes these channels, resulting in cell depolarization. The 5-HT3 receptor family is stimulatory and the only non-G-protein coupled receptor. Activation of 5-HT3 receptors opens a non-selective Na+/K+ ion channel that depolarizes neurons and increases neurotransmitter release. The 5-HT4, 5-HT6, and 5-HT7 receptors are stimulatory and increase neuronal excitability. They are coupled to Gs protein and signal via activation of adenylyl cyclase and elevation in cAMP levels. Finally, 5-HT5 receptors are inhibitory, and, as for the 5-HT1 receptor family members, are coupled to Gi/o proteins that suppress adenylyl cyclase and decrease cAMP levels.Reference Aghajanian and Sanders-Bush43, Reference Glennon, Dukat and Westkaemper44 In the hippocampus, cells often co-express several types of 5-HT receptors that can have either complementary or opposing effects on cell function (Figure 1 and Table 1). Moreover, 5-HT receptors can form homodimers or heterodimers with other G-protein coupled receptors, which adds further complexity to 5-HT signaling.Reference Herrick-Davis45 For example, heterodimers of 5-HT1A-5HT7 receptors and 5-HT2A-mGlu2 (metabotropic glutamate 2) receptors have been shown to have characteristics that differ from their individual counterparts.Reference Herrick-Davis45Reference Naumenko, Popova, Lacivita, Leopoldo and Ponimaskin47 Below we describe the expression patterns of 5-HT receptors and discuss their effects on hippocampal circuitry and hippocampus-mediated behavioral responses based on published results and our data in rodents. We chose to only include behavioral models of memory and learning, since these models have the best link to hippocampal function.Reference Jarrard48Reference Knierim and Hamilton50

Figure 2 Expression of several classes of 5-HT receptors and the 5-HT reuptake transporter (SERT) by ex vivo autoradiography in the rat hippocampus. Autoradiographic images representing total (left panels) and non-specific binding (right panels) for each of 5 separate serotonergic targets in coronal brain sections (20 µm in thickness). 5-HT1A receptors were mapped using 3 nM [3H]8-OH-DPAT (A) alone or (B) in combination with 1 µM of the 5-HT1A receptor selective antagonist WAY100635 to determine the level of nonspecific binding. 5-HT1B/1D receptors were mapped using 1 nM [3H] GR125743 (C) alone or (D) in combination with 1 µM of the 5-HT1B receptor preferring SB216641 to determine the level of nonspecific binding. 5-HT3 receptors were mapped using 3 nM [3H] LY278584 (E) alone or (F) in combination with 1 µM ondansetron to determine the level of nonspecific binding. 5-HT7 receptors were mapped using 4.5 nM [3H]SB269970 (G) alone or (H) in combination with 1 µM of unlabeled SB269970 to determine the level of nonspecific binding. Finally, SERT was mapped using 4.5 nM [3H] escitalopram (I) alone or (J) in combination with 1 µM paroxetine to determine the level of nonspecific binding. Scale bars represent 5 mm.

Table 1 5-HT receptor subtypes in the rodent hippocampus

* Affinities for 5-HT were calculated from pKi/pKd data obtained from the IUPHAR data base. For further details and references see http://www.iuphar-db.org.

** 5-HT5a. Expression strength is indicated by –: absent, +: low, ++: moderate, +++: strong, ?: unknown.

Abbreviations used: GPCR: G-protein-coupled receptor; I: inhibitory; S: stimulatory; : increase; : decrease; Pyr: pyramidal; Gran: granule; IN: interneuron; PV: parvalbumin; CCK: cholecystokinin; Calbin: calbindin; Calre: calretinin.

5-HT1A receptors

Among all 5-HT receptor subtypes, 5-HT1A receptors have the highest affinity for 5-HT (Table 1). In the hippocampus, they are found on non-serotonergic cells as heteroreceptors and inhibit cellular activity via activating GIRK channels.Reference Luscher, Jan, Stoffel, Malenka and Nicoll51 They are defined as heteroreceptors because they control release of neurotransmitters other than 5-HT. 5-HT1A receptors are moderately to highly expressed throughout the hippocampusReference Pompeiano, Palacios and Mengod52 (Figure 2A). They have been detected on both glutamatergic principal cells and at least two subtypes of GABAergic interneurons (Table 1).

Activation of 5-HT1A receptors primarily leads to inhibition of hippocampal pyramidal cells.Reference Andrade and Nicoll53Reference Tada, Kasamo, Suzuki, Matsuzaki and Kojima59 Interestingly, in the prefrontal cortex, 5-HT1A receptor agonists produce both excitatory and inhibitory effects on cortical pyramidal cells.Reference Llado-Pelfort, Santana, Ghisi, Artigas and Celada60 This might be ascribed to a difference in the distribution of 5-HT1A receptors on interneurons versus pyramidal cells in these 2 brain regions.

The function of 5-HT1A receptors has been extensively studied in multiple behavioral studies, 16 of which are listed in Table 2. Modulation of 5-HT1A receptor activity in animal models of memory and learning has produced inconsistent results that range from impairment to improvement (Table 2). Some of these inconsistencies might be due to differences in experimental design across studies. Multiple factors, such as drug dose, length of treatment (acute vs chronic), whether the drug was administered before or after the training period, and also the age and stain of animals, could all influence the behavioral outcome. However, in some studies (eg, Haider et al Reference Haider, Khaliq, Tabassum and Haleem61), opposing results were obtained with different doses of the same compound under the same experimental conditions. This suggests that the variability in results might be partially due to the complex effects of 5-HT1A receptors expressed on different cell types. For instance, 5-HT1A receptors can inhibit both principal (glutamatergic) neurons and GABAergic interneurons. Inhibition of GABAergic interneurons would disinhibit principal cells and thus counteract the direct effects of 5-HT1A receptors expressed on principal neurons. Therefore, selectively targeting 5-HT1A receptors may not be an optimal strategy for modulating hippocampal function, unless these receptors could be targeted in a regional or cell-specific manner.Reference Newman-Tancredi, Martel and Assie62

Table 2 Effects of serotonergic manipulations on hippocampal dependent memory tests in rodents

÷: no effect; : increase/improve; : decrease/impair; Ø: prevented/reduced deficits.

Abbreviations used: MWM: Morris Water Maze; RAM: Radial Arm Maze; BM: Barnes Maze; OP: Object Placement (Preference); CFC: Contextual Fear Conditioning; PS: Pattern Separation; SA: Spontaneous Alternation; FA: Forced Alternation; DA: Delayed Alternation.

5-HT1B and 5-HT1D receptors

5-HT1B heteroreceptors are found throughout the hippocampus at levels ranging from low to very high.Reference Boschert, Amara, Segu and Hen63Reference Sari, Lefevre and Bancila65 They are expressed on axonal terminals and dendrites of principal cells, which include pyramidal cells in Ammon’s horn and granule cells in the DG (Table 1). The highest expression is found in the dorsal subiculum, which might originate from axonal terminals of CA1 pyramidal cells that project to that region (Figure 2C).Reference Sari, Lefevre and Bancila65 Interestingly, in our experiments, the subiculum had the strongest signal for 5-HT1B receptor expression in the rodent forebrain.

Much less is known about the 5-HT1D receptor. 5-HT1D receptors are generally thought to be expressed at much lower levels than 5-HT1B receptors in the rodent brain.Reference Bruinvels, Landwehrmeyer and Gustafson64, Reference Bruinvels, Palacios and Hoyer66 5-HT1D and 5-HT1B receptors are often expressed in the same brain regions.Reference Bruinvels, Landwehrmeyer and Gustafson64 However, no 5-HT1D receptor-specific binding has been detected in the dorsal subiculum, where 5-HT1B receptor-specific binding is very strong.Reference Bruinvels, Palacios and Hoyer66 Interestingly, Xie et al Reference Xie, Lee, O’Dowd and George67 suggest that when 5-HT1B and 5-HT1D receptors are co-expressed, they might exist in a heterodimerized state. Thus, it can be questioned if the effects of 5-HT1B and 5-HT1D receptors should be considered separately.

Activation of 5-HT1B receptors attenuates glutamate transmission in the subiculum and CA1 regions of the hippocampus.Reference Boeijinga and Boddeke68, Reference Mlinar, Falsini and Corradetti69 The effect of 5-HT1B receptor modulation in behavioral models of memory and learning has been far less studied than that for 5-HT1A receptors (Table 2). In general, several studies suggest that 5-HT1B receptor stimulation may negatively affect performance in hippocampal-dependent memory tests. Antagonism of 5-HT1B receptors, in spite of its associated increase in extracellular ACh levels in the dorsal hippocampus,Reference Hu, Wang, Stenfors, Ogren and Kehr70 does not seem be effective in these models (Table 2).

5-HT2A and 5-HT2C receptors

5-HT2A receptors are broadly present within the hippocampus, but little is known about the expression of 5-HT2C receptors.Reference Bombardi71, Reference Cornea-Hebert, Riad, Wu, Singh and Descarries72 5-HT2A receptors are expressed on both principal glutamatergic cells (on their somatic and dendritic regions) and on all known subtypes of hippocampal interneurons (Table 1). There is also some evidence suggesting that 5-HT2A receptors are expressed on mossy fibers in the CA3 region.Reference Bombardi71, Reference Luttgen, Ove Ogren and Meister73 Thus, since 5-HT2A receptors are stimulatory and are expressed on both principal cells and GABAergic interneurons, it would be expected that they would have mixed effects on the firing of principal cells. However, data from 2 electrophysiological studies in brain slices suggest that the effects that 5-HT2 receptors have on interneurons might overwhelm their direct excitatory effects on principal cells.Reference Shen and Andrade74, Reference Wang and Arvanov75 Further research is needed to confirm these observations and clarify which of the 2 effects of 5-HT2 receptors (indirect inhibition or direct excitation of principal neurons) prevails in physiological conditions.

Studies of selective 5-HT2 receptor ligands in behavioral models show variable effects (Table 2), possibly reflecting differences in experimental design across studies and the fact that 5-HT2 receptors are expressed on multiple cell types in the hippocampus (Table 1). Little is known about the effects of 5-HT2C receptor modulation due to a lack of selective compounds. Thus, as with the 5-HT1A receptor, selective targeting of 5-HT2 receptors may not be an optimal strategy for modulating hippocampal function due to their varying functions in this brain region.

5-HT3 receptors

5-HT3 receptors are also found throughout the hippocampus.Reference Morales, Battenberg, de Lecea and Bloom76, Reference Morales, Battenberg, de Lecea, Sanna and Bloom77 Autoradiographic data from our laboratory suggest that 5-HT3 receptors have a distinct expression gradient within the hippocampus, with the highest expression observed in the caudal and ventral portions (Figure 2E). Interestingly, histological evidence in the rodent forebrain suggests that 5-HT3 receptors are almost exclusively expressed on GABAergic interneurons.Reference Bloom and Morales78, Reference Morales and Bloom79 5-HT3 receptor-expressing interneurons are generally immunopositive for cholecystokinin, and for the calcium binding proteins calretinin and calbindin.Reference Morales and Bloom79

Based on this histological evidence, it can be hypothesized that 5-HT3 receptors provide a fast excitatory drive onto hippocampal GABAergic interneurons and inhibit hippocampal principal cells. Consistent with this hypothesis, pharmacological activation of 5-HT3 receptors depolarizes hippocampal interneuronsReference Kawa80, Reference McMahon and Kauer81 and increases inhibitory drive onto CA1 pyramidal cells.Reference Dale, Zhang and Leiser82Reference Turner, Mokler and Luebke85 Conversely, 5-HT3 receptor antagonists inhibit hippocampal interneurons, increase the firing rate of pyramidal cells, and enhance long-term potentiation (LTP) in in vivo electrophysiology recordings in rats.Reference Reznic and Staubli86Reference Staubli and Xu88 Taken together, these mechanistic findings might point to a pro-cognitive effect of 5-HT3 receptor antagonism. However, behavioral studies of selective 5-HT3 receptor antagonists in models of memory and learning have again shown inconsistent results (Table 2).

5-HT4 receptors

Autoradiographic studies have demonstrated the presence of 5-HT4 receptors throughout the hippocampus.Reference Vilaro, Cortes and Mengod89, Reference Waeber, Sebben, Nieoullon, Bockaert and Dumuis90 In general, protein expression is low-to-moderate, with the highest levels found in the stratum oriens and pyramidale of Ammon’s horn, subiculum, and the molecular layer of the DG (Table 1). 5-HT4 receptor mRNA has been detected in hippocampal pyramidal cells.Reference Peñas-Cazorla and Vilaró91 Interestingly, 5-HT4 receptor mRNA was not found in cells expressing glutamic acid decarboxylase 65 (GAD65), which is thought to be a selective marker of GABAergic neurons.Reference Peñas-Cazorla and Vilaró91 Thus, it appears that 5-HT4 receptors preferentially act to stimulate pyramidal neurons, without directly modulating GABA neurotransmission. In support of this hypothesis, 2 electrophysiology studies have shown that stimulation of 5-HT4 receptors increases the excitability of CA1 pyramidal cells.Reference Chapin, Haj-Dahmane, Torres and Andrade92, Reference Mlinar, Mascalchi, Mannaioni, Morini and Corradetti93

5-HT4 receptors have been shown to modulate the cholinergic system. In microdialysis recordings, application of the 5-HT4 receptor agonist SC53116 causes a release of ACh, and this effect is blocked by the 5-HT4 receptor antagonist GR113808.Reference Matsumoto, Togashi and Mori94 Thus in theory, 5-HT4 receptor agonists should be pro-cognitive. This hypothesis has been investigated in preclinical models, but the results to date have been disappointing (Table 2).

5-HT5 receptors

Immunohistochemical expression studies have shown that 5-HT5 receptors are present in some portions of the hippocampus. For example, Oliver et al Reference Oliver, Kinsey, Wainwright and Sirinathsinghji95 observed moderate immunoreactivity levels in CA1, CA2, and CA3 regions and weak immunoreactivity levels in the DG. This study also reported that 5-HT5 receptors were present on pyramidal and granule principal cells and on interneurons in the DG.Reference Oliver, Kinsey, Wainwright and Sirinathsinghji95 Due to the lack of selective compounds, no mechanistic and behavioral studies targeting 5-HT5 receptors have been performed in rodents.

5-HT6 receptors

Histochemical studies suggest that 5-HT6 receptors are expressed at moderate-to-high levels in all subfields of the rodent hippocampus.Reference Kinsey, Wainwright, Heavens, Sirinathsinghji and Oliver96, Reference Gerard, Martres and Lefevre97 The expression is particularly strong in the molecular layer of the DG and in the stratum oriens and stratum radiatum of CA1, where 5-HT6 receptors are thought to be expressed on dendritic processes of pyramidal cells.Reference Gerard, Martres and Lefevre97 Moderate levels of 5-HT6 receptor immunoreactivity have been observed in CA2 and CA3 regions. There has been also one report showing the expression of 5-HT6 receptors on a subset of calretinin- and calbindin-positive hippocampal interneurons.Reference de Jong and Helboe98

Pharmacological stimulation of 5-HT6 receptors by the 5-HT6 receptor agonist WAY-181187 increases GABA transmission and attenuates LTP in the CA1 area of the hippocampus.Reference West, Marcy, Marino and Schaffhauser99 Both of these effects were blocked by the selective 5-HT6 receptor antagonists SB-399885.Reference West, Marcy, Marino and Schaffhauser99 Furthermore, systemic administration of WAY-181187 increases GABA levels in several brain regions, including the dorsal hippocampus.Reference Schechter, Lin and Smith100 Consistent with its enhancing effect on GABA transmission, antagonism of 5-HT6 receptors increases extracellular glutamate levels in the frontal cortex and dorsal hippocampus.Reference Dawson, Nguyen and Li101 However, although the potentiating effects of 5-HT6 receptors on GABA transmission have been well documented, it is not clear whether these responses are due to direct effects of 5-HT6 receptors on GABAergic interneurons.Reference Pehrson and Sanchez102

Several 5-HT6 receptor antagonists are currently in clinical development for the treatment of Alzheimer’s disease.Reference Wilkinson, Windfeld and Colding-Jorgensen103 However, in rodent hippocampus-dependent behavioral models, the effects of 5-HT6 receptor antagonists have been only investigated in a small number of studies with variable results (Table 2).

5-HT7 receptors

Immunohistochemical data suggest that 5-HT7 receptors are expressed throughout Ammon’s horn, especially on the soma and dendrites of pyramidal neurons.Reference Neumaier, Sexton, Yracheta, Diaz and Brownfield104 There is also weak 5-HT7 receptor expression in the DG.Reference Neumaier, Sexton, Yracheta, Diaz and Brownfield104 It is important to note that in the rodent brain expression of 5-HT7 receptors changes during development. It is the highest during the first 2 post-natal weeks and progressively decreases with age.Reference Garcia-Alcocer, Segura, Garcia Pena, Martinez-Torres and Miledi105Reference Muneoka and Takigawa107 Our autoradiographic data using the selective 5-HT7 receptor antagonist [3H]SB269970 did not show strong binding in the hippocampal sections taken from adult rats (Figure 2G). This result raises questions regarding the level of expression of 5-HT7 receptors in the adult rodent hippocampus.

5-HT7 receptor stimulation increases the firing of pyramidal neurons and glutamatergic transmission in hippocampal brain slices.Reference Ciranna108Reference Tokarski, Zahorodna, Bobula and Hess111 Stimulation of 5-HT7 receptors also enhances inhibitory transmission in the hippocampus.Reference Tokarski, Kusek and Hess112 This suggests that 5-HT7 receptors might be expressed on GABAergic interneurons; however, there are no histological data available to support this notion. In behavioral studies, both 5-HT7 agonists and antagonists have shown both memory-enhancing and memory-impairing properties depending on the animal model and test conditions (such as 5-HT tone) (Table 2).Reference Gasbarri and Pompili113, Reference Meneses114 Thus, additional research is needed to determine the role of 5-HT7 receptors on cognition and memory function.

Effect of 5-HT and SSRIs on CA1 Pyramidal Cells and Hippocampal Function

Given that most 5-HT receptors are expressed on both excitatory cells and inhibitory interneurons and can function in either a stimulatory or inhibitory manner depending on the receptor subtype, it would be expected that the net effect of 5-HT on hippocampal function depends on local 5-HT concentration, the ratio of different 5-HT receptor subtypes expressed, and the density of 5-HT receptors in a particular population of cells. In general, 5-HT inhibits CA1 pyramidal cells and thereby decreases hippocampal output in rodents (reviewed by CirannaReference Ciranna108). Stimulation of the serotonergic projection from the dorsal raphe nucleus to the hippocampus decreases the firing rate of CA1 pyramidal cells in anesthetized animals.Reference Chaput, Araneda and Andrade55, Reference Chaput, Lesieur and de Montigny115Reference Segal118 In a similar manner, application of 5-HT to hippocampal brain slices inhibits the function of pyramidal neurons by hyperpolarizing their membrane potential and increasing local GABA transmission.Reference Andrade and Nicoll53, Reference Shen and Andrade74, Reference Passani, Pugliese, Azzurrini and Corradetti83Reference Turner, Mokler and Luebke85 There have also been reports of excitatory effects of 5-HT on pyramidal cell function, but the magnitude of excitation was much smaller than the 5-HT-induced inhibition.Reference Andrade and Nicoll53, Reference Andrade and Chaput119 However, one has to be careful in interpreting the ex vivo results obtained in brain slices; in most of these studies, 5-HT was exogenously applied at moderately high concentrations (15–50 micromolar), which might be higher than physiologically relevant concentrations of 5-HT in the brain. Therefore, these studies might exaggerate a contribution of certain subtypes of 5-HT receptors to its overall response. In summary, it seems that the overall effect of 5-HT on the hippocampal circuit in rodents is to inhibit pyramidal cell output. However, a majority of the studies that have led to this conclusion were either done in anesthetized animals or in brain slice preparations, and conclusions from such studies should be therefore interpreted with caution.

The inhibitory effect of 5-HT in the hippocampus is mediated via its actions on 5-HT1A, 5-HT1B, 5-HT2A/5-HT2C, 5-HT3, 5-HT6, and possibly 5-HT7 receptors.Reference Andrade and Nicoll53Reference Chaput, Araneda and Andrade55, Reference Shen and Andrade74, Reference McMahon and Kauer81Reference Ropert and Guy84, Reference West, Marcy, Marino and Schaffhauser99, Reference Tokarski, Kusek and Hess112, Reference Piguet and Galvan120 Activation of 5-HT1A receptors has a direct inhibitory effect on pyramidal cell firing by hyperpolarizing their membrane potential via activating a potassium conductance.Reference Andrade and Nicoll53Reference Chaput, Araneda and Andrade55 Other 5-HT receptors subtypes decrease the activity of pyramidal cells indirectly by mainly activating interneurons and enhancing GABA transmission onto pyramidal cells.Reference Shen and Andrade74, Reference McMahon and Kauer81Reference Ropert and Guy84, Reference West, Marcy, Marino and Schaffhauser99, Reference Tokarski, Kusek and Hess112, Reference Piguet and Galvan120 Multiple synaptic connections from hippocampal interneurons onto pyramidal cells might further amplify the inhibitory effect of 5-HT in the hippocampus.Reference Pehrson and Sanchez102

SSRIs, which act through the inhibition of the 5-HT transporter, are among the most studied serotonergic agents. Microdialysis studies in rodents have shown that systemic administration of SSRIs rapidly enhances extracellular 5-HT concentrations in multiple brain regions, including the ventral hippocampus.Reference Cremers, Rea and Bosker121Reference Hervas and Artigas123 Although SSRIs are selective for the serotonergic system, they do not show selectivity for 5-HT receptor subtypes and could, in theory, simultaneously activate all 5-HT receptors. However, since 5-HT receptors have different affinities for 5-HT, with 5-HT1A receptors being the most sensitive type (Table 1), local 5-HT concentrations in the brain would determine which 5-HT receptor subtypes become engaged upon SSRI treatment. In addition, chronic treatment with SSRIs, which is often required to achieve clinical efficacy, can desensitize and change expression patterns of 5-HT receptor subtypes.Reference Le Poul, Boni and Hanoun124 For instance, chronic treatment with paroxetine desensitizes presynaptic 5-HT1A autoreceptors in the raphe nuclei, which leads to increased serotonergic transmission.Reference Blier, de Montigny and Chaput125, Reference Chaput, de Montigny and Blier126

In electrophysiology studies, the effect of SSRIs in the hippocampus has been mostly studied in relationship to hippocampal LTP, which is thought to be important for learning and memory.Reference Malenka and Nicoll127 In a majority of studies in normal animals, both the application of 5-HT and acute and chronic treatments with SSRIs inhibit hippocampal LTP.Reference Staubli and Otaky87, Reference Corradetti, Ballerini, Pugliese and Pepeu128Reference Stewart and Reid132 Interestingly, exposure to stress also impairs LTP in the CA1 and DG regions of the hippocampus (reviewed by Pittenger et al Reference Pittenger and Duman133 and Popoli et al Reference Popoli, Gennarelli and Racagni134). Chronic treatments with SSRIs can reverse these stress-induced deficits in LTP,Reference Holderbach, Clark, Moreau, Bischofberger and Normann135, Reference Matsumoto, Tachibana and Togashi136 which suggests that SSRIs can restore hippocampal function in disease-like conditions. The positive effects of SSRIs are thought to be mediated, at least in part, by increasing the expression of brain-derived neurotrophic factor (BDNF) and neurogenesis in the hippocampal and cortical circuits.Reference O’Leary and Cryan23, Reference Pittenger and Duman133

Consistent with the hypothesis that SSRIs might stimulate multiple hippocampal 5-HT receptors expressed on different cell types, their net effects in behavioral cognition models have been limited and variable. The clinical experience with SSRIs is aligned with the preclinical data. Thus, while fMRI studies reveal that the neural systems important for emotional processing are adequately normalized by SSRIs in the treatment of depression,Reference Godlewska, Norbury, Selvaraj, Cowen and Harmer137 SSRIs are unable to correct the over-activation of the frontolimbic circuitry important for the non-emotional cognition.Reference Walsh, Williams and Brammer11 Although there are studies showing that SSRIs can remediate “hippocampal-related” cognitive deficits in patients with depression,Reference Vythilingam, Vermetten and Anderson19 a recent study by Herzallah et al Reference Herzallah, Moustafa and Natsheh138 indicates that SSRIs can also impair hippocampus-dependent generalization of past learning to novel contexts.

In conclusion, the regulation of hippocampal function by 5-HT is complex, involving multiple receptor subtypes and diverse expression patterns of 5-HT receptors on principal glutamatergic cells and GABAergic interneurons. Thus, administration of an SSRI may not be a rational approach to achieve enhanced hippocampal output and subsequent improvement of cognitive function in patients with MDD due to the potential of activation of multiple receptor subtypes, which may have opposing effects on cell function. On the other hand, targeting a single 5-HT receptor subtype may not be a viable strategy either due to redundancies in the serotonergic system. The consequences of modulating one 5-HT receptor may be attenuated by effects through other 5-HT receptor subtypes. Drugs designed to target single 5-HT receptors have so far not yielded new pharmacological treatments. For example, the selective 5-HT1A receptor agonist flesinoxan was under development for the treatment of generalized anxiety disorder for many years, but its clinical program was stopped in the late 1990s after it failed to show efficacy in 2 large phase-3 clinical trials.Reference Olivier139 The 5-HT1B/1D receptor antagonist elzasonan (CP-448187) was recently under development for the treatment of MDD and was tested in several phase-2 clinical trials, but its development program was also discontinued.Reference Kirchhoff, Nguyen, Soczynska, Woldeyohannes and McIntyre140 An alternative approach to targeting a single receptor subtype could be to target a combination of 5-HT receptor subtypes that would work in a concerted manner. The multimodal antidepressant vortioxetine is an example of such an approach.

Effects of the Multimodal Antidepressant Vortioxetine on Hippocampus Function

Vortioxetine is a 5-HT3, 5-HT7, and 5-HT1D receptor antagonist, a 5-HT1B receptor partial agonist, a 5-HT1A receptor agonist, and a SERT inhibitor in cellular assays.Reference Westrich, Pehrson and Zhong141, Reference Bang-Andersen, Ruhland and Jorgensen142 Vortioxetine has been approved for the treatment of MDD in the US, the EU, Australia and several other countries. Furthermore, clinical studies with cognitive outcome measures have shown that vortioxetine significantly improves cognitive function in MDD patients compared with placebo treatment. The efficacy of vortioxetine on cognitive function has been demonstrated in 3 randomized, double-blinded, placebo-controlled studies in MDD patients.Reference Katona, Hansen and Olsen143Reference Mahableshwarkar, Zajecka, Jacobson, Chen and Keefe145 One clinical trial was conducted in elderly MDD patients with cognition as a secondary pre-defined outcome and included 128 patients in the placebo group, 136 patients in the vortioxetine-treated group, and 128 patients in the duloxetine-treated group.Reference Katona, Hansen and Olsen143 The other 2 clinical trials were designed to compare the efficacy of vortioxetine to that of placebo on cognitive function as the primary efficacy outcome and on depressive symptoms as the secondary efficacy outcome.Reference McIntyre, Lophaven and Olsen144, Reference Mahableshwarkar, Zajecka, Jacobson, Chen and Keefe145 The study by McIntyre et al Reference McIntyre, Lophaven and Olsen144 included 196 patients in the placebo group, 195 patients in the 10 mg vortioxetine group, and 207 patients in the 20 mg vortioxetine group. The study by Mahableshwarkar et al Reference Mahableshwarkar, Zajecka, Jacobson, Chen and Keefe145 had 194 patients in the placebo group, 198 patients in the vortioxetine-treated group, and 210 patients in the duloxetine-treated group. These clinical studies demonstrated that vortioxetine improves objective measures of processing speed, executive function, attention and learning, and memory, including hippocampus-dependent memory measures.Reference Katona, Hansen and Olsen143, Reference McIntyre, Lophaven and Olsen144, Reference Mahableshwarkar, Zajecka, Jacobson, Chen and Keefe145 Path analyses suggested that the effect on cognitive function was largely independent of its effect on improvements in mood symptoms, supporting the hypothesis that these domains do not necessarily track together. Furthermore, an fMRI study showed that vortioxetine reduced neural activity in the left hippocampus during a working memory task in patients remitted from depression.Reference Browning, Smith and Conen146 This indicates that vortioxetine, unlike SSRIs, might restore compensatory over-activation in the hippocampus by increasing neural efficiency.Reference Walsh, Williams and Brammer11

Consistent with clinical findings, vortioxetine has shown antidepressant as well as pro-cognitive activities in a number of preclinical animal models.Reference Sanchez, Asin and Artigas147 Furthermore, in several behavioral and mechanistic studies that engaged hippocampal and cortical circuitry, vortioxetine’s effects differentiated from those of SSRIs and SNRIs.Reference Sanchez, Asin and Artigas147 In the following paragraphs, we review these preclinical results and discuss how vortioxetine might modulate hippocampal function and affect hippocampus-dependent cognitive behaviors in rodents.

Acute and chronic treatments with vortioxetine increase extracellular 5-HT levels in the rat ventral hippocampus to a much greater extent than those observed with SSRIs.Reference Mork, Pehrson and Brennum148, Reference Pehrson, Cremers and Betry149 Interestingly, combining an SSRI with the 5-HT3 receptor antagonist ondansetron resulted in a similar potentiating effect on 5-HT levels.Reference Mork, Pehrson and Brennum148 This suggests that the effect of vortioxetine was at least partially due to its 5-HT3 receptor antagonism. Since 5-HT3 receptors are expressed on GABAergic neurons,Reference Morales, Battenberg, de Lecea and Bloom76, Reference Puig, Santana, Celada, Mengod and Artigas150 it was hypothesized that vortioxetine, through its blockade of 5-HT3 receptors, reduces GABA release and thereby attenuates the inhibitory effect that GABA exerts on 5-HT release in the hippocampus.Reference Riga, Celada, Sanchez and Artigas151 Recent data by Riga et al Reference Riga, Celada, Sanchez and Artigas151 support this hypothesis. In their study, local application of ondansetron to the ventral hippocampus augmented the effect of the SSRI escitalopram on increasing extracellular 5‐HT levels. This effect was reversed by the local application of the GABAB receptor agonist baclofen, which restored GABAB receptor tone in the hippocampus.Reference Riga, Celada, Sanchez and Artigas151 Locally applied baclofen also attenuated the potentiating effect of vortioxetine on extracellular 5‐HT. Taken together, these results indicate that 5-HT3 receptor antagonism plays a prominent role in the vortioxetine’s effect on 5-HT levels in the hippocampus.

Although 5-HT3 receptor antagonism is important in the pharmacology of vortioxetine, contributions from its other receptor activities cannot be ruled out. For instance, an in vivo electrophysiology study of pyramidal neurons in the CA3 area of the hippocampus by El Mansari et al Reference El Mansari, Lecours and Blier152 showed that vortioxetine acts as a partial agonist of 5-HT1B receptors and can function as either an agonist or an antagonist depending on the endogenous 5-HT tone. Vortioxetine enhanced the inhibitory effect of the stimulation of the 5-HT bundle at a high, but not at a low frequency, and reversed the inhibitory effect of the 5-HT1B receptor agonist CP 94253.Reference El Mansari, Lecours and Blier152 Thus, vortioxetine also modulates the intra-hippocampal circuitry through its effects at 5-HT1B receptors. 5-HT1B receptors are densely expressed in the subiculum, the main output area of the hippocampus (Figure 2C), and are believed to play an important role in memory function.Reference O’Mara, Sanchez-Vives, Brotons-Mas and O’Hare153, Reference Sari154 Thus, vortioxetine, through its partial agonism at 5-HT1B receptors, might have a positive outcome on memory processing. Additional studies are needed to confirm this hypothesis, as well as to test the potential role of vortioxetine’s other receptor activities on hippocampal function.

Acute and sub-chronic treatments with vortioxetine also increase extracellular levels of NE and HA in the ventral hippocampus.Reference Pehrson, Cremers and Betry149, Reference Smagin, Song, Budac, Pehrson, Li and Sanchez155 Increases in DA and ACh levels have also been observed, but only after the acute treatment.Reference Mork, Pehrson and Brennum148, Reference Pehrson, Cremers and Betry149 Furthermore, a study in rat hippocampal slices showed that vortioxetine disinhibited CA1 pyramidal neurons in response to 5-HT, again most likely through its 5-HT3 receptor antagonism, whereas escitalopram had no effect on this measure.Reference Dale, Zhang and Leiser82 In line with these findings, several results indicate that vortioxetine promotes glutamate-dependent neuronal plasticity in the hippocampus to a greater degree than SSRIs. For example, vortioxetine, unlike escitalopram, produced a significant increase in LTP in rat hippocampal slices.Reference Dale, Zhang and Leiser82 Furthermore, in mice aged 12 months, chronic treatment with vortioxetine activated neuronal plasticity–related genes and improved hippocampus-dependent, visual-spatial memory deficits, whereas fluoxetine had no effect on these readouts.Reference Li, Abdourahman and Tamm156 In another study performed in rats, vortioxetine increased cell proliferation in the hippocampal DG faster than fluoxetine (3 days for vortioxetine compared to 10 days for fluoxetine).Reference Bétry, Etiévant, Pehrson, Sanchez and Haddjeri157 Vortioxetine also produced a larger degree of hippocampal dendritic branching than fluoxetine after 2 weeks of dosing in mice.Reference Guilloux, Mendez-David and Pehrson158 In line with these mechanistic data, vortioxetine showed pro-cognitive effects in hippocampus-dependent cognition models in rodents, such as footshock-induced fear conditioning and spontaneous alternation (Table 2). However, these findings are relatively recent, and vortioxetine has been studied less than other serotonergic drugs and receptors. For instance, there have been only 3 behavioral studies on the effects of vortioxetine on hippocampal-dependent memory versus 16 studies on 5-HT1A receptors and 10 studies on 5-HT2 receptors with different pharmacological and genetic approaches (Table 2). Thus, confirmation and expansion of these results are important.

Taken together, vortioxetine’s effects in the hippocampus support the notion that its antidepressant activities and pro-cognitive effects are mediated, at least to some extent, through increased glutamate neurotransmission and increased neuroplasticity. It is important to note that although increased glutamate neurotransmission is thought to favor neuronal plasticity, it is also clear that excessive glutamate release (for instance, in relation to stress) can be neurotoxic (reviewed in Sanacora and BanasrReference Sanacora and Banasr159 and Sanacora et al Reference Sanacora, Treccani and Popoli160). Vortioxetine’s effects on glutamate are limited to enhanced neuronal function. This has been shown in microdialysis studies, where the treatment with vortioxetine did not result in measureable changes in extracellular glutamate in the ventral hippocampus and prefrontal cortex.Reference Pehrson and Sanchez161

In conclusion, vortioxetine’s combined inhibition of 5-HT reuptake and 5-HT receptor modulation results in a differentiated effect on hippocampus function and hippocampal-dependent behavior compared to SSRIs. The full implication of vortioxetine’s modulation of multiple neurotransmitter systems on its antidepressant and pro-cognitive potential is complex and remains to be elucidated in future studies.

Overall Conclusion and Future Directions

There is considerable evidence to support the notion that the hippocampus plays an important role in emotional and cognitive processing, and that both of these functions are affected in patients with MDD. SSRIs and SNRIs are the predominant pharmacotherapies for treating MDD, and their enhancing effects on 5-HT levels are believed to be important for their therapeutic efficacy. However, the biological processes that lead to the recovery from the depressive state remain poorly understood. Furthermore, despite several decades of extensive research, the role of 5-HT in regulating hippocampal function in normal or disease states is not well understood, probably due to the high degree of complexity of the serotonergic system.

Multiple classes of 5-HT receptors are often co-expressed on the same cell types with functions that can either be complementary or opposing, and little is known about the interactions between different 5-HT receptors subtypes. Furthermore, the majority of 5-HT receptors in the hippocampus are found on both principal glutamatergic cells and GABAergic interneurons. The 2 known exceptions are the 5-HT3 receptor subclass, which has only been found on interneurons, and the 5-HT4 receptor subclass, which has only been found on pyramidal cells. 5-HT3 receptors are also the only non–G-protein-coupled receptors that function as a ligand-gated ion channel. The impact of the unique expression pattern and effector system of the 5-HT3 receptor remains to be elucidated. However, given the key role that 5-HT3 receptor antagonism appears to have in mediating the pharmacological effects of vortioxetine, at least in preclinical behavioral, electrophysiology, and microdialysis studies,Reference Mork, Pehrson and Brennum148, Reference Bétry, Etiévant, Pehrson, Sanchez and Haddjeri157, Reference Bétry, Pehrson, Etiévant, Ebert, Sánchez and Haddjeri162 this receptor subtype might have an important role in the hippocampus. 5-HT4 receptors have been less studied, and understanding their function in the hippocampus remains to be elucidated in further detail.

The effect of SSRIs on hippocampal function remains poorly defined. While SSRIs have the potential to normalize hippocampal plasticity under stress conditions and to treat mood symptoms in MDD, their effects on cognitive function are less clear. Since SSRIs do not possess selectivity for 5-HT receptor subtypes, their efficacy might be weakened due to opposing activities of different 5-HT receptor subtypes. In this context, multitarget drugs or combination therapies might be a better strategy to modulate both emotional and cognitive processes in the hippocampus. Future insights into interactions between different serotonergic subtypes might therefore lead to novel treatment options for the treatment of MDD.

Disclosures

All authors are full-time employees of H. Lundbeck A/S.

Footnotes

We would like to thank Dr. Bjarke Ebert (H Lundbeck A/S) and Simon Reid (Fusion Animation, London, United Kingdom) for assistance in preparing Figure 1 of the manuscript. This research was funded by Takeda Pharmaceuticals and H. Lundbeck A/S.

References

1.Femenia, T, Gomez-Galan, M, Lindskog, M, Magara, S. Dysfunctional hippocampal activity affects emotion and cognition in mood disorders. Brain Res. 2012; 1476: 5870.Google Scholar
2.Small, SA, Schobel, SA, Buxton, RB, Witter, MP, Barnes, CA. A pathophysiological framework of hippocampal dysfunction in ageing and disease. Nat Rev Neurosci. 2011; 12(10): 585601.CrossRefGoogle ScholarPubMed
3.Bell-McGinty, S, Butters, MA, Meltzer, CC, Greer, PJ, Reynolds, CF 3rd, Becker, JT. Brain morphometric abnormalities in geriatric depression: long-term neurobiological effects of illness duration. Am J Psychiatry. 2002; 159(8): 14241427.CrossRefGoogle ScholarPubMed
4.Bremner, JD, Narayan, M, Anderson, ER, Staib, LH, Miller, HL, Charney, DS. Hippocampal volume reduction in major depression. Am J Psychiatry. 2000; 157(1): 115118.Google Scholar
5.Frodl, TS, Koutsouleris, N, Bottlender, R, et al. Depression-related variation in brain morphology over 3 years: effects of stress? Arch Gen Psychiatry. 2008; 65(10): 11561165.Google Scholar
6.Sheline, YI, Gado, MH, Kraemer, HC. Untreated depression and hippocampal volume loss. Am J Psychiatry. 2003; 160(8): 15161518.CrossRefGoogle ScholarPubMed
7.Videbech, P, Ravnkilde, B. Hippocampal volume and depression: a meta-analysis of MRI studies. Am J Psychiatry. 2004; 161(11): 19571966.Google Scholar
8.MacQueen, GM, Campbell, S, McEwen, BS, et al. Course of illness, hippocampal function, and hippocampal volume in major depression. Proc Natl Acad Sci U S A. 2003; 100(3): 13871392.CrossRefGoogle ScholarPubMed
9.Gorwood, P, Corruble, E, Falissard, B, Goodwin, GM. Toxic effects of depression on brain function: impairment of delayed recall and the cumulative length of depressive disorder in a large sample of depressed outpatients. Am J Psychiatry. 2008; 165(6): 731739.CrossRefGoogle Scholar
10.Harvey, PO, Fossati, P, Pochon, JB, et al. Cognitive control and brain resources in major depression: an fMRI study using the n-back task. Neuroimage. 2005; 26(3): 860869.CrossRefGoogle ScholarPubMed
11.Walsh, ND, Williams, SC, Brammer, MJ, et al. A longitudinal functional magnetic resonance imaging study of verbal working memory in depression after antidepressant therapy. Biol Psychiatry. 2007; 62(11): 12361243.Google Scholar
12.Hammar, A, Ardal, G. Cognitive functioning in major depression—a summary. Front Hum Neurosci. 2009; 3: 26.Google Scholar
13.Lee, RS, Hermens, DF, Porter, MA, Redoblado-Hodge, MA. A meta-analysis of cognitive deficits in first-episode major depressive disorder. J Affect Disord. 2012; 140(2): 113124.CrossRefGoogle ScholarPubMed
14.Porter, RJ, Bourke, C, Gallagher, P. Neuropsychological impairment in major depression: its nature, origin and clinical significance. Aust N Z J Psychiatry. 2007; 41(2): 115128.CrossRefGoogle ScholarPubMed
15.Hasselbalch, BJ, Knorr, U, Kessing, LV. Cognitive impairment in the remitted state of unipolar depressive disorder: a systematic review. J Affect Disord. 2011; 134(1–3): 2031.Google Scholar
16.Campbell, S, MacQueen, G. The role of the hippocampus in the pathophysiology of major depression. J Psychiatry Neurosci. 2004; 29(6): 417426.Google Scholar
17.Biringer, E, Rongve, A, Lund, A. A review of modern antidepressants’ effects on neurocognitive function. Cur Psychiatr Rev. 2009; 5(3): 164174.Google Scholar
18.McIntyre, RS, Cha, DS, Soczynska, JK, et al. Cognitive deficits and functional outcomes in major depressive disorder: determinants, substrates, and treatment interventions. Depress Anxiety. 2013; 30(6): 515527.Google Scholar
19.Vythilingam, M, Vermetten, E, Anderson, GM, et al. Hippocampal volume, memory, and cortisol status in major depressive disorder: effects of treatment. Biol Psychiatry. 2004; 56(2): 101112.Google Scholar
20.Fanselow, MS, Dong, HW. Are the dorsal and ventral hippocampus functionally distinct structures? Neuron. 2010; 65(1): 719.Google Scholar
21.Moser, MB, Moser, EI. Functional differentiation in the hippocampus. Hippocampus. 1998; 8(6): 608619.Google Scholar
22.Poppenk, J, Evensmoen, HR, Moscovitch, M, Nadel, L. Long-axis specialization of the human hippocampus. Trends Cogn Sci. 2013; 17(5): 230240.Google Scholar
23.O’Leary, OF, Cryan, JF. A ventral view on antidepressant action: roles for adult hippocampal neurogenesis along the dorsoventral axis. Trends Pharmacol Sci. 2014; 35(12): 675687.Google Scholar
24.Moser, E, Moser, MB, Andersen, P. Spatial learning impairment parallels the magnitude of dorsal hippocampal lesions, but is hardly present following ventral lesions. J Neurosci. 1993; 13(9): 39163925.CrossRefGoogle ScholarPubMed
25.Pothuizen, HH, Zhang, WN, Jongen-Relo, AL, Feldon, J, Yee, BK. Dissociation of function between the dorsal and the ventral hippocampus in spatial learning abilities of the rat: a within-subject, within-task comparison of reference and working spatial memory. Eur J Neurosci. 2004; 19(3): 705712.CrossRefGoogle Scholar
26.Bannerman, DM, Grubb, M, Deacon, RM, Yee, BK, Feldon, J, Rawlins, JN. Ventral hippocampal lesions affect anxiety but not spatial learning. Behav Brain Res. 2003; 139(1–2): 197213.CrossRefGoogle Scholar
27.Bannerman, DM, Rawlins, JN, McHugh, SB, et al. Regional dissociations within the hippocampus—memory and anxiety. Neurosci Biobehav Rev. 2004; 28(3): 273283.CrossRefGoogle ScholarPubMed
28.Kjelstrup, KG, Tuvnes, FA, Steffenach, HA, Murison, R, Moser, EI, Moser, MB. Reduced fear expression after lesions of the ventral hippocampus. Proc Natl Acad Sci U S A. 2002; 99(16): 1082510830.Google Scholar
29.Rudy, JW, Matus-Amat, P. The ventral hippocampus supports a memory representation of context and contextual fear conditioning: implications for a unitary function of the hippocampus. Behav Neurosci. 2005; 119(1): 154163.Google Scholar
30.Ramon y, Cajal S. Histologie du Systeme Nerveux de l’Homme et des Vertbré Vol II. Paris: Maloine; 1911.Google Scholar
31.Lorente de Nó, R. Studies of the structure of the cerebral cortex. II. Continuation of the study of the ammonic system. Journal für Psychologie und Neurologie. 1934; 46: 113177.Google Scholar
32.Lavenex, P. Functional anatomy, development and pathology of the hippocampus. In: Bartsch T ed. The Clinical Neurobiology of the Hippocampus: An Integrative View. Oxford, UK: Oxford University Press; 2012: 1038.Google Scholar
33.Freund, TF, Buzsáki, G. Interneurons of the hippocampus. Hippocampus. 1996; 6(4): 347470.Google Scholar
34.Parra, P, Gulyas, AI, Miles, R. How many subtypes of inhibitory cells in the hippocampus? Neuron. 1998; 20(5): 983993.Google Scholar
35.Ribak, CE, Nitsch, R, Seress, L. Proportion of parvalbumin-positive basket cells in the GABAergic innervation of pyramidal and granule cells of the rat hippocampal formation. J Comp Neurol. 1990; 300(4): 449461.Google Scholar
36.Sloviter, RS. Calcium-binding protein (calbindin-D28k) and parvalbumin immunocytochemistry: localization in the rat hippocampus with specific reference to the selective vulnerability of hippocampal neurons to seizure activity. J Comp Neurol. 1989; 280(2): 183196.Google Scholar
37.Aznar, S, Qian, Z, Shah, R, Rahbek, B, Knudsen, GM. The 5-HT1A serotonin receptor is located on calbindin- and parvalbumin-containing neurons in the rat brain. Brain Res. 2003; 959(1): 5867.CrossRefGoogle ScholarPubMed
38.Gulyas, AI, Hajos, N, Freund, TF. Interneurons containing calretinin are specialized to control other interneurons in the rat hippocampus. J Neurosci. 1996; 16(10): 33973411.Google Scholar
39.Berumen, LC, Rodriguez, A, Miledi, R, Garcia-Alcocer, G. Serotonin receptors in hippocampus. ScientificWorldJournal. 2012; 2012: 823493.CrossRefGoogle ScholarPubMed
40.Oleskevich, S, Descarries, L, Watkins, KC, Seguela, P, Daszuta, A. Ultrastructural features of the serotonin innervation in adult rat hippocampus: an immunocytochemical description in single and serial thin sections. Neuroscience. 1991; 42(3): 777791.Google Scholar
41.Umbriaco, D, Garcia, S, Beaulieu, C, Descarries, L. Relational features of acetylcholine, noradrenaline, serotonin and GABA axon terminals in the stratum radiatum of adult rat hippocampus (CA1). Hippocampus. 1995; 5(6): 605620.Google Scholar
42.Vizi, ES, Kiss, JP. Neurochemistry and pharmacology of the major hippocampal transmitter systems: synaptic and nonsynaptic interactions. Hippocampus. 1998; 8(6): 566607.3.0.CO;2-W>CrossRefGoogle ScholarPubMed
43.Aghajanian, GK, Sanders-Bush, E. Serotonin. In: Davis KL, Charney D, Coyle JT, Nemeroff C eds. Neuropsychopharmacology: The Fifth Generation of Progress. Philadelphia: Lippincott, Williams, & Wilkins; 2002.Google Scholar
44.Glennon, RA, Dukat, M, Westkaemper, RB. Serotonin receptor subtypes and ligands. In: Bloom FE, Kupfer DJ eds. Psychopharmacology: The Fourth Generation of Progress. New York: Raven Press; 2000.Google Scholar
45.Herrick-Davis, K. Functional significance of serotonin receptor dimerization. Exp Brain Res. 2013; 230(4): 375386.Google Scholar
46.Gonzalez-Maeso, J, Ang, RL, Yuen, T, et al. Identification of a serotonin/glutamate receptor complex implicated in psychosis. Nature. 2008; 452(7183): 9397.CrossRefGoogle ScholarPubMed
47.Naumenko, VS, Popova, NK, Lacivita, E, Leopoldo, M, Ponimaskin, EG. Interplay between serotonin 5-HT1A and 5-HT7 receptors in depressive disorders. CNS Neurosci Ther. 2014; 20(7): 582590.Google Scholar
48.Jarrard, LE. On the role of the hippocampus in learning and memory in the rat. Behav Neural Biol. 1993; 60(1): 926.CrossRefGoogle ScholarPubMed
49.Squire, LR. Memory and the hippocampus: a synthesis from findings with rats, monkeys, and humans. Psychol Rev. 1992; 99(2): 195231.Google Scholar
50.Knierim, JJ, Hamilton, DA. Framing spatial cognition: neural representations of proximal and distal frames of reference and their roles in navigation. Physiological Rev. 2011; 91(4): 12451279.Google Scholar
51.Luscher, C, Jan, LY, Stoffel, M, Malenka, RC, Nicoll, RA. G protein-coupled inwardly rectifying K+ channels (GIRKs) mediate postsynaptic but not presynaptic transmitter actions in hippocampal neurons. Neuron. 1997; 19(3): 687695.Google Scholar
52.Pompeiano, M, Palacios, JM, Mengod, G. Distribution and cellular localization of mRNA coding for 5-HT1A receptor in the rat brain: correlation with receptor binding. J Neurosci. 1992; 12(2): 440453.Google Scholar
53.Andrade, R, Nicoll, RA. Pharmacologically distinct actions of serotonin on single pyramidal neurones of the rat hippocampus recorded in vitro. J Physiol. 1987; 394: 99124.Google Scholar
54.Beck, SG, Clarke, WP, Goldfarb, J. Spiperone differentiates multiple 5-hydroxytryptamine responses in rat hippocampal slices in vitro. Eur J Pharmacol. 1985; 116(1–2): 195197.Google Scholar
55.Chaput, Y, Araneda, RC, Andrade, R. Pharmacological and functional analysis of a novel serotonin receptor in the rat hippocampus. Eur J Pharmacol. 1990; 182(3): 441456.CrossRefGoogle ScholarPubMed
56.Haddjeri, N, Blier, P, de Montigny, C. Long-term antidepressant treatments result in a tonic activation of forebrain 5-HT1A receptors. J Neursci. 1998; 18(23): 1015010156.CrossRefGoogle Scholar
57.Johnston, A, McBain, CJ, Fisahn, A. 5-Hydroxytryptamine1A receptor-activation hyperpolarizes pyramidal cells and suppresses hippocampal gamma oscillations via Kir3 channel activation. J Physiol. 2014; 592(Pt 19): 41874199.CrossRefGoogle ScholarPubMed
58.Kasamo, K, Suzuki, T, Tada, K, et al. Endogenous 5-HT tonically inhibits spontaneous firing activity of dorsal hippocampus CA1 pyramidal neurons through stimulation of 5-HT(1A) receptors in quiet awake rats: in vivo electrophysiological evidence. Neuropsychopharmacology. 2001; 24(2): 141151.Google Scholar
59.Tada, K, Kasamo, K, Suzuki, T, Matsuzaki, Y, Kojima, T. Endogenous 5-HT inhibits firing activity of hippocampal CA1 pyramidal neurons during conditioned fear stress-induced freezing behavior through stimulating 5-HT1A receptors. Hippocampus. 2004; 14(2): 143147.Google Scholar
60.Llado-Pelfort, L, Santana, N, Ghisi, V, Artigas, F, Celada, P. 5-HT1A receptor agonists enhance pyramidal cell firing in prefrontal cortex through a preferential action on GABA interneurons. Cereb Cortex. 2012; 22(7): 14871497.CrossRefGoogle ScholarPubMed
61.Haider, S, Khaliq, S, Tabassum, S, Haleem, DJ. Role of somatodendritic and postsynaptic 5-HT(1)A receptors on learning and memory functions in rats. Neurochem Res. 2012; 37(10): 21612166.Google Scholar
62.Newman-Tancredi, A, Martel, JC, Assie, MB, et al. Signal transduction and functional selectivity of F15599, a preferential post-synaptic 5-HT1A receptor agonist. Br J Pharmacol. 2009; 156(2): 338353.CrossRefGoogle ScholarPubMed
63.Boschert, U, Amara, DA, Segu, L, Hen, R. The mouse 5-hydroxytryptamine1B receptor is localized predominantly on axon terminals. Neuroscience. 1994; 58(1): 167182.Google Scholar
64.Bruinvels, AT, Landwehrmeyer, B, Gustafson, EL, et al. Localization of 5-HT1B, 5-HT1D alpha, 5-HT1E and 5-HT1F receptor messenger RNA in rodent and primate brain. Neuropharmacology. 1994; 33(3–4): 367386.Google Scholar
65.Sari, Y, Lefevre, K, Bancila, M, et al. Light and electron microscopic immunocytochemical visualization of 5-HT1B receptors in the rat brain. Brain Res. 1997; 760(1–2): 281286.CrossRefGoogle ScholarPubMed
66.Bruinvels, AT, Palacios, JM, Hoyer, D. Autoradiographic characterisation and localisation of 5-HT1D compared to 5-HT1B binding sites in rat brain. Naunyn Schmiedebergs Arch Pharmacol. 1993; 347(6): 569582.Google Scholar
67.Xie, Z, Lee, SP, O’Dowd, BF, George, SR. Serotonin 5-HT1B and 5-HT1D receptors form homodimers when expressed alone and heterodimers when co-expressed. FEBS Lett. 1999; 456(1): 6367.CrossRefGoogle ScholarPubMed
68.Boeijinga, PH, Boddeke, HW. Serotonergic modulation of neurotransmission in the rat subicular cortex in vitro: a role for 5-HT1B receptors. Naunyn Schmiedebergs Arch Pharmacol. 1993; 348(6): 553557.Google Scholar
69.Mlinar, B, Falsini, C, Corradetti, R. Pharmacological characterization of 5-HT(1B) receptor-mediated inhibition of local excitatory synaptic transmission in the CA1 region of rat hippocampus. Br J Pharmacol. 2003; 138(1): 7180.CrossRefGoogle ScholarPubMed
70.Hu, XJ, Wang, FH, Stenfors, C, Ogren, SO, Kehr, J. Effects of the 5-HT1B receptor antagonist NAS-181 on extracellular levels of acetylcholine, glutamate and GABA in the frontal cortex and ventral hippocampus of awake rats: a microdialysis study. Eur Neuropsychopharmacol. 2007; 17(9): 580586.CrossRefGoogle ScholarPubMed
71.Bombardi, C. Neuronal localization of 5-HT2A receptor immunoreactivity in the rat hippocampal region. Brain Res Bull. 2012; 87(2–3): 259273.Google Scholar
72.Cornea-Hebert, V, Riad, M, Wu, C, Singh, SK, Descarries, L. Cellular and subcellular distribution of the serotonin 5-HT2A receptor in the central nervous system of adult rat. J Comp Neurol. 1999; 409(2): 187209.Google Scholar
73.Luttgen, M, Ove Ogren, S, Meister, B. Chemical identity of 5-HT2A receptor immunoreactive neurons of the rat septal complex and dorsal hippocampus. Brain Res. 2004; 1010(1–2): 156165.Google Scholar
74.Shen, RY, Andrade, R. 5-Hydroxytryptamine2 receptor facilitates GABAergic neurotransmission in rat hippocampus. J Pharmacol Exp Ther. 1998; 285(2): 805812.Google Scholar
75.Wang, RY, Arvanov, VL. M100907, a highly selective 5-HT2A receptor antagonist and a potential atypical antipsychotic drug, facilitates induction of long-term potentiation in area CA1 of the rat hippocampal slice. Brain Res. 1998; 779(1–2): 309313.Google Scholar
76.Morales, M, Battenberg, E, de Lecea, L, Bloom, FE. The type 3 serotonin receptor is expressed in a subpopulation of GABAergic neurons in the rat neocortex and hippocampus. Brain Res. 1996; 731(1–2): 199202.Google Scholar
77.Morales, M, Battenberg, E, de Lecea, L, Sanna, PP, Bloom, FE. Cellular and subcellular immunolocalization of the type 3 serotonin receptor in the rat central nervous system. Brain Res Mol Brain Res. 1996; 36(2): 251260.Google Scholar
78.Bloom, FE, Morales, M. The central 5-HT3 receptor in CNS disorders. Neurochem Res. 1998; 23(5): 653659.Google Scholar
79.Morales, M, Bloom, FE. The 5-HT3 receptor is present in different subpopulations of GABAergic neurons in the rat telencephalon. J Neurosci. 1997; 17(9): 31573167.Google Scholar
80.Kawa, K. Distribution and functional properties of 5-HT3 receptors in the rat hippocampal dentate gyrus: a patch-clamp study. J Neurophysiol. 1994; 71(5): 19351947.Google Scholar
81.McMahon, LL, Kauer, JA. Hippocampal interneurons are excited via serotonin-gated ion channels. J Neurophysiol. 1997; 78(5): 24932502.Google Scholar
82.Dale, E, Zhang, H, Leiser, SC, et al. Vortioxetine disinhibits pyramidal cell function and enhances synaptic plasticity in the rat hippocampus. J Psychopharmacol. 2014; 28(10): 891902.CrossRefGoogle ScholarPubMed
83.Passani, MB, Pugliese, AM, Azzurrini, M, Corradetti, R. Effects of DAU 6215, a novel 5-hydroxytryptamine3 (5-HT3) antagonist on electrophysiological properties of the rat hippocampus. Br J Pharmacol. 1994; 112(2): 695703.Google Scholar
84.Ropert, N, Guy, N. Serotonin facilitates GABAergic transmission in the CA1 region of rat hippocampus in vitro. J Physiol. 1991; 441(1): 121136.CrossRefGoogle ScholarPubMed
85.Turner, TJ, Mokler, DJ, Luebke, JI. Calcium influx through presynaptic 5-HT3 receptors facilitates GABA release in the hippocampus: in vitro slice and synaptosome studies. Neuroscience. 2004; 129(3): 703718.CrossRefGoogle ScholarPubMed
86.Reznic, J, Staubli, U. Effects of 5-HT3 receptor antagonism on hippocampal cellular activity in the freely moving rat. J Neurophysiol. 1997; 77(1): 517521.Google Scholar
87.Staubli, U, Otaky, N. Serotonin controls the magnitude of LTP induced by theta bursts via an action on NMDA-receptor-mediated responses. Brain Res. 1994; 643(1–2): 1016.Google Scholar
88.Staubli, U, Xu, FB. Effects of 5-HT3 receptor antagonism on hippocampal theta rhythm, memory, and LTP induction in the freely moving rat. J Neurosci. 1995; 15(3 Pt 2): 24452452.Google Scholar
89.Vilaro, MT, Cortes, R, Mengod, G. Serotonin 5-HT4 receptors and their mRNAs in rat and guinea pig brain: distribution and effects of neurotoxic lesions. J Comp Neurol. 2005; 484(4): 418439.CrossRefGoogle ScholarPubMed
90.Waeber, C, Sebben, M, Nieoullon, A, Bockaert, J, Dumuis, A. Regional distribution and ontogeny of 5-HT4 binding sites in rodent brain. Neuropharmacology. 1994; 33(3–4): 527541.Google Scholar
91.Peñas-Cazorla, R, Vilaró, MT. Serotonin 5-HT receptors and forebrain cholinergic system: receptor expression in identified cell populations. Brain Struct Funct. In press. DOI: 10.1007/s00429-014-0864-z.Google Scholar
92.Chapin, EM, Haj-Dahmane, S, Torres, G, Andrade, R. The 5-HT(4) receptor-induced depolarization in rat hippocampal neurons is mediated by cAMP but is independent of I(h). Neurosci Lett. 2002; 324(1): 14.CrossRefGoogle Scholar
93.Mlinar, B, Mascalchi, S, Mannaioni, G, Morini, R, Corradetti, R. 5-HT4 receptor activation induces long-lasting EPSP-spike potentiation in CA1 pyramidal neurons. Eur J Nneurosci. 2006; 24(3): 719731.Google Scholar
94.Matsumoto, M, Togashi, H, Mori, K, et al. Evidence for involvement of central 5-HT(4) receptors in cholinergic function associated with cognitive processes: behavioral, electrophysiological, and neurochemical studies. J Pharmacol Exp Ther. 2001; 296(3): 676682.Google Scholar
95.Oliver, KR, Kinsey, AM, Wainwright, A, Sirinathsinghji, DJ. Localization of 5-ht(5A) receptor-like immunoreactivity in the rat brain. Brain Res. 2000; 867(1–2): 131142.CrossRefGoogle ScholarPubMed
96.Kinsey, AM, Wainwright, A, Heavens, R, Sirinathsinghji, DJ, Oliver, KR. Distribution of 5-ht(5A), 5-ht(5B), 5-ht(6) and 5-HT(7) receptor mRNAs in the rat brain. Brain Res Mol Brain Res. 2001; 88(1–2): 194198.Google Scholar
97.Gerard, C, Martres, MP, Lefevre, K, et al. Immuno-localization of serotonin 5-HT6 receptor-like material in the rat central nervous system. Brain Res. 1997; 746(1–2): 207219.CrossRefGoogle ScholarPubMed
98.de Jong, I, Helboe, L. Distribution of serotonin receptor 5-HT6 mRNA in selected neuronal populations in rat brain: a double-labelling in situ hybridization study. Alzheimer’s & Dementia. 2014; 10(4 Suppl): 925926.Google Scholar
99.West, PJ, Marcy, VR, Marino, MJ, Schaffhauser, H. Activation of the 5-HT(6) receptor attenuates long-term potentiation and facilitates GABAergic neurotransmission in rat hippocampus. Neuroscience. 2009; 164(2): 692701.Google Scholar
100.Schechter, LE, Lin, Q, Smith, DL, et al. Neuropharmacological profile of novel and selective 5-HT6 receptor agonists: WAY-181187 and WAY-208466. Neuropsychopharmacol. 2008; 33(6): 13231335.Google Scholar
101.Dawson, LA, Nguyen, HQ, Li, P. The 5-HT(6) receptor antagonist SB-271046 selectively enhances excitatory neurotransmission in the rat frontal cortex and hippocampus. Neuropsychopharmacology. 2001; 25(5): 662668.Google Scholar
102.Pehrson, AL, Sanchez, C. Altered gamma-aminobutyric acid neurotransmission in major depressive disorder: a critical review of the supporting evidence and the influence of serotonergic antidepressants. Drug Des Devel Ther. 2015; 9: 603624.Google Scholar
103.Wilkinson, D, Windfeld, K, Colding-Jorgensen, E. Safety and efficacy of idalopirdine, a 5-HT6 receptor antagonist, in patients with moderate Alzheimer’s disease (LADDER): a randomised, double-blind, placebo-controlled phase 2 trial. Lancet Neurol. 2014; 13(11): 10921099.Google Scholar
104.Neumaier, JF, Sexton, TJ, Yracheta, J, Diaz, AM, Brownfield, M. Localization of 5-HT(7) receptors in rat brain by immunocytochemistry, in situ hybridization, and agonist stimulated cFos expression. J Chem Neuroanat. 2001; 21(1): 6373.Google Scholar
105.Garcia-Alcocer, G, Segura, LC, Garcia Pena, M, Martinez-Torres, A, Miledi, R. Ontogenetic distribution of 5-HT2C, 5-HT5A, and 5-HT7 receptors in the rat hippocampus. Gene Expr. 2006; 13(1): 5357.Google Scholar
106.Kobe, F, Guseva, D, Jensen, TP, et al. 5-HT7R/G12 signaling regulates neuronal morphology and function in an age-dependent manner. J Neurosci. 2012; 32(9): 29152930.CrossRefGoogle Scholar
107.Muneoka, KT, Takigawa, M. 5-Hydroxytryptamine7 (5-HT7) receptor immunoreactivity-positive ‘stigmoid body’-like structure in developing rat brains. Int J Dev Neurosci. 2003; 21(3): 133143.Google Scholar
108.Ciranna, L. Serotonin as a modulator of glutamate- and GABA-mediated neurotransmission: implications in physiological functions and in pathology. Curr Neuropharmacol. 2006; 4(2): 101114.Google Scholar
109.Costa, L, Trovato, C, Musumeci, SA, Catania, MV, Ciranna, L. 5-HT(1A) and 5-HT(7) receptors differently modulate AMPA receptor-mediated hippocampal synaptic transmission. Hippocampus. 2012; 22(4): 790801.Google Scholar
110.Gill, CH, Soffin, EM, Hagan, JJ, Davies, CH. 5-HT7 receptors modulate synchronized network activity in rat hippocampus. Neuropharmacology. 2002; 42(1): 8292.Google Scholar
111.Tokarski, K, Zahorodna, A, Bobula, B, Hess, G. 5-HT7 receptors increase the excitability of rat hippocampal CA1 pyramidal neurons. Brain Res. 2003; 993(1–2): 230234.Google Scholar
112.Tokarski, K, Kusek, M, Hess, G. 5-HT7 receptors modulate GABAergic transmission in rat hippocampal CA1 area. J Physiol Pharmacol. 2011; 62(5): 535540.Google Scholar
113.Gasbarri, A, Pompili, A. Serotonergic 5-HT7 receptors and cognition. Rev Neurosci. 2014; 25(3): 311323.Google Scholar
114.Meneses, A. Memory formation and memory alterations: 5-HT6 and 5-HT7 receptors, novel alternative. Rev Neurosci. 2014; 25(3): 325356.Google Scholar
115.Chaput, Y, Lesieur, P, de Montigny, C. Effects of short-term serotonin depletion on the efficacy of serotonin neurotransmission: electrophysiological studies in the rat central nervous system. Synapse. 1990; 6(4): 328337.Google Scholar
116.Ferraro, G, Montalbano, ME, Sardo, P, La Grutta, V. Lateral habenula and hippocampus: a complex interaction raphe cells-mediated. J Neural Transm. 1997; 104(6–7): 615631.Google Scholar
117.Segal, M. Physiological and pharmacological evidence for a serotonergic projection to the hippocampus. Brain Res. 1975; 94(1): 115131.Google Scholar
118.Segal, M. 5-HT antagonists in rat hippocampus. Brain Res. 1976; 103(1): 161166.Google Scholar
119.Andrade, R, Chaput, Y. 5-Hydroxytryptamine4-like receptors mediate the slow excitatory response to serotonin in the rat hippocampus. J Pharmacol Exp Ther. 1991; 257(3): 930937.Google Scholar
120.Piguet, P, Galvan, M. Transient and long-lasting actions of 5-HT on rat dentate gyrus neurones in vitro. J Physiol. 1994; 481(Pt 3): 629639.Google Scholar
121.Cremers, TI, Rea, K, Bosker, FJ, et al. Augmentation of SSRI effects on serotonin by 5-HT2C antagonists: mechanistic studies. Neuropsychopharmacology. 2007; 32(7): 15501557.Google Scholar
122.Haider, S, Khaliq, S, Ahmed, SP, Haleem, DJ. Long-term tryptophan administration enhances cognitive performance and increases 5HT metabolism in the hippocampus of female rats. Amino Acids. 2006; 31(4): 421425.Google Scholar
123.Hervas, I, Artigas, F. Effect of fluoxetine on extracellular 5-hydroxytryptamine in rat brain. Role of 5-HT autoreceptors. Eur J Pharmacol. 1998; 358(1): 918.CrossRefGoogle ScholarPubMed
124.Le Poul, E, Boni, C, Hanoun, N, et al. Differential adaptation of brain 5-HT1A and 5-HT1B receptors and 5-HT transporter in rats treated chronically with fluoxetine. Neuropharmacology. 2000; 39(1): 110122.Google Scholar
125.Blier, P, de Montigny, C, Chaput, Y. A role for the serotonin system in the mechanism of action of antidepressant treatments: preclinical evidence. J Clin Psychiatry. 1990; 51(Suppl): 1420; discussion 21.Google Scholar
126.Chaput, Y, de Montigny, C, Blier, P. Presynaptic and postsynaptic modifications of the serotonin system by long-term administration of antidepressant treatments. An in vivo electrophysiologic study in the rat. Neuropsychopharmacology. 1991; 5(4): 219229.Google Scholar
127.Malenka, RC, Nicoll, RA. Long-term potentiation—a decade of progress? Science. 1999; 285(5435): 18701874.Google Scholar
128.Corradetti, R, Ballerini, L, Pugliese, AM, Pepeu, G. Serotonin blocks the long-term potentiation induced by primed burst stimulation in the CA1 region of rat hippocampal slices. Neuroscience. 1992; 46(3): 511518.Google Scholar
129.Mnie-Filali, O, El Mansari, M, Espana, A, Sanchez, C, Haddjeri, N. Allosteric modulation of the effects of the 5-HT reuptake inhibitor escitalopram on the rat hippocampal synaptic plasticity. Neurosci Lett. 2006; 395(1): 2327.Google Scholar
130.Ryan, B, Musazzi, L, Mallei, A, et al. Remodelling by early-life stress of NMDA receptor-dependent synaptic plasticity in a gene-environment rat model of depression. Int J Neuropsychopharmacol. 2009; 12(4): 553559.CrossRefGoogle Scholar
131.Shakesby, AC, Anwyl, R, Rowan, MJ. Overcoming the effects of stress on synaptic plasticity in the intact hippocampus: rapid actions of serotonergic and antidepressant agents. J Neurosci. 2002; 22(9): 36383644.Google Scholar
132.Stewart, CA, Reid, IC. Repeated ECS and fluoxetine administration have equivalent effects on hippocampal synaptic plasticity. Psychopharmacology (Berl). 2000; 148(3): 217223.Google Scholar
133.Pittenger, C, Duman, RS. Stress, depression, and neuroplasticity: a convergence of mechanisms. Neuropsychopharmacology. 2008; 33(1): 88109.Google Scholar
134.Popoli, M, Gennarelli, M, Racagni, G. Modulation of synaptic plasticity by stress and antidepressants. Bipolar Disord. 2002; 4(3): 166182.Google Scholar
135.Holderbach, R, Clark, K, Moreau, JL, Bischofberger, J, Normann, C. Enhanced long-term synaptic depression in an animal model of depression. Biol Psychiatry. 2007; 62(1): 92100.Google Scholar
136.Matsumoto, M, Tachibana, K, Togashi, H, et al. Chronic treatment with milnacipran reverses the impairment of synaptic plasticity induced by conditioned fear stress. Psychopharmacology (Berl). 2005; 179(3): 606612.Google Scholar
137.Godlewska, BR, Norbury, R, Selvaraj, S, Cowen, PJ, Harmer, CJ. Short-term SSRI treatment normalises amygdala hyperactivity in depressed patients. Psychol Med. 2012; 42(12): 26092617.Google Scholar
138.Herzallah, MM, Moustafa, AA, Natsheh, JY, et al. Depression impairs learning, whereas the selective serotonin reuptake inhibitor, paroxetine, impairs generalization in patients with major depressive disorder. J Affect Disord. 2013; 151(2): 484492.Google Scholar
139.Olivier, B. Serotonin: a never-ending story. Eur J Pharmacol. 2015; 753: 218.Google Scholar
140.Kirchhoff, VD, Nguyen, HT, Soczynska, JK, Woldeyohannes, HO, McIntyre, RS. Discontinued psychiatric drugs in 2008. Expert Opin Investig Drugs. 2009; 18(10): 14311443.Google Scholar
141.Westrich, L, Pehrson, A, Zhong, H, et al. In vitro and in vivo effects of the multimodal antidepressant vortioxetine (Lu AA21004) at human and rat targets. Int J Psych Clin Pract. 2012; 5(Suppl 1): 47.Google Scholar
142.Bang-Andersen, B, Ruhland, T, Jorgensen, M, et al. Discovery of 1-[2-(2,4-dimethylphenylsulfanyl)phenyl]piperazine (Lu AA21004): a novel multimodal compound for the treatment of major depressive disorder. J Med Chem. 2011; 54(9): 32063221.Google Scholar
143.Katona, C, Hansen, T, Olsen, CK. A randomized, double-blind, placebo-controlled, duloxetine-referenced, fixed-dose study comparing the efficacy and safety of Lu AA21004 in elderly patients with major depressive disorder. Int Clin Psychopharmacol. 2012; 27(4): 215223.Google Scholar
144.McIntyre, RS, Lophaven, S, Olsen, CK. A randomized, double-blind, placebo-controlled study of vortioxetine on cognitive function in depressed adults. Int J Neuropsychopharmacol. 2014; 17(10): 15571567.Google Scholar
145.Mahableshwarkar, AR, Zajecka, J, Jacobson, W, Chen, Y, Keefe, RS. A randomized, placebo-controlled, active-reference, double-blind, flexible-dose study of the efficacy of vortioxetine on cognitive function in major depressive disorder. Neuropsychopharmacology. 2015; 40(8): 20252037.Google Scholar
146.Browning, M, Smith, J, Conen, S, et al. Vortioxetine reduces BOLD signal during performance of the N-Back task in subjects remitted from depression and healthy control participants. Neuropsychopharm. 2014; 39(Supplement 1): S480.Google Scholar
147.Sanchez, C, Asin, KE, Artigas, F. Vortioxetine, a novel antidepressant with multimodal activity: Review of preclinical and clinical data. Pharmacol Ther. 2015; 145: 4357.Google Scholar
148.Mork, A, Pehrson, A, Brennum, LT, et al. Pharmacological effects of Lu AA21004: a novel multimodal compound for the treatment of major depressive disorder. J Pharmacol Exp Ther. 2012; 340(3): 666675.Google Scholar
149.Pehrson, AL, Cremers, T, Betry, C, et al. Lu AA21004, a novel multimodal antidepressant, produces regionally selective increases of multiple neurotransmitters—a rat microdialysis and electrophysiology study. Eur Neuropsychopharmacol. 2013; 23(2): 133145.Google Scholar
150.Puig, MV, Santana, N, Celada, P, Mengod, G, Artigas, F. In vivo excitation of GABA interneurons in the medial prefrontal cortex through 5-HT3 receptors. Cereb Cortex. 2004; 14(12): 13651375.CrossRefGoogle Scholar
151.Riga, MS, Celada, P, Sanchez, C, Artigas, F. Cortical and hippocampal microcircuits involved in the mechanism of action of the new antidepressant drug vortioxetine. Neuropsychopharm. 2014; 39(Supplement 1): S632.Google Scholar
152.El Mansari, M, Lecours, M, Blier, P. Effects of acute and sustained administration of vortioxetine on the serotonin system in the hippocampus: electrophysiological studies in the rat brain. Psychopharmacology (Berl). 2015; 232(13): 23432352.Google Scholar
153.O’Mara, SM, Sanchez-Vives, MV, Brotons-Mas, JR, O’Hare, E. Roles for the subiculum in spatial information processing, memory, motivation and the temporal control of behaviour. Prog Neuropsychopharmacol Biol Psychiatry. 2009; 33(5): 782790.Google Scholar
154.Sari, Y. Serotonin1B receptors: from protein to physiological function and behavior. Neurosci Biobehav Rev. 2004; 28(6): 565582.Google Scholar
155.Smagin, G, Song, D, Budac, DP, Pehrson, A, Li, Y, Sanchez, C. Chronic treatment with vortioxetine activates the central histaminergic system: a microdialysis study in rats. Biol Psychiatry. 2014; 75(9), Supplement, 391S.Google Scholar
156.Li, Y, Abdourahman, A, Tamm, JA, et al. Reversal of age-associated cognitive deficits is accompanied by increased plasticity-related gene expression after chronic antidepressant administration in middle-aged mice. Pharmacol Biochem Behav. 2015; 135: 7082.CrossRefGoogle ScholarPubMed
157.Bétry, C, Etiévant, A, Pehrson, A, Sanchez, C, Haddjeri, N. Effect of the multimodal acting antidepressant vortioxetine on rat hippocampal plasticity and recognition memory. Prog Neuropsychopharmacol Biol Psychiatry. 2015; 58: 3846.Google Scholar
158.Guilloux, JP, Mendez-David, I, Pehrson, A, et al. Antidepressant and anxiolytic potential of the multimodal antidepressant vortioxetine (Lu AA21004) assessed by behavioural and neurogenesis outcomes in mice. Neuropharmacology. 2013; 73: 147159.Google Scholar
159.Sanacora, G, Banasr, M. From pathophysiology to novel antidepressant drugs: glial contributions to the pathology and treatment of mood disorders. Biol Psychiatry. 2013; 73(12): 11721179.Google Scholar
160.Sanacora, G, Treccani, G, Popoli, M. Towards a glutamate hypothesis of depression: an emerging frontier of neuropsychopharmacology for mood disorders. Neuropharmacology. 2012; 62(1): 6377.Google Scholar
161.Pehrson, AL, Sanchez, C. Vortioxetine reverses social recognition memory impairments induced by acetylcholine or glutamate dysregulation in rats. Eur Neuropsychopharmacol. 2014; 24(Supplement 2): S369.Google Scholar
162.Bétry, C, Pehrson, AL, Etiévant, A, Ebert, B, Sánchez, C, Haddjeri, N. The rapid recovery of 5-HT cell firing induced by the antidepressant vortioxetine involves 5-HT(3) receptor antagonism. Int J Neuropsychopharmacol. 2013; 16(5): 11151127.Google Scholar
163.Pompeiano, M, Palacios, JM, Mengod, G. Distribution of the serotonin 5-HT2 receptor family mRNAs: comparison between 5-HT2A and 5-HT2C receptors. Brain Res Mol Brain Res. 1994; 23(1–2): 163178.Google Scholar
164.Haider, S, Khaliq, S, Haleem, DJ. Enhanced serotonergic neurotransmission in the hippocampus following tryptophan administration improves learning acquisition and memory consolidation in rats. Pharmacol Rep. 2007; 59(1): 5357.Google Scholar
165.Levkovitz, Y, Richter-Levin, G, Segal, M. Effect of 5-hydroxytryptophane on behavior and hippocampal physiology in young and old rats. Neurobiol Aging. 1994; 15(5): 635641.Google Scholar
166.Egashira, N, Mishima, K, Katsurabayashi, S, et al. Involvement of 5-hydroxytryptamine neuronal system in Delta(9)-tetrahydrocannabinol-induced impairment of spatial memory. Eur J Pharmacol. 2002; 445(3): 221229.Google Scholar
167.Inoue, T, Kitaichi, Y, Koyama, T. SSRIs and conditioned fear. Prog Neuropsychopharmacol Biol Psychiatry. 2011; 35(8): 18101819.Google Scholar
168.Karabeg, MM, Grauthoff, S, Kollert, SY, et al. 5-HTT deficiency affects neuroplasticity and increases stress sensitivity resulting in altered spatial learning performance in the Morris water maze but not in the Barnes maze. PloS one. 2013; 8(10): e78238.Google Scholar
169.Nonkes, LJ, de Pooter, M, Homberg, JR. Behavioural therapy based on distraction alleviates impaired fear extinction in male serotonin transporter knockout rats. J Psychiatry Neurosci. 2012; 37(4): 224230.Google Scholar
170.Luoni, A, Hulsken, S, Cazzaniga, G, Racagni, G, Homberg, JR, Riva, MA. Behavioural and neuroplastic properties of chronic lurasidone treatment in serotonin transporter knockout rats. Int J Neuropsychopharmacol. 2013; 16(6): 13191330.Google Scholar
171.Ampuero, E, Stehberg, J, Gonzalez, D, et al. Repetitive fluoxetine treatment affects long-term memories but not learning. Behav Brain Res. 2013; 247: 92100.Google Scholar
172.Sass, A, Wortwein, G. The effect of subchronic fluoxetine treatment on learning and memory in adolescent rats. Behav Brain Res. 2012; 228(1): 169175.Google Scholar
173.Gumuslu, E, Mutlu, O, Sunnetci, D, et al. The effects of tianeptine, olanzapine and fluoxetine on the cognitive behaviors of unpredictable chronic mild stress-exposed mice. Drug Res. 2013; 63(10): 532539.Google Scholar
174.Valluzzi, JA, Chan, K. Effects of fluoxetine on hippocampal-dependent and hippocampal-independent learning tasks. Behav Pharmacol. 2007; 18(5–6): 507513.Google Scholar
175.Heinen, M, Hettich, MM, Ryan, DP, Schnell, S, Paesler, K, Ehninger, D. Adult-onset fluoxetine treatment does not improve behavioral impairments and may have adverse effects on the Ts65Dn mouse model of Down syndrome. Neural Plasticity. 2012; 2012: Article 467251.Google Scholar
176.Myint, AM, O’Mahony, S, Kubera, M, et al. Role of paroxetine in interferon-alpha-induced immune and behavioural changes in male Wistar rats. J Psychopharmacol. 2007; 21(8): 843850.Google Scholar
177.Wang, Y, Neumann, M, Hansen, K, et al. Fluoxetine increases hippocampal neurogenesis and induces epigenetic factors but does not improve functional recovery after traumatic brain injury. J Neurotrauma. 2011; 28(2): 259268.Google Scholar
178.Naudon, L, Hotte, M, Jay, TM. Effects of acute and chronic antidepressant treatments on memory performance: a comparison between paroxetine and imipramine. Psychopharmacology (Berl). 2007; 191(2): 353364.Google Scholar
179.Egashira, N, Matsumoto, Y, Mishima, K, et al. Low dose citalopram reverses memory impairment and electroconvulsive shock-induced immobilization. Pharmacol Biochem Behav. 2006; 83(1): 161167.Google Scholar
180.Montezinho, LP, Miller, S, Plath, N, et al. The effects of acute treatment with escitalopram on the different stages of contextual fear conditioning are reversed by atomoxetine. Psychopharmacology (Berl). 2010; 212(2): 131143.Google Scholar
181.Bridoux, A, Laloux, C, Derambure, P, Bordet, R, Monaca Charley, C. The acute inhibition of rapid eye movement sleep by citalopram may impair spatial learning and passive avoidance in mice. J Neural Transm. 2013; 120(3): 383389.Google Scholar
182.Jensen, JB, du Jardin, KG, Song, D, et al. Vortioxetine, but not escitalopram or duloxetine, reverses memory impairment induced by central 5-HT depletion in rats: evidence for direct 5-HT receptor modulation. Eur Neuropsychopharmacol. 2014; 24(1): 148159.Google Scholar
183.Mørk, A, Montezinho, LP, Miller, S, et al. Vortioxetine (Lu AA21004), a novel multimodal antidepressant, enhances memory in rats. Pharmacol Biochem Behav. 2013; 105: 4150.Google Scholar
184.du Jardin, KG, Jensen, JB, Sanchez, C, Pehrson, AL. Vortioxetine dose-dependently reverses 5-HT depletion-induced deficits in spatial working and object recognition memory: a potential role for 5-HT1A receptor agonism and 5-HT3 receptor antagonism. Eur Neuropsychopharmacol. 2014; 24(1): 160171.Google Scholar
185.Adams, W, Kusljic, S, van den Buuse, M. Serotonin depletion in the dorsal and ventral hippocampus: effects on locomotor hyperactivity, prepulse inhibition and learning and memory. Neuropharmacology. 2008; 55(6): 10481055.CrossRefGoogle ScholarPubMed
186.Lehmann, O, Bertrand, F, Jeltsch, H, et al. 5,7-DHT-induced hippocampal 5-HT depletion attenuates behavioural deficits produced by 192 IgG-saporin lesions of septal cholinergic neurons in the rat. Eur J Neurosci. 2002; 15(12): 19912006.Google Scholar
187.Lehmann, O, Jeltsch, H, Lazarus, C, Tritschler, L, Bertrand, F, Cassel, JC. Combined 192 IgG-saporin and 5,7-dihydroxytryptamine lesions in the male rat brain: a neurochemical and behavioral study. Pharmacol Biochem Behav. 2002; 72(4): 899912.Google Scholar
188.Lehmann, O, Jeltsch, H, Lehnardt, O, Pain, L, Lazarus, C, Cassel, JC. Combined lesions of cholinergic and serotonergic neurons in the rat brain using 192 IgG-saporin and 5,7-dihydroxytryptamine: neurochemical and behavioural characterization. Eur J Neurosci. 2000; 12(1): 6779.Google Scholar
189.Murtha, SJ, Pappas, BA. Neurochemical, histopathological and mnemonic effects of combined lesions of the medial septal and serotonin afferents to the hippocampus. Brain Res. 1994; 651(1–2): 1626.Google Scholar
190.Volpe, BT, Hendrix, CS, Park, DH, Towle, AC, Davis, HP. Early post-natal administration of 5,7-dihydroxytryptamine destroys 5-HT neurons but does not affect spatial memory. Brain Res. 1992; 589(2): 262267.Google Scholar
191.Gutierrez-Guzman, BE, Hernandez-Perez, JJ, Lopez-Vazquez, MA, Fregozo, CS, Guevara, MA, Olvera-Cortes, ME. Serotonin depletion of supramammillary/posterior hypothalamus nuclei produces place learning deficiencies and alters the concomitant hippocampal theta activity in rats. Eur J Pharmacol. 2012; 682(1–3): 99109.Google Scholar
192.Richter-Levin, G, Greenberger, V, Segal, M. The effects of general and restricted serotonergic lesions on hippocampal electrophysiology and behavior. Brain Res. 1994; 642(1–2): 111116.Google Scholar
193.Kenton, L, Boon, F, Cain, DP. Combined but not individual administration of beta-adrenergic and serotonergic antagonists impairs water maze acquisition in the rat. Neuropsychopharmacology. 2008; 33(6): 12981311.Google Scholar
194.Galani, R, Berthel, MC, Lazarus, C, et al. The behavioral effects of enriched housing are not altered by serotonin depletion but enrichment alters hippocampal neurochemistry. Neurobiol Learn Mem. 2007; 88(1): 110.Google Scholar
195.Lieben, CK, van Oorsouw, K, Deutz, NE, Blokland, A. Acute tryptophan depletion induced by a gelatin-based mixture impairs object memory but not affective behavior and spatial learning in the rat. Behav Brain Res. 2004; 151(1–2): 5364.Google Scholar
196.Uchida, S, Umeeda, H, Kitamoto, A, Masushige, S, Kida, S. Chronic reduction in dietary tryptophan leads to a selective impairment of contextual fear memory in mice. Brain Res. 2007; 1149: 149156.Google Scholar
197.Dai, JX, Han, HL, Tian, M, et al. Enhanced contextual fear memory in central serotonin-deficient mice. Proc Natl Acad Sci U S A. 2008; 105(33): 1198111986.Google Scholar
198.Bert, B, Dere, E, Wilhelmi, N, et al. Transient overexpression of the 5-HT1A receptor impairs water-maze but not hole-board performance. Neurobiol Learn Mem. 2005; 84(1): 5768.Google Scholar
199.Bert, B, Voigt, JP, Kusserow, H, Theuring, F, Rex, A, Fink, H. Increasing the number of 5-HT(1A)-receptors in cortex and hippocampus does not induce mnemonic deficits in mice. Pharmacol Biochem Behav. 2009; 92(1): 7681.Google Scholar
200.Wolff, M, Costet, P, Gross, C, Hen, R, Segu, L, Buhot, MC. Age-dependent effects of serotonin-1A receptor gene deletion in spatial learning abilities in mice. Brain Res Mol Brain Res. 2004; 130(1–2): 3948.Google Scholar
201.Klemenhagen, KC, Gordon, JA, David, DJ, Hen, R, Gross, CT. Increased fear response to contextual cues in mice lacking the 5-HT1A receptor. Neuropsychopharmacology. 2006; 31(1): 101111.Google Scholar
202.Malá, H, Arnberg, K, Chu, D, Nedergaard, SK, Witmer, J, Mogensen, J. Only repeated administration of the serotonergic agonist 8-OH-DPAT improves place learning of rats subjected to fimbria-fornix transection. Pharmacol Biochem Behav. 2013; 109: 5058.Google Scholar
203.Winter, JC, Petti, DT. The effects of 8-hydroxy-2-(di-n-propylamino)tetralin and other serotonergic agonists on performance in a radial maze: a possible role for 5-HT1A receptors in memory. Pharmacol Biochem Behav. 1987; 27(4): 625628.Google Scholar
204.Helsley, S, Siegel, TL, Fiorella, D, Rabin, RA, Winter, JC. WAY-100635 reverses 8-OH-DPAT-induced performance impairment in the radial maze. Prog Neuropsychopharmacol Biol Psychiatry. 1998; 22(7): 11791184.Google Scholar
205.Egashira, N, Yano, A, Ishigami, N, et al. Investigation of mechanisms mediating 8-OH-DPAT-induced impairment of spatial memory: involvement of 5-HT1A receptors in the dorsal hippocampus in rats. Brain Res. 2006; 1069(1): 5462.Google Scholar
206.Inui, K, Egashira, N, Mishima, K, et al. The serotonin1A receptor agonist 8-OHDPAT reverses delta 9-tetrahydrocannabinol-induced impairment of spatial memory and reduction of acetylcholine release in the dorsal hippocampus in rats. Neurotox Res. 2004; 6(2): 153158.Google Scholar
207.Buhot, MC, Patra, SK, Naili, S. Spatial memory deficits following stimulation of hippocampal 5-HT1B receptors in the rat. Eur J Pharmacol. 1995; 285(3): 221228.Google Scholar
208.Bauer, EP. Serotonin in fear conditioning processes. Behav Brain Res. 2015; 277c: 6877.Google Scholar
209.Youn, J, Misane, I, Eriksson, TM, et al. Bidirectional modulation of classical fear conditioning in mice by 5-HT(1A) receptor ligands with contrasting intrinsic activities. Neuropharmacology. 2009; 57(5–6): 567576.Google Scholar
210.Hirst, WD, Andree, TH, Aschmies, S, et al. Correlating efficacy in rodent cognition models with in vivo 5-hydroxytryptamine1a receptor occupancy by a novel antagonist, (R)-N-(2-methyl-(4-indolyl-1-piperazinyl)ethyl)-N-(2-pyridinyl)-cyclohexane carboxamide (WAY-101405). J Pharmacol Exp Ther. 2008; 325(1): 134145.Google Scholar
211.McDevitt, RA, Hiroi, R, Mackenzie, SM, et al. Serotonin 1B autoreceptors originating in the caudal dorsal raphe nucleus reduce expression of fear and depression-like behavior. Biol Psychiatry. 2011; 69(8): 780787.Google Scholar
212.Wolff, M, Savova, M, Malleret, G, Hen, R, Segu, L, Buhot, MC. Serotonin 1B knockout mice exhibit a task-dependent selective learning facilitation. Neurosci Lett. 2003; 338(1): 14.Google Scholar
213.Malleret, G, Hen, R, Guillou, JL, Segu, L, Buhot, MC. 5-HT1B receptor knock-out mice exhibit increased exploratory activity and enhanced spatial memory performance in the Morris water maze. J Neurosci. 1999; 19(14): 61576168.Google Scholar
214.Ahlander-Luttgen, M, Madjid, N, Schott, PA, Sandin, J, Ogren, SO. Analysis of the role of the 5-HT1B receptor in spatial and aversive learning in the rat. Neuropsychopharmacology. 2003; 28(9): 16421655.Google Scholar
215.Fedotova, YO, Ordyan, NE. Blockade of 5-HT2A/2C-type receptors impairs learning in female rats in the course of estrous cycle. Bull Exp Biol Med. 2010; 150(1): 68.Google Scholar
216.Naghdi, N, Harooni, HE. The effect of intrahippocampal injections of ritanserin (5HT2A/2C antagonist) and granisetron (5HT3 antagonist) on learning as assessed in the spatial version of the water maze. Behav Brain Res. 2005; 157(2): 205210.Google Scholar
217.Levin, E, Icenogle, L, Farzad, A. Ketanserin attenuates nicotine-induced working memory improvement in rats. Pharmacol Biochem Behav. 2005; 82(2): 289292.Google Scholar
218.Cohen, H. Anxiolytic effect and memory improvement in rats by antisense oligodeoxynucleotide to 5-hydroxytryptamine-2A precursor protein. Depress Anxiety. 2005; 22(2): 8493.Google Scholar
219.Zhang, G, Ásgeirsdóttir, HN, Cohen, SJ, Munchow, AH, Barrera, MP, Stackman, RW Jr. Stimulation of serotonin 2A receptors facilitates consolidation and extinction of fear memory in C57BL/6J mice. Neuropharmacology. 2013; 64: 403413.Google Scholar
220.Tecott, LH, Logue, SF, Wehner, JM, Kauer, JA. Perturbed dentate gyrus function in serotonin 5-HT2C receptor mutant mice. Proc Natl Acad Sci U S A. 1998; 95(25): 1502615031.Google Scholar
221.Burghardt, NS, Bush, DE, McEwen, BS, LeDoux, JE. Acute selective serotonin reuptake inhibitors increase conditioned fear expression: blockade with a 5-HT(2C) receptor antagonist. Biol Psychiatry. 2007; 62(10): 11111118.Google Scholar
222.Hodges, H, Sowinski, P, Sinden, JD, Netto, CA, Fletcher, A. The selective 5-HT3 receptor antagonist, WAY100289, enhances spatial memory in rats with ibotenate lesions of the forebrain cholinergic projection system. Psychopharmacology (Berl). 1995; 117(3): 318332.Google Scholar
223.Pitsikas, N, Brambilla, A, Borsini, F. Effect of DAU 6215, a novel 5-HT3 receptor antagonist, on scopolamine-induced amnesia in the rat in a spatial learning task. Pharmacol Biochem Behav. 1994; 47(1): 9599.Google Scholar
224.Harrell, AV, Allan, AM. Improvements in hippocampal-dependent learning and decremental attention in 5-HT(3) receptor overexpressing mice. Learn Mem. 2003; 10(5): 410419.Google Scholar
225.Segu, L, Lecomte, MJ, Wolff, M, et al. Hyperfunction of muscarinic receptor maintains long-term memory in 5-HT4 receptor knock-out mice. PloS one. 2010; 5(3): e9529.Google Scholar
226.Cachard-Chastel, M, Devers, S, Sicsic, S, et al. Prucalopride and donepezil act synergistically to reverse scopolamine-induced memory deficit in C57Bl/6j mice. Behav Brain Res. 2008; 187(2): 455461.Google Scholar
227.Orsetti, M, Dellarole, A, Ferri, S, Ghi, P. Acquisition, retention, and recall of memory after injection of RS67333, a 5-HT(4) receptor agonist, into the nucleus basalis magnocellularis of the rat. Learn Mem. 2003; 10(5): 420426.Google Scholar
228.Fontana, DJ, Daniels, SE, Wong, EH, Clark, RD, Eglen, RM. The effects of novel, selective 5-hydroxytryptamine (5-HT)4 receptor ligands in rat spatial navigation. Neuropharmacology. 1997; 36(4–5): 689696.Google Scholar
229.Woolley, ML, Bentley, JC, Sleight, AJ, Marsden, CA, Fone, KC. A role for 5-ht6 receptors in retention of spatial learning in the Morris water maze. Neuropharmacology. 2001; 41(2): 210219.Google Scholar
230.Rogers, DC, Hagan, JJ. 5-HT6 receptor antagonists enhance retention of a water maze task in the rat. Psychopharmacology (Berl). 2001; 158(2): 114119.Google Scholar
231.Stean, TO, Hirst, WD, Thomas, DR, et al. Pharmacological profile of SB-357134: a potent, selective, brain penetrant, and orally active 5-HT(6) receptor antagonist. Pharmacol Biochem Behav. 2002; 71(4): 645654.Google Scholar
232.Lindner, MD, Hodges, DB Jr., Hogan, JB, et al. An assessment of the effects of serotonin 6 (5-HT6) receptor antagonists in rodent models of learning. J Pharmacol Exp Ther. 2003; 307(2): 682691.Google Scholar
233.Roberts, AJ, Hedlund, PB. The 5-HT(7) receptor in learning and memory. Hippocampus. 2012; 22(4): 762771.Google Scholar
234.Gasbarri, A, Cifariello, A, Pompili, A, Meneses, A. Effect of 5-HT(7) antagonist SB-269970 in the modulation of working and reference memory in the rat. Behav Brain Res. 2008; 195(1): 164170.Google Scholar
Figure 0

Figure 1 Schematic illustration of the rat hippocampal circuit with 5-HT receptor localization. The main areas of the hippocampus, including the dentate gyrus (DG), CA3, CA2, CA1, and the subiculum regions, and synaptic connections between them are indicated. Principal (granule and pyramidal) cells are shown in blue, and interneurons are shown in green. Expression of 5-HT receptor subtypes on hippocampal CA1 and CA3 pyramidal cells, granule cells, and interneurons are shown. References for 5-HT receptor localization are listed Table 1. At least 16 subtypes of interneurons have been identified in the hippocampus; one representative interneuron is shown for illustrative purposes. Note that the 5-HT1A heteroreceptor is expressed at high levels throughout the hippocampus. The 5-HT1B receptor is found at highest levels in the subiculum. Based on histology data, the 5-HT3 receptor is only expressed on the interneurons, and the 5-HT4 receptor is only expressed on pyramidal cells. Other 5-HT receptors subtypes are found on both principal cells and interneurons.

Figure 1

Figure 2 Expression of several classes of 5-HT receptors and the 5-HT reuptake transporter (SERT) by ex vivo autoradiography in the rat hippocampus. Autoradiographic images representing total (left panels) and non-specific binding (right panels) for each of 5 separate serotonergic targets in coronal brain sections (20 µm in thickness). 5-HT1A receptors were mapped using 3 nM [3H]8-OH-DPAT (A) alone or (B) in combination with 1 µM of the 5-HT1A receptor selective antagonist WAY100635 to determine the level of nonspecific binding. 5-HT1B/1D receptors were mapped using 1 nM [3H] GR125743 (C) alone or (D) in combination with 1 µM of the 5-HT1B receptor preferring SB216641 to determine the level of nonspecific binding. 5-HT3 receptors were mapped using 3 nM [3H] LY278584 (E) alone or (F) in combination with 1 µM ondansetron to determine the level of nonspecific binding. 5-HT7 receptors were mapped using 4.5 nM [3H]SB269970 (G) alone or (H) in combination with 1 µM of unlabeled SB269970 to determine the level of nonspecific binding. Finally, SERT was mapped using 4.5 nM [3H] escitalopram (I) alone or (J) in combination with 1 µM paroxetine to determine the level of nonspecific binding. Scale bars represent 5 mm.

Figure 2

Table 1 5-HT receptor subtypes in the rodent hippocampus

Figure 3

Table 2 Effects of serotonergic manipulations on hippocampal dependent memory tests in rodents