Skip to main content
Translational Neuroscience logoLink to Translational Neuroscience
. 2016 May 9;7(1):35–49. doi: 10.1515/tnsci-2016-0007

The serotonergic system and cognitive function

Dubravka Švob Štrac 1,, Nela Pivac 1, Dorotea Mück-Šeler 1
PMCID: PMC5017596  PMID: 28123820

Abstract

Symptoms of cognitive dysfunction like memory loss, poor concentration, impaired learning and executive functions are characteristic features of both schizophrenia and Alzheimer’s disease (AD). The neurobiological mechanisms underlying cognition in healthy subjects and neuropsychiatric patients are not completely understood. Studies have focused on serotonin (5-hydroxytryptamine, 5-HT) as one of the possible cognitionrelated biomarkers. The aim of this review is to provide a summary of the current literature on the role of the serotonergic (5-HTergic) system in cognitive function, particularly in AD and schizophrenia.

The role of the 5-HTergic system in cognition is modulated by the activity and function of 5-HT receptors (5-HTR) classified into seven groups, which differ in structure, action, and localization. Many 5-HTR are located in the regions linked to various cognitive processes. Preclinical studies using animal models of learning and memory, as well as clinical in vivo (neuroimaging) and in vitro (post-mortem) studies in humans have shown that alterations in 5-HTR activity influence cognitive performance.

The current evidence implies that reduced 5-HT neurotransmission negatively influences cognitive functions and that normalization of 5-HT activity may have beneficial effects, suggesting that 5-HT and 5-HTR represent important pharmacological targets for cognition enhancement and restoration of impaired cognitive performance in neuropsychiatric disorders.

Keywords: Alzheimer’s disease, Cognitive function, Receptors, Schizophrenia, Serotonin

1. Introduction

Cognitive functions represent a spectrum of mental abilities and complex processes related to attention, memory, judgment and evaluation, problem-solving and decisionmaking, as well as to comprehension and language synthesis. As normal aging is frequently associated with a decline in memory and cognitive abilities, cognitive impairment is one of the major challenges of our rapidly aging society. Moreover, cognitive deficits are prominent features of many psychiatric and neurodegenerative disorders including schizophrenia and Alzheimer’s disease (AD). In spite of extensive research, the neurobiological underpinnings of cognitive flexibility in both healthy subjects and neuropsychiatric patients are still unclear. A great deal of attention has been directed towards the role of serotonin (5-hydroxytryptamine, 5-HT) in various emotional states and mood disorders. More recently, studies have focused on 5-HT as one of the possible cognition-related biomarkers. This mini-review assesses the literature on the involvement of 5-HT receptors (5-HTR) in multiple aspects of cognitive performance and particularly emphasizes 5-HTRs potential for therapeutic intervention of cognitive deficits in AD and schizophrenia.

2. 5-HT and cognition

As a neurotransmitter, 5-HT not only regulates many important physiological processes such as body temperature, sleep, appetite, pain and motor activity [1], but also modulates higher brain functions, including cognition and emotional behaviour [2]. Widespread distribution of serotonergic (5-HTergic) neurons allows modulation of various neuronal networks located in distant brain regions whose coordinated activity is required for most cognitive functions [3]. A high density of 5-HTergic projections in the hippocampus and prefrontal cortex [4-6] underlines the anatomical and neurochemical linkage of the 5-HTergic system with brain areas most commonly associated with learning and memory [7]. While the 5-HTergic system in the hippocampus is involved in different memory processes, spatial navigation, decision making and social relationships [8-10], in the prefrontal cortex 5-HT plays a major role in working memory, attention, decision-making and reversal learning [11,12].

The neuromodulatory action of 5-HT on cognitive functions in both physiological and pathological states largely depends on the action of enzymes, transporters, and specific subtypes of expressed receptors (5-HTR), and their localization, which regulate local 5-HT concentration and neurotransmission [13,14]. In addition, part of 5-HT’s role in the neurobiology of learning and memory might be attributed to complex interactions between the 5-HTergic system and other neurotransmitters such as acetylcholine, dopamine, GABA and glutamate [15]. Preclinical and clinical studies suggest that the activity of the 5-HTergic system is associated with short- and long-term memory and cognitive performance, during aging [16] as well as in many psychiatric (schizophrenia, depression, alcoholism) and neurological (AD, epilepsy) disorders [4,17,18].

Cognitive-attentional dysfunction is one of the key features of schizophrenia and it is related to poor psychosocial outcomes [19]. Major cognitive dimensions affected in schizophrenia include speed of processing, working memory, vigilance, attention, reasoning and problem solving, sound cognition, as well as both visual and verbal learning and memory [20]. In schizophrenia, ascending 5-HTergic pathways from the dorsal raphe nuclei to the substantia nigra and from the rostral raphe nuclei to the neocortex, limbic regions, and basal ganglia are upregulated, leading to dopaminergic hypofunction [21]. It is believed that the symptoms of schizophrenia are at least in part due to this interconnectivity between 5-HTergic and dopaminergic systems [22]. On the other hand, the disruption of serotonergic / cholinergic / GABAergic interactions in the frontal cortex [23], as well as synaptic disorganization in the hippocampus [24], suggests that synchronization of neural activity between the prefrontal cortex and the hippocampus might be crucial for the complex cognitive behaviour in schizophrenia [3].

The most pronounced symptom of AD is the progressive decline of various cognitive domains, primarily affecting episodic, semantic and working memory, but also executive functions such as attention, linguistic and visiospatial skills [25]. The hippocampus, the most affected brain area in AD, is involved in cognitive changes and impaired memory observed in the disease [26]. A reduced numbers and activity of 5-HTergic neurons [27,28], associated with a decrease in the concentration of 5-HT and its main metabolite 5-hydroxyindoleacetic acid (5-HIAA) in the post-mortem brain [29], cerebrospinal fluid [30], and blood platelets [31], suggest extensive serotonergic denervation in AD [32]. The association of 5-HT signalling with accumulation of amyloid-β (Aβ) plaques [27,33], and expression or processing of amyloid precursor protein (APP), as well as the improvement of cognitive functions following treatment with 5-HT modulators [32,34,35], indicate that the 5-HTergic system is a potential target for AD therapy. The alterations in the 5-HTergic system could be also associated with the development of non-cognitive symptoms in AD, called behavioural and psychological symptoms of dementia (BPSD), which affect up to 90% of dementia patients [36], and include aggressive behaviour, depression, psychotic symptoms (hallucinations, delusions), apathy, as well as changes in sleep and appetite [37].

3. Serotonin receptors (5-HTR)

A variety of 5-HTergic functions is accomplished by the release of 5-HT in targeted areas and its action via at least 14 different pre- and postsynaptic 5-HTR [38]. As shown in Tables 1 and 2, 5-HTR are subdivided according to their distribution, molecular structure, cell response and function into seven groups from 5-HTR1 to 5-HTR7 [39, 40]. Except 5-HTR3 which are ligand-gated ion channels, all other 5-HTR are G-protein-coupled receptors influencing different transduction pathways [39].

Table 1.

Classification, distribution and function of 5-HTR.

Receptor family Subtype Distribution Mechanism Cellular response
5-HT1 1A, 1B, 1D, 1E, 1F CNS, blood vessels Adenylate cyclase Inhibitory
5-HT2 2A, 2B, 2C CNS, PNS, platelets, blood vessels, smooth muscle Phospholipase C Excitatory
5-HT3 3A, 3B CNS, PNS; GI tract Ligand-gated ion channel Excitatory
5-HT4 CNS, PNS Adenylate cyclase Excitatory
5-HT5 CNS Adenylate cyclase Inhibitory
5-HT6 CNS Adenylate cyclase Excitatory
5-HT7 CNS, GI tract, blood vessels Adenylate cyclase Excitatory

Abbreviations: CNS = central nervous system; PNS = peripheral nervous system, GI tract = gastrointestinal tract.

Table 2.

5-HTR in the brain.

Receptor family Distribution in the brain
5-HT1 Pituitary gland, rostral raphe nuclei, hippocampus, prefrontal cortex cerebellum, basal ganglia, amygdala, globus pallidus, putamen, caudate nucleus
5-HT2 Cerebral cortex, basal ganglia, amygdala, choroid plexus, hypothalamus, hippocampus, caudate nucleus, putamen, globus pallidus, substantia nigra
5-HT3 Area postrema, tractus solitarius, limbic system, hippocampus, cerebral cortex
5-HT4 Prefrontal cortex, caudate nucleus, putamen, globus pallidus, hippocampus, substantia nigra
5-HT5 Cerebral cortex, amygdala, cerebellum, hypothalamus, hippocampus
5-HT6 Dentate gyrus, hippocampus, olfactory tubercle, nucleus accumbens, amygdala, cerebellum
5-HT7 Thalamus

Although 5-HTR are widespread in the central nervous system (CNS) and to a lesser extent in some peripheral organs, the prefrontal cortex and hippocampus are the two main targets of 5-HTergic neurons and express almost all 5-HTR [41, 42]. 5-HTR subtypes found in these brain regions include 5-HT1′, 5-HT2′, 5-HT3′, 5-HT4′, 5-HT5′, 5-HT6′, and 5-HT7′ class of receptors [43, 44]. The activation of different 5-HTR subtypes through the action of distinct neuronal networks, within the same brain region, or even within the same local synapse can have opposite outcomes [13, 45]. The changes in the 5-HTR related to cognition are given in Table 3. Since both agonists and antagonists of specific 5-HTR affect cognitive processes, 5-HTR emerged as attractive potential drug targets for the treatment of cognitive deficits.

Table 3.

Changes in the 5-HTR related to cognition.

Receptor Findings reported Reference
5-HT1A Decrease in receptor activity and density with aging in healthy subjects 54
Stimulation of receptors by antipsychotics increase prefrontal cortical dopamine release 61
Negative correlation between receptor binding potential and cognitive function in healthy persons 55
Decreased receptor number or density in elderly with MCI and AD 57
Relationship between receptor number and BPSD 58
No correlation between receptor binding potential and cognitive function in healthy persons 56
Receptors affect declarative and non-declarative memory functions via glutamatergic, cholinergic and GABAergic neurons 49
Receptors regulate different kinases and immediate early genes implicated in memory formation 49
Decreased receptor binding in amygdala of patients with schizophrenia 60
Increased receptor binding in prefrontal cortex of patients with schizophrenia 60
No change in brain receptor binding in patients with schizophrenia 60
Activation of postsynaptic receptors in rodents impairs emotional memory through attenuation of neuronal activity 53
Activation of presynaptic receptors reduces 5-HT release and exerts pro-cognitive effects on passive avoidance retention 53
Potential of 5-HTR1A and DR2 heterodimers in the frontal cortex for cognitive enhancement 62
5-HT1B Receptor agonist reduces cognitive function in experimental animals 66
Decrease in receptor binding potential with aging in healthy subjects 65
Role of receptor inverse agonists and antagonists in treatment of damaged memory and cognitive processes 66
Decrease in receptor density in frontal and temporal cortex associated with cognitive dysfunction in patients with AD 67
Decrease in receptor binding in brain stem associated with good response to psychotherapy in depressive patients 69
Positive correlations between creative ability, a measure of divergent thinking, and average receptor availability in grey matter 64
Increased receptor mRNA levels in the hippocampal formation in patients with schizophrenia 70
Receptor agonists induce potentiation of latent inhibition, a characteristic of antipsychotics 71
5-HT2A Decreased receptor number in hippocampus and frontal cortex with aging correlates with cognitive decline 27
Severity of cognitive impairment in AD patients correlates with the decrease in receptor binding 77,78
Receptor antagonism improves cognition in schizophrenia 85
Receptor antagonist improves working memory function in young and aged monkeys 82
Receptor low affinity of the atypical antipsychotics is more beneficial for cognition and social function than high affinity 87
Decrease in receptor density with aging correlates with cognitive decline 73
Conflicting results in vivo studies of receptor binding on schizophrenia patients 83, 84
Changes in receptor expression associated with pathological and progressive accumulation of Aâ in animal model of AD 79
Decrease in brain receptor density in patients with AD 74-76
Decrease in number of prefrontal receptors in brain of schizophrenia patients 60
Receptor activation with high affinity agonist enhanced working memory in rats 80
Intrahippocampal injections of receptor antagonist increase rat spatial learning and memory 81
5-HT2C Receptor agonism, rather than antagonism has beneficial effects on cognitive functions in schizophrenia 99
Both receptor agonists and antagonists may have positive effects on cognitive functions in schizophrenia 90
Reducing receptor activity facilitates reversal learning in mouse by reducing influence of previously non-rewarded associations 92
Receptor agonist treatment ameliorated impairments in cognitive flexibility and reversal learning in the mutant mice 94
Administration of selective receptor antagonist, prior to environmental stress, prevented tau hyperphosphorylation and repaired defects in hippocampal LTP and spatial memory 93
Stimulation with receptor agonist in vitro and in vivo reduces Aâ production 95, 96
Increase of receptors in NK-cells linked with cognitive deficits in AD
5-HT3 Ondansetron blocked scopolamine-induced learning deficits in learning 104
Ondansetron improved radial arm maze performance in MK801-impaired rats 103
Antagonist itasetron showed memory-enhancing effects 105
Receptor antagonists improve memory 102
Receptor antagonists provide improvement in cognitive symptoms of schizophrenia 115
Ondansetron as potential adjunctive treatment for schizophrenia particularly for negative symptoms and cognitive impairments 116-118
Improvements in verbal memory after tropisetron therapy 119
Link between gene variant of the 5-HTR3E subunit and sustained attention in schizophrenic patients 114
Blockade of receptors protects neurons against Ab-induced neurotoxicity by inhibition and stimulation of glutamate and acetylcholine release, respectively 106, 107, 108
Neuroprotective effects of synthetic compounds targeting 5-HTR3 with acetylcholinesterase or with alpha-7 nicotinic receptor activity 110-112
5-HT4 Decreased receptor number in hippocampus and cortex from AD patients 58, 140
Receptor agonists and antagonists modulate short-term and long-term memory in rats 128
Beneficial effects of receptor activation on cognition in rodents and primates 122, 129, 130
Receptor agonists acutely improved performance on learning and memory tests 132-134
Receptor agonists reversed age-related or pharmacologically-induced cognitive deficits 86, 135-137
Chronic partial agonist improved memory performance in mice 138
Receptor agonists stimulate acetylcholine release, regulate memory performance, have neuroprotective and neurotrophic effects 141-143
No change in the receptor number during aging in humans 139
Receptor role in cognitive processes and expression of genes that regulate synaptic plasticity 131
Receptor agonists decrease production of neurotoxic Aâ 142-144
Receptor expression not changed in schizophrenic patients 146
Receptor gene haplotype associated with schizophrenia 147
5-HT5 Receptor blockade impairs short- and long-term memory, while its stimulation facilitate it 150
Receptor antagonist improves positive symptoms and cognitive impairment in animal models of schizophrenia, aged rats and mice with memory deficit 151, 152
5-HT6 Decreased receptor density in temporal and frontal cortex in AD patients 67
Decrease in the number of neurons expressing receptors in AD patients 74
Receptor antagonists enhance cognitive performance 158, 159
Receptor agonists and antagonists regulate learning and memory 153-155
High affinity receptor compounds are investigated in vitro or in pre-clinical and clinical trials for the improvement of cognitive functions in AD 155-157
Recruitment of mammalian target of rapamycin (mTOR) by receptors in the prefrontal cortex (PFC) contributes to perturbed cognition in schizophrenia 162
Compounds with high receptor affinity are enrolled in preclinical and clinical investigations 155-157
Improvements following administration of receptor antagonists in preclinical tests for episodic memory, social cognition, executive function, working memory 163
Receptor antagonist in the AD mouse model counteracts memory impairment by attenuating the generation of Aâ 160
Receptor agonism facilitate the emotional learning by promoting the neuronal plasticity in caudate putamen, hippocampus, and PFC 161
Combination of receptor antagonist with low doses of prazosin enhances memory and demonstrates potential in treatment of schizophrenia 164
5-HT7 Receptors associated with hippocampus-dependent cognitive processes 166
Increase in recognition memory and antipsychotic efficacy of receptor antagonist 182
Receptor agonists could be useful in the treatment of memory decline in AD 175, 176
Decrease in the receptor number in hippocampus and prefrontal cortex of schizophrenia patients 178, 179
Association between receptor gene haplotype and schizophrenia 180
Increase in the receptor number in rats treated with typical antipsychotic haloperidol 178
High affinity of several atypical antipsychotics for receptors 178, 181
Receptor antagonism have procognitive effects 167
Receptor antagonist attenuated phencyclidine (PCP) and scopolamine-induced learning deficits and improved reference memory 168-171
Receptor antagonist attenuated MK-801, scopolamine and PCP-induced impairments in learning and memory 172, 173
Receptor antagonism facilitates memory retention 53
Selective receptor agonist rescues alterations in motor coordination, spatial reference memory and synaptic plasticity in mouse model of Rett syndrome 174

3.1. 5-HT1A receptors

The 5-HTR type 1A (5-HTR1A), probably the most investigated 5-HT receptor class, are highly abundant in cortical and limbic brain regions, associated with cognitive functions [46]. The involvement of 5-HTR1A autoreceptors in cognitive performance has been underlined by their important role in the regulation of the activity of the entire 5-HTergic system. 5-HTR1A autoreceptors, located on the soma of 5-HTergic neurons, are key components of the negative feedback loop that inhibits neuronal signalling and 5-HT release [47]. 5-HTR1A heteroreceptors located on postsynaptic 5-HTergic and non-5-HTergic neurons [39], particularly those in the limbic system, are involved in the control of cognitive functions, mood and emotional states [48]. 5-HTR1A can affect declarative and non-declarative memory functions by exerting their influence on the activity of glutamatergic, cholinergic and GABAergic neurons in the cerebral cortex, hippocampus and the septohippocampal projection [49]. Moreover, 5-HTR1A activation increases dopamine release in the medial prefrontal cortex, striatum, and hippocampus [50, 51]. In addition to cooperation with other neurotransmitter systems [52], 5-HTR1A regulate the G-protein dependent and independent signalling pathways that target immediate early genes implicated in memory formation [49].

In rodents, activation of postsynaptic 5-HTR1A impairs emotional memory through attenuation of neuronal activity, whereas presynaptic 5-HTR1A activation reduces 5-HT release and exerts procognitive effects on passive avoidance retention [53]. Human studies as well as experimentation in animals both suggest an association of 5-HTR1A with cognition. In healthy subjects during aging the activity and density of 5-HTR1A declines 10% every ten years [54]. Positron emission tomography (PET) studies have found a negative [55] or a lack of [56] correlation between 5-HTR1A binding potential and cognitive function in healthy individuals. The discrepancies are probably due to the differences in specificity and sensitivity of cognitive tests and in methodologies that were used, as well as ethnic diversity of the subjects [56].

A progressive decline in the density of 5-HTR1A in the hippocampus and dorsal raphe has been found in elderly subjects with mild cognitive impairment (MCI) and AD [57]. A relationship between the number of 5-HTR1A in the temporal cortex and aggressive behaviour [58] suggests their role in development of BPSD. In addition, results from behavioural and pharmacological studies with antagonists or inverse agonists imply that 5-HTR1A could be an important target for the new compounds used in AD treatment [59].

In addition to the association of the 5-HTR1A and cognitive processes, reported in animal models, healthy subjects, and patients with MCI and AD, there are also data regarding their role in schizophrenia. Although in vivo imaging studies of 5-HTR1A distribution in schizophrenia patients showed inconsistent results, it appears that ligand binding to 5-HTR1A is enhanced in the cortical areas, while it is diminished or unchanged in the amygdala [60]. In addition, a recent meta-analysis study has reported a significant increase in post-mortem 5-HTR1A binding in the prefrontal cortex of patients with schizophrenia [60]. Direct stimulation of 5-HTR1A increases prefrontal cortical dopamine release. It is therefore possible that the antipsychotic effects of clozapine, ziprasidone and aripiprazole are in part due to their agonist effects on 5-HTR1A [61]. The recent detection of 5-HTR1A and dopamine receptors type 2 (DR2) heterodimers in the frontal cortex and their potential for cognitive enhancement, has implications for the development of improved pharmacotherapy for schizophrenia or other disorders [62].

3.2. 5-HT1B receptors

Activated presynaptic 5-HTR type 1B (5-HTR1B) inhibit the release of 5-HT and other neurotransmitters (GABA, glutamate, noradrenaline), while modulating the release of acetylcholine [63]. Postsynaptic 5-HTR1B are found on non-5-HTergic neurons within the basal ganglia, striatum, hippocampus and cortex [39]. Positive correlations between creative ability, a measure of divergent thinking, and average 5-HTR1B levels in the grey matter have been reported in the study of Varrone et al. [64]. A moderate age-related decline in the brain’s 5-HTR1B binding potential of 8% every 10 years has also been observed in healthy subjects [65]. The suggested role of 5-HTR1B in memory and learning is consistent with the findings that 5-HTR1B agonists reduce cognitive function in experimental animals [66].

Post mortem analysis of brains from AD patients showed a reduced density of 5-HTR1B in the frontal and temporal cortex, associated with cognitive dysfunction [67]. Since these receptors inhibit acetylcholine release [68], when they are in a heteroreceptor context, lower expression of 5-HTR1B observed in AD [67] might represent a compensatory mechanism aimed to repair downregulated acetylcholine levels. These results suggest that disrupted cognition in AD might be improved by the administration of inverse agonists and antagonists of 5-HTR1B [66]. The suggested importance of 5-HTR1B in cognition has been confirmed in a recent clinical PET study in patients with major depressive disorders before and after psychotherapy [69]. Increased 5-HTR1B mRNA levels in the hippocampal formation were also observed in patients with schizophrenia [70]. The up-regulation of 5-HTR1B in concert with the downregulation of 5-HTR2A, could lead to a reduction in GABAergic activity and consequently enhanced hippocampal glutamatergic output in schizophrenia [70]. On the other hand, agonists of 5-HTR1B might provide an alternative therapeutic approach for schizophrenia, as they induce potentiation of latent inhibition, which is a characteristic of many effective antipsychotics [71].

3.3. 5-HT2A receptors

5-HTR type 2A (5-HTR2A) are located mostly in different parts of the cortex, basal ganglia and slightly less in the hippocampus [39], where they enhance the release of dopamine, glutamate and GABA, and inhibit the release of noradrenaline [72]. An age-dependent decrease in the number of 5-HTR2A was found in the hippocampus and frontal cortex of healthy individuals [27]. The observed 12% reduction in the density of 5-HTR2A every decade, also correlates with cognitive decline [73].

Imunohistochemical [74], post-mortem [75] and imaging [76] studies revealed reduced brain 5-HTR2A density in patients with AD. The severity of cognitive impairment in AD patients seems to also correlate with the decrease in neocortical temporal 5-HTR2A [77, 78]. Studies on the AD mouse model that overexpresses Aβ and has high age-dependent levels of amyloid plaques [79], suggested that changes in 5-HTR2A expression are associated with the pathological and progressive accumulation of Aβ.

Various animal studies investigated the effects 5-HTR2A agonists or antagonists on cognitive performance. The activation of 5-HTR2A with the high affinity agonist TCB-2 enhanced working memory in rats [80], while intrahippocampal injections of ritanserin (5-HTR2A/2C antagonist) increased spatial learning and memory, also in rats [81]. Improved working memory was also observed in younger and older monkeys following treatment with 5-HTR2A antagonist EMD 281014 [82].

The role of 5-HTR2A in the most common cognitive deficits in schizophrenia, such as attention, executive functions, and spatial working memory is not clear [83]. A recent meta-analysis [60] found a reduced number of prefrontal 5-HTR2A in the post-mortem brain of schizophrenia patients. On the other hand, in vivo studies of 5-HTR2A binding reported conflicting results [83, 84]. The observed discrepancies might be due to the fact that post-mortem studies included patients treated with antipsychotics - 5-HTR2 antagonists, which may decrease the density of 5-HTR2A. Although 5-HTR2A antagonism was reported to improve cognition in schizophrenia [85], the adverse side effects of 5-HTR2A antagonists, such as atypical antipsychotics olanzapine and clozapine, limit their clinical use to shortterm treatment of BPSD in patients with most severe symptoms [86]. However, some studies suggest that 5-HTR2A affinity plays an important role in the modulation of the cognitive effects of atypical antipsychotics, indicating that low affinity to 5-HTR2A is more beneficial for cognitive and social performance than high affinity [87]. Further development of highly selective 5-HTR2A ligands is essential for elucidating the critical involvement of these receptors in different cognitive functions [88].

3.4. 5-HT2C receptors

5-HT activity in different brain regions is modulated by 5-HTR type 2C (5-HTR2C), which are found throughout the CNS [89]. RNA editing generates at least 14 functionally distinct 5-HTR2C isoforms, any of which could be a potential target for improved therapeutic and side effect profiles [90]. Moreover, the negligible presence of 5-HTR2C in cardiac and vascular tissues makes these receptors ideal targets for treatment of brain disorders, due to their limited peripheral side effects [91].

In various animal models, 5-HTR2C antagonism seems to improve cognitive flexibility. 5-HTR2C blocking with the antagonist SB242084 promoted reversal learning in mice [92], whereas administration of the selective 5-HTR2C antagonist RS-102,221 prevented tau hyperphosphorylation and repaired the defects in hippocampal long-term potentiation and spatial memory [93], suggesting a beneficial effect on disrupted hippocampal synaptic plasticity. Treatment with the 5-HTR2C agonist CP809,101 improved impairments in cognitive flexibility and reversal learning in mutant mice with a genetically engineered 5-HT-synthesizing enzyme (tryptophanhydroxylase-2) [94]. Pharmacological stimulation of 5-HTR2C with another agonist dexnorfenfluramine in vitro [95] and in vivo [96] enhanced secretion of the APP metabolite and reduced Aβ production, suggesting that one of the strategies for AD therapy development could be to target 5-HTR2C. The significant increase of 5-HTR2C in NK-cells has been linked with cognitive deficits in AD [97], suggesting that it may serve as a biomarker for diagnosing dementia.

The presence of 5-HTR2C in the limbic system, frontal cortex, and hippocampus suggests their possible involvement in schizophrenia [98]. Preclinical data revealed that 5-HTR2C modulation of cognitive symptoms might have beneficial effects in schizophrenia, as well [99]. Several studies indicated that both 5-HTR2C agonists and antagonists may have positive effects on cognitive functions in schizophrenia [for review see 90]. Further research focused on the 5-HTR2C modulation is necessary to assess whether 5-HTR2C agonism or antagonism improve cognitive defects in schizophrenia [90].

3.5. 5-HT3 receptors

In contrast to all other 5-HTR, 5-HTR type 3 (5-HTR3) are ligand-gated channels that regulate permeability to sodium, potassium and calcium ions in the CNS and peripheral nervous system. These receptors induce rapid membrane depolarization and consequently the release of 5-HT, acetylcholine, dopamine, GABA and peptides [100]. While the location of 5-HTR3 on presynaptic neurons in cortical regions, amygdala and striatum, and on postsynaptic neurons in the hippocampus [39], suggest a potential role in cognition, studies investigating the involvement of 5-HT3 receptors in cognitive functions are few.

The 5-HTR3 antagonists, such as ondansetron, are mainly used for the treatment of chemotherapy-induced emesis [101]. However, they have been also shown to improve cognition in different models of memory impairment [102]. Boast et al. [103] reported that ondansetron improves radial arm maze performance in MK801-impaired rats, suggesting its cognition enhancing properties. Ondansetron also blocked scopolamineinduced learning deficits [104], whereas another 5-HTR3 antagonist itasetron showed memory-enhancing effects [105].

Several pharmacological studies suggested the role of 5-HTR3 in AD. 5-HTR3 antagonists MDL72222 and Y25130 reduced Aβ proteininduced neurotoxicity in cultured rat cortical neurons [106]. Moreover, the inhibition of 5-HTR3 with tropisetron alleviated the spatial memory deficit in the rat model of AD. It also protected neurons from Aβ- induced inflammation and neurotoxicity by inhibiting and stimulating glutamate and acetylcholine release, respectively [107,108], or by inhibiting calcineurin activity in the hippocampus (107). Other 5-HTR3 antagonists which interact with multiple targets have also been investigated [109]. Synthetic dual action compounds targeting 5-HTR3 (antagonist) and acetylcholinesterase activity (inhibitor) [110, 111], or 5-HTR3 (inhibitor) and alpha-7 nicotinic receptor (activator) [112] had neuroprotective effects. 5-HTR3 antagonists could therefore be used as building blocks in the development of new neuroprotective drugs.

While no significant changes in the number and affinity of 5-HTR3 in amygdala of schizophrenic patients have been observed in one post-mortem study [113], another report noted a link between a coding variant of the Translational Neuroscience 41 5-HTR3E subunit and sustained attention in schizophrenic patients [114], suggesting the involvement of 5-HTR3 in cognitive deficits in schizophrenia. Several recent studies also found that 5-HTR3 antagonists can provide significant amelioration of cognitive symptoms of schizophrenia [115]. Ondansetron has shown some promise in treatments for schizophrenia, particularly for negative symptoms and cognitive impairments [116, 117]. Akhondzadeh et al. [116] and Levkovitz et al. [118] reported improved visio-spatial learning and memory following treatment with ondansetron, whereas Zhang et al. [119] observed improvements in verbal memory after tropisetron therapy. These findings suggest that 5-HTR3 antagonists possibly have therapeutic potential for the management of cognitive deficits in both AD and schizophrenia.

3.6. 5-HT4 receptors

5-HTR type 4 (5-HTR4) have been found in different brain regions such as the hypothalamus, hippocampus, nucleus accumbens, ventral pallidum, amygdala, basal ganglia, olfactory bulbs, frontal cortex and substantia nigra [120, 121]. These receptors are cleary highly expressed in brain structures involved in memory processes, including cell bodies and nerve endings of GABA neurons in the limbic system, as well as cholinergic neurons in the cortex where they modulate acetylcholine release [122]. In addition to acetylcholine [123, 124], activation of 5-HTR4 increases the release of dopamine [125, 126] and 5-HT [127].

Both 5-HTR4 agonists and antagonists modulate short-term and long-term memory in rats [128]. Various studies reported beneficial effects of 5-HTR4 activation on cognition in rodents and primates [122, 129, 130]. 5-HTR4 have also been implicated in the expression of genes that regulate synaptic plasticity [131]. 5-HTR4 agonists, administered acutely, improved performance on learning and memory tests [132-134], and reversed agerelated or pharmacologically-induced cognitive deficits [86, 135-137]. Chronic activation of 5-HTR4 also seems promising strategy for the treatment of memory deficits, as long-term administration of the partial agonist RS-67333 improved recognition memory in mice (138).

Although some studies reported that aging does not change the number of 5-HTR4 in the human brain [139], reduced levels of 5-HTR4 have been measured post-mortem in the hippocampus and cortex of AD patients [58,140]. Both, in vivo and in vitro studies imply that activation of 5-HTR4 by agonists like 5-HT itself has a beneficial effect in AD. 5-HTR4 agonists have been shown to stimulate acetylcholine release [141], regulate memory performance and have neuroprotective and neurotrophic effects [142, 143]. Additionally, they stimulate the non-amyloid-forming metabolism of APP and thus decrease the production of neurotoxic Aβ, involved in the AD etiology [142-144].

Since they modulate cognitive functions, 5-HTR4 receptors are attractive target candidates for therapeutic strategies aimed at curbing cognitive symptoms of schizophrenia [145]. 5-HTR4 expression does not appear to change in schizophrenic patients [146], but 5-HTR4 gene variants have been associated with schizophrenia [147]. Overall, selective 5-HTR4 ligands may provide novel approaches for the development of new cognitive enhancers, which would be useful for treatments of both AD and schizophrenia [138].

3.7. 5-HT5 receptors

5-HTR type 5 (5-HTR5) are expressed in different brain regions like the cerebral cortex, hippocampus, nucleus accumbens, amygdala, and hypothalamus [148, 149]. A preclinical study demonstrated that blocking and stimulation of 5-HTR5A might impair and facilitate short- and long-term memory, respectively [150]. On the other hand, the 5-HTR5A antagonist ASP5736 improved positive and cognitive symptoms in a schizophrenia mouse model as well as in aged rats and mice with scopolamine-induced working memory deficit [151, 152]. These data suggest that ASP5736 may be used in the treatment of cognitive defects in schizophrenic patients. As far as we know, no data are yet available on 5-HTR5 and AD.

3.8. 5-HT6 receptors

5-HTR type 6 (5-HTR6) are located on postsynaptic 5-HT neurons in the basal ganglia, cortex and limbic system and on cholinergic and GABAergic neurons in the striatum [39]. Preclinical studies on 5-HTR6 suggest their role in regulation of learning and memory, mediated probably by stimulation of glutamatergic and cholinergic transmission [153-155].

The density of 5-HTR6 in the temporal and frontal cortex [67], as well as the number of neurons expressing 5-HTR6 [74], are reduced in AD patients, suggesting that these receptors might also be a good target for antidementia medication. Several compounds with high affinity to 5-HTR6, especially 5-HTR6 antagonists, have been synthesized and are currently being investigated in vitro or are in different phases of pre-clinical and clinical trials for the improvement of cognitive functions in AD [155-157]. 5-HTR6 antagonists were shown to enhance cognitive performance in different learning and memory tests performed on rodents and primates [158, 159]. For instance, in the AD mouse model, the 5-HTR6 antagonist, SB271036, counteracts memory deficiencies probably by reducing Aβ formation via the inhibition of γ-secretase activity and the inactivation of astrocytes and microglia [160]. 5-HTR6 agonism has also been reported to facilitate emotional learning by promoting the neuronal plasticity in the caudate putamen, hippocampus, and prefrontal cortex [161].

In animal models of schizophrenia, the activation of 5-HTR6 was associated with the stimulation of the mTOR (mammalian Target of Rapamycin) signalling pathway in prefrontal cortex. Rapamycin, the mTOR inhibitor, prevented cognitive deficits induced by 5-HTR6 agonists [162]. Improvements in episodic memory, social cognition, executive function, and working memory were also observed following administration of 5-HTR6 antagonists in preclinical trials [163]. There are also reports showing that the combination of 5-HTR6 antagonist PRX-07034 with low doses of prazosin enhances memory which could be used for the treatment of schizophrenia [164].

3.9. 5-HT7 receptors

5-HTR type 7 (5-HTR7) are the most recently discovered 5-HTR. They are located mostly in the hippocampus, hypothalamus and thalamus, and somewhat less in the cortex, as well as in the amygdala and the dorsal raphe nucleus [39, 165]. Although 5-HTR7 are associated with hippocampus-dependent cognitive processes [166], their role in memory and cognition is still unclear, mainly due to the lack of selective agonists and antagonists. Most studies are therefore done with partially specific drugs that also target other receptors.

Preclinical data show pro-cognitive effects of a 5-HTR7 antagonist, alone or in combination with antidepressants, indicating that 5-HTR7 antagonist represent new targets for the treatment of cognitive deficits in stressrelated neuropsychiatric disorders [167]. The 5-HTR7 antagonist SB-269970 attenuated PCP and scopolamine-induced learning deficits [168-170], and improved reference memory [171]. Another antagonist, lurasidone, attenuated MK-801, scopolamine and PCPinduced impairments in learning and memory [172,173].

In the mice model of Rett syndrome, treatment with LP-211, a selective 5-HTR7 agonist, improved motor coordination, spatial reference memory, and synaptic plasticity, suggesting a potential therapeutic potential of 5-HTR7 agonism in therapies for this neurological disorder [174]. Recent findings in rodents suggested that antagonism of the 5-HTR1A assists memory retention, likely through activation of 5-HTR7, which facilitate emotional memory [53].

In addition, preclinical study proposed that 5-HTR7 agonists could be useful in the treatment of impaired memory associated with aging or AD [175]. Therapeutic effects of 5-HTR7 agonists on cognitive symptoms in AD have also been examined in clinical trials [176]. It therefore seems that promnesic or anti-amnesic effects of both 5-HTR7 agonists and antagonists depend on whether the basal performance is normal or impaired [177].

The lower expression of 5-HTR7 in the hippocampus and prefrontal cortex of schizophrenia patients [178, 179], a positive association between a 5-HTR7 gene haplotype and the incidence of schizophrenia [180], the affinity of several atypical antipsychotics for 5-HTR7 [178, 181], and the increase of the 5-HTR7 number in rats treated with typical antipsychotic haloperidol [178], all suggest the involvement of these 5-HTR in the neurobiological alterations in schizophrenia. In line with the potential contribution of 5-HTR7 to cognitive dysfunction in schizophrenic patients, a preclinical study suggested an improvement in recognition memory and antipsychotic efficacy after treatment with a 5-HTR7 antagonist in animal model of psychosis and cognition [182].

4. Conclusion

Evidence from the literature suggests that the 5-HTergic system plays a significant role in cognitive performance. Pro-cognitive and neuroprotective effects of 5-HT have been observed in both humans and animals [183-186]. The involvement of 5-HTR in learning and memory is consistent with high expression of those receptors in limbic areas, the prefrontal cortex and basal ganglia, which are all brain regions involved in the regulation of various cognitive processes [187] and are innervated by 5-HTergic projections from the raphe nuclei [5]. Modulatory effects of different 5-HTR on cognition are additionally mediated by interactions with other neurotransmitter systems such as cholinergic, dopaminergic, GABAergic and glutamatergic.

Although agonists and/or antagonists of 5-HTR1A/1B, 5-HTR2A/2C, 5-HTR3, 5-HTR4, 5-HTR6 and 5-HTR7 have a potential for the treatment of cognitive deficits in healthy elderly people and patients with AD and schizophrenia [187], results of studies attempting to identify individual 5-HTergic targets for cognitive enhancement have been somewhat disappointing.

Contradictory findings might be due to variability in the choice of protocols for training/testing, behavioral tasks and animal models, as well as drugs that were used. Moreover, dysfunction in distinct cognitive areas, observed during aging or in diseases such as schizophrenia and AD, might have different underlying mechanisms. This might suggest that subjects with different cognitive impairments could benefit from customized therapeutic strategies. As the effects of a drug on a single 5-HTR could be counterbalanced by changes in the other receptors in order to maintain homeostasis, the development of medications which act on multiple 5-HTergic targets may be more promising for treatments of cognitive impairments caused by 5-HTergic dysfunction.

An example of such a multitarget drug is the antidepressant vortioxetine [188, 189], which modulates various 5-HTergic components, acting as a 5-HTR3, 5-HTR7, and 5-HTR1D antagonist, and a 5-HTR1B partial agonist, a 5-HTR1A agonist and also a 5-HT transporter inhibitor. It produces more robust effects on cognitive function by activation of multiple neurotransmitter systems (noradrenergic, dopaminergic, cholinergic, histaminergic, etc.), or possibly some other factors such as brain-derived neurotrophic factor (BDNF), which are critical for neural plasticity and cognitive processing [13].

Overcoming the big challenges in the development of these drugs, is an extremely worthy endeavor as it would result in the substantial improvement of the quality of life of patients with age-related cognitive impairments and other diseases with a significant component of memory dysfunction, such as AD and schizophrenia. New pharmacological, genetic and epigenetic tools, including selective 5-HTR antagonists and agonists, as well as novel transgenic, molecular and neuroimaging techniques might offer important insights into the role of 5-HTergic system in cognitive performance including memory formation, amnesia, or related behavioral/psychiatric alterations. Investigating changes in the 5-HTergic and other neurotransmitter systems at a circuit level, as well as potential neurobiological markers (5-HTR protein or mRNA expression, signaling cascades, etc.) may prove particularly valuable in elucidating normal and impaired cognition and exploring the full potential of 5-HTergic drugs as cognition enhancers.

Acknowledgments

Conflict of interest statement: The authors declare no conflict of interest. The authors thank Marta Radman Livaja and Donald C. Carleton Jr. for editing the English language.

References

  • [1].Olivier B.. Serotonin: a never-ending story. Eur. J. Pharmacol. 2015;753:2–18. doi: 10.1016/j.ejphar.2014.10.031. [DOI] [PubMed] [Google Scholar]
  • [2].Ciranna L.. Serotonin as a modulator of glutamate- and GABAmediated neurotransmission: implications in physiological functions and in pathology. Curr. Neuropharmacol. 2006;4:101–114. doi: 10.2174/157015906776359540. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [3].Puig M., Gener T.. Serotonin modulation of prefronto-hippocampal rhythms in health and disease. ACS Chem. Neurosci. 2015;6:1017–1025. doi: 10.1021/cn500350e. [DOI] [PubMed] [Google Scholar]
  • [4].Mück-Šeler D., Pivac N.. Serotonin. Period. Biol. 2011;113:29–41. [Google Scholar]
  • [5].Charnay Y., Léger L.. Brain serotonergic circuitries. Dialogues Clin. Neurosci. 2010;12:471–487. doi: 10.31887/DCNS.2010.12.4/ycharnay. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [6].Boureau Y.L., Dayan P.. Opponency revisited: competition and cooperation between dopamine and serotonin. Neuropsychopharmacology. 2011;36:74–97. doi: 10.1038/npp.2010.151. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [7].Harvey JA.. Role of the serotonin 5-HT2A receptor in learning. Learn. Mem. 2003;10:355–362. doi: 10.1101/lm.60803. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [8].Glikmann-Johnston Y., Saling M.M., Reutens D.C., Stout J.C.. Hippocampal 5-HT1A receptor and spatial learning and memory. Front. Pharmacol. 2015;6:289. doi: 10.3389/fphar.2015.00289. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [9].Buzsáki G., Moser E.I.. Memory, navigation and theta rhythm in the hippocampal-entorhinal system. Nat. Neurosci. 2013;16:130–138. doi: 10.1038/nn.3304. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [10].Rubin R.D., Watson P.D., Duff M.C., Cohen N.J.. The role of the hippocampus in flexible cognition and social behavior. Front. Hum. Neurosci. 2014;8:742. doi: 10.3389/fnhum.2014.00742. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [11].Robbins T.W.. From arousal to cognition: the integrative position of the prefrontal cortex. Prog. Brain Res. 2000;126:469–483. doi: 10.1016/S0079-6123(00)26030-5. [DOI] [PubMed] [Google Scholar]
  • [12].Clark L., Cools R., Robbins T.W.. The neuropsychology of ventral prefrontal cortex: decision-making and reversal learning. Brain Cogn. 2004;55:41–53. doi: 10.1016/S0278-2626(03)00284-7. [DOI] [PubMed] [Google Scholar]
  • [13].Leiser S.C., Li Y., Pehrson A.L., Dale E., Smagin G., Sanchez C.. Serotonergic regulation of prefrontal cortical circuitries involved in cognitive processing: a review of individual 5-HT receptor mechanisms and concerted effects of 5-HT receptors exemplified by the multimodal antidepressant vortioxetine. ACS Chem. Neurosci. 2015;6:970–986. doi: 10.1021/cn500340j. [DOI] [PubMed] [Google Scholar]
  • [14].Meneses A.. Serotonin, neural markers, and memory. Front. Pharmacol. 2015;6:143. doi: 10.3389/fphar.2015.00143. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [15].Seyedabadi M., Fakhfouri G., Ramezani V., Mehr S.E., Rahimian R.. The role of serotonin in memory: interactions with neurotransmitters and downstream signaling. Exp. Brain Res. 2014;232:723–738. doi: 10.1007/s00221-013-3818-4. [DOI] [PubMed] [Google Scholar]
  • [16].Rodriguez J.J., Noristani H.N., Verkhratsky A.. The serotonergic system in ageing and Alzheimer’s disease. Prog. Neurobiol. 2012;99:15–41. doi: 10.1016/j.pneurobio.2012.06.010. [DOI] [PubMed] [Google Scholar]
  • [17].Lin S.H., Lee L.T., Yang Y.K.. Serotonin and mental disorders: a concise review on molecular neuroimaging evidence. Clin. Psychopharmacol. Neurosci. 2014;12:196–202. doi: 10.9758/cpn.2014.12.3.196. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [18].Terry A.V. Jr, Buccafusco J.J., Wilson C.. Cognitive dysfunction in neuropsychiatric disorders: selected serotonin receptor subtypes as therapeutic targets. Behav. Brain Res. 2008;195:30–38. doi: 10.1016/j.bbr.2007.12.006. [DOI] [PubMed] [Google Scholar]
  • [19].Green M.F., Kern R.S., Braff D.L., Mintz J.. Neurocognitive deficits and functional outcome in schizophrenia: are we measuring the “right stuff”? Schizophr. Bull. 2000;26:119–136. doi: 10.1093/oxfordjournals.schbul.a033430. [DOI] [PubMed] [Google Scholar]
  • [20].Charney D., Nestler E. Neurobiology of mental illness. 3rd ed. Oxford University Press; New York: 2011. [Google Scholar]
  • [21].Kapur S., Remington G.. Serotonin-dopamine interaction and its relevance to schizophrenia. Am. J. Psychiatry. 1996;153:466–476. doi: 10.1176/ajp.153.4.466. [DOI] [PubMed] [Google Scholar]
  • [22].Stephan K.E., Friston K.J., Frith C.D.. Dysconnection in schizophrenia: from abnormal synaptic plasticity to failures of self-monitoring. Schizophr. Bull. 2009;35:509–527. doi: 10.1093/schbul/sbn176. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [23].Dean B.. A predicted cortical serotonergic/cholinergic/GABAergic interface as a site of pathology in schizophrenia. Clin. Exp. Pharmacol. Physiol. 2001;28:74–78. doi: 10.1046/j.1440-1681.2001.03401.x. [DOI] [PubMed] [Google Scholar]
  • [24].Boyer P., Phillips J.L., Rousseau F.L., Ilivitsky S.. Hippocampal abnormalities and memory deficits: new evidence of a strong pathophysiological link in schizophrenia. Brain Res. Rev. 2007;54:92–112. doi: 10.1016/j.brainresrev.2006.12.008. [DOI] [PubMed] [Google Scholar]
  • [25].Joubert S., Gour N., Guedj E., Didic M., Guériot C., Koric L.. Earlyonset and late-onset Alzheimer’s disease are associated with distinct patterns of memory impairment. Cortex. 2016;74:217–232. doi: 10.1016/j.cortex.2015.10.014. [DOI] [PubMed] [Google Scholar]
  • [26].Moodley K.K., Chan D.. The hippocampus in neurodegenerative disease. Front. Neurol. Neurosci. 2014;34:95–108. doi: 10.1159/000356430. [DOI] [PubMed] [Google Scholar]
  • [27].Meltzer C.C., Smith G., DeKosky S., Pollock B.G., Mathis C.A., Moore R.Y.. Serotonin in aging, late-life depression, and Alzheimer’s disease: the emerging role of functional imaging. Neuropsychopharmacology. 1998;18:407–430. doi: 10.1016/S0893-133X(97)00194-2. [DOI] [PubMed] [Google Scholar]
  • [28].Aletrino M.A., Vogels O.J., Van Domburg P.H., Ten Donkelaar H.J.. Cell loss in the nucleus raphes dorsalis in Alzheimer’s disease. Neurobiol. Aging. 1992;13:461–468. doi: 10.1016/0197-4580(92)90073-7. [DOI] [PubMed] [Google Scholar]
  • [29].Garcia-Alloza M., Gil-Bea F.J., Diez-Ariza M., Chen C.P., Francis P.T., Lasheras B.. Cholinergic–serotonergic imbalance contributes to cognitive and behavioural symptoms in Alzheimer’s disease. Neuropsychologia. 2005;43:442–449. doi: 10.1016/j.neuropsychologia.2004.06.007. [DOI] [PubMed] [Google Scholar]
  • [30].Tohgi H., Abe T., Takahashi S., Kimura M., Takahashi J., Kikuchi T.. Concentrations of serotonin and its related substances in the cerebrospinal fluid in patients with Alzheimer type dementia. Neurosci. Lett. 1992;141:9–12. doi: 10.1016/0304-3940(92)90322-x. [DOI] [PubMed] [Google Scholar]
  • [31].Mück-Šeler D., Presečki P., Mimica N., Mustapić M., Pivac N., Babić A.. Platelet serotonin concentration and monoamine oxidase type B activity in female patients in early, middle and late phase of Alzheimer’s disease. Prog. Neuropsychopharmacol. Biol. Psychiatry. 2009;33:1226–1231. doi: 10.1016/j.pnpbp.2009.07.004. [DOI] [PubMed] [Google Scholar]
  • [32].Ramirez M.J., Lai M.K., Tordera R.M., Francis P.T.. Serotonergic therapies for cognitive symptoms in Alzheimer’s disease: rationale and current status. Drugs. 2014;74:729–736. doi: 10.1007/s40265-014-0217-5. [DOI] [PubMed] [Google Scholar]
  • [33].Cirrito J.R., Disabato B.M., Restivo J.L., Verges D.K., Goebel W.D., Sathyan A.. Serotonin signaling is associated with lower amyloid-β levels and plaques in transgenic mice and humans. Proc. Natl. Acad. Sci. USA. 2011;108:14968–14973. doi: 10.1073/pnas.1107411108. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [34].Payton S., Cahill C.M., Randall J.D., Gullans S.R., Rogers J.T.. Drug discovery targeted to the Alzheimer’s APP mRNA 5’-untranslated region: the action of paroxetine and dimercaptopropanol. J. Mol. Neurosci. 2003;20:267–275. doi: 10.1385/JMN:20:3:267. [DOI] [PubMed] [Google Scholar]
  • [35].Geldenhuys W.J., Van der Schyf C.J.. Role of serotonin in Alzheimer’s disease: a new therapeutic target? CNS Drugs. 2011;25:765–781. doi: 10.2165/11590190-000000000-00000. [DOI] [PubMed] [Google Scholar]
  • [36].Lanari A., Amenta F., Silvestrelli G., Tomassoni D., Parnetti L.. Neurotransmitter deficits in behavioural and psychological symptoms of Alzheimer’s disease. Mech. Ageing Dev. 2006;127:158–165. doi: 10.1016/j.mad.2005.09.016. [DOI] [PubMed] [Google Scholar]
  • [37].Cerejeira J., Lagarto L., Mukaetova-Ladinska E.B.. Behavioral and psychological symptoms of dementia. Front. Neurol. 2012;3:73. doi: 10.3389/fneur.2012.00073. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [38].Nichols D.E., Nichols C.D.. Serotonin receptors. Chem. Rev. 2008;108:1614–1641. doi: 10.1021/cr078224o. [DOI] [PubMed] [Google Scholar]
  • [39].Hoyer D., Hannon J.P., Martin G.R.. Molecular, pharmacological and functional diversity of 5-HT receptors. Pharmacol. Biochem. Behav. 2002;71:533–554. doi: 10.1016/s0091-3057(01)00746-8. [DOI] [PubMed] [Google Scholar]
  • [40].McCorvy J.D., Roth B.L.. Structure and function of serotonin G proteincoupled receptors. Pharmacol. Ther. 2015;150:129–142. doi: 10.1016/j.pharmthera.2015.01.009. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [41].Berumen L.C., Rodríguez A., Miledi R., García-Alcocer G.. Serotonin receptors in hippocampus. Scientific World Journal. 2012;2012:823493. doi: 10.1100/2012/823493. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [42].Celada P., Puig M.V., Artigas F.. Serotonin modulation of cortical neurons and networks. Front. Integr. Neurosci. 2013;7:25. doi: 10.3389/fnint.2013.00025. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [43].Barnes N.M., Sharp T.. A review of central 5-HT receptors and their function. Neuropharmacology. 1999;38:1083–1152. doi: 10.1016/s0028-3908(99)00010-6. [DOI] [PubMed] [Google Scholar]
  • [44].Meneses A.. Involvement of 5-HT2A/2B/2C receptors on memory formation: simple agonism, antagonism, or inverse agonism? Cell. Mol. Neurobiol. 2002;22:675–688. doi: 10.1023/a:1021800822997. [DOI] [PubMed] [Google Scholar]
  • [45].Ciranna L.. Serotonin as a modulator of glutamate- and GABAmediated neurotransmission: implications in physiological functions and in pathology. Curr. Neuropharmacol. 2006;4:101–114. doi: 10.2174/157015906776359540. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [46].Hall H., Lundkvist C., Halldin C., Farde L., Pike V.W., McCarron J.A.. Autoradiographic localization of 5-HT1A receptors in the postmortem human brain using [3H]WAY-100635 and [11C]WAY-100635. Brain Res. 1997;745:96–108. doi: 10.1016/s0006-8993(96)01131-6. [DOI] [PubMed] [Google Scholar]
  • [47].Hjorth S., Bengtsson H.J., Kullberg A., Carlzon D., Peilot H., Auerbach S.B.. Serotonin autoreceptor function and antidepressant drug action. J. Psychopharmacol. 2000;14:177–185. doi: 10.1177/026988110001400208. [DOI] [PubMed] [Google Scholar]
  • [48].Popova N.K., Naumenko V.S.. 5-HT1A receptor as a key player in the brain 5-HT system. Rev. Neurosci. 2013;24:191–204. doi: 10.1515/revneuro-2012-0082. [DOI] [PubMed] [Google Scholar]
  • [49].Ogren S.O., Eriksson T.M., Elvander-Tottie E., D’Addario C., Ekström J.C., Svenningsson P.. The role of 5-HT1A receptors in learning and memory. Behav. Brain Res. 2008;195:54–77. doi: 10.1016/j.bbr.2008.02.023. [DOI] [PubMed] [Google Scholar]
  • [50].Li Z., Ichikawa J., Dai J., Meltzer H.Y.. Aripiprazole, a novel antipsychotic drug, preferentially increases dopamine release in the prefrontal cortex and hippocampus in rat brain. Eur. J. Pharmacol. 2004;493:75–83. doi: 10.1016/j.ejphar.2004.04.028. [DOI] [PubMed] [Google Scholar]
  • [51].Bantick R.A., De Vries M.H., Grasby P.M.. The effect of a 5-HT1A receptor agonist on striatal dopamine release. Synapse. 2005;57:67–75. doi: 10.1002/syn.20156. [DOI] [PubMed] [Google Scholar]
  • [52].Chilmonczyk Z., Bojarski A.J., Pilc A., Sylte I.. Functional selectivity and antidepressant activity of serotonin 1A receptor ligands. Int. J. Mol. Sci. 2015;16:18474–18506. doi: 10.3390/ijms160818474. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [53].Stiedl O., Pappa E., Konradsson-Geuken Å., Ögren S.O.. The role of the serotonin receptor subtypes 5-HT1A and 5-HT7 and its interaction in emotional learning and memory. Front. Pharmacol. 2015;6:162. doi: 10.3389/fphar.2015.00162. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [54].Tauscher J., Verhoeff N.P., Christensen B.K., Hussey D., Meyer J.H., Kecojevic A.. Serotonin 5-HT1A receptor binding potential declines with age as measured by [11C]WAY-100635 and PET. Neuropsychopharmacology. 2001;24:522–530. doi: 10.1016/S0893-133X(00)00227-X. [DOI] [PubMed] [Google Scholar]
  • [55].Yasuno F., Suhara T., Nakayama T., Ichimiya T., Okubo Y., Takano A.. Inhibitory effect of hippocampal 5-HT1A receptors on human explicit memory. Am. J. Psychiatry. 2003;160:334–340. doi: 10.1176/appi.ajp.160.2.334. [DOI] [PubMed] [Google Scholar]
  • [56].Borg J.. Molecular imaging of the 5-HT1A receptor in relation to human cognition. Behav. Brain. Res. 2008;195:103–111. doi: 10.1016/j.bbr.2008.06.011. [DOI] [PubMed] [Google Scholar]
  • [57].Kepe V., Barrio J.R., Huang S.C., Ercoli L., Siddarth P., Shoghi-Jadid K.. Serotonin 1A receptors in the living brain of Alzheimer’s disease patients. Proc. Natl. Acad. Sci. USA. 2006;103:702–707. doi: 10.1073/pnas.0510237103. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [58].Lai M.K., Tsang S.W., Francis P.T., Esiri M.M., Hope T., Lai O.F.. [3H] GR113808 binding to serotonin 5-HT4 receptors in the postmortem neocortex of Alzheimer disease: a clinicopathological study. J. Neural Transm. 2003;110:779–788. doi: 10.1007/s00702-003-0825-9. [DOI] [PubMed] [Google Scholar]
  • [59].Pessoa-Mahana H., Recabarren-Gajardo G., Temer J.F., Zapata-Torres G., Pessoa-Mahana C.D., Saitz Barría C.. Synthesis, docking studies and biological evaluation of benzo[b]thiophen-2-yl-3-(4-arylpiperazin-1-yl)-propan-1-one derivatives on 5-HT1A serotonin receptors. Molecules. 2012;17:1388–1407. doi: 10.3390/molecules17021388. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [60].Selvaraj S., Arnone D., Cappai A., Howes O.. Alterations in the serotonin system in schizophrenia: a systematic review and metaanalysis of postmortem and molecular imaging studies. Neurosci. Biobehav. Rev. 2014;45:233–245. doi: 10.1016/j.neubiorev.2014.06.005. [DOI] [PubMed] [Google Scholar]
  • [61].Ichikawa J., Ishii H., Bonaccorso S., Fowler W.L., O’Laughlin I.A., Meltzer H.Y.. 5-HT2A and D2 receptor blockade increases cortical DA release via 5-HT1A receptor activation: a possible mechanism of atypical antipsychotic-induced cortical dopamine release. J. Neurochem. 2001;76:1521–1531. doi: 10.1046/j.1471-4159.2001.00154.x. [DOI] [PubMed] [Google Scholar]
  • [62].Łukasiewicz S., Błasiak E., Szafran-Pilch K., Dziedzicka-Wasylewska M.. Dopamine D2 and serotonin 5-HT1A receptor interaction in the context of the effects of antipsychotics in vitro studies. J. Neurochem. 2016 doi: 10.1111/jnc.13582. [Epub ahead of print] [DOI] [PubMed] [Google Scholar]
  • [63].Pauwels P.J.. 5-HT1B/D receptor antagonists. Gen. Pharmacol. 1997;29:293–303. doi: 10.1016/s0306-3623(96)00460-0. [DOI] [PubMed] [Google Scholar]
  • [64].Varrone A., Svenningsson P., Marklund P., Fatouros-Bergman H., Forsberg A., Halldin C.. 5-HT1B receptor imaging and cognition: a positron emission tomography study in control subjects and Parkinson’s disease patients. Synapse. 2015;69:365–374. doi: 10.1002/syn.21823. [DOI] [PubMed] [Google Scholar]
  • [65].Matuskey D., Pittman B., Planeta-Wilson B., Walderhaug E., Henry S., Gallezot J-D.. Age effects on the serotonin 1B receptor as assessed by PET imaging. J. Nucl. Med. 2012;53:1411–1414. doi: 10.2967/jnumed.112.103598. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [66].Meneses A., Hong E.. Role of 5-HT1B, 5-HT2A and 5-HT2C receptors in learning. Behav. Brain Res. 1997;87:105–110. doi: 10.1016/s0166-4328(96)02266-8. [DOI] [PubMed] [Google Scholar]
  • [67].Garcia-Alloza M., Hirst W.D., Chen C.P., Lasheras B., Francis P.T., Ramirez M.J.. Differential involvement of 5-HT1B/1D and 5-HT6 receptors in cognitive and non-cognitive symptoms in Alzheimer’s disease. Neuropsychopharmacology. 2004;29:410–416. doi: 10.1038/sj.npp.1300330. [DOI] [PubMed] [Google Scholar]
  • [68].Raiteri M., Marchi M., Maura G., Bonanno G.. Presynaptic regulation of acetylcholine release in the CNS. Cell. Biol. Int. Rep. 1989;13:1109–1118. doi: 10.1016/0309-1651(89)90024-6. Translational Neuroscience 45. [DOI] [PubMed] [Google Scholar]
  • [69].Tiger M., Rück C., Forsberg A., Varrone A., Lindefors N., Halldin C.. Reduced 5-HT1B receptor binding in the dorsal brain stem after cognitive behavioural therapy of major depressive disorder. Psychiatry Res. 2014;223:164–170. doi: 10.1016/j.pscychresns.2014.05.011. [DOI] [PubMed] [Google Scholar]
  • [70].López-Figueroa A.L., Norton C.S., López-Figueroa M.O., Armellini-Dodel D., Burke S., Akil H.. Serotonin 5-HT1A, 5-HT1B, and 5-HT2A receptor mRNA expression in subjects with major depression, bipolar disorder, and schizophrenia. Biol. Psychiatry. 2004;55:225–233. doi: 10.1016/j.biopsych.2003.09.017. [DOI] [PubMed] [Google Scholar]
  • [71].Boulenguez P., Peters S.L., Mitchell S.N., Chauveau J., Gray J.A., Joseph M.H.. Dopamine release in the nucleus accumbens and latent inhibition in the rat following microinjections of a 5-HT1B agonist into the dorsal subiculum: implications for schizophrenia. J. Psychopharmacol. 1998;12:258–267. doi: 10.1177/026988119801200305. [DOI] [PubMed] [Google Scholar]
  • [72].Fink K.B., Gothert M.. 5-HT receptor regulation of neurotransmitter release. Pharmacol. Rev. 2007;59:360–417. doi: 10.1124/pr.107.07103. [DOI] [PubMed] [Google Scholar]
  • [73].Hasselbalch S.G., Madsen K., Svarer C., Pinborg L.H., Holm S., Paulson O.B.. Reduced 5-HT2A receptor binding in patients with mild cognitive impairment. Neurobiol. Aging. 2008;29:1830–1838. doi: 10.1016/j.neurobiolaging.2007.04.011. [DOI] [PubMed] [Google Scholar]
  • [74].Lorke D.E., Lu G., Cho E., Yew D.T.. Serotonin 5-HT2A and 5-HT6 receptors in the prefrontal cortex of Alzheimer and normal aging patients. BMC Neurosci. 2006;7:36. doi: 10.1186/1471-2202-7-36. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [75].Tsang S.W., Keene J., Hope T., Spence I., Frances P.T., Wong P.T.H.. A serotoninergic basis for hyperphagic eating changes in Alzheimer’s disease. J. Neurol. Sci. 2010;288:151–155. doi: 10.1016/j.jns.2009.08.066. [DOI] [PubMed] [Google Scholar]
  • [76].Marner L., Frokjaer V.G., Kalbitzer J., Lehel S., Madsen K., Baare W.F.C.. Loss of serotonin 2A receptors exceeds loss of serotonergic projections in early Alzheimer’s disease: a combined [11C] DASB and [18F] altanserin-PET study. Neurobiol. Aging. 2012;33:479–487. doi: 10.1016/j.neurobiolaging.2010.03.023. [DOI] [PubMed] [Google Scholar]
  • [77].Versijpt J., Van Laere K.J., Dumont F., Decoo D., Vandecapelle M., Santens P.. Imaging of the 5-HT2A system: age-, gender-, and Alzheimer’s disease-related findings. Neurobiol. Aging. 2003;24:553–561. doi: 10.1016/s0197-4580(02)00137-9. [DOI] [PubMed] [Google Scholar]
  • [78].Lai MK, Tsang SW, Alder JT, Keene J, Hope T, Esiri MM, Francis PT, Chen CP. Loss of serotonin 5-HT2A receptors in the postmortem temporal cortex correlates with rate of cognitive decline in Alzheimer’s disease. Psychopharmacology. 2005;179:673–677. doi: 10.1007/s00213-004-2077-2. [DOI] [PubMed] [Google Scholar]
  • [79].Holm P., Ettrup A., Klein A.B., Santini M.A., El-Sayed M., Elvang A.B.. Plaque deposition dependent decrease in 5-HT2A serotonin receptor in AbPPswe/PS1DE9 amyloid overexpressing mice. J. Alzheimers Dis. 2010;20:1201–1213. doi: 10.3233/JAD-2010-100117. [DOI] [PubMed] [Google Scholar]
  • [80].Li L.B., Zhang L., Sun Y.N., Han L.N., Wu Z.H., Zhang Q.J.. Activation of serotonin 2A receptors in the medial septum-diagonal band of Broca complex enhanced working memory in the hemiparkinsonian rats. Neuropharmacology. 2015;91:23–33. doi: 10.1016/j.neuropharm.2014.11.025. [DOI] [PubMed] [Google Scholar]
  • [81].Naghdi N., Harooni H.E.. 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;157:205–210. doi: 10.1016/j.bbr.2004.06.024. [DOI] [PubMed] [Google Scholar]
  • [82].Terry A.V., Buccafusco J.J., Bartoszyk G.D.. Selective serotonin 5-HT2A receptor antagonist EMD 281014 improves delayed matching performance in young and aged rhesus monkeys. Psychopharmacology. 2005;179:725–732. doi: 10.1007/s00213-004-2114-1. [DOI] [PubMed] [Google Scholar]
  • [83].Rasmussen H., Erritzoe D., Andersen R., Ebdrup B.H., Aggernaes B., Oranje B.. Decreased frontal serotonin 2A receptor binding in antipsychotic-naive patients with first-episode schizophrenia. Arch. Gen. Psychiatry. 2010;67:9–16. doi: 10.1001/archgenpsychiatry.2009.176. [DOI] [PubMed] [Google Scholar]
  • [84].Okubo Y., Suhara T., Suzuki K., Kobayashi K., Inoue O., Terasaki O.. Serotonin 5-HT2 receptors in schizophrenic patients studied by positron emission tomography. Life Sci. 2000;66:2455–2464. doi: 10.1016/s0024-3205(00)80005-3. [DOI] [PubMed] [Google Scholar]
  • [85].Roth B.L., Hanizavareh S.M., Blum A.E.. Serotonin receptors represent highly favorable molecular targets for cognitive enhancement in schizophrenia and other disorders. Psychopharmacology. 2004;174:17–24. doi: 10.1007/s00213-003-1683-8. [DOI] [PubMed] [Google Scholar]
  • [86].Liperoti R., Pedone C., Corsonello A.. Antipsychotics for the treatment of behavioral and psychological symptoms of dementia (BPSD) Curr. Neuropharmacol. 2008;6:117–124. doi: 10.2174/157015908784533860. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [87].Tyson P.J., Laws K.R., Flowers K.A., Tyson A., Mortimer A.M.. Cognitive function and social abilities in patients with schizophrenia: relationship with atypical antipsychotics. Psychiatry Clin. Neurosci. 2006;60:473–479. doi: 10.1111/j.1440-1819.2006.01534.x. [DOI] [PubMed] [Google Scholar]
  • [88].Zhang G., Stackman R.W. Jr. The role of serotonin 5-HT2A receptors in memory and cognition. Front. Pharmacol. 2015;6:225. doi: 10.3389/fphar.2015.00225. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [89].Di Giovanni G., De Deurwaerdère P.. New therapeutic opportunities for 5-HT2C receptor ligands in neuropsychiatric disorders. Pharmacol. Ther. 2016;157:125–162. doi: 10.1016/j.pharmthera.2015.11.009. [DOI] [PubMed] [Google Scholar]
  • [90].Jensen N.H., Cremers T.I., Sotty F.. Therapeutic potential of 5-HT2C receptor ligands. ScientificWorldJournal. 2010;10:1870–1885. doi: 10.1100/tsw.2010.180. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [91].Cheng J., Kozikowski A.P.. We Need 2C but Not 2B: Developing serotonin 2C (5-HT2C) receptor agonists for the treatment of CNS disorders. Chem. Med. Chem. 2015;10:1963–1967. doi: 10.1002/cmdc.201500437. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [92].Nilsson S.R., Ripley T.L., Somerville E.M., Clifton P.G.. Reduced activity at the 5-HT2C receptor enhances reversal learning by decreasing the influence of previously non-rewarded associations. Psychopharmacology. 2012;224:241–254. doi: 10.1007/s00213-012-2746-5. [DOI] [PubMed] [Google Scholar]
  • [93].Busceti C.L., Di Pietro P., Riozzi B., Traficante A., Biagioni F., Nisticò R.. 5-HT2C serotonin receptor blockade prevents tau protein hyperphosphorylation and corrects the defect in hippocampal synaptic plasticity caused by a combination of environmental stressors in mice. Pharmacol. Res. 2015;99:258–268. doi: 10.1016/j.phrs.2015.06.017. [DOI] [PubMed] [Google Scholar]
  • [94].Del’Guidice T., Lemay F., Lemasson M., Levasseur-Moreau J., Manta S., Etievant A.. Stimulation of 5-HT2C receptors improves cognitive deficits induced by human tryptophan hydroxylase 2 loss of function mutation. Neuropsychopharmacology. 2014;39:1125–1134. doi: 10.1038/npp.2013.313. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [95].Nitsch R.M., Deng M., Growdon J.H., Wurtman R.J.. Serotonin 5-HT2A and 5-HT2C receptors stimulate amyloid precursor protein ectodomain secretion. J. Biol. Chem. 1996;271:4188–4194. doi: 10.1074/jbc.271.8.4188. [DOI] [PubMed] [Google Scholar]
  • [96].Arjona A.A., Pooler A.M., Lee R.K., Wurtman R.J.. Effect of a 5-HT2C serotonin agonist, dexnorfenfluramine, on amyloid precursor protein metabolism in guinea pigs. Brain Res. 2002;951:135–140. doi: 10.1016/s0006-8993(02)03153-0. [DOI] [PubMed] [Google Scholar]
  • [97].Martins L.C., Rocha N.P., Torres K.C., Dos Santos R.R., França G.S., de Moraes E.N.. Disease-specific expression of the serotoninreceptor 5-HT2C in natural killer cells in Alzheimer’s dementia. J. Neuroimmunol. 2012;251:73–79. doi: 10.1016/j.jneuroim.2012.06.003. Translational Neuroscience 46. [DOI] [PubMed] [Google Scholar]
  • [98].Chagraoui A., Thibaut F., Skiba M., Thuillez C., Bourin M.. 5-HT2C receptors in psychiatric disorders: a review. Prog. Neuropsychopharmacol. Biol. Psychiatry. 2016;66:120–135. doi: 10.1016/j.pnpbp.2015.12.006. [DOI] [PubMed] [Google Scholar]
  • [99].Clemett D.A., Punhani T., Duxon M.S., Blackburn T.P., Fone K.C.. Immunohistochemical localisation of the 5-HT2C receptor protein in the rat CNS. Neuropharmacology. 2000;39:123–132. doi: 10.1016/s0028-3908(99)00086-6. [DOI] [PubMed] [Google Scholar]
  • [100].Faerber L., Drechsler S., Ladenburger S., Gschaidmeier H., Fischer W.. The neuronal 5-HT3 receptor network after 20 years of research - evolving concepts in management of pain and inflammation. Eur. J. Pharmacol. 2007;560:1–8. doi: 10.1016/j.ejphar.2007.01.028. [DOI] [PubMed] [Google Scholar]
  • [101].Herrstedt J., Dombernowsky P.. Anti-emetic therapy in cancer chemotherapy: current status. Basic Clin. Pharmacol. Toxicol. 2007;101:143–150. doi: 10.1111/j.1742-7843.2007.00122.x. [DOI] [PubMed] [Google Scholar]
  • [102].Pehrson A., Gaarn du Jardin Nielsen K., Jensen J.B., Sanchez C.. The novel multimodal antidepressant Lu AA21004 improves memory performance in 5-HT depleted rats via 5-HT3 and 5-HT1A receptor mechanisms. Eur. Neuropsychopharmacol. 2012;22:S269–S269. [Google Scholar]
  • [103].Boast C., Bartolomeo A.C., Morris H., Moyer J.A.. 5HT antagonists attenuate MK801-impaired radial arm maze performance in rats. Neurobiol. Learn. Mem. 1999;71:259–271. doi: 10.1006/nlme.1998.3886. [DOI] [PubMed] [Google Scholar]
  • [104].Carey G.J., Costall B., Domeney A.M., Gerrard P.A., Jones D.N., Naylor R.J.. Ondansetron and arecoline prevent scopolamineinduced cognitive deficits in the marmoset. Pharmacol. Biochem. Behav. 1992;42:75–83. doi: 10.1016/0091-3057(92)90449-p. [DOI] [PubMed] [Google Scholar]
  • [105].Pitsikas N., Borsini F.. Itasetron (DAU 6215) prevents age-related memory deficits in the rat in a multiple choice avoidance task. Eur. J. Pharmacol. 1996;311:115–119. doi: 10.1016/0014-2999(96)00586-9. [DOI] [PubMed] [Google Scholar]
  • [106].Ju Yeon Ban, Yeon Hee Seong. Blockade of 5-HT3 receptor with MDL 72222 and Y 25130 reduces Ab25-35-induced neurotoxicity in cultured rat cortical neurons. Eur. J. Pharmacol. 2005;520:12–21. doi: 10.1016/j.ejphar.2005.07.021. [DOI] [PubMed] [Google Scholar]
  • [107].Rahimian R., Fakhfouri G., Ejtemaei Mehr S., Ghia J.E., Genazzani A.A., Payandemehr B.. Tropisetron attenuates Ab–induced inflammatory and apoptotic responses in rats. Eur. J. Clin. Invest. 2013;43:1039–1051. doi: 10.1111/eci.12141. [DOI] [PubMed] [Google Scholar]
  • [108].Spilman P., Descamps O., Gorostiza O., Peters-Libeu C., Poksay K.S., Matalis A.. The multi-functional drug tropisetron binds APP and normalizes cognition in a murine Alzheimer’s model. Brain Res. 2014;1551:25–44. doi: 10.1016/j.brainres.2013.12.029. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [109].Fakhfouri G., Mousavizadeh K., Mehr S.E., Dehpour A.R., Zirak M.R., Ghia J.E.. From chemotherapy-induced emesis to neuroprotection: therapeutic opportunities for 5-HT3 receptor antagonists. Mol. Neurobiol. 2015;52:1670–1679. doi: 10.1007/s12035-014-8957-5. [DOI] [PubMed] [Google Scholar]
  • [110].Fakhfouri G., Rahimian R., Ghia J-E., Khan W.I., Dehpour A.R.. Impact of 5-HT3 receptor antagonists on peripheral and central diseases. Drug Discov. Today. 2012;17:741–747. doi: 10.1016/j.drudis.2012.02.009. [DOI] [PubMed] [Google Scholar]
  • [111].Cappelli A., Gallelli A., Manini M., Anzini M., Mennuni L., Makovec F.. Further studies on the interaction of the 5-hydroxytryptamine 3 (5-HT3) receptor with arylpiperazine ligands. Development of a new 5-HT3 receptor ligand showing potent acetylcholinesterase inhibitory properties. J. Med. Chem. 2005;48:3564–3575. doi: 10.1021/jm0493461. [DOI] [PubMed] [Google Scholar]
  • [112].Rezvani A.H., Kholdebarin E., Brucato F.H., Callahan P.M., Lowe D.A., Levin E.D.. Effect of R3487/MEM3454, a novel nicotinic a7 receptor partial agonist and 5-HT3 antagonist on sustained attention in rats. Prog. Neuropsychopharmacol. Biol. Psychiatry. 2009;33:269–275. doi: 10.1016/j.pnpbp.2008.11.018. [DOI] [PubMed] [Google Scholar]
  • [113].Abi-Dargham A., Laruelle M., Lipska B., Jaskiw G.E., Wong D.T., Robertson D.W.. Serotonin 5-HT3 receptors in schizophrenia: a postmortem study of the amygdala. Brain Res. 1993;616:53–57. doi: 10.1016/0006-8993(93)90191-o. [DOI] [PubMed] [Google Scholar]
  • [114].Lennertz L., Wagner M., Frommann I., Schulze-Rauschenbach S., Schuhmacher A., Kühn K.U.. A coding variant of the novel serotonin receptor subunit 5-HT3E influences sustained attention in schizophrenia patients. Eur. Neuropsychopharmacol. 2010;20:414–420. doi: 10.1016/j.euroneuro.2010.02.012. [DOI] [PubMed] [Google Scholar]
  • [115].Ellenbroek B.A., Prinssen E.P.. Can 5-HT3 antagonists contribute toward the treatment of schizophrenia? Behav. Pharmacol. 2015;26:33–44. doi: 10.1097/FBP.0000000000000102. [DOI] [PubMed] [Google Scholar]
  • [116].Akhondzadeh S., Mohammadi N., Noroozian M., Karamghadiri N., Ghoreishi A, Jamshidi A.H.. Added ondansetron for stable schizophrenia: a double blind, placebo controlled trial. Schizophr. Res. 2009;107:206–212. doi: 10.1016/j.schres.2008.08.004. [DOI] [PubMed] [Google Scholar]
  • [117].Zhang Z.J., Kang W.H., Li Q., Wang X.Y., Yao S.M., Ma A.Q.. Beneficial effects of ondansetron as an adjunct to haloperidol for chronic, treatment-resistant schizophrenia: a double-blind, randomized, placebo-controlled study. Schizophr. Res. 2006;88:102–110. doi: 10.1016/j.schres.2006.07.010. [DOI] [PubMed] [Google Scholar]
  • [118].Levkovitz Y., Arnest G., Mendlovic S., Treves I., Fennig S.. The effect of ondansetron on memory in schizophrenic patients. Brain Res. Bull. 2005;65:291–295. doi: 10.1016/j.brainresbull.2003.09.022. [DOI] [PubMed] [Google Scholar]
  • [119].Zhang X.Y., Liu L., Liu S., Hong X., Chen da C., Xiu M.H.. Shortterm tropisetron treatment and cognitive and P50 auditory gating deficits in schizophrenia. Am. J. Psychiatry. 2012;169:974–981. doi: 10.1176/appi.ajp.2012.11081289. [DOI] [PubMed] [Google Scholar]
  • [120].Bockaert J., Claeysen S., Compan V., Dumuis A.. 5-HT4 receptors. Curr. Drug Targets CNS Neurol. Disord. 2004;3:39–51. doi: 10.2174/1568007043482615. [DOI] [PubMed] [Google Scholar]
  • [121].Vilaró M.T., Cortés 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;484:418–439. doi: 10.1002/cne.20447. [DOI] [PubMed] [Google Scholar]
  • [122].Bockaert J.I., Claeysen S., Compan V., Dumuis A.. 5-HT4 receptors: history, molecular pharmacology and brain functions. Neuropharmacology. 2008;55:922–931. doi: 10.1016/j.neuropharm.2008.05.013. [DOI] [PubMed] [Google Scholar]
  • [123].Consolo S., Arnaboldi S., Giorgi S., Russi G., Ladinsky H.. 5-HT4 receptor stimulation facilitates acetylcholine release in rat frontal cortex. Neuroreport. 1994;5:1230–1232. doi: 10.1097/00001756-199406020-00018. [DOI] [PubMed] [Google Scholar]
  • [124].Kilbinger H., Wolf D.. Effects of 5-HT4 receptor stimulation on basal and electrically evoked release of acetylcholine from guinea-pig myenteric plexus. Naunyn Schmiedebergs Arch. Pharmacol. 1992;345:270–275. doi: 10.1007/BF00168686. [DOI] [PubMed] [Google Scholar]
  • [125].Lucas G., Di Matteo V., De Deurwaerdère P., Porras G., Martín-Ruiz R., Artigas F.. Neurochemical and electrophysiological evidence that 5-HT4 receptors exert a state-dependent facilitatory control in vivo on nigrostriatal, but not mesoaccumbal, dopaminergic function. Eur. J. Neurosci. 2001;13:889–898. doi: 10.1046/j.0953-816x.2000.01453.x. [DOI] [PubMed] [Google Scholar]
  • [126].Steward L.J., Ge J., Stowe R.L., Brown D.C., Bruton R.K., Stokes P.R.. Ability of 5-HT4 receptor ligands to modulate rat striatal dopamine release in vitro and in vivo. Br. J. Pharmacol. 1996;117:55–62. doi: 10.1111/j.1476-5381.1996.tb15154.x. Translational Neuroscience 47. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [127].Ge J., Barnes N.M.. 5-HT4 receptor-mediated modulation of 5-HT release in the rat hippocampus in vivo. Br. J. Pharmacol. 1996;117:1475–1480. doi: 10.1111/j.1476-5381.1996.tb15309.x. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [128].Letty S., Child R., Dumuis A., Pantaloni A., Bockaert J., Rondouin G.. 5-HT4 receptors improve social olfactory memory in the rat. Neuropharmacology. 1997;36:681–687. doi: 10.1016/s0028-3908(96)00169-4. [DOI] [PubMed] [Google Scholar]
  • [129].King M.V., Marsden C.A., Fone K.C.. A role for the 5-HT1A, 5-HT4 and 5-HT6 receptors in learning and memory. Trends Pharmacol. Sci. 2008;29:482–492. doi: 10.1016/j.tips.2008.07.001. [DOI] [PubMed] [Google Scholar]
  • [130].Marchetti E., Jacquet M., Jeltsch H., Migliorati M., Nivet E., Cassel J.C.. Complete recovery of olfactory associative learning by activation of 5-HT4 receptors after dentate granule cell damage in rats. Neurobiol. Learn. Mem. 2008;90:185–191. doi: 10.1016/j.nlm.2008.03.010. [DOI] [PubMed] [Google Scholar]
  • [131].Vidal R., Pilar-Cuéllar F., dos Anjos S., Linge R., Treceño B., Vargas VI.. New strategies in the development of antidepressants: towards the modulation of neuroplasticity pathways. Curr. Pharm. Des. 2011;17:521–533. doi: 10.2174/138161211795164086. [DOI] [PubMed] [Google Scholar]
  • [132].Lelong V., Dauphin F., Boulouard M.. RS 67333 and D-cycloserine accelerate learning acquisition in the rat. Neuropharmacology. 2001;41:517–522. doi: 10.1016/s0028-3908(01)00085-5. [DOI] [PubMed] [Google Scholar]
  • [133].Mohler E.G., Shacham S., Noiman S., Lezoualch F., Robert S., Gastineau M.. VRX-03011, a novel 5-HT4 agonist, enhances memory and hippocampal acetylcholine efflux. Neuropharmacology. 2007;53:563–573. doi: 10.1016/j.neuropharm.2007.06.016. [DOI] [PubMed] [Google Scholar]
  • [134].Levallet G., Hotte M., Boulouard M., Dauphin F.. Increased particulate phosphodiesterase 4 in the prefrontal cortex supports 5-HT4 receptor-induced improvement of object recognition memory in the rat. Psychopharmacology. 2009;202:125–139. doi: 10.1007/s00213-008-1283-8. [DOI] [PubMed] [Google Scholar]
  • [135].Fontana D.J., Daniels S.E., Wong E.H., Clark R.D., Eglen R.M.. The effects of novel, selective 5-hydroxytryptamine 5-HT4 receptor ligands in rat spatial navigation. Neuropharmacology. 1997;36:689–696. doi: 10.1016/s0028-3908(97)00055-5. [DOI] [PubMed] [Google Scholar]
  • [136].Lamirault L., Simon H.. Enhancement of place and object recognition memory in young adult and old rats by RS 67333, a partial agonist of 5-HT4 receptors. Neuropharmacology. 2001;41:844–853. doi: 10.1016/s0028-3908(01)00123-x. [DOI] [PubMed] [Google Scholar]
  • [137].Lelong V., Lhonneur L., Dauphin F., Boulouard M.. BIMU 1 and RS 67333, two 5-HT4 receptor agonists, modulate spontaneous alternation deficits induced by scopolamine in the mouse. Naunyn Schmiedebergs Arch. Pharmacol. 2003;367:621–628. doi: 10.1007/s00210-003-0743-2. [DOI] [PubMed] [Google Scholar]
  • [138].Quiedeville A., Boulouard M., Hamidouche K., Da Silva Costa-Aze V., Nee G., Rochais C.. Chronic activation of 5-HT4 receptors or blockade of 5-HT6 receptors improve memory performances. Behav. Brain Res. 2015;293:10–17. doi: 10.1016/j.bbr.2015.07.020. [DOI] [PubMed] [Google Scholar]
  • [139].Madsen K., Haahr M.T., Marner L., Keller S.H., Baare W.F., Svarer C.. Age and sex effects on 5-HT4 receptors in the human brain: a [11C]SB207145 PET study. J. Cereb. Blood Flow Metab. 2011;31:1475–1481. doi: 10.1038/jcbfm.2011.11. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [140].Reynolds G.P., Mason S.L., Meldrum A., De Keczer S., Parnes H., Eglen R.M.. 5-Hydroxytryptamine 5-HT4 receptors in post mortem human brain tissue: distribution, pharmacology and effects of neurodegenerative diseases. Br. J. Pharmacol. 1995;114:993–998. doi: 10.1111/j.1476-5381.1995.tb13303.x. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [141].Matsumoto M., Togashi H., Mori K., Ueno K-I., Ohashi S., Kojima T.. Evidence for involvement of central 5-HT4 receptors in cholinergic function associated with cognitive processes: behavioral, electrophysiological, and neurochemical studies. J. Pharmacol. Exp. Ther. 2001;296:676–682. [PubMed] [Google Scholar]
  • [142].Cho S., Hu Y.. Activation of 5-HT4 receptors inhibits secretion of b-amyloid peptides and increases neuronal survival. Exp. Neurol. 2007;203:274–278. doi: 10.1016/j.expneurol.2006.07.021. [DOI] [PubMed] [Google Scholar]
  • [143].Giannoni P., Gaven F., de Bundel D., Baranger K., Marchetti-Gauthier E., Roman F.S.. Early administration of RS 67333, a specific 5-HT4 receptor agonist, prevents amyloidogenesis and behavioral deficits in the 5XFAD mouse model of Alzheimer’s disease. Front. Aging Neurosci. 2013;5:96. doi: 10.3389/fnagi.2013.00096. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [144].Cochet M., Donneger R., Cassier E., Gaven F., Lichtenthaler S.F., Marin P.. 5-HT4 receptors constitutively promote the nonamyloidogenic pathway of APP cleavage and interact with ADAM10. ACS Chem. Neurosci. 2013;4:130–140. doi: 10.1021/cn300095t. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [145].Shimizu S., Mizuguchi Y., Ohno Y.. Improving the treatment of schizophrenia: role of 5-HT receptors in modulating cognitive and extrapyramidal motor functions. CNS Neurol. Disord. Drug Targets. 2013;12:861–869. doi: 10.2174/18715273113129990088. [DOI] [PubMed] [Google Scholar]
  • [146].Dean B., Tomaskovic-Crook E., Opeskin K., Keks N., Copolov D.. No change in the density of the serotonin1A receptor, the serotonin 4 receptor or the serotonin transporter in the dorsolateral prefrontal cortex from subjects with schizophrenia. Neurochem. Int. 1999;34:109–115. doi: 10.1016/s0197-0186(98)00074-6. [DOI] [PubMed] [Google Scholar]
  • [147].Suzuki T., Iwata N., Kitamura Y., Kitajima T., Yamanouchi Y., Ikeda M.. Association of a haplotype in the serotonin 5-HT4 receptor gene (HTR4) with Japanese schizophrenia. Am. J. Med. Genet. B Neuropsychiatr. Genet. 2003;121B:7–13. doi: 10.1002/ajmg.b.20060. [DOI] [PubMed] [Google Scholar]
  • [148].Pasqualetti M., Ori M., Nardi I., Castagna M., Cassano G.B., Marazziti D.. Distribution of the 5-HT5A serotonin receptor mRNA in the human brain. Mol. Brain Res. 1998;56:1–8. doi: 10.1016/s0169-328x(98)00003-5. [DOI] [PubMed] [Google Scholar]
  • [149].Oliver K.R., Kinsey A.M., Wainwright A., Sirinathsinghji D.J.. Localization of 5-HT5A receptor-like immunoreactivity in the rat brain. Brain Res. 2000;867:131–142. doi: 10.1016/s0006-8993(00)02273-3. [DOI] [PubMed] [Google Scholar]
  • [150].Gonzalez R., Chavez-Pascacio K., Meneses A.. Role of 5-HT5A receptors in the consolidation of memory. Behav. Brain Res. 2013;252:246–251. doi: 10.1016/j.bbr.2013.05.051. [DOI] [PubMed] [Google Scholar]
  • [151].Yamazaki M., Harada K., Yamamoto N., Yarimizu J., Okabe M., Shimada T.. ASP5736, a novel 5-HT5A receptor antagonist, ameliorates positive symptoms and cognitive impairment in animal models of schizophrenia. Eur. Neuropsychopharmacol. 2014;24:1698–1708. doi: 10.1016/j.euroneuro.2014.07.009. [DOI] [PubMed] [Google Scholar]
  • [152].Yamazaki M., Okabe M., Yamamoto N., Yarimizu J., Harada K.. Novel 5-HT5A receptor antagonists ameliorate scopolamineinduced working memory deficit in mice and reference memory impairment in aged rats. J. Pharmacol. Sci. 2015;127:362–369. doi: 10.1016/j.jphs.2015.02.006. [DOI] [PubMed] [Google Scholar]
  • [153].Fone K.C.. An update on the role of the 5-HT6 receptor in cognitive function. Neuropharmacology. 2008;55:1015–1022. doi: 10.1016/j.neuropharm.2008.06.061. Translational Neuroscience 48. [DOI] [PubMed] [Google Scholar]
  • [154].Woods S., Clarke N.N., Layfield R., Fone K.C.. 5-HT6 receptor agonists and antagonists enhance learning and memory in a conditioned emotion response paradigm by modulation of cholinergic and glutamatergic mechanisms. Br. J. Pharmacol. 2012;167:436–449. doi: 10.1111/j.1476-5381.2012.02022.x. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [155].Bali A., Singh S.. Serotonergic 5-HT6 receptor antagonists. Heterocyclic chemistry and potential therapeutic significance. Curr. Top. Med. Chem. 2015;15:1643–1662. doi: 10.2174/1568026615666150427110420. [DOI] [PubMed] [Google Scholar]
  • [156].Liu K.G., Robichaud A.J.. 5-HT6 antagonists as potential treatment for cognitive dysfunction. Drug Dev. Res. 2009;70:145–168. [Google Scholar]
  • [157].Ramirez M.J.. 5-HT6 receptors and Alzheimer’s disease. Alzheimer’s Res. Ther. 2013;5:15. doi: 10.1186/alzrt169. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [158].Foley A.G., Murphy K.J., Hirst W.D., Gallagher H.C., Hagan J.J., Upton N.. The 5-HT6 receptor antagonist SB-271046 reverses scopolaminedisrupted consolidation of a passive avoidance task and ameliorates spatial task deficits in aged rats. Neuropsychopharmacology. 2004;29:93–100. doi: 10.1038/sj.npp.1300332. [DOI] [PubMed] [Google Scholar]
  • [159].Upton N., Chuang T.T., Hunter A.J., Virley D.J.. 5-HT6 receptor antagonists as novel cognitive enhancing agents for Alzheimer’s disease. Neurotherapeutics. 2008;5:458–469. doi: 10.1016/j.nurt.2008.05.008. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [160].Yun H.M., Park K.R., Kim E.C., Kim S., Hong J.T.. Serotonin 6 receptor controls Alzheimer’s disease and depression. Oncotarget. 2015;6:26716–26728. doi: 10.18632/oncotarget.5777. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [161].Pereira M., Martynhak B.J., Andreatini R., Svenningsson P.. 5-HT6 receptor agonism facilitates emotional learning. Front. Pharmacol. 2015;6:200. doi: 10.3389/fphar.2015.00200. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [162].Meffre J., Chaumont-Dubel S., Mannoury la Cour C., Loiseau F., Watson D.J., Dekeyne A.. 5-HT6 receptor recruitment of mTOR as a mechanism for perturbed cognition in schizophrenia. EMBO Mol. Med. 2012:41043–1056. doi: 10.1002/emmm.201201410. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [163].de Bruin N.M., Kruse C.G.. 5-HT6 receptor antagonists: potential efficacy for the treatment of cognitive impairment in schizophrenia. Curr. Pharm. Des. 2015;21:3739–3759. doi: 10.2174/1381612821666150605112105. [DOI] [PubMed] [Google Scholar]
  • [164].Abraham R., Nirogi R., Shinde A., Irupannanavar S.. Low-dose prazosin in combination with 5-HT6 antagonist PRX-07034 has antipsychotic effects. Can. J. Physiol. Pharmacol. 2015;93:13–21. doi: 10.1139/cjpp-2014-0254. [DOI] [PubMed] [Google Scholar]
  • [165].Beaudet G., Bouet V., Jozet-Alves C., Schumann-Bard P., Dauphin F., Paizanis E.. Spatial memory deficit across aging: current insights of the role of 5-HT7 receptors. Front. Behav. Neurosci. 2015;8:448. doi: 10.3389/fnbeh.2014.00448. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [166].Roberts A.J., Hedlund P.B.. The 5-HT7 receptor in learning and memory. Importance of the hippocampus. Hippocampus. 2012;22:762–771. doi: 10.1002/hipo.20938. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [167].Nikiforuk A.. Selective blockade of 5-HT7 receptors facilitates attentional set-shifting in stressed and control rats. Behav. Brain Res. 2012;226:118–123. doi: 10.1016/j.bbr.2011.09.006. [DOI] [PubMed] [Google Scholar]
  • [168].McLean S.L., Woolley M.L., Thomas D., Neill J.C.. Role of 5-HT receptor mechanisms in sub-chronic PCP-induced reversal learning deficits in the rat. Psychopharmacology. 2009;206:403–414. doi: 10.1007/s00213-009-1618-0. [DOI] [PubMed] [Google Scholar]
  • [169].Meneses A.. Effects of the 5-HT7 receptor antagonists SB-269970 and DR 4004 in autoshaping Pavlovian/instrumental learning task. Behav. Brain Res. 2004;155:275–282. doi: 10.1016/j.bbr.2004.04.026. [DOI] [PubMed] [Google Scholar]
  • [170].Horiguchi M., Huang M., Meltzer H.Y.. Interaction of mGlu2/3 agonism with clozapine and lurasidone to restore novel object recognition in subchronic phencyclidine-treated rats. Psychopharmacology. 2011;217:13–24. doi: 10.1007/s00213-011-2251-2. [DOI] [PubMed] [Google Scholar]
  • [171].Gasbarri A., Cifariello A., Pompili A., Meneses A.. Effect of 5-HT7 antagonist SB-269970 in the modulation of working and reference memory in the rat. Behav. Brain Res. 2008;195:164–170. doi: 10.1016/j.bbr.2007.12.020. [DOI] [PubMed] [Google Scholar]
  • [172].Enomoto T., Ishibashi T., Tokuda K., Ishiyama T., Toma S., Ito A.. Lurasidone reverses MK-801-induced impairment of learning and memory in the Morris water maze and radial-arm maze tests in rats. Behav. Brain Res. 2008;186:197–207. doi: 10.1016/j.bbr.2007.08.012. [DOI] [PubMed] [Google Scholar]
  • [173].Horiguchi M., Huang M., Meltzer H.Y.. The role of 5-hydroxytryptamine 7 receptors in the phencyclidine-induced novel object recognition deficit in rats. J. Pharmacol. Exp. Ther. 2011;338:605–614. doi: 10.1124/jpet.111.180638. [DOI] [PubMed] [Google Scholar]
  • [174].De Filippis B., Chiodi V., Adriani W., Lacivita E., Mallozzi C., Leopoldo M.. Long-lasting beneficial effects of central serotonin receptor 7 stimulation in female mice modeling Rett syndrome. Front. Behav. Neurosci. 2015;9:86. doi: 10.3389/fnbeh.2015.00086. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [175].Perez-Garcia G.S., Meneses A.. Effects of the potential 5-HT7 receptor agonist AS 19 in an autoshaping learning task. Behav. Brain Res. 2005;163:136–140. doi: 10.1016/j.bbr.2005.04.014. [DOI] [PubMed] [Google Scholar]
  • [176].Werner F.M., Coveñas R.. Serotonergic drugs: agonists/antagonists at specific serotonergic subreceptors for the treatment of cognitive, depressant and psychotic symptoms in Alzheimer’s disease. Curr. Pharm. Des. 2016 doi: 10.2174/1381612822666160127113524. [Epub ahead of print] [DOI] [PubMed] [Google Scholar]
  • [177].Meneses A.. Memory formation and memory alterations: 5-HT6 and 5-HT7 receptors, novel alternative. Rev. Neurosci. 2014;25:325–356. doi: 10.1515/revneuro-2014-0001. [DOI] [PubMed] [Google Scholar]
  • [178].Dean B., Pavey G., Thomas D., Scarr E.. Cortical serotonin7, 1D and 1F receptors: effects of schizophrenia, suicide and antipsychotic drug treatment. Schizophr. Res. 2006;88:265–274. doi: 10.1016/j.schres.2006.07.003. [DOI] [PubMed] [Google Scholar]
  • [179].East S.Z., Burnet P.W., Kerwin R.W., Harrison P.J.. An RT-PCR study of 5-HT6 and 5-HT7 receptor mRNAs in the hippocampal formation and prefrontal cortex in schizophrenia. Schizophr. Res. 2002;57:15–26. doi: 10.1016/s0920-9964(01)00323-1. [DOI] [PubMed] [Google Scholar]
  • [180].Ikeda M., Iwata N., Kitajima T., Suzuki T., Yamanouchi Y., Kinoshita Y.. Positive association of the serotonin 5-HT7 receptor gene with schizophrenia in a Japanese population. Neuropsychopharmacology. 2006;31:866–871. doi: 10.1038/sj.npp.1300901. [DOI] [PubMed] [Google Scholar]
  • [181].Roth B.L., Craigo S.C., Choudhary M.S., Uluer A., Monsma F.J. Jr, Shen Y.. Binding of typical and atypical antipsychotic agents to 5-hydroxytryptamine-6 and 5-hydroxytryptamine-7 receptors. J. Pharmacol. Exp. Ther. 1994;268:1403–1410. [PubMed] [Google Scholar]
  • [182].Waters K.A., Stean T.O., Hammond B., Virley J.D., Upton N., Kew J.N.C.. Effects of the selective 5-HT7 receptor antagonist SB-269970 in animal models of psychosis and cognition. Behav. Brain Res. 2012;228:211–218. doi: 10.1016/j.bbr.2011.12.009. [DOI] [PubMed] [Google Scholar]
  • [183].Haider S., Khaliq S., Ahmed S.P., Haleem D.J.. Long-term tryptophan administration enhances cognitive performance and increases 5-HT metabolism in the hippocampus of female rats. Amino Acids. 2006;31:421–425. doi: 10.1007/s00726-005-0310-x. Translational Neuroscience 49. [DOI] [PubMed] [Google Scholar]
  • [184].Levkovitz Y., Ophir-Shaham O., Bloch Y., Treves I., Fennig S., Grauer E.. Effect of L-tryptophan on memory in patients with schizophrenia. J. Nerv. Ment. Dis. 2003;191:568–573. doi: 10.1097/01.nmd.0000087182.29781.e0. [DOI] [PubMed] [Google Scholar]
  • [185].Porter R.J., Lunn B.S., O’Brien J.T.. Effects of acute tryptophan depletion on cognitive function in Alzheimer’s disease and in the healthy elderly. Psychol. Med. 2003;33:41–49. doi: 10.1017/s0033291702006906. [DOI] [PubMed] [Google Scholar]
  • [186].Schmitt J.A., Wingen M., Ramaekers J.G., Evers E.A., Riedel W.J.. Serotonin and human cognitive performance. Curr. Pharm. Des. 2006;12:2473–2486. doi: 10.2174/138161206777698909. [DOI] [PubMed] [Google Scholar]
  • [187].Meneses A.. 5-HT systems: emergent targets for memory formation and memory alterations. Rev. Neurosci. 2013;24:629–64. doi: 10.1515/revneuro-2013-0026. [DOI] [PubMed] [Google Scholar]
  • [188].Sanchez C., Asin K.E., Artigas F.. Vortioxetine, a novel antidepressant with multimodal activity: review of preclinical and clinical data. Pharmacol. Ther. 2015;145:43–57. doi: 10.1016/j.pharmthera.2014.07.001. [DOI] [PubMed] [Google Scholar]
  • [189].Stahl S.M.. Modes and nodes explain the mechanism of action of vortioxetine, a multimodal agent (MMA): enhancing serotonin release by combining serotonin (5HT) transporter inhibition with actions at 5HT receptors (5HT1A, 5HT1B, 5HT1D, 5HT7 receptors) CNS Spectr. 2015;20:93–97. doi: 10.1017/S1092852915000139. [DOI] [PubMed] [Google Scholar]

Articles from Translational Neuroscience are provided here courtesy of De Gruyter

RESOURCES