Hostname: page-component-586b7cd67f-tf8b9 Total loading time: 0 Render date: 2024-11-24T23:25:25.596Z Has data issue: false hasContentIssue false

Translating the promise of 5HT4 receptor agonists for the treatment of depression

Published online by Cambridge University Press:  03 April 2020

Susannah E Murphy*
Affiliation:
University Department of Psychiatry, Warneford Hospital, University of Oxford, OX3 7JX, UK Oxford Health NHS Foundation Trust, Warneford Hospital, Oxford, UK
Angharad N de Cates
Affiliation:
University Department of Psychiatry, Warneford Hospital, University of Oxford, OX3 7JX, UK Oxford Health NHS Foundation Trust, Warneford Hospital, Oxford, UK
Amy L Gillespie
Affiliation:
University Department of Psychiatry, Warneford Hospital, University of Oxford, OX3 7JX, UK Oxford Health NHS Foundation Trust, Warneford Hospital, Oxford, UK
Beata R Godlewska
Affiliation:
University Department of Psychiatry, Warneford Hospital, University of Oxford, OX3 7JX, UK Oxford Health NHS Foundation Trust, Warneford Hospital, Oxford, UK
Jessica C Scaife
Affiliation:
University Department of Psychiatry, Warneford Hospital, University of Oxford, OX3 7JX, UK Oxford Health NHS Foundation Trust, Warneford Hospital, Oxford, UK
Lucy C Wright
Affiliation:
University Department of Psychiatry, Warneford Hospital, University of Oxford, OX3 7JX, UK Oxford Health NHS Foundation Trust, Warneford Hospital, Oxford, UK
Philip J Cowen
Affiliation:
University Department of Psychiatry, Warneford Hospital, University of Oxford, OX3 7JX, UK Oxford Health NHS Foundation Trust, Warneford Hospital, Oxford, UK
Catherine J Harmer
Affiliation:
University Department of Psychiatry, Warneford Hospital, University of Oxford, OX3 7JX, UK Oxford Health NHS Foundation Trust, Warneford Hospital, Oxford, UK
*
Author for correspondence: Susannah E Murphy, E-mail: [email protected]
Rights & Permissions [Opens in a new window]

Abstract

Animal experimental studies suggest that 5-HT4 receptor activation holds promise as a novel target for the treatment of depression and cognitive impairment. 5-HT4 receptors are post-synaptic receptors that are located in striatal and limbic areas known to be involved in cognition and mood. Consistent with this, 5-HT4 receptor agonists produce rapid antidepressant effects in a number of animal models of depression, and pro-cognitive effects in tasks of learning and memory. These effects are accompanied by molecular changes, such as the increased expression of neuroplasticity-related proteins that are typical of clinically useful antidepressant drugs. Intriguingly, these antidepressant-like effects have a fast onset of their action, raising the possibility that 5-HT4 receptor agonists may be a particularly useful augmentation strategy in the early stages of SSRI treatment. Until recently, the translation of these effects to humans has been challenging. Here, we review the evidence from animal studies that the 5-HT4 receptor is a promising target for the treatment of depression and cognitive disorders, and outline a potential pathway for the efficient and cost-effective translation of these effects into humans and, ultimately, to the clinic.

Type
Invited Review
Creative Commons
Creative Common License - CCCreative Common License - BY
This is an Open Access article, distributed under the terms of the Creative Commons Attribution licence (http://creativecommons.org/licenses/by/4.0/), which permits unrestricted re-use, distribution, and reproduction in any medium, provided the original work is properly cited.
Copyright
Copyright © The Author(s) 2020

Introduction

The potentiation of monoamine activity, and in particular serotonergic neurotransmission, has been the dominant target of antidepressant treatments since the serendipitous discovery of the antidepressant effects of monoamine oxidase inhibitor and tricyclics in the 1950s. Whilst drugs such as selective serotonin reuptake inhibitors (SSRIs) are more effective than placebo (Cipriani et al., Reference Cipriani, Furukawa, Salanti, Chaimani, Atkinson, Ogawa and Geddes2018), there are significant limitations to current treatment approaches. Only about one third of patients achieve remission after 14 weeks of SSRI treatment and around 30% of patients remain ‘treatment resistant’, even after trying multiple treatment approaches (Rush et al., Reference Rush, Warden, Wisniewski, Fava, Trivedi, Gaynes and Nierenberg2009; Trivedi et al., Reference Trivedi, Fava, Wisniewski, Thase, Quitkin, Warden and Rush2006). When SSRIs are effective, they require chronic administration to achieve therapeutic efficacy and it can take several weeks for depressive symptoms to fully resolve (Frazer & Benmansour, Reference Frazer and Benmansour2002; Mitchell, Reference Mitchell2006). SSRI treatment is also associated with adverse effects which can compromise treatment adherence, including gastrointestinal symptoms, drowsiness, weight gain, sexual dysfunction and emotional blunting (Goethe, Woolley, Cardoni, Woznicki, & Piez, Reference Goethe, Woolley, Cardoni, Woznicki and Piez2007; Goodwin, Price, De Bodinat, & Laredo, Reference Goodwin, Price, De Bodinat and Laredo2017). Given that depression is now ranked as the single largest contributor to global disability (World Health Organisation, 2017), there is a pressing need for novel treatment approaches that are more effective, better tolerated and faster acting.

SSRIs selectively block serotonin (5-hydroxytryptamine, 5-HT) transporters, which results in an increase in 5-HT levels at all post-synaptic serotonin receptor subtypes. This indirect activation of a broad range of receptors is likely to contribute not only to the therapeutic action, but also to the unwanted aspects of SSRIs. In particular, early activation of 5-HT1A autoreceptors by SSRIs results in a paradoxical decrease in 5-HT cell firing, and is thought to contribute to the slow onset of the clinical action of SSRIs (Artigas, Reference Artigas2013). Equally, activation of post-synaptic 5-HT2C receptors has been associated with the increase in anxiety that can characterise initiation of SSRI treatment (Burghardt, Bush, McEwen, & LeDoux, Reference Burghardt, Bush, McEwen and LeDoux2007).

More selective activation of specific 5-HT receptors has the potential to produce better tolerated, faster acting treatments. Preclinical studies suggest that the 5-HT4 receptor is a particularly promising therapeutic target for this approach. In animal models, 5-HT4 receptor activation has antidepressant effects across a range of behavioural paradigms. Intriguingly, the onset of antidepressant effects following 5-HT4 receptor agonism is more rapid than that seen with SSRIs, and is accompanied by early molecular and cellular changes that mirror those seen following chronic administration of conventional antidepressants. This profile of effects suggests that 5-HT4 receptor agonists may hold potential as novel antidepressants, and may be useful adjuncts to SSRI treatment to increase the speed of onset of therapeutic effects. In addition, 5-HT4 receptor agonists produce pro-cognitive effects in tasks of learning and memory in rodents, highlighting a potential role in targeting cognitive deficits associated with depression, which are not well addressed by current antidepressant medications (Shilyansky et al., Reference Shilyansky, Williams, Gyurak, Harris, Usherwood and Etkin2016).

To date, the evidence that the 5-HT4 receptor might be a useful treatment target mainly comes from preclinical studies, and it is important that these findings are translated into human studies. Human PET imaging studies using the [11C]-SB 207145 ligand have provided some initial support for a role of the 5-HT4 receptor in the pathophysiology of depression (Madsen et al., Reference Madsen, Torstensen, Holst, Haahr, Knorr, Frokjaer and Knudsen2014). However, it remains to be established whether activation of 5-HT4 receptors in humans has effects that mirror the antidepressant and pro-cognitive effects seen in animals, and therefore whether this is a viable antidepressant target. Until recently, such experimental studies in humans have not possible due to a lack of suitable compounds. With the licensing of the 5-HT4 partial receptor agonist, prucalopride (Resolor), for the treatment of constipation, there is a timely opportunity to establish the effects of 5-HT4 activation on mood and cognition in humans, using an experimental medicine framework. Here, we review the evidence from animal studies that the 5-HT4 receptor is a promising target for the treatment of depression and cognitive disorders, and outline a potential pathway for the efficient translation of these effects into humans and, ultimately, to the clinic.

5-HT4 receptors are expressed in the basal ganglia and limbic system

Since its first description 30 years ago (Dumuis, Bouhelal, Sebben, Cory, & Bockaert, Reference Dumuis, Bouhelal, Sebben, Cory and Bockaert1988), the 5-HT4 receptor has been discovered both in the brain and in various sites in the periphery. Peripherally, 5-HT4 receptors are found in the heart (Kaumann, Reference Kaumann1990), the intestine (Craig & Clarke, Reference Craig and Clarke1990), the adrenal glands (Idres, Delarue, Lefebvre, & Vaudry, Reference Idres, Delarue, Lefebvre and Vaudry1991) and the bladder (Tonini & Candura, Reference Tonini and Candura1996). In the brain, 5-HT4 receptors are located post-synaptically and are expressed in the hypothalamus, hippocampus, basal ganglia, amygdala, olfactory bulb, septal area and neocortex (Cai, Flores-Hernandez, Feng, & Yan, Reference Cai, Flores-Hernandez, Feng and Yan2002; Compan et al., Reference Compan, Daszuta, Salin, Sebben, Bockaert and Dumuis1996; Roychowdhury, Haas, & Anderson, Reference Roychowdhury, Haas and Anderson1994; Vilaró, Cortés, & Mengod, Reference Vilaró, Cortés and Mengod2005; Waeber, Sebben, Nieoullon, Bockaert, & Dumuis, Reference Waeber, Sebben, Nieoullon, Bockaert and Dumuis1994). In rodents, 5-HT4 receptors are mainly situated on gamma-aminobutyric acid (GABA) neurones in the striatum and nucleus accumbens (Compan et al., Reference Compan, Daszuta, Salin, Sebben, Bockaert and Dumuis1996), glutamate pyramidal cells in the medial prefrontal cortex (PFC) and hippocampus (Cai et al., Reference Cai, Flores-Hernandez, Feng and Yan2002; Vilaró et al., Reference Vilaró, Cortés and Mengod2005) and hippocampal granule cells (Vilaró et al., Reference Vilaró, Cortés and Mengod2005). 5-HT4 receptors have also been described in the rat dorsal raphe nucleus (DRN) in autoradiographic studies (Vilaró et al., Reference Vilaró, Cortés and Mengod2005; Waeber et al., Reference Waeber, Sebben, Nieoullon, Bockaert and Dumuis1994), although 5-HT4 receptor mRNA has not been found suggesting that the 5-HT4 receptors are located on DRN afferent neurons rather than on DRN 5-HT cells (Vilaró et al., Reference Vilaró, Cortés and Mengod2005).

Evidence from PET studies in humans confirms that 5-HT4 receptors have a high density in the caudate, putamen and nucleus accumbens, with lower expression across a broad range of cortical and limbic areas, including the hippocampus and amygdala (Beliveau et al., Reference Beliveau, Ganz, Feng, Ozenne, Hojgaard, Fisher and Knudsen2017). Similarly, the highest levels of human 5-HT4 receptor mRNA have been found in the caudate and putamen, with lower levels in the amygdala temporal cortex and hippocampus (Medhurst, Lezoualc'h, Fischmeister, Middlemiss, & Sanger, Reference Medhurst, Lezoualc'h, Fischmeister, Middlemiss and Sanger2001). Post-mortem human brain studies have also confirmed a high density of the 5-HT4 receptor in the basal ganglia (caudate nucleus, putamen, nucleus accumbens, globus pallidus and substantia nigra), as well as the hippocampus (particularly in the CA1 region) and an intermediate density in the neocortex and amygdala (Bonaventure et al., Reference Bonaventure, Hall, Gommeren, Cras, Langlois, Jurzak and Leysen2000; Varnas, Halldin, Pike, & Hall, Reference Varnas, Halldin, Pike and Hall2003). In addition, low levels of 5-HT4 binding sites have been reported in the human thalamus and raphe nuclei (Beliveau et al., Reference Beliveau, Ganz, Feng, Ozenne, Hojgaard, Fisher and Knudsen2017; Varnas et al., Reference Varnas, Halldin, Pike and Hall2003).

The neurochemical and molecular effects of 5-HT4 receptor activation

5HT4 receptors are transmembrane-spanning G protein-coupled receptors, and their activation results in increased neuronal excitability that is mediated through the stimulation of adenylyl cyclase and an increase in cyclic adenosine monophosphate (Bockaert, Claeysen, Compan, & Dumuis, Reference Bockaert, Claeysen, Compan and Dumuis2008; Dumuis et al., Reference Dumuis, Bouhelal, Sebben, Cory and Bockaert1988). In addition, 5-HT4 receptor activation initiates important G protein-independent pathways, including the activation of tyrosine kinase Src and, in turn, neuronal extracellular-signal-regulated kinase (ERK) (Barthet et al., Reference Barthet, Framery, Gaven, Pellissier, Reiter, Claeysen and Dumuis2007), which is known to play a critical role in hippocampal synaptic plasticity (Norman, Thiels, Barrionuevo, & Klann, Reference Norman, Thiels, Barrionuevo and Klann2000).

5-HT4 receptors also indirectly affect neuronal function through their modulation of neurotransmitter release, including GABA (Bijak & Misgeld, Reference Bijak and Misgeld1997), acetylcholine (Consolo, Arnaboldi, Giorgi, Russi, & Ladinsky, Reference Consolo, Arnaboldi, Giorgi, Russi and Ladinsky1994; Johnson et al., Reference Johnson, Drummond, Grimwood, Sawant-Basak, Miller, Tseng and Siok2012; Siniscalchi, Badini, Beani, & Bianchi, Reference Siniscalchi, Badini, Beani and Bianchi1999), dopamine (Bockaert, Claeysen, Compan, & Dumuis, Reference Bockaert, Claeysen, Compan and Dumuis2004; Bockaert et al., Reference Bockaert, Claeysen, Compan and Dumuis2008; Bonhomme, De Deurwaerdere, Le Moal, & Spampinato, Reference Bonhomme, De Deurwaerdere, Le Moal and Spampinato1995), histamine (Johnson et al., Reference Johnson, Drummond, Grimwood, Sawant-Basak, Miller, Tseng and Siok2012) and serotonin (Ge & Barnes, Reference Ge and Barnes1996; Licht et al., Reference Licht, Kirkegaard, Zueger, Chourbaji, Gass, Aznar and Knudsen2010). Of particular interest is the enhanced release of acetylcholine in the frontal cortex and hippocampus following 5-HT4 receptor activation, which has been linked to its facilitatory role in memory and cognition (Hagena & Manahan-Vaughan, Reference Hagena and Manahan-Vaughan2017; King, Marsden, & Fone, Reference King, Marsden and Fone2008). In support of this 5-HT4 receptor agonists have been shown to increase electrically-evoked [3H]choline efflux in guinea pig brain slices in the cerebral cortex, hippocampus and nucleus basalis magnocellularis (NbM) (Siniscalchi et al., Reference Siniscalchi, Badini, Beani and Bianchi1999), and facilitate the release of ACh selectively in the frontal cortex (Consolo et al., Reference Consolo, Arnaboldi, Giorgi, Russi and Ladinsky1994). Importantly, both of these effects are blocked by 5-HT4 receptor antagonists, which further supports a specific role for the 5-HT4 receptor in cholinergic neurotransmission. Given that 5-HT4 receptors are not present on cholinergic basal forebrain neurons, it is thought that the enhancement of ACh release induced by 5-HT4 receptor agonists is likely to be mediated by GABAergic and/or glutamatergic cell populations (Penas-Cazorla & Vilaro, Reference Penas-Cazorla and Vilaro2015).

Importantly, 5-HT4 receptors in the PFC control the firing rate of midbrain serotonergic cells. Consistent with this, the basal firing of 5-HT cells in the DRN is reduced by over 50% in 5-HT4 receptor knockout mice compared with wildtypes (Conductier et al., Reference Conductier, Dusticier, Lucas, Cote, Debonnel, Daszuta and Compan2006). The facilitatory control exerted by 5-HT4 receptors on serotonergic firing is thought to operate via a positive PFC–DRN feedback loop in which the activation of postsynaptic 5-HT4 receptors located on medial PFC pyramidal cells has an excitatory effect on serotonin neurons in the DRN dorsal raphe, resulting in an increase in the release of serotonin in target structures such as the PFC and hippocampus. Consistent with this model, overexpression of 5-HT4 receptors in the mPFC, but not in the striatum or hippocampus, increases DRN 5-HT neuronal activity (Lucas et al., Reference Lucas, Compan, Charnay, Neve, Nestler, Bockaert and Debonnel2005). In addition, pharmacological studies have demonstrated that administration of the selective 5-HT4 receptor agonists, RS67333 and prucalopride, increases DRN 5-HT cell firing (Faye et al., Reference Faye, Hen, Guiard, Denny, Gardier, Mendez-David and David2019; Lucas et al., Reference Lucas, Compan, Charnay, Neve, Nestler, Bockaert and Debonnel2005; Lucas & Debonnel, Reference Lucas and Debonnel2002; Moha ou Maati et al., Reference Moha ou Maati, Bourcier-Lucas, Veyssiere, Kanzari, Heurteaux, Borsotto and Lucas2016). Intriguingly, this increase in firing can be seen within 30 min of drug administration, suggesting that 5-HT4 receptor agonists hold potential as rapid onset antidepressant agents (Lucas et al., Reference Lucas, Compan, Charnay, Neve, Nestler, Bockaert and Debonnel2005; Lucas & Debonnel, Reference Lucas and Debonnel2002). This is in stark contrast to the decrease in raphe cell firing that is characteristic of SSRIs, and which has been implicated in their delayed onset of action (Blier, Piñeyro, el Mansari, Bergeron, & de Montigny, Reference Blier, Piñeyro, el Mansari, Bergeron and de Montigny1998). Importantly, the excitatory effect of 5-HT4 receptor agonists on the firing of midbrain 5-HT cells is abolished by bilateral electrolytic lesions of the mPFC, demonstrating a causal role of the mPFC in the facilitation of 5-HT cell firing by 5-HT4 receptor agonists (Moha ou Maati et al., Reference Moha ou Maati, Bourcier-Lucas, Veyssiere, Kanzari, Heurteaux, Borsotto and Lucas2016).

Short-term 5-HT4 receptor activation has also been shown to increase hippocampal cell proliferation and the expression of neuroplasticity-related proteins such as CREB and BDNF (Ishizuka, Goshima, Ozawa, & Watanabe, Reference Ishizuka, Goshima, Ozawa and Watanabe2014; Lucas et al., Reference Lucas, Rymar, Du, Mnie-Filali, Bisgaard, Manta and Debonnel2007; Pascual-Brazo et al., Reference Pascual-Brazo, Castro, Díaz, Valdizán, Pilar-Cuéllar, Vidal and Pazos2012). These mirror those seen with chronic antidepressant administration (Malberg, Eisch, Nestler, & Duman, Reference Malberg, Eisch, Nestler and Duman2000; Warner-Schmidt & Duman, Reference Warner-Schmidt and Duman2006), but occur more rapidly. For example, 3 days administration of the 5-HT4 receptor agonist, RS67333, increases hippocampal cell proliferation in the dentate gyrus and the expression of neuroplasticity-related proteins, such as BDNF and CREB (Lucas et al., Reference Lucas, Rymar, Du, Mnie-Filali, Bisgaard, Manta and Debonnel2007; Pascual-Brazo et al., Reference Pascual-Brazo, Castro, Díaz, Valdizán, Pilar-Cuéllar, Vidal and Pazos2012). After 7 days of administration the level of these neuroplastic changes are comparable with those seen after 2–3 weeks of SSRI administration (Malberg et al., Reference Malberg, Eisch, Nestler and Duman2000; Pascual-Brazo et al., Reference Pascual-Brazo, Castro, Díaz, Valdizán, Pilar-Cuéllar, Vidal and Pazos2012; Samuels et al., Reference Samuels, Mendez-David, Faye, David, Pierz, Gardier and David2016). Further, long-term 5-HT4 receptor activation (4 weeks) facilitates the maturation of newborn neurons in the adult rat hippocampus (Mendez-David et al., Reference Mendez-David, David, Darcet, Wu, Kerdine-Römer, Gardier and Hen2014).

Furthermore, 5-HT4 receptors are involved in fine-tuning of the key cellular processes underlying synaptic plasticity, long-term potentiation (LTP) and long-term depression (LTD), which is likely to play a key role in their effects on long term hippocampal-dependent memory. Activation of the 5-HT4 receptor has been shown to prevent LTD in all the main hippocampal subfields (Kemp & Manahan-Vaughan, Reference Kemp and Manahan-Vaughan2005; Twarkowski, Hagena, & Manahan-Vaughan, Reference Twarkowski, Hagena and Manahan-Vaughan2016). The effect of 5-HT4 receptor activation on LTP is complex and specific to individual hippocampal subfields, which may relate to differences in the expression of different receptor isoforms (Hagena & Manahan-Vaughan, Reference Hagena and Manahan-Vaughan2017). A number of studies have demonstrated that stimulation of 5-HT4 receptors in freely behaving rats promotes synaptic information-encoding through LTP at both the CA1 region (Kemp & Manahan-Vaughan, Reference Kemp and Manahan-Vaughan2005) and dentate gyrus (Kulla & Manahan-Vaughan, Reference Kulla and Manahan-Vaughan2002; Twarkowski et al., Reference Twarkowski, Hagena and Manahan-Vaughan2016). Conversely, LTP at mossy fibre synapses of the CA3 region is suppressed by 5-HT4 receptor activation (Twarkowski et al., Reference Twarkowski, Hagena and Manahan-Vaughan2016). The effect of 5-HT4 receptor activation on LTP has also been shown to be time-dependent; for example, the long lasting sustainment of LTP (i.e. after 48 h) in the dentate gyrus can be impaired by 5-HT4 receptor stimulation (Twarkowski et al., Reference Twarkowski, Hagena and Manahan-Vaughan2016). Taken together, this pattern suggests that 5-HT4 receptor activation acts to promote information encoding through LTP in CA1 and dentate gyrus at the expense of information coding through LTD, which is likely to impact upon the content of information encoding (Hagena & Manahan-Vaughan, Reference Hagena and Manahan-Vaughan2017; Twarkowski et al., Reference Twarkowski, Hagena and Manahan-Vaughan2016). The effect of 5-HT4 receptor activation on dentate gyrus plasticity is particularly interesting in light of recent evidence that LTP induction in the dentate gyrus is sufficient to induce antidepressant-like effects in rat models within a rapid timeframe (Kanzari et al., Reference Kanzari, Bourcier-Lucas, Freyssin, Abrous, Haddjeri and Lucas2018), suggesting that the facilitatory effect of 5-HT4 receptor agonists on DG plasticity may, at least in part, account for their fast-acting antidepressant-like properties.

Preclinical studies support a role for the 5-HT4 receptor in the aetiology of depression and anxiety

Preclinical studies provide support for the involvement of the 5-HT4 receptor in the aetiology of depression. Changes in 5-HT4 receptor expression have been reported in a number of rodent models of depression, although the direction of effect varies by model. In the olfactory bulbectomy (OBX) and glucocorticoid receptor heterozygous [GR(±)] models, an increase in 5-HT4 receptor binding has been reported in the ventral hippocampus (OBX) and striatum [GR(±)] (Licht et al., Reference Licht, Kirkegaard, Zueger, Chourbaji, Gass, Aznar and Knudsen2010). In contrast, in the Flinders-sensitive line rat model of depression, a decrease in hippocampal 5-HT4 receptor binding was reported (Licht et al., Reference Licht, Marcussen, Wegener, Overstreet, Aznar and Knudsen2009). 5-HT4 receptor mRNA and protein has also been shown to be down-regulated in rats subjected to maternal deprivation (but not chronic unpredictable stress), with a strong correlation between levels of hippocampal 5-HT4 receptor mRNA and anhedonic-like behaviour in these animals (Bai et al., Reference Bai, Zhu, Zhang, Zhang, Zhang, Xue and Zhang2014).

5-HT4 receptor knock out mice do not show a clear depressed-like phenotype, with no differences in behaviour on the forced swim test, which may be due to the considerable adaptations in the serotoninergic system that occur across development in these animals (Amigo et al., Reference Amigo, Diaz, Pilar-Cuellar, Vidal, Martin, Compan and Castro2016; Conductier et al., Reference Conductier, Dusticier, Lucas, Cote, Debonnel, Daszuta and Compan2006). However, the knock out animals do show some evidence of anhedonic- and apathetic-like behaviour, with a reduction in sucrose consumption and nesting behaviour, and decreased central time in the open field (OF), which is suggestive of increased anxiety (Amigo et al., Reference Amigo, Diaz, Pilar-Cuellar, Vidal, Martin, Compan and Castro2016). Interestingly, a recent study suggests that focal overexpression of the 5-HT4 receptor in the medial PFC produces an ‘anti-depressed’ and anxiolytic behavioural phenotype, which is consistent with the central role ascribed to descending inputs from the mPFC to the DRN in mediating increased raphe firing (Castello et al., Reference Castello, LeFrancois, Flajolet, Greengard, Friedman and Rebholz2018).

5-HT4 receptors are involved in the mechanism of action of SSRIs

There is also convincing evidence that 5-HT4 receptors play a role in the action of SSRIs in animal models. Chronic administration of monoaminergic antidepressants, such as fluoxetine, paroxetine and venlafaxine (but not reboxetine), decreases 5-HT4 receptor density in the basal ganglia and hippocampus in the rat brain (Licht et al., Reference Licht, Marcussen, Wegener, Overstreet, Aznar and Knudsen2009; Vidal, Valdizan, Mostany, Pazos, & Castro, Reference Vidal, Valdizan, Mostany, Pazos and Castro2009; Vidal, Valdizan, Vilaró, Pazos, & Castro, Reference Vidal, Valdizan, Vilaró, Pazos and Castro2010). In contrast, expression of the 5-HT4 receptor has been shown to be upregulated after chronic fluoxetine administration in corticostriatal pyramidal cells expressing the adaptor protein p11, located in layer 5 of the cerebral cortex (Schmidt et al., Reference Schmidt, Warner-Schmidt, Otopalik, Pickett, Greengard and Heintz2012).

More direct evidence for a role for the 5-HT4 receptor in the action of conventional antidepressants comes from studies in which 5-HT4 receptor ligands have been shown to modify the effects of monoaminergic antidepressants. For example, administration of the 5-HT4 receptor agonist RS67333 has been shown to augment the acute effect of paroxetine on extracellular serotonin levels in the rat ventral hippocampus (Licht et al., Reference Licht, Kirkegaard, Zueger, Chourbaji, Gass, Aznar and Knudsen2010). Perhaps the most convincing evidence comes from studies which demonstrate that disrupting 5-HT4 receptor function can abolish the behavioural effects of SSRIs. For example, the 5-HT4 receptor antagonist GR125487 blocks the effects of fluoxetine on a range of depression- and anxiety-related paradigms in a corticosterone model (CORT) of anxiety/depression (Mendez-David et al., Reference Mendez-David, David, Darcet, Wu, Kerdine-Römer, Gardier and Hen2014), suggesting that 5-HT4 receptor activation is necessary for the antidepressant and anxiolytic effects of SSRIs (although see Cryan and Lucki, Reference Cryan and Lucki2000). A similar effect is found in 5-HT4 receptor knock out mice, where chronic fluoxetine administration has been shown to reverse the behavioural effects of OBX in wild type but not knock out animals (Amigo et al., Reference Amigo, Diaz, Pilar-Cuellar, Vidal, Martin, Compan and Castro2016).

Interestingly, there is also evidence to suggest that the 5-HT4 receptor contributes to the neurotrophic effects of chronic SSRI treatment. For example, the usual increase in cell proliferation in the dentate gyrus following chronic administration of fluoxetine is not present in 5-HT4 receptor knock out mice (Imoto et al., Reference Imoto, Kira, Sukeno, Nishitani, Nagayasu, Nakagawa and Segi-Nishida2015), and is partially blocked by 5-HT4 receptor antagonism (Mendez-David et al., Reference Mendez-David, David, Darcet, Wu, Kerdine-Römer, Gardier and Hen2014), suggesting that the neurogenic effects of SSRIs may be partly mediated by the 5-HT4 receptor. Further support for this comes from a study which demonstrates that the induction of immature like granule cells by chronic fluoxetine treatment (‘dematuration’) is blocked in 5-HT4 receptor knock-out mice (Kobayashi et al., Reference Kobayashi, Ikeda, Sakai, Yamasaki, Haneda, Miyakawa and Suzuki2010).

Short-term administration of 5-HT4 receptor agonists has antidepressant and anxiolytic-like effects in animals

Studies in rodents have demonstrated that 5-HT4 receptor agonists exert antidepressant-like effects on behavioural paradigms that mirror those seen with conventional SSRIs, but with a more rapid onset of action (Lucas et al., Reference Lucas, Rymar, Du, Mnie-Filali, Bisgaard, Manta and Debonnel2007; Mendez-David et al., Reference Mendez-David, David, Darcet, Wu, Kerdine-Römer, Gardier and Hen2014; Pascual-Brazo et al., Reference Pascual-Brazo, Castro, Díaz, Valdizán, Pilar-Cuéllar, Vidal and Pazos2012). Using the classic forced swim paradigm, the 5-HT4 receptor partial agonists RS67333 and prucalopride have been shown to reduce immobility following acute (Lucas et al., Reference Lucas, Rymar, Du, Mnie-Filali, Bisgaard, Manta and Debonnel2007) and 3–5 day administration (Gomez-Lazaro et al., Reference Gomez-Lazaro, Garmendia, Beitia, Perez-Tejada, Azpiroz and Arregi2012; Pascual-Brazo et al., Reference Pascual-Brazo, Castro, Díaz, Valdizán, Pilar-Cuéllar, Vidal and Pazos2012). These effects parallel those seen with acute SSRI administration, which also induces a reduction in immobility on this task, although the effect of the 5-HT4 agonists was almost double that seen with acute citalopram (Lucas et al., Reference Lucas, Rymar, Du, Mnie-Filali, Bisgaard, Manta and Debonnel2007).

Short-term administration of 5-HT4 agonists also induces behavioural changes in paradigms that require chronic administration of SSRIs to shift. For example, 3 days administration of RS67333 was sufficient to increase sucrose consumption in the chronic mild stress model of depression (Lucas et al., Reference Lucas, Rymar, Du, Mnie-Filali, Bisgaard, Manta and Debonnel2007) and in CORT rats (Pascual-Brazo et al., Reference Pascual-Brazo, Castro, Díaz, Valdizán, Pilar-Cuéllar, Vidal and Pazos2012), and decrease hyperlocomotion in the OBX model (Lucas et al., Reference Lucas, Rymar, Du, Mnie-Filali, Bisgaard, Manta and Debonnel2007). Within 7 days of RS67333 a complete reversal of the effects of corticosterone on sucrose consumption was seen, paralleling the effect of 21-day treatment with fluoxetine (Pascual-Brazo et al., Reference Pascual-Brazo, Castro, Díaz, Valdizán, Pilar-Cuéllar, Vidal and Pazos2012), and suggesting a rapid reversal of anhedonic behaviour following 5-HT4 receptor agonism. These effects are consistent with the neurobiological adaptations that have been reported following 3 days of 5-HT4 receptor agonism, including desensitisation of 5-HT1A autoreceptors, increased CREB phosphorylation and increased hippocampal cell proliferation (Lucas et al., Reference Lucas, Rymar, Du, Mnie-Filali, Bisgaard, Manta and Debonnel2007), and suggest that 5-HT4 receptor agonists have a unique, rapid-acting profile of effects.

Intriguingly, co-administration of 5-HT4 receptor agonists with SSRIs prevents the attenuation in raphe firing that is typically seen at the onset of SSRI administration (Lucas et al., Reference Lucas, Du, Romeas, Mnie-Filali, Haddjeri, Piñeyro and Debonnel2010), suggesting that 5-HT4 receptor agonists may have promise as an augmentation strategy for SSRI treatment. In support of this proposal, co-administration of RS67333 and the SSRI citalopram led to a synergistic inhibition of hippocampal firing, while the same combination potentiated the ability of RS67333 to phosphorylate CREB in the hippocampus and reduce immobility in the forced swim test (Lucas et al., Reference Lucas, Du, Romeas, Mnie-Filali, Haddjeri, Piñeyro and Debonnel2010).

5-HT4 receptor agonists have also been associated with rapid effects on anxiety. Seven days administration of RS67333 reduces latency to feed in the novelty suppressed feeding (NSF) paradigm, whereas 14 days administration of fluoxetine is required to see the same effect (Pascual-Brazo et al., Reference Pascual-Brazo, Castro, Díaz, Valdizán, Pilar-Cuéllar, Vidal and Pazos2012). A similar effect was seen in the mouse CORT model of anxiety/depression, where 7 day administration of RS67333, but not fluoxetine, reduced anxiety-related behaviour in the OF test and the elevated plus maze (EPM), although the effects of subchronic RS67333 on the NSF were not replicated in this model (Mendez-David et al., Reference Mendez-David, David, Darcet, Wu, Kerdine-Römer, Gardier and Hen2014). Interestingly, the effects of RS67333 on the OF and EPM tests were also evident in animals who had undergone hippocampal X-irradiation, suggesting that they are not dependent on hippocampal neurogenesis (Mendez-David et al., Reference Mendez-David, David, Darcet, Wu, Kerdine-Römer, Gardier and Hen2014).

Consistent with a fast onset of action, acute administration of RS67333 is sufficient to induce an anxiolytic-like effect on the elevated zero-maze (Bell, Duke, Gilmore, Page, & Begue, Reference Bell, Duke, Gilmore, Page and Begue2014), and reverse the anxiogenic effects of chronic cannabinoid exposure during adolescence (Abboussi et al., Reference Abboussi, Said, Fifel, Lakehayli, Tazi and El Ganouni2016). A recent report confirmed an anxiolytic-like effect of acute systemic infusion of RS67333 on the EPM, the OF test and the NSF test (Faye et al., Reference Faye, Hen, Guiard, Denny, Gardier, Mendez-David and David2019). Intriguingly, a similar profile was seen when RS67333 was directly infused into the mPFC, and the anxiolytic-like effects of RS67333 were attenuated by optogenetic inhibition of the mPFC terminals in the DRN, suggesting that direct activation of 5-HT4 receptors in the mPFC is responsible for the fast anxiolytic effects of 5-HT4 receptor agonists, via the mPFC-DRN circuit (Faye et al., Reference Faye, Hen, Guiard, Denny, Gardier, Mendez-David and David2019).

The effects of 5HT4 receptor antagonism on animal depression- or anxiety-like behaviour are less well established. Acute administration of the 5-HT4 receptor antagonists SDZ205-557, GR113808 and SB204070 does not influence anxiety-related behaviour in the light/dark choice test in mice (Costall & Naylor, Reference Costall and Naylor1997). Four weeks administration of the antagonist GR125487 has also been shown not to have an effect on behavioural models of anxiety in a corticosterone model of depression/anxiety, although the antagonist did block the effects of an SSRI in this model (Mendez-David et al., Reference Mendez-David, David, Darcet, Wu, Kerdine-Römer, Gardier and Hen2014). In contrast, two studies have reported an anxiolytic effect of acute administration of SB204070, GR113808 (Silvestre, Fernandez, & Palacios, Reference Silvestre, Fernandez and Palacios1996) and SB207266A (Kennett, Bright, Trail, Blackburn, & Sanger, Reference Kennett, Bright, Trail, Blackburn and Sanger1997; Silvestre et al., Reference Silvestre, Fernandez and Palacios1996) on the EPM test. It is unclear what explains these contradictory findings, although there are differences in dosage, timing of antagonist administration and behavioural outcome measures used in the studies, which may be important.

5-HT4 Receptor activation has a facilitatory effect on rodent tests of learning and memory

A large body of evidence demonstrates a facilitatory effect of 5-HT4 receptor activation on rodent and primate behavioural tests of cognition, and in particular learning and memory (Hagena & Manahan-Vaughan, Reference Hagena and Manahan-Vaughan2017; King et al., Reference King, Marsden and Fone2008). Early studies using relatively non-selective compounds, such as the mixed 5-HT4 receptor agonist/5-HT3 receptor antagonist BIMU1 demonstrated a facilitation of autoshaping (Meneses & Hong, Reference Meneses and Hong1997) and associative memory (Marchetti-Gauthier, Roman, Dumuis, Bockaert, & Soumireu-Mourat, Reference Marchetti-Gauthier, Roman, Dumuis, Bockaert and Soumireu-Mourat1997), which was attributed to 5-HT4 receptor activation since both effects were blocked by co-administration of a 5-HT4 receptor antagonist. Subsequent studies using more selective agonists confirmed that acute 5-HT4 receptor activation improves a wide of range of behavioural tests of learning and memory, including place recognition (Lamirault & Simon, Reference Lamirault and Simon2001), object recognition (Lamirault & Simon, Reference Lamirault and Simon2001; Levallet, Hotte, Boulouard, & Dauphin, Reference Levallet, Hotte, Boulouard and Dauphin2009; Moser et al., Reference Moser, Bergis, Jegham, Lochead, Duconseille, Terranova and Scatton2002), delayed matching-to-sample (Terry et al., Reference Terry, Buccafusco, Jackson, Prendergast, Fontana, Wong and Eglen1998), Morris water maze (Lelong, Dauphin, & Boulouard, Reference Lelong, Dauphin and Boulouard2001), delayed spontaneous alternation (Mohler et al., Reference Mohler, Shacham, Noiman, Lezoualc'h, Robert, Gastineau and Ragozzino2007) and olfactory association learning (Marchetti et al., Reference Marchetti, Chaillan, Dumuis, Bockaert, Soumireu-Mourat and Roman2004; Marchetti et al., Reference Marchetti, Jacquet, Escoffier, Miglioratti, Dumuis, Bockaert and Roman2011) tasks. The positive effect of 5-HT4 receptor agonism on object recognition is also seen after chronic (14 day) activation of the receptor (Quiedeville et al., Reference Quiedeville, Boulouard, Hamidouche, Da Silva Costa-Aze, Nee, Rochais and Bouet2015). Critically, 5-HT4 receptor antagonism has consistently been shown to block these agonist-induced pro-cognitive effects, confirming a specific role for the 5-HT4 receptor in learning and memory (Fontana, Daniels, Wong, Clark, & Eglen, Reference Fontana, Daniels, Wong, Clark and Eglen1997; Lamirault & Simon, Reference Lamirault and Simon2001; Mohler et al., Reference Mohler, Shacham, Noiman, Lezoualc'h, Robert, Gastineau and Ragozzino2007; Moser et al., Reference Moser, Bergis, Jegham, Lochead, Duconseille, Terranova and Scatton2002). Whilst 5-HT4 receptor activation has particularly strong effects on hippocampal-dependent learning and memory, it also facilitates other forms of cognition, including attention (as measured by the five choice serial reaction time task, Hille, Bate, Davis, and Gonzalez, Reference Hille, Bate, Davis and Gonzalez2008) and social memory (Letty et al., Reference Letty, Child, Dumuis, Pantaloni, Bockaert and Rondouin1997).

The timing of 5-HT4 receptor activation seems to be critical to the profile of effects on memory processes, with a particularly clear role for the receptor in learning and memory acquisition (Hagena & Manahan-Vaughan, Reference Hagena and Manahan-Vaughan2017). Consistent with this, studies in which an agonist is administered prior to training have typically shown a facilitatory effect of 5-HT4 receptor activation (Lamirault & Simon, Reference Lamirault and Simon2001; Meneses & Hong, Reference Meneses and Hong1997; Orsetti, Dellarole, Ferri, & Ghi, Reference Orsetti, Dellarole, Ferri and Ghi2003), whereas the effect of post-training agonist administration is less clear, with reports of both enhanced (Lamirault & Simon, Reference Lamirault and Simon2001; Orsetti et al., Reference Orsetti, Dellarole, Ferri and Ghi2003) and impaired (Meneses & Hong, Reference Meneses and Hong1997; Nasehi, Tabatabaie, Khakpai, & Zarrindast, Reference Nasehi, Tabatabaie, Khakpai and Zarrindast2015) memory consolidation.

Interestingly, the cognition enhancing effects of 5-HT4 receptor activation have also been demonstrated in the chronic corticosterone model of anxiety/depression (Darcet, Gardier, David, & Guilloux, Reference Darcet, Gardier, David and Guilloux2016), suggesting that this may be a promising strategy for treating the cognitive impairments associated with stress-related disorders. Chronic administration of the 5-HT4 receptor agonist RS67333 restored corticosterone-induced deficits on a broad range of tests of learning and memory, in contrast to chronic fluoxetine, which had a much narrower profile of effects. The 5-HT4 receptor agonist RS67333 has also been shown to restore emotional memory performance in a passive avoidance test in the Flinders sensitive line rat model of depression (Eriksson et al., Reference Eriksson, Delagrange, Spedding, Popoli, Mathe, Ogren and Svenningsson2012). Together these findings suggest that 5-HT4 receptor agonists may be a potential target for the improvement of the cognitive and emotional processing deficits associated with depression.

In contrast to the clear pro-cognitive effects of 5-HT4 receptor activation, the effect of 5-HT4 receptor antagonism is less well characterised and findings are mixed. Whilst the antagonists SDZ205557 and RS67532 have been shown to impair olfactory association memory formation and passive memory formation (Galeotti, Ghelardini, & Bartolini, Reference Galeotti, Ghelardini and Bartolini1998; Marchetti, Dumuis, Bockaert, Soumireu-Mourat, & Roman, Reference Marchetti, Dumuis, Bockaert, Soumireu-Mourat and Roman2000), other studies report no effect of 5-HT4 receptor antagonism on memory processes (Moser et al., Reference Moser, Bergis, Jegham, Lochead, Duconseille, Terranova and Scatton2002; Orsetti et al., Reference Orsetti, Dellarole, Ferri and Ghi2003). It has been suggested that this may be due to a low level of behaviourally-mediated exogenous serotonin release, which would lessen the effects of receptor blockade compared with receptor activation (Orsetti et al., Reference Orsetti, Dellarole, Ferri and Ghi2003). Likewise, 5-HT4 receptor knock-out mice do not have clear behavioural deficits in learning and memory (Segu et al., Reference Segu, Lecomte, Wolff, Santamaria, Hen, Dumuis and Compan2010). However, these animals do show an impairment in long-term spatial memory on the Morris water maze following scopolamine administration at a dose ineffective in wild type controls, suggesting that developmental adaptations in the cholinergic system may compensate for the absence of 5-HT4 receptors under baseline conditions (Segu et al., Reference Segu, Lecomte, Wolff, Santamaria, Hen, Dumuis and Compan2010).

The memory enhancing properties of 5-HT4 receptor activation are often attributed to a facilitation of central cholinergic activity. This is supported by studies demonstrating that 5-HT4 receptor agonists enhance the brain ACh output under both baseline conditions (Consolo et al., Reference Consolo, Arnaboldi, Giorgi, Russi and Ladinsky1994) and during mnemonic demands (Mohler et al., Reference Mohler, Shacham, Noiman, Lezoualc'h, Robert, Gastineau and Ragozzino2007), and that 5-HT4 receptor agonists reverse the cognitive impairments induced by muscarinic antagonists (Cachard-Chastel et al., Reference Cachard-Chastel, Devers, Sicsic, Langlois, Lezoualc'h, Gardier and Belzung2008; Fontana et al., Reference Fontana, Daniels, Wong, Clark and Eglen1997; Lo et al., Reference Lo, De Maeyer, Vermaercke, Callaerts-Vegh, Schuurkes and D'Hooge2014; Marchetti-Gauthier et al., Reference Marchetti-Gauthier, Roman, Dumuis, Bockaert and Soumireu-Mourat1997; Moser et al., Reference Moser, Bergis, Jegham, Lochead, Duconseille, Terranova and Scatton2002). For example, acute 5-HT4 receptor activation reverses the impairments in spatial learning and memory that are induced by atropine and scopolamine on the Morris water maze (Fontana et al., Reference Fontana, Daniels, Wong, Clark and Eglen1997; Lo et al., Reference Lo, De Maeyer, Vermaercke, Callaerts-Vegh, Schuurkes and D'Hooge2014) and the deficits in passive avoidance behaviour that are induced by scopolamine on the passive avoidance test (Galeotti et al., Reference Galeotti, Ghelardini and Bartolini1998; Lo et al., Reference Lo, De Maeyer, Vermaercke, Callaerts-Vegh, Schuurkes and D'Hooge2014). More specifically, local infusion of the 5-HT4 receptor agonist RS67333 into the NbM enhances the acquisition and consolidation of place recognition memory, which has been suggested to be due to a potentiation of the activity of the cholinergic NbM-cortical pathways (Orsetti et al., Reference Orsetti, Dellarole, Ferri and Ghi2003; although see Penas-Cazorla & Vilaro, Reference Penas-Cazorla and Vilaro2015). Further support for the idea that the memory-enhancing effects of 5-HT4 receptor activation are mediated via an interaction with cholinergic systems comes from studies that demonstrate that acetylcholinesterase inhibitors (AChEI) potentiate the effects of 5-HT4 agonists on cognition (Cachard-Chastel et al., Reference Cachard-Chastel, Lezoualc'h, Dewachter, Deloménie, Croes, Devijver and Gardier2007; Lamirault, Guillou, Thal, & Simon, Reference Lamirault, Guillou, Thal and Simon2003; Mohler et al., Reference Mohler, Shacham, Noiman, Lezoualc'h, Robert, Gastineau and Ragozzino2007; Moser et al., Reference Moser, Bergis, Jegham, Lochead, Duconseille, Terranova and Scatton2002). For example, sub-efficacious doses of the 5-HT4 receptor agonist prucalopride and the AChEI donepezil reverse scopolamine-induced deficits in spatial learning and memory when given in combination (Cachard-Chastel et al., Reference Cachard-Chastel, Lezoualc'h, Dewachter, Deloménie, Croes, Devijver and Gardier2007).

Establishing the effects of 5-HT4 receptor activation in humans

Taken together, this preclinical evidence suggests that the 5-HT4 receptor may be a promising target for the treatment of depression and cognitive disorder in humans. However, there have been relatively few studies in humans to establish the translation of these effects, and to characterise the profile of 5-HT4 receptor activation on human cognition and emotion.

There is some initial evidence to support a role for the 5-HT4 receptor in the neurobiology of depression. Healthy individuals with a family history of depression have been shown to have lower 5-HT4 receptor binding in the striatum, and this reduction is most pronounced in people with more than one first degree relative with a history of depression (Madsen et al., Reference Madsen, Torstensen, Holst, Haahr, Knorr, Frokjaer and Knudsen2014). Conversely, an increase in the density and functionality of 5-HT4 receptors in cortical and striatal areas has been reported in an analysis of post-mortem brains from depressed violent suicide victims (Rosel et al., Reference Rosel, Arranz, Urretavizcaya, Oros, San and Navarro2004). Polymorphisms in the splice variant region of the gene encoding the 5-HT4 receptor have been reported to be associated with unipolar depression (Ohtsuki et al., Reference Ohtsuki, Ishiguro, Detera-Wadleigh, Toyota, Shimizu, Yamada and Arinami2002), although this was not confirmed in a recent genome wide association study (Wray et al., Reference Wray, Ripke, Mattheisen, Trzaskowski, Byrne, Abdellaoui and Consortium2018). An involvement of the 5-HT4 receptor in human memory is also supported by a PET study that reported an inverse correlation of 5-HT4 receptor binding in the hippocampus and performance on the Auditory Verbal Learning Task (Haahr et al., Reference Haahr, Fisher, Holst, Madsen, Jensen, Marner and Hasselbalch2013). Interestingly, the direction of this effect is opposite to the memory enhancing effects of 5-HT4 receptor agonism seen in preclinical studies, which may be due to the binding potential representing a composite measure of receptor density and affinity and/or that lower 5-HT4 receptor binding could also be reflective of higher chronic endogenous serotonin levels (Haahr et al., Reference Haahr, Fisher, Jensen, Frokjaer, Mahon, Madsen and Knudsen2014).

These divergent correlational findings highlight the importance of further investigation in humans using an experimental medicine approach. Establishing the cognitive effects of compounds targeting the 5-HT4 receptor is particularly important in light of the limited predictive validity of animal models and the disappointing failure to predict the efficacy of pro-cognitive and antidepressant drugs in humans from drug discovery studies in rodents (Hyman, Reference Hyman2012; Millan et al., Reference Millan, Agid, Brune, Bullmore, Carter, Clayton and Young2012). We have previously demonstrated that acute administration of clinically active antidepressants in healthy volunteers and depressed patients produces changes in information processing that can be measured using behavioural and neuroimaging paradigms (Godlewska & Harmer, Reference Godlewska and Harmer2020). Importantly, these changes are predictive of later therapeutic effects and these measures can therefore be used as surrogate markers of clinical effects to screen and understand the mechanisms of candidate agents early in development. Testing the effects of compounds targeting the 5HT4 receptor in humans using this approach could help clarify the role of the 5HT4 system in cognition and emotion and translate the potentially promising results from preclinical models to clinical application.

Experimental medicine often takes a stepwise approach, first testing the effects of the compound in healthy participants to gain information on relevant dosing, duration of treatment and specific cognitive and emotional processes which are sensitive to this manipulation. This information can then be used to guide the most appropriate design for testing in clinical groups including those with depression, anxiety and cognitive impairment. Again, experimental medicine markers (such as measures of learning or emotional processing) can be useful prior to clinical testing of efficacy (symptom improvement) to characterise likely mechanisms, target and patient groups. Such a stepwise approach may improve decision making about a compound's development and reduce the high failure rate associated with drug development in this area (Wong, Siah, & Lo, Reference Wong, Siah and Lo2019).

Until recently, further elucidation of the role of the 5-HT4 receptor in humans has been difficult because of the lack of suitable compounds to manipulate 5-HT4 receptor function. The development of 5-HT4 receptor agonists for use as antidepressants in humans is complicated by the important role played by the 5-HT4 receptor in the periphery, in particular in the gastrointestinal tract and the heart, and the potential for peripheral side effects. Indeed, two 5-HT4 agonists, cisapride and tegaserod, that were developed for the treatment of gastrointestinal problems have both been restricted in use or withdrawn due to concerns about cardiac side effects. However, the particular concern with these drugs was risk of ventricular arrhythmias, which is probably due to their interaction with the human ether-a-go-go related gene potassium channel (De Maeyer, Lefebvre, & Schuurkes, Reference De Maeyer, Lefebvre and Schuurkes2008). Newer, more selective partial agonists, such as prucalopride which is licensed for the treatment of constipation, have good cardiac safety profiles. Prucalopride has good brain penetration (Johnson et al., Reference Johnson, Drummond, Grimwood, Sawant-Basak, Miller, Tseng and Siok2012), has pro-cognitive and antidepressant-like effects in animal models (Cachard-Chastel et al., Reference Cachard-Chastel, Lezoualc'h, Dewachter, Deloménie, Croes, Devijver and Gardier2007; Lucas et al., Reference Lucas, Rymar, Du, Mnie-Filali, Bisgaard, Manta and Debonnel2007) and therefore offers a unique opportunity to investigate the role of the 5-HT4 receptor in human cognition.

We recently explored the effect of a single dose of prucalopride in healthy volunteers and found consistent pro-cognitive effects across measures of learning memory including declarative memory, reward learning and emotional encoding (Murphy, Wright, Browning, Cowen, & Harmer, Reference Murphy, Wright, Browning, Cowen and Harmer2019). These findings replicate the findings in preclinical studies and support further clinical assessment of these compounds in depression and for disorders involving memory impairment. In line with data in animal models that 5HT4 agonists may be beneficial in depression, and also in combination with an SSRI, we are currently exploring the effects of a 5HT4 partial agonist (PF-04995274) on emotional and cognitive function in unmedicated depressed patients (NCT03516604) and patients who have not showed a sufficient response to current SSRI treatment (NCT03515733). This kind of experimental medicine approach can provide early proof-of-concept evidence to support larger clinical trials, and may have value in selecting the best treatment, for the right subgroup of depressed patients, prior to full randomised clinical trials of efficacy.

Conclusions

There has been much interest in the psychotropic effects of 5-HT4 receptor activation, stimulated by consistent evidence that 5-HT4 receptor agonists have antidepressant-like and pro-cognitive effects on a range of animal models. However, the translation of these effects to humans has been slow, and, to date, no 5-HT4 receptor agonists have been licensed for any indication other than gastrointestinal disorders. Whilst there had been some concern about the safety of 5-HT4 receptor agonists, due to this receptor's important roles outside of the central nervous system (for example in the heart), prucalopride has proved to be a safe approach to the treatment of chronic constipation. Experimental medicine offers a cost-effective way to efficiently translate these effects into humans and explore the possibility that 5-HT4 receptor activation might be a useful adjunct to antidepressant treatment, both to speed the onset of clinical antidepressant effects and to target the cognitive symptoms that are not well addressed by current treatments.

Acknowledgements

The study was funded by the Medical Research Council and supported by the NIHR Oxford Health Biomedical Research Centre and NIHR Oxford Cognitive Health Clinical Research Facility. The views expressed are those of the authors and not necessarily those of the National Health Service (NHS), the NIHR or the Department of Health. The research materials supporting this publication can be accessed by contacting the corresponding author.

Disclosures

CJH has received consultancy fees from P1vital Ltd., Janssen Pharmaceuticals, Sage Therapeutics, Pfizer and Lundbeck. SEM has received consultancy fees from P1vital Ltd. and Janssen Pharmaceuticals. CJH and SEM hold grant income from UCB Pharma and Janssen Pharmaceuticals. CJH, SEM and PJC hold grant income from a collaborative research project with Pfizer. ANdC is funded by a Wellcome Trust Clinical Doctoral Research Fellowship.

References

Abboussi, O., Said, N., Fifel, K., Lakehayli, S., Tazi, A., & El Ganouni, S. (2016). Behavioral effects of D3 receptor inhibition and 5-HT4 receptor activation on animals undergoing chronic cannabinoid exposure during adolescence. Metabolic Brain Disease, 31(2), 321327. doi:10.1007/s11011-015-9753-2.CrossRefGoogle ScholarPubMed
Amigo, J., Diaz, A., Pilar-Cuellar, F., Vidal, R., Martin, A., Compan, V., … Castro, E. (2016). The absence of 5-HT4 receptors modulates depression- and anxiety-like responses and influences the response of fluoxetine in olfactory bulbectomised mice: Adaptive changes in hippocampal neuroplasticity markers and 5-HT1A autoreceptor. Neuropharmacology, 111, 4758. doi:10.1016/j.neuropharm.2016.08.037.CrossRefGoogle Scholar
Artigas, F. (2013). Serotonin receptors involved in antidepressant effects. Pharmacology and Therapeutics, 137(1), 119131. doi:10.1016/j.pharmthera.2012.09.006.CrossRefGoogle ScholarPubMed
Bai, M., Zhu, X. Z., Zhang, Y., Zhang, S., Zhang, L., Xue, L., … Zhang, X. (2014). Anhedonia was associated with the dysregulation of hippocampal HTR4 and microRNA Let-7a in rats. Physiology and Behavior, 129, 135141. doi:10.1016/j.physbeh.2014.02.035.CrossRefGoogle ScholarPubMed
Barthet, G., Framery, B., Gaven, F., Pellissier, L., Reiter, E., Claeysen, S., … Dumuis, A. (2007). 5-hydroxytryptamine 4 receptor activation of the extracellular signal-regulated kinase pathway depends on Src activation but not on G protein or beta-arrestin signaling. Molecular Biology of the Cell, 18(6), 19791991. doi:10.1091/mbc.e06-12-1080.CrossRefGoogle ScholarPubMed
Beliveau, V., Ganz, M., Feng, L., Ozenne, B., Hojgaard, L., Fisher, P. M., … Knudsen, G. M. (2017). A high-resolution in vivo atlas of the human brain's serotonin system. Journal of Neuroscience, 37(1), 120128. doi:10.1523/jneurosci.2830-16.2016.Google ScholarPubMed
Bell, R., Duke, A. A., Gilmore, P. E., Page, D., & Begue, L. (2014). Anxiolytic-like effects observed in rats exposed to the elevated zero-maze following treatment with 5-HT2/5-HT3/5-HT4 ligands. Scientific Reports, 4, 3881. doi:10.1038/srep03881.CrossRefGoogle Scholar
Bijak, M., & Misgeld, U. (1997). Effects of serotonin through serotonin1A and serotonin4 receptors on inhibition in the Guinea-pig dentate gyrus in vitro. Neuroscience, 78(4), 10171026. doi:10.1016/s0306-4522(96)00666-5.CrossRefGoogle ScholarPubMed
Blier, P., Piñeyro, G., el Mansari, M., Bergeron, R., & de Montigny, C. (1998). Role of somatodendritic 5-HT autoreceptors in modulating 5-HT neurotransmission. Annals of the New York Academy of Sciences, 861, 204216.10.1111/j.1749-6632.1998.tb10192.xCrossRefGoogle ScholarPubMed
Bockaert, J., Claeysen, S., Compan, V., & Dumuis, A. (2004). 5-HT4 receptors. Current Drug Targets: CNS and Neurological Disorders, 3(1), 3951.Google ScholarPubMed
Bockaert, J., Claeysen, S., Compan, V., & Dumuis, A. (2008). 5-HT(4) receptors: History, molecular pharmacology and brain functions. Neuropharmacology, 55(6), 922931. doi:10.1016/j.neuropharm.2008.05.013.CrossRefGoogle ScholarPubMed
Bonaventure, P., Hall, H., Gommeren, W., Cras, P., Langlois, X., Jurzak, M., & Leysen, J. E. (2000). Mapping of serotonin 5-HT(4) receptor mRNA and ligand binding sites in the post-mortem human brain. Synapse (New York, N.Y.), 36(1), 3546. doi:10.1002/(sici)1098-2396(200004)36:1<35::aid-syn4>3.0.co;2-y.3.0.CO;2-Y>CrossRefGoogle ScholarPubMed
Bonhomme, N., De Deurwaerdere, P., Le Moal, M., & Spampinato, U. (1995). Evidence for 5-HT4 receptor subtype involvement in the enhancement of striatal dopamine release induced by serotonin: A microdialysis study in the halothane-anesthetized rat. Neuropharmacology, 34(3), 269279. doi:10.1016/0028-3908(94)00145-i.CrossRefGoogle ScholarPubMed
Burghardt, N. S., Bush, D. E., McEwen, B. S., & LeDoux, J. E. (2007). Acute selective serotonin reuptake inhibitors increase conditioned fear expression: Blockade with a 5-HT(2C) receptor antagonist. Biological Psychiatry, 62(10), 11111118. doi:10.1016/j.biopsych.2006.11.023.CrossRefGoogle ScholarPubMed
Cachard-Chastel, M., Devers, S., Sicsic, S., Langlois, M., Lezoualc'h, F., Gardier, A. M., & Belzung, C. (2008). Prucalopride and donepezil act synergistically to reverse scopolamine-induced memory deficit in C57Bl/6j mice. Behavioural Brain Research, 187(2), 455461. doi:10.1016/j.bbr.2007.10.008.CrossRefGoogle ScholarPubMed
Cachard-Chastel, M., Lezoualc'h, F., Dewachter, I., Deloménie, C., Croes, S., Devijver, H., … Gardier, A. M. (2007). 5-HT4 receptor agonists increase sAPPalpha levels in the cortex and hippocampus of male C57BL/6j mice. British Journal of Pharmacology, 150(7), 883892. doi:10.1038/sj.bjp.0707178.CrossRefGoogle ScholarPubMed
Cai, X., Flores-Hernandez, J., Feng, J., & Yan, Z. (2002). Activity-dependent bidirectional regulation of GABA(A) receptor channels by the 5-HT(4) receptor-mediated signalling in rat prefrontal cortical pyramidal neurons. Journal of Physiology, 540(Pt 3), 743759.10.1113/jphysiol.2001.013391CrossRefGoogle Scholar
Castello, J., LeFrancois, B., Flajolet, M., Greengard, P., Friedman, E., & Rebholz, H. (2018). CK2 regulates 5-HT4 receptor signaling and modulates depressive-like behavior. Molecular Psychiatry, 23(4), 872882. doi:10.1038/mp.2017.240.CrossRefGoogle ScholarPubMed
Cipriani, A., Furukawa, T. A., Salanti, G., Chaimani, A., Atkinson, L. Z., Ogawa, Y., … Geddes, J. R. (2018). Comparative efficacy and acceptability of 21 antidepressant drugs for the acute treatment of adults with major depressive disorder: A systematic review and network meta-analysis. Lancet (London, England), 391(10128), 13571366. doi:10.1016/S0140-6736(17)32802-7.CrossRefGoogle ScholarPubMed
Compan, V., Daszuta, A., Salin, P., Sebben, M., Bockaert, J., & Dumuis, A. (1996). Lesion study of the distribution of serotonin 5-HT4 receptors in rat basal ganglia and hippocampus. European Journal of Neuroscience, 8(12), 25912598.10.1111/j.1460-9568.1996.tb01553.xCrossRefGoogle ScholarPubMed
Conductier, G., Dusticier, N., Lucas, G., Cote, F., Debonnel, G., Daszuta, A., … Compan, V. (2006). Adaptive changes in serotonin neurons of the raphe nuclei in 5-HT(4) receptor knock-out mouse. European Journal of Neuroscience, 24(4), 10531062. doi:10.1111/j.1460-9568.2006.04943.x.CrossRefGoogle ScholarPubMed
Consolo, S., Arnaboldi, S., Giorgi, S., Russi, G., & Ladinsky, H. (1994). 5-HT4 Receptor stimulation facilitates acetylcholine release in rat frontal cortex. Neuroreport, 5(10), 12301232.CrossRefGoogle ScholarPubMed
Costall, B., & Naylor, R. J. (1997). The influence of 5-HT2 and 5-HT4 receptor antagonists to modify drug induced disinhibitory effects in the mouse light/dark test. British Journal of Pharmacology, 122(6), 11051118. doi:10.1038/sj.bjp.0701513.CrossRefGoogle ScholarPubMed
Craig, D. A., & Clarke, D. E. (1990). Pharmacological characterization of a neuronal receptor for 5-hydroxytryptamine in Guinea pig ileum with properties similar to the 5-hydroxytryptamine receptor. Journal of Pharmacology and Experimental Therapeutics, 252(3), 13781386.Google Scholar
Cryan, J. F., & Lucki, I. (2000). 5-HT4 receptors do not mediate the antidepressant-like behavioral effects of fluoxetine in a modified forced swim test. European Journal of Pharmacology, 409(3), 295299. doi:10.1016/s0014-2999(00)00858-x.CrossRefGoogle ScholarPubMed
Darcet, F., Gardier, A. M., David, D. J., & Guilloux, J. P. (2016). Chronic 5-HT4 receptor agonist treatment restores learning and memory deficits in a neuroendocrine mouse model of anxiety/depression. Neuroscience Letters, 616, 197203. doi:10.1016/j.neulet.2016.01.055.CrossRefGoogle Scholar
De Maeyer, J. H., Lefebvre, R. A., & Schuurkes, J. A. (2008). 5-HT4 Receptor agonists: Similar but not the same. Neurogastroenterology and Motility, 20(2), 99112. doi:10.1111/j.1365-2982.2007.01059.x.CrossRefGoogle Scholar
Dumuis, A., Bouhelal, R., Sebben, M., Cory, R., & Bockaert, J. (1988). A nonclassical 5-hydroxytryptamine receptor positively coupled with adenylate cyclase in the central nervous system. Molecular Pharmacology, 34(6), 880887.Google ScholarPubMed
Eriksson, T. M., Delagrange, P., Spedding, M., Popoli, M., Mathe, A. A., Ogren, S. O., & Svenningsson, P. (2012). Emotional memory impairments in a genetic rat model of depression: Involvement of 5-HT/MEK/Arc signaling in restoration. Molecular Psychiatry, 17(2), 173184. doi:10.1038/mp.2010.131.CrossRefGoogle Scholar
Faye, C., Hen, R., Guiard, B., Denny, C., Gardier, A., Mendez-David, I., & David, D. (2019). Rapid anxiolytic effects of RS67333, a serotonin type 4 receptor agonist, and diazepam, a benzodiazepine, are mediated by projections from the prefrontal cortex to the dorsal raphe nucleus. Biological Psychiatry 87(6):514-525. doi:https://doi.org/10.1016/j.biopsych.2019.08.009.Google ScholarPubMed
Fontana, D. J., Daniels, S. E., Wong, E. H., Clark, R. D., & Eglen, R. M. (1997). The effects of novel, selective 5-hydroxytryptamine (5-HT)4 receptor ligands in rat spatial navigation. Neuropharmacology, 36(4-5), 689696.CrossRefGoogle ScholarPubMed
Frazer, A., & Benmansour, S. (2002). Delayed pharmacological effects of antidepressants. Molecular Psychiatry, 7 (Suppl 1), S23S28. doi:10.1038/sj.mp.4001015.CrossRefGoogle ScholarPubMed
Galeotti, N., Ghelardini, C., & Bartolini, A. (1998). Role of 5-HT4 receptors in the mouse passive avoidance test. Journal of Pharmacology and Experimental Therapeutics, 286(3), 11151121.Google ScholarPubMed
Ge, J., & Barnes, N. M. (1996). 5-HT4 receptor-mediated modulation of 5-HT release in the rat hippocampus in vivo. British Journal of Pharmacology, 117(7), 14751480. doi:10.1111/j.1476-5381.1996.tb15309.x.CrossRefGoogle ScholarPubMed
Godlewska, B. R., & Harmer, C. J. (2020). Cognitive neuropsychological theory of antidepressant action: A modern-day approach to depression and its treatment. Psychopharmacology. doi:10.1007/s00213-019-05448-0.Google Scholar
Goethe, J. W., Woolley, S. B., Cardoni, A. A., Woznicki, B. A., & Piez, D. A. (2007). Selective serotonin reuptake inhibitor discontinuation: Side effects and other factors that influence medication adherence. Journal of Clinical Psychopharmacology, 27(5), 451458. doi:10.1097/jcp.0b013e31815152a5.CrossRefGoogle ScholarPubMed
Gomez-Lazaro, E., Garmendia, L., Beitia, G., Perez-Tejada, J., Azpiroz, A., & Arregi, A. (2012). Effects of a putative antidepressant with a rapid onset of action in defeated mice with different coping strategies. Progress in Neuro-Psychopharmacology and Biological Psychiatry, 38(2), 317327. doi:10.1016/j.pnpbp.2012.04.019.CrossRefGoogle ScholarPubMed
Goodwin, G. M., Price, J., De Bodinat, C., & Laredo, J. (2017). Emotional blunting with antidepressant treatments: A survey among depressed patients. Journal of Affective Disorders, 221, 3135. doi:10.1016/j.jad.2017.05.048.CrossRefGoogle ScholarPubMed
Haahr, M. E., Fisher, P., Holst, K., Madsen, K., Jensen, C. G., Marner, L., & Hasselbalch, S. (2013). The 5-HT4 receptor levels in hippocampus correlates inversely with memory test performance in humans. Human Brain Mapping, 34(11), 30663074. doi:10.1002/hbm.22123.CrossRefGoogle ScholarPubMed
Haahr, M. E., Fisher, P. M., Jensen, C. G., Frokjaer, V. G., Mahon, B. M., Madsen, K., & Knudsen, G. M. (2014). Central 5-HT4 receptor binding as biomarker of serotonergic tonus in humans: A [11C]SB207145 PET study. Molecular Psychiatry, 19(4), 427432. doi:10.1038/mp.2013.147.CrossRefGoogle ScholarPubMed
Hagena, H., & Manahan-Vaughan, D. (2017). The serotonergic 5-HT4 receptor: A unique modulator of hippocampal synaptic information processing and cognition. Neurobiology of Learning and Memory, 138, 145153. doi:10.1016/j.nlm.2016.06.014.CrossRefGoogle ScholarPubMed
Hille, C., Bate, S., Davis, J., & Gonzalez, M. I. (2008). 5-HT4 Receptor agonism in the five-choice serial reaction time task. Behavioural Brain Research, 195(1), 180186. doi:10.1016/j.bbr.2008.08.007.CrossRefGoogle ScholarPubMed
Hyman, S. E. (2012). Revolution stalled. Science Translational Medicine, 4(155), 155cm111. doi:10.1126/scitranslmed.3003142.CrossRefGoogle ScholarPubMed
Idres, S., Delarue, C., Lefebvre, H., & Vaudry, H. (1991). Benzamide derivatives provide evidence for the involvement of a 5-HT4 receptor type in the mechanism of action of serotonin in frog adrenocortical cells. Brain Research: Molecular Brain Research, 10(3), 251258. doi:10.1016/0169-328x(91)90068-9.Google ScholarPubMed
Imoto, Y., Kira, T., Sukeno, M., Nishitani, N., Nagayasu, K., Nakagawa, T., … Segi-Nishida, E. (2015). Role of the 5-HT4 receptor in chronic fluoxetine treatment-induced neurogenic activity and granule cell dematuration in the dentate gyrus. Molecular Brain, 8, 29. doi:10.1186/s13041-015-0120-3.CrossRefGoogle ScholarPubMed
Ishizuka, T., Goshima, H., Ozawa, A., & Watanabe, Y. (2014). Stimulation of 5-HT4 receptor enhances differentiation of mouse induced pluripotent stem cells into neural progenitor cells. Clinical and Experimental Pharmacology and Physiology, 41(5), 345350. doi:10.1111/1440-1681.12224.CrossRefGoogle ScholarPubMed
Johnson, D. E., Drummond, E., Grimwood, S., Sawant-Basak, A., Miller, E., Tseng, E., … Siok, C. J. (2012). The 5-hydroxytryptamine4 receptor agonists prucalopride and PRX-03140 increase acetylcholine and histamine levels in the rat prefrontal cortex and the power of stimulated hippocampal theta oscillations. Journal of Pharmacology and Experimental Therapeutics, 341(3), 681691. doi:10.1124/jpet.112.192351.CrossRefGoogle Scholar
Kanzari, A., Bourcier-Lucas, C., Freyssin, A., Abrous, D. N., Haddjeri, N., & Lucas, G. (2018). Inducing a long-term potentiation in the dentate gyrus is sufficient to produce rapid antidepressant-like effects. Molecular Psychiatry, 23(3), 587596. doi:10.1038/mp.2017.94.CrossRefGoogle ScholarPubMed
Kaumann, A. J. (1990). Piglet sinoatrial 5-HT receptors resemble human atrial 5-HT4-like receptors. Naunyn-Schmiedebergs Archives of Pharmacology, 342(5), 619622. doi:10.1007/bf00169055.CrossRefGoogle ScholarPubMed
Kemp, A., & Manahan-Vaughan, D. (2005). The 5-hydroxytryptamine4 receptor exhibits frequency-dependent properties in synaptic plasticity and behavioural metaplasticity in the hippocampal CA1 region in vivo. Cerebral Cortex, 15(7), 10371043. doi:10.1093/cercor/bhh204.CrossRefGoogle ScholarPubMed
Kennett, G. A., Bright, F., Trail, B., Blackburn, T. P., & Sanger, G. J. (1997). Anxiolytic-like actions of the selective 5-HT4 receptor antagonists SB 204070A and SB 207266A in rats. Neuropharmacology, 36(4-5), 707712. doi:10.1016/s0028-3908(97)00037-3.CrossRefGoogle ScholarPubMed
King, M. V., Marsden, C. A., & Fone, K. C. (2008). A role for the 5-HT(1A), 5-HT4 and 5-HT6 receptors in learning and memory. Trends in Pharmacological Sciences, 29(9), 482492.CrossRefGoogle ScholarPubMed
Kobayashi, K., Ikeda, Y., Sakai, A., Yamasaki, N., Haneda, E., Miyakawa, T., & Suzuki, H. (2010). Reversal of hippocampal neuronal maturation by serotonergic antidepressants. Proceedings of the National Academy of Sciences of the United States of America, 107(18), 84348439. doi:10.1073/pnas.0912690107.CrossRefGoogle ScholarPubMed
Kulla, A., & Manahan-Vaughan, D. (2002). Modulation by serotonin 5-HT(4) receptors of long-term potentiation and depotentiation in the dentate gyrus of freely moving rats. Cerebral Cortex, 12(2), 150162. doi:10.1093/cercor/12.2.150.CrossRefGoogle ScholarPubMed
Lamirault, L., Guillou, C., Thal, C., & Simon, H. (2003). Combined treatment with galanthaminium bromide, a new cholinesterase inhibitor, and RS 67333, a partial agonist of 5-HT4 receptors, enhances place and object recognition in young adult and old rats. Progress in Neuro-Psychopharmacology and Biological Psychiatry, 27(1), 185195. doi:10.1016/s0278-5846(02)00351-2.CrossRefGoogle ScholarPubMed
Lamirault, L., & Simon, H. (2001). Enhancement of place and object recognition memory in young adult and old rats by RS 67333, a partial agonist of 5-HT4 receptors. Neuropharmacology, 41(7), 844853.10.1016/S0028-3908(01)00123-XCrossRefGoogle ScholarPubMed
Lelong, V., Dauphin, F., & Boulouard, M. (2001). RS 67333 And D-cycloserine accelerate learning acquisition in the rat. Neuropharmacology, 41(4), 517522.10.1016/S0028-3908(01)00085-5CrossRefGoogle ScholarPubMed
Letty, S., Child, R., Dumuis, A., Pantaloni, A., Bockaert, J., & Rondouin, G. (1997). 5-HT4 receptors improve social olfactory memory in the rat. Neuropharmacology, 36(4-5), 681687.CrossRefGoogle ScholarPubMed
Levallet, G., Hotte, M., Boulouard, M., & Dauphin, F. (2009). Increased particulate phosphodiesterase 4 in the prefrontal cortex supports 5-HT4 receptor-induced improvement of object recognition memory in the rat. Psychopharmacology, 202(1–3), 125139. doi:10.1007/s00213-008-1283-8.CrossRefGoogle ScholarPubMed
Licht, C. L., Kirkegaard, L., Zueger, M., Chourbaji, S., Gass, P., Aznar, S., & Knudsen, G. M. (2010). Changes in 5-HT4 receptor and 5-HT transporter binding in olfactory bulbectomized and glucocorticoid receptor heterozygous mice. Neurochemistry International, 56(4), 603610. doi:10.1016/j.neuint.2010.01.003.CrossRefGoogle ScholarPubMed
Licht, C. L., Marcussen, A. B., Wegener, G., Overstreet, D. H., Aznar, S., & Knudsen, G. M. (2009). The brain 5-HT4 receptor binding is down-regulated in the flinders sensitive line depression model and in response to paroxetine administration. Journal of Neurochemistry, 109(5), 13631374. doi:10.1111/j.1471-4159.2009.06050.x.CrossRefGoogle ScholarPubMed
Lo, A. C., De Maeyer, J. H., Vermaercke, B., Callaerts-Vegh, Z., Schuurkes, J. A., & D'Hooge, R. (2014). SSP-002392, a new 5-HT4 receptor agonist, dose-dependently reverses scopolamine-induced learning and memory impairments in C57Bl/6 mice. Neuropharmacology, 85, 178189. doi:10.1016/j.neuropharm.2014.05.013.CrossRefGoogle ScholarPubMed
Lucas, G., Compan, V., Charnay, Y., Neve, R. L., Nestler, E. J., Bockaert, J., … Debonnel, G. (2005). Frontocortical 5-HT4 receptors exert positive feedback on serotonergic activity: Viral transfections, subacute and chronic treatments with 5-HT4 agonists. Biological Psychiatry, 57(8), 918925. doi:10.1016/j.biopsych.2004.12.023.CrossRefGoogle ScholarPubMed
Lucas, G., & Debonnel, G. (2002). 5-HT4 Receptors exert a frequency-related facilitatory control on dorsal raphé nucleus 5-HT neuronal activity. European Journal of Neuroscience, 16(5), 817822.10.1046/j.1460-9568.2002.02150.xCrossRefGoogle ScholarPubMed
Lucas, G., Du, J., Romeas, T., Mnie-Filali, O., Haddjeri, N., Piñeyro, G., & Debonnel, G. (2010). Selective serotonin reuptake inhibitors potentiate the rapid antidepressant-like effects of serotonin4 receptor agonists in the Rat. PLoS ONE, 5(2):e9253.10.1371/journal.pone.0009253CrossRefGoogle ScholarPubMed
Lucas, G., Rymar, V. V., Du, J., Mnie-Filali, O., Bisgaard, C., Manta, S., … Debonnel, G. (2007). Serotonin(4) (5-HT(4)) receptor agonists are putative antidepressants with a rapid onset of action. Neuron, 55(5), 712725. doi:10.1016/j.neuron.2007.07.041.CrossRefGoogle Scholar
Madsen, K., Torstensen, E., Holst, K. K., Haahr, M. E., Knorr, U., Frokjaer, V. G., … Knudsen, G. M. (2014). Familial risk for major depression is associated with lower striatal 5-HT(4) receptor binding. International Journal of Neuropsychopharmacology, 18(1) pii: pyu034. doi:10.1093/ijnp/pyu034.Google Scholar
Malberg, J. E., Eisch, A. J., Nestler, E. J., & Duman, R. S. (2000). Chronic antidepressant treatment increases neurogenesis in adult rat hippocampus. Journal of Neuroscience, 20(24), 91049110.10.1523/JNEUROSCI.20-24-09104.2000CrossRefGoogle ScholarPubMed
Marchetti-Gauthier, E., Roman, F. S., Dumuis, A., Bockaert, J., & Soumireu-Mourat, B. (1997). BIMU1 Increases associative memory in rats by activating 5-HT4 receptors. Neuropharmacology, 36(4–5), 697706.10.1016/S0028-3908(97)00058-0CrossRefGoogle ScholarPubMed
Marchetti, E., Chaillan, F. A., Dumuis, A., Bockaert, J., Soumireu-Mourat, B., & Roman, F. S. (2004). Modulation of memory processes and cellular excitability in the dentate gyrus of freely moving rats by a 5-HT4 receptors partial agonist, and an antagonist. Neuropharmacology, 47(7), 10211035. doi:10.1016/j.neuropharm.2004.06.033.CrossRefGoogle Scholar
Marchetti, E., Dumuis, A., Bockaert, J., Soumireu-Mourat, B., & Roman, F. S. (2000). Differential modulation of the 5-HT(4) receptor agonists and antagonist on rat learning and memory. Neuropharmacology, 39(11), 20172027. doi:10.1016/s0028-3908(00)00038-1.CrossRefGoogle ScholarPubMed
Marchetti, E., Jacquet, M., Escoffier, G., Miglioratti, M., Dumuis, A., Bockaert, J., & Roman, F. S. (2011). Enhancement of reference memory in aged rats by specific activation of 5-HT(4) receptors using an olfactory associative discrimination task. Brain Research, 1405, 4956. doi:10.1016/j.brainres.2011.06.020.CrossRefGoogle ScholarPubMed
Medhurst, A. D., Lezoualc'h, F., Fischmeister, R., Middlemiss, D. N., & Sanger, G. J. (2001). Quantitative mRNA analysis of five C-terminal splice variants of the human 5-HT4 receptor in the central nervous system by TaqMan real time RT-PCR. Brain Research: Molecular Brain Research, 90(2), 125134.Google ScholarPubMed
Mendez-David, I., David, D. J., Darcet, F., Wu, M. V., Kerdine-Römer, S., Gardier, A. M., & Hen, R. (2014). Rapid anxiolytic effects of a 5-HT₄ receptor agonist are mediated by a neurogenesis-independent mechanism. Neuropsychopharmacology, 39(6), 13661378. doi:10.1038/npp.2013.332.CrossRefGoogle ScholarPubMed
Meneses, A., & Hong, E. (1997). Effects of 5-HT4 receptor agonists and antagonists in learning. Pharmacology, Biochemistry and Behavior, 56(3), 347351. doi:10.1016/s0091-3057(96)00224-9.CrossRefGoogle ScholarPubMed
Millan, M. J., Agid, Y., Brune, M., Bullmore, E. T., Carter, C. S., Clayton, N. S., … Young, L. J. (2012). Cognitive dysfunction in psychiatric disorders: Characteristics, causes and the quest for improved therapy. Nature Reviews. Drug Discovery, 11(2), 141168. doi:10.1038/nrd3628.CrossRefGoogle ScholarPubMed
Mitchell, A. J. (2006). Two-week delay in onset of action of antidepressants: New evidence. British Journal of Psychiatry, 188, 105106. England.CrossRefGoogle ScholarPubMed
Moha ou Maati, H., Bourcier-Lucas, C., Veyssiere, J., Kanzari, A., Heurteaux, C., Borsotto, M., … Lucas, G. (2016). The peptidic antidepressant spadin interacts with prefrontal 5-HT(4) and mGluR(2) receptors in the control of serotonergic function. Brain Structure & Function, 221(1), 2137. doi:10.1007/s00429-014-0890-x.CrossRefGoogle Scholar
Mohler, E. G., Shacham, S., Noiman, S., Lezoualc'h, F., Robert, S., Gastineau, M., … Ragozzino, M. E. (2007). VRX-03011, a novel 5-HT4 agonist, enhances memory and hippocampal acetylcholine efflux. Neuropharmacology, 53(4), 563573. doi:10.1016/j.neuropharm.2007.06.016.CrossRefGoogle ScholarPubMed
Moser, P. C., Bergis, O. E., Jegham, S., Lochead, A., Duconseille, E., Terranova, J. P., … Scatton, B. (2002). SL65.0155, A novel 5-hydroxytryptamine(4) receptor partial agonist with potent cognition-enhancing properties. Journal of Pharmacology and Experimental Therapeutics, 302(2), 731741. doi:10.1124/jpet.102.034249.CrossRefGoogle ScholarPubMed
Murphy, S. E., Wright, L. C., Browning, M., Cowen, P. J., & Harmer, C. J. (2019). A role for 5-HT4 receptors in human learning and memory. Psychological Medicine, 19. doi:10.1017/s0033291719002836.Google ScholarPubMed
Nasehi, M., Tabatabaie, M., Khakpai, F., & Zarrindast, M. R. (2015). The effects of CA1 5HT4 receptors in MK801-induced amnesia and hyperlocomotion. Neuroscience Letters, 587, 7378. doi:10.1016/j.neulet.2014.12.019.CrossRefGoogle ScholarPubMed
Norman, E. D., Thiels, E., Barrionuevo, G., & Klann, E. (2000). Long-term depression in the hippocampus in vivo is associated with protein phosphatase-dependent alterations in extracellular signal-regulated kinase. Journal of Neurochemistry, 74(1), 192198. doi:10.1046/j.1471-4159.2000.0740192.x.CrossRefGoogle ScholarPubMed
Ohtsuki, T., Ishiguro, H., Detera-Wadleigh, S. D., Toyota, T., Shimizu, H., Yamada, K., … Arinami, T. (2002). Association between serotonin 4 receptor gene polymorphisms and bipolar disorder in Japanese case-control samples and the NIMH genetics initiative bipolar pedigrees. Molecular Psychiatry, 7(9), 954961. doi:10.1038/sj.mp.4001133.CrossRefGoogle ScholarPubMed
Orsetti, M., Dellarole, A., Ferri, S., & Ghi, P. (2003). Acquisition, retention, and recall of memory after injection of RS67333, a 5-HT(4) receptor agonist, into the nucleus basalis magnocellularis of the rat. Learning and Memory, 10(5), 420426. doi:10.1101/lm.67303.CrossRefGoogle ScholarPubMed
Pascual-Brazo, J., Castro, E., Díaz, A., Valdizán, E. M., Pilar-Cuéllar, F., Vidal, R., … Pazos, A. (2012). Modulation of neuroplasticity pathways and antidepressant-like behavioural responses following the short-term (3 and 7 days) administration of the 5-HT₄ receptor agonist RS67333. International Journal of Neuropsychopharmacology, 15(5), 631643. doi:10.1017/S1461145711000782.CrossRefGoogle ScholarPubMed
Penas-Cazorla, R., & Vilaro, M. T. (2015). Serotonin 5-HT4 receptors and forebrain cholinergic system: Receptor expression in identified cell populations. Brain Structure & Function, 220(6), 34133434. doi:10.1007/s00429-014-0864-z.CrossRefGoogle ScholarPubMed
Quiedeville, A., Boulouard, M., Hamidouche, K., Da Silva Costa-Aze, V., Nee, G., Rochais, C., & Bouet, V. (2015). Chronic activation of 5-HT4 receptors or blockade of 5-HT6 receptors improve memory performances. Behavioural Brain Research, 293, 1017. doi:10.1016/j.bbr.2015.07.020.CrossRefGoogle ScholarPubMed
Rosel, P., Arranz, B., Urretavizcaya, M., Oros, M., San, L., & Navarro, M. A. (2004). Altered 5-HT2A and 5-HT4 postsynaptic receptors and their intracellular signalling systems IP3 and cAMP in brains from depressed violent suicide victims. Neuropsychobiology, 49(4), 189195. doi:10.1159/000077365.CrossRefGoogle ScholarPubMed
Roychowdhury, S., Haas, H., & Anderson, E. G. (1994). 5-HT1A and 5-HT4 receptor colocalization on hippocampal pyramidal cells. Neuropharmacology, 33(3–4), 551557.10.1016/0028-3908(94)90086-8CrossRefGoogle ScholarPubMed
Rush, A. J., Warden, D., Wisniewski, S. R., Fava, M., Trivedi, M. H., Gaynes, B. N., & Nierenberg, A. A. (2009). STAR*D: Revising conventional wisdom. CNS Drugs, 23(8), 627647. doi:10.2165/00023210-200923080-00001.Google ScholarPubMed
Samuels, B. A., Mendez-David, I., Faye, C., David, S. A., Pierz, K. A., Gardier, A. M., … David, D. J. (2016). Serotonin 1A and serotonin 4 receptors: Essential mediators of the neurogenic and behavioral actions of antidepressants. Neuroscientist, 22(1), 2645. doi:10.1177/1073858414561303.CrossRefGoogle ScholarPubMed
Schmidt, E. F., Warner-Schmidt, J. L., Otopalik, B. G., Pickett, S. B., Greengard, P., & Heintz, N. (2012). Identification of the cortical neurons that mediate antidepressant responses. Cell, 149(5), 11521163. doi:10.1016/j.cell.2012.03.038.CrossRefGoogle ScholarPubMed
Segu, L., Lecomte, M. J., Wolff, M., Santamaria, J., Hen, R., Dumuis, A., … Compan, V. (2010). Hyperfunction of muscarinic receptor maintains long-term memory in 5-HT4 receptor knock-out mice. PLoS ONE, 5(3), e9529. doi:10.1371/journal.pone.0009529.CrossRefGoogle ScholarPubMed
Shilyansky, C., Williams, L. M., Gyurak, A., Harris, A., Usherwood, T., & Etkin, A. (2016). Effect of antidepressant treatment on cognitive impairments associated with depression: A randomised longitudinal study. The Lancet. Psychiatry, 3(5), 425435. doi:10.1016/s2215-0366(16)00012-2.CrossRefGoogle ScholarPubMed
Silvestre, J. S., Fernandez, A. G., & Palacios, J. M. (1996). Effects of 5-HT4 receptor antagonists on rat behaviour in the elevated plus-maze test. European Journal of Pharmacology, 309(3), 219222. doi:10.1016/0014-2999(96)00457-8.CrossRefGoogle ScholarPubMed
Siniscalchi, A., Badini, I., Beani, L., & Bianchi, C. (1999). 5-HT4 Receptor modulation of acetylcholine outflow in Guinea pig brain slices. Neuroreport, 10(3), 547551.10.1097/00001756-199902250-00019CrossRefGoogle ScholarPubMed
Terry, A. V. Jr., Buccafusco, J. J., Jackson, W. J., Prendergast, M. A., Fontana, D. J., Wong, E. H., … Eglen, R. M. (1998). Enhanced delayed matching performance in younger and older macaques administered the 5-HT4 receptor agonist, RS 17017. Psychopharmacology, 135(4), 407415. doi:10.1007/s002130050529.CrossRefGoogle ScholarPubMed
Tonini, M., & Candura, S. M. (1996). 5-HT4 Receptor agonists and bladder disorders. Trends in Pharmacological Sciences, 17, 314316. England.CrossRefGoogle ScholarPubMed
Trivedi, M. H., Fava, M., Wisniewski, S. R., Thase, M. E., Quitkin, F., Warden, D., … Rush, A. J. (2006). Medication augmentation after the failure of SSRIs for depression. New England Journal of Medicine, 354(12), 12431252. doi:10.1056/NEJMoa052964.CrossRefGoogle ScholarPubMed
Twarkowski, H., Hagena, H., & Manahan-Vaughan, D. (2016). The 5-hydroxytryptamine4 receptor enables differentiation of informational content and encoding in the hippocampus. Hippocampus, 26(7), 875891. doi:10.1002/hipo.22569.CrossRefGoogle ScholarPubMed
Varnas, K., Halldin, C., Pike, V. W., & Hall, H. (2003). Distribution of 5-HT4 receptors in the postmortem human brain – an autoradiographic study using [125I]SB 207710. European Neuropsychopharmacology, 13(4), 228234.10.1016/S0924-977X(03)00009-9CrossRefGoogle Scholar
Vidal, R., Valdizan, E. M., Mostany, R., Pazos, A., & Castro, E. (2009). Long-term treatment with fluoxetine induces desensitization of 5-HT4 receptor-dependent signalling and functionality in rat brain. Journal of Neurochemistry, 110(3), 11201127. doi:10.1111/j.1471-4159.2009.06210.x.CrossRefGoogle ScholarPubMed
Vidal, R., Valdizan, E., Vilaró, M., Pazos, A., & Castro, E. (2010). Reduced signal transduction by 5-HT4 receptors after long-term venlafaxine treatment in rats. British Journal of Pharmacology, 161(3), 695706. doi:10.1111/j.1476-5381.2010.00903.x.CrossRefGoogle ScholarPubMed
Vilaró, M. T., Cortés, R., & Mengod, G. (2005). Serotonin 5-HT4 receptors and their mRNAs in rat and Guinea pig brain: Distribution and effects of neurotoxic lesions. Journal of Comparative Neurology, 484(4), 418439. doi:10.1002/cne.20447.CrossRefGoogle ScholarPubMed
Waeber, C., Sebben, M., Nieoullon, A., Bockaert, J., & Dumuis, A. (1994). Regional distribution and ontogeny of 5-HT4 binding sites in rodent brain. Neuropharmacology, 33(3–4), 527541. doi:10.1016/0028-3908(94)90084-1.CrossRefGoogle ScholarPubMed
Warner-Schmidt, J. L., & Duman, R. S. (2006). Hippocampal neurogenesis: Opposing effects of stress and antidepressant treatment. Hippocampus, 16(3), 239249. doi:10.1002/hipo.20156.CrossRefGoogle ScholarPubMed
Wong, C. H., Siah, K. W., & Lo, A. W. (2019). Estimation of clinical trial success rates and related parameters. Biostatistics, 20(2), 273286.10.1093/biostatistics/kxx069CrossRefGoogle ScholarPubMed
World Health Organisation (2017). Depression and other common mental disorders. Global Health Estimates.Google Scholar
Wray, N. R., Ripke, S., Mattheisen, M., Trzaskowski, M., Byrne, E. M., Abdellaoui, A., … Consortium, M. D. D. W. G. O. T. P. G. (2018). Genome-wide association analyses identify 44 risk variants and refine the genetic architecture of major depression. Nature Genetics, 50(5), 668681. doi:10.1038/s41588-018-0090-3.CrossRefGoogle ScholarPubMed