Submitted Successfully!
To reward your contribution, here is a gift for you: A free trial for our video production service.
Thank you for your contribution! You can also upload a video entry or images related to this topic.
Version Summary Created by Modification Content Size Created at Operation
1 -- 2992 2023-03-23 11:39:29 |
2 Reference format revised. Meta information modification 2992 2023-03-24 04:41:49 |

Video Upload Options

Do you have a full video?

Confirm

Are you sure to Delete?
Cite
If you have any further questions, please contact Encyclopedia Editorial Office.
Fukuyama, K.; Motomura, E.; Okada, M. Therapeutic Potential of Serotonin Type 7 Receptor Modulation. Encyclopedia. Available online: https://encyclopedia.pub/entry/42470 (accessed on 01 July 2024).
Fukuyama K, Motomura E, Okada M. Therapeutic Potential of Serotonin Type 7 Receptor Modulation. Encyclopedia. Available at: https://encyclopedia.pub/entry/42470. Accessed July 01, 2024.
Fukuyama, Kouji, Eishi Motomura, Motohiro Okada. "Therapeutic Potential of Serotonin Type 7 Receptor Modulation" Encyclopedia, https://encyclopedia.pub/entry/42470 (accessed July 01, 2024).
Fukuyama, K., Motomura, E., & Okada, M. (2023, March 23). Therapeutic Potential of Serotonin Type 7 Receptor Modulation. In Encyclopedia. https://encyclopedia.pub/entry/42470
Fukuyama, Kouji, et al. "Therapeutic Potential of Serotonin Type 7 Receptor Modulation." Encyclopedia. Web. 23 March, 2023.
Therapeutic Potential of Serotonin Type 7 Receptor Modulation
Edit

Although a number of mood-stabilising atypical antipsychotics and antidepressants modulate serotonin type 7 receptor (5-HT7), the detailed contributions of 5-HT7 function to clinical efficacy and pathophysiology have not been fully understood. The mood-stabilising antipsychotic agent, lurasidone, and the serotonin partial agonist reuptake inhibitor, vortioxetine, exhibit higher binding affinity to 5-HT7 than other conventional antipsychotics and antidepressants. The initially expected rapid onset of antidepressant effects—in comparison with conventional antidepressants or mood-stabilising antipsychotics—due to 5-HT7 inhibition has not been observed with lurasidone and vortioxetine; however, several clinical studies suggest that 5-HT7 inhibition likely contributes to quality of life of patients with schizophrenia and mood disorders via the improvement of cognition. Furthermore, it reported that 5-HT7 inhibition might mitigate antipsychotic-induced weight gain and metabolic complication by blocking other monoamine receptors. Further preclinical studies for the development of 5-HT7 modulation against neurodevelopmental disorders and neurodegenerative diseases have been ongoing. Various findings from various preclinical studies indicate the possibility that 5-HT7 modifications can provide two independent strategies. The first is that 5-HT7 inhibition ameliorates the dysfunction of inter-neuronal transmission in mature networks. The other is that activation of 5-HT7 can improve transmission dysfunction due to microstructure abnormality in the neurotransmission network—which could be unaffected by conventional therapeutic agents—via modulating intracellular signalling during the neurodevelopmental stage or via loss of neural networks with aging.

antipsychotics antidepressants mood stabilising schizophrenia serotonin type 7 receptor

1. Introduction

Serotonin (5-HT) receptor type 7 (5-HT7) is one of the most recently (1993) identified members of the 5-HT receptor family [1][2][3][4][5]. It has been demonstrated that 5-HT7 is highly expressed in functionally relevant regions of the brain [6][7]. Indeed, in the central nervous system, 5-HT7 is most predominantly expressed in the thalamus, hypothalamus, hippocampus, prefrontal cortex, basal ganglia, amygdala and dorsal raphe nucleus [8][9][10][11][12][13][14]. The predominant expression of 5-HT7 in the limbic regions provides a candidate hypothesis that 5-HT7 contributes to the regulation of memory processing, cognition and emotional perception [9][10][11][12][15][16]. The expression of 5-HT7 has been also observed in the kidney, liver, pancreas, spleen, stomach and smooth muscle cells of the arteries and gastrointestinal tract [17]. Based on these findings, 5-HT7 modulation is also considered to be a possible therapeutic target for the treatment of peripheral organs [18][19][20][21].
A number of preclinical studies have reported that 5-HT7 plays important roles in the regulation of mood, memory processing, cognition and emotional perception by following various experiments using selective 5-HT7 modulators and 5-HT7 knockout mice models [15][22][23][24][25][26]. Although modulating 5-HT7 is one of the targets for the treatment of schizophrenia and mood and anxiety disorders in current psychopharmacology, unfortunately, the clinical application of selective 5-HT7 receptor modulators has not yet been achieved [16]. However, several conventional mood-stabilising atypical antipsychotics, such as aripiprazole, brexpiprazole, clozapine, lurasidone, olanzapine, quetiapine, risperidone and zotepine are known to be inhibitors of 5-HT7 [12][27][28][29][30][31][32][33][34][35][36][37][38][39][40] (Table 1). Lurasidone is an antipsychotic agent with the highest binding affinity to 5-HT7 among mood-stabilising atypical antipsychotics [16][27] (Table 1). Furthermore, a novel antidepressant, vortioxetine, which is categorized as a 5-HT partial agonist reuptake inhibitor (SPARI), exhibits distinct pharmacodynamic profiles compared to other monoamine transporter-inhibiting antidepressants, since vortioxetine acutely and chronically suppresses the function of 5-HT7 [38][39][41][42].
Table 1. Receptor-binding profiles of antipsychotics and antidepressants.
Receptor LUR APZ Brex CLZ OLZ PMZ QTP RIS ZTP VTX
5-HT1A 6.8 5.6 0.12 124 >1000 650 432 423 471 15.0
5-HT2A 2.0 8.7 0.47 5.4 2.3 48.4 100 0.2 2.7  
5-HT3 >1000 630   241 57 >1000 >1000 >1000 472 3.7
5-HT7 0.5 10.3 3.7 18.0 365 0.5 307 6.6 12.0 19.0
H1 >1000 27.6 19 1.13 1.2 692 11 20.1 3.21  
D1 262 >1000 160 266 100 >1000 712 244 71.0  
D2 1.7 3.3 0.3 157 52.3 0.3 245 3.6 25.0  
Reference [27] [28][29] [30] [31][32] [33][43] [34] [35] [29][36] [37] [38]
Notes: lurasidone (LUR), aripiprazole (APZ), brexpiprazole (Brex), clozapine (CLZ), olanzapine (OLZ), pimozide (PMZ), quetiapine (QTP), risperidone (RIS), zotepine (ZTP) and antidepressant vortioxetine (VTX) against serotonin (5-HT) type 1A (5-HT1A), type 2A (5-HT2A), type 3 (5-HT3), and type 7 (5-HT7) receptor, histamine H1 (H1) receptor, dopamine receptors type 1 (D1) and 2 (D2). Data are equilibrium-constant (Ki) values (nM).

2. Preclinical Findings about Therapeutic Potential of 5-HT7 Modulation

2.1. Depression

The earliest identified physiological function of 5-HT7 was regulation of circadian rhythms [2]. In this context, 5-HT7 was found to be expressed in the suprachiasmatic nucleus [44], which is a major regulatory region of circadian rhythms [45]. The influence of 5-HT7 on sleep regulation is complicated because the 5-HT7 inverse agonist SB269970 [40][42][46][47] increased latency in the onset but decreased the total amount of time spent in rapid eye movement sleep [48]. A number of antidepressants also increased latency in the onset but decreased the total amount of time spent in rapid eye movement sleep, similar to SB269970 [49].

The 5-HT7 knockout mice displayed a reduction of immobility times in both a forced swim test and a tail suspension test, common pharmacological behaviour tests for the screening of antidepressants (decreasing immobility time is considered to correlate to antidepressant action in humans) [22][50]. Based on this finding, 5-HT7 inhibitors/antagonists were actively explored and developed as antidepressants in the early years of this century. SB269970, an inverse agonist of 5-HT7, has been shown to reduce immobility time in forced swim and tail suspension tests; as a result, it became a candidate for use as an antidepressant [22][23][51]. Additionally, the administration of subeffective concentration of SB269970 enhanced the anti-immobility action of subeffective doses of of desipramine, citalopram and imipramine in the forced swim and tail suspension tests [51][52][53]. Most notably, SB269970 generated significantly rapid onset of antidepressant-like effects in olfactory bulbectomised models, when compared to fluoxetine [54]. As a result, 5-HT7 inhibitors were anticipated to join the rapid onset antidepressant class, since it is recognised that one of the major problems of monoamine transporter-inhibiting antidepressants, a currently/commonly prescribed antidepressant class, is that they require 2-4 weeks for the onset of antidepressive effects. The target regions for antidepressant effects of 5-HT7 inhibitors remain controversial. Injection of SB269970 into the hippocampus generated antidepressant-like activity in the forced swim test [55]. Injection of SB269970 into the lateral habenular nucleus lesion model using 6-hydroxydoapmine also exhibited antidepressant-like activity, whereas injection into the medial prefrontal cortex lesion of a model, conversely, displayed the enhancement of depressive-like activity [56][57]. AS19, a 5-HT7 agonist, demonstrated the opposite region-dependent effect against SB269970 [56][57].

2.2. Anxiety

The potential function of 5-HT7 on anxiety demonstrated the inconsistent results between pharmacological and molecular biological studies, similar to the case of depression. Expression of 5-HT7 mRNA increased by acute restraint stress but not by chronic variable stress in the rat hippocampus, indicating that 5-HT7 contributes to the regulation of response to stress [58]; however, the anxiety-like behaviour in the light/dark transfer or elevated plus maze tests did not affect both 5-HT7 knockdown or knockout models [50][59][60]. On the other hand, the anxiolytic-like activity of SB269970 (both systemic and intra-hippocampal local administrations) was demonstrated by the Vogel drinking test, the elevated plus maze test and the four plates test [23][55]. Both SB269970 and 5-HT7 knockout mice demonstrated anxiolytic-like activity in the marble-burying test, which is a model of anxiety and obsessive–compulsive disorder [61]. These inconsistent results among molecular biological and pharmacological experiments suggest that the level of 5-HT7 inactivation required for anxiolytic effects is probably dependent on the model utilized. In other words, appropriate 5-HT7 inhibition may be beneficial for anxiolytic effects.

2.3. Schizophrenia

A number of pharmacological studies reported the therapeutic potential in components of positive and negative symptoms and cognitive dysfunction of schizophrenia using chemical-induced schizophrenia models. Compared with wild-type mice, 5-HT7 knockout mice were less susceptible to prepulse inhibition deficits of phencyclidine [62]. SB-269970 suppressed hyperactivity induced by ketamine, phencyclidine and amphetamine [63][64], whereas the effects of 5-HT7 on prepulse inhibition deficits were inconsistent. SB-269970 improved amphetamine-induced prepulse inhibition deficits [64], but did not affect those induced by an NMDA/glutamate inhibitor, phencyclidine or ketamine [62][65]; however, SB-258741, a 5-HT7 antagonist, did not affect the amphetamine-induced prepulse inhibition deficits, but suppressed phencyclidine-induced prepulse inhibition deficits [66]. Ketamine-induced negative symptoms associated with social withdrawal were attenuated by SB269970, but not affected by SB-258741 [65][66]. The effects of SB269970 on positive symptoms are possibly involved in dopaminergic transmission but not in glutamatergic transmission, whereas, conversely, the effects of SB-258741 on positive symptoms are possibly involved in glutamatergic transmission but not in dopaminergic transmission. Furthermore, social withdrawal induced by NMDA/glutamate receptor inhibition is prevented by SB269970 but not by SB258741. These discrepancies between SB269970 and SB258741 could not be explained by their receptor-binding profiles alone, since these compounds displayed binding affinity to 5-HT7, D2 and D3 receptors [66]. These findings suggest that the effects between SB269970 and SB258741 are probably dependent upon the materials (pharmacological and molecular biological models) and compounds employed.
On the other hand, the effects of 5-HT7 inhibition on neurocognitive dysfunction (procognitive effects) demonstrated its promise. SB269970 attenuates amnesia in short-term memory induced by ketamine and MK801 [67][68], and this effect was suppressed by AS19 [69]. The new valuable tool for exploring the neurobiological bases of cognitive dysfunction in schizophrenia, five-choice serial reaction time task (including attention, response inhibition, cognitive flexibility and processing speed), demonstrated that SB269970 improved the impairment of working memory and impulsivity, without affecting premature responding induced by MK801 [70].

3. Clinical Evaluation of 5-HT7 Modulators

3.1. Vortioxetine

Vortioxetine is an antidepressant belonging to the family of monoamine transporter-inhibiting antidepressants; its antidepressant effect is thought to arise from not only its monoamine transporter inhibition but also 5-HT7 inhibition. The binding affinity of vortioxetine to 5-HT7 is relatively low compared to serotonin transporter, 5-HT1A and 5-HT3 [38], but the relevant therapeutic concentration of vortioxetine functionally suppresses 5-HT7 [39][41].

3.2. Lurasidone

A mood-stabilising antipsychotic agent, lurasidone possesses the highest binding affinity to 5-HT7 among antipsychotics [27] (Table 1). Several meta-analyses reported that lurasidone significantly improves positive and negative depressive symptoms [71][72][73][74]. Therefore, the general efficacy of lurasidone for the treatment of schizophrenia is considered to be comparable to that of other atypical antipsychotics [16]. Meta-analyses also demonstrated that lurasidone has antidepressant effects comparable to those of other mood-stabilising antipsychotics against major depressive disorder and bipolar depression [75][76]. The efficacy of lurasidone in the acute treatment of bipolar depression, as both monotherapy and adjunctive therapy to lithium/valproate, has been reported in clinical trials [77][78][79]. Like vortioxetine, the rapid onset of the antidepressive effects of lurasidone have not been demonstrated [75][80][81][82][83].

4. Intracellular Signalling Associated with 5-HT7

Four splice variants of 5-HT7 were identified. These were distinct in their carboxyl terminals due to introns in the 5-HT7 gene, including 5-HT7a, 5-HT7b and 5-HT7c in rodents, and 5-HT7a, 5-HT7b and 5-HT7d in humans [6][84][85][86][87][88]. All of these four splicing variants directly affect three intracellular signalling pathways via activations of Gαs, Gα12 and metalloproteinase-9 [4][89][90]. Significant differences among 5-HT7 splicing variants in localisation, ligand-binding affinity and adenylate cyclase activity have not been observed [6], whereas 5-HT7a isoform specifically activates the abovementioned cAMP-dependent signalling through the activation of type 1 and 8 adenylyl cyclase via Ca2+/calmodulin-dependent and Gs-independent signalling [91].
Activation of 5-HT7 enhances synthesis of cyclic adenosine monophosphate (cAMP) via activation of adenylyl cyclase [4]. Increasing cAMP activates signalling of both protein kinase A (PKA) and the exchange protein directly activated by cAMP (EPAC) [10][90][92]. These two signalling pathways affect various signalling transductions via phosphorylation of target proteins, leading to the propagation of the signalling to the next biochemical events. Subsequently, enhanced PKA stimulates cyclin-dependent kinase 5 (Cdk5) [10][90] and Ras [93][94], resulting in serine/threonine extracellular signal-regulated kinases (Erk) signalling activation [93][95]. Activation of EPAC also indirectly enhances Erk signalling [93][95]. The second signalling pathway regulates adenosine monophosphate-dependent protein kinase (AMPK) [42][96][97] via inhibitory PKA and stimulatory EPAC [98]
Two other types of signalling associated with 5-HT7 were also identified: Gα12 [90] and metalloproteinase-9 (MMP9) [89]. It has been shown that 5-HT7/Gα12 activates cell division cycle protein 42 (Cdc42) [90][99] and activates signalling pathways associated with Gα12 [90]. In addition, it is recognized that 5-HT7/Gα12 activates both Ras homolog gene family member A (RhoA) and cell division cycle protein 42 (Cdc42) [90][99]
It has been established that the serotonergic system plays crucial roles in the organisation of the neural system, such as generation of neurogenesis, cell migration, axon guidance, dendritogenesis, synaptogenesis and brain wiring during the development of the mature brain [100]. During the embryonic stage, 5-HT7 leads neurite outgrowth through activations of Cdk5 with mTOR [94][101]. Therefore, 5-HT7 contributes to the establishment and maintenance of neural connectivity and synaptic plasticity in early developmental stages [102]. In other words, the reorganization of dendritic morphology induced by 5-HT7 plays important roles in new synapse growth and initial neuronal network formation, which is the target of event-related structural and functional plasticity in the early developmental stage [103][104]

5. Effects of 5-HT7 on Neuronal Transmission

Behavioural study demonstrated that 5-HT7 knockout mice displayed impairments of contextual learning, seeking behaviour and allocentric spatial memory [105]. Electrophysiological study also demonstrated that 5-HT7 knockout mice exhibited impaired hippocampal long-term potentiation [59]. In addition, 5-HT7-induced activation of PKA signalling enhanced N-methyl-D-aspartate (NMDA)-evoked currents, resulting in the enhancement of population spike amplitude and bursting frequency in hippocampal CA1 and CA3 regions, respectively [106][107][108]. Furthermore, 5-HT7 activates hippocampal transmission postsynaptically due to enhanced phosphorylation of the AMPA/glutamate receptor induced by cAMP/cAMP response element-binding protein (CREB) signalling [109][110]. Additionally, 5-HT7 reversed long-term depression associated with metabotropic glutamate receptors (mGluR) [26].
Activation of 5-HT7 during adolescence induced persistent upregulation of 5-HT7 [13]. Chronic exposure to methylphenidate during postnatal life and adolescence probably provides persistent structural rearrangements of the brain’s reward pathways associated with 5-HT7 [111]. During the pre- and postnatal periods, exposure to selective serotonin reuptake inhibitors generates long-term anxiety in adulthood without affecting the morphological alterations of the brain [112][113]. The molecular mechanism underlying 5-HT7-induced remodelling increases neurites and dendritic spine elongation via MMP-9/CD44 with Cdc42 in reversal learning and neuronal regeneration [89]. Therefore, the impacts of 5-HT7 on neuronal plasticity during early development are not limited to embryonic and early postnatal development but can also persist in adolescence and adulthood.
Although activation of hippocampal 5-HT7 contributes to the formation of learning and memory, suppression of 5-HT7, conversely, improved other cognitive components, such as executive function, which play important roles in quality of life. Traditionally, enhanced mesocortical dopaminergic transmission or dopamine release in the frontal cortex induced by blockade of D2 in the ventral tegmental area are considered to be key players in the improvement of positive and negative symptoms of schizophrenia [32]. Additionally, thalamocortical glutamatergic transmission are considered to play important roles in neurocognitive function [32][114][115][116].
Tonic hyperactivation of thalamocortical glutamatergic transmission was observed in schizophrenia, ADHD and autism [12][39][115][116][117][118][119][120][121][122][123][124][125][126]. Other 5-HT7 molecules, such as group II and III mGluRs and α2 adrenoceptor, which compensate thalamocortical glutamatergic transmission, have also been identified [116][117][118][120][125]. The behavioural importance of neuronal networks is affirmed by lesion studies demonstrating that lesions to hub regions are associated with task impairments across multiple functional domains [127][128][129]. Notably, the prediction of the thalamocortical pathway on task-specific cortical activity patterns was outperformed compared to the prediction of the cortical and hippocampal pathway [127][128][129]. The mediodorsal thalamic nucleus is reciprocally connected with the medial prefrontal cortex and receives glutamatergic inputs from the hippocampus [40][41][124][130][131]. The glutamatergic neurons in the mediodorsal thalamic nucleus were mainly suppressed but were activated by propagation of ripple burst during the non-rapid eye movement sleep phase [130][131][132]. Therefore, the mediodorsal thalamic nucleus plays important roles in memory processing during sleep and sensory integration during wakefulness [16][133][134][135][136].

6. Therapeutic Potential in Other Disease and Disorders Based on the Preclinical Findings

6.1. Neurodevelopmental Disorders

Activation of 5-HT7 was a potential candidate target for relieving symptoms in patients with Rett syndrome. Rett syndrome is the second most common cause of mental retardation in females and plays a role in severe neurodevelopmental disorders such as breathing dysfunction, loss of coordination, abnormal eye and hand movements, epilepsy, aberrant sleeping behaviour and cognitive impairment [137]. The prime pathogenesis of Rett syndrome is known to be various genetic mutations in methyl CpG-binding protein 2 gene (MeCP2) on the X chromosome, cyclin-dependent kinase-like 5 (CDKL5), forkhead box G1 (FOXG1), WD repeat domain 45 (WDR45) or syntaxin binding protein 1 (STXBP1) [138][139]. Restoring MeCP2 function can normalise functional abnormalities of MeCP2 knockout mice, whereas overexpression of the MeCP2 gene led to neurological defects [140]. Therefore, recent preclinical studies explored the targets in MeCP2 downstream effectors and other signalling, including 5-HT7. Based on the reduced 5-HT7 expression in Rett syndrome models, the systemic administration of 5-HT7 agonist relieved the related symptoms, anxiety, environment-related exploratory behaviour and motor learning ability of Rett syndrome mice models [141][142].

6.2. Neurodegenerative Diseases

Non-selectively activation of 5-HT receptors using 5-HT transporter inhibitors has shown little clinical benefit in neurodegenerative disorders [143][144]; however, several preclinical studies reported the attractive findings that 5-HT7 is a therapeutic candidate target for neurodegenerative disorders. Administration of 5-HT7 agonist also suppressed impairment of long-term potentiation and apoptosis in hippocampal streptozotocin-mediated neurodegeneration in murine models [145]. Amyloid-β induces neurotoxicity through several mechanisms including apoptosis, excitotoxicity and oxidative stress [146]; these were reverted by selective 5-HT7 agonist [147]. Amyloid-β is a key player in pathomechanisms of various neurogenerative diseases, such as Alzheimer’s disease, frontotemporal dementia, cerebral amyloid angiopathy, and cerebral amyloidosis, through multiple pathways [146][148]; however, the detailed roles of 5-HT7 in pathomechanisms of neurodegenerative disease associated with amyloid-β remain to be clarified [149][150].

6.3. Epilepsy

Various studies have reported on antiseizure activities—using audiogenic seizures in DBA/2J mice, or the absence of seizures in WAG/Rij rats—following the administration of selective and non-selective antagonists [151][152]. These studies demonstrated that SB269970 and AS-19 decreased and increased the frequency of pilocarpine-induced temporal lobe epilepsy seizures in rat models, respectively [153].

6.4. Cartinoma

There are some findings indicating that 5-HT7 modulation may be therapeutic targets for the treatment of cancer. Notably, it has been anticipated that 5-HT7 antagonists would be useful for the treatment of hepatocellular cancer and small-intestinal neuroendocrine neoplasms, since 5-HT7 contributes to hepatocyte proliferation [18][19][20]. Serotonergic function activates proliferation of hepatocellular cancer and the expression of β-catenin, which plays an important role in the pathomechanisms of hepatocellular cancer via the activation of 5-HT7. The expression of 5-HT7 increased in hepatocellular cancer cell lines, but the 5-HT7 antagonist SB258719 suppressed 5-HT-induced increase in β-catenin levels and cell viability. Furthermore, SB258719 also suppressed proliferation and tumour growth. When small-intestinal neuroendocrine neoplasms, which are tumours derived from enterochromaffin cells, metastasize to the liver, the liver produces 5-HT, resulting in hyperactivation of 5-HT7 in the liver [20]. Hyperactivation of 5-HT7 in the liver leads to secretion of the growth factor IGF-1, which accelerates proliferation of metastatic tumour cells. Based on these findings, 5-HT7 inhibition is considered to be a candidate target for the treatment of hepatic metastasis cancers. It has been reported that 5-HT7 expression increased in certain types of breast cancers; this increase was amplified with tumour grade [154]. Indeed, SB269970 suppressed tumour formation in an MDA-MB-231 cancer cell [21].

References

  1. Bard, J.A.; Zgombick, J.; Adham, N.; Vaysse, P.; Branchek, T.A.; Weinshank, R.L. Cloning of a novel human serotonin receptor (5-ht7) positively linked to adenylate cyclase. J. Biol. Chem. 1993, 268, 23422–23426.
  2. Lovenberg, T.W.; Baron, B.M.; de Lecea, L.; Miller, J.D.; Prosser, R.A.; Rea, M.A.; Foye, P.E.; Racke, M.; Slone, A.L.; Siegel, B.W. A novel adenylyl cyclase-activating serotonin receptor (5-ht7) implicated in the regulation of mammalian circadian rhythms. Neuron 1993, 11, 449–458.
  3. Ruat, M.; Traiffort, E.; Leurs, R.; Tardivel-Lacombe, J.; Diaz, J.; Arrang, J.-M.; Schwartz, J.-C. Molecular cloning, characterization, and localization of a high-affinity serotonin receptor (5-ht7) activating camp formation. Proc. Natl. Acad. Sci. USA 1993, 90, 8547–8551.
  4. Shen, Y.; Monsma, F.; Metcalf, M.; Jose, P.; Hamblin, M.; Sibley, D. Molecular cloning and expression of a 5-hydroxytryptamine7 serotonin receptor subtype. J. Biol. Chem. 1993, 268, 18200–18204.
  5. Blattner, K.M.; Canney, D.J.; Pippin, D.A.; Blass, B.E. Pharmacology and therapeutic potential of the 5-ht7 receptor. ACS Chem. Neurosci. 2018, 10, 89–119.
  6. Gellynck, E.; Heyninck, K.; Andressen, K.W.; Haegeman, G.; Levy, F.O.; Vanhoenacker, P.; Van Craenenbroeck, K. The serotonin 5-ht 7 receptors: Two decades of research. Exp. Brain Res. 2013, 230, 555–568.
  7. Matthys, A.; Haegeman, G.; Van Craenenbroeck, K.; Vanhoenacker, P. Role of the 5-ht7 receptor in the central nervous system: From current status to future perspectives. Mol. Neurobiol. 2011, 43, 228–253.
  8. L’Estrade, E.T.; Erlandsson, M.; Edgar, F.G.; Ohlsson, T.; Knudsen, G.M.; Herth, M.M. Towards selective cns pet imaging of the 5-ht7 receptor system: Past, present and future. Neuropharmacology 2020, 172, 107830.
  9. Lippiello, P.; Hoxha, E.; Speranza, L.; Volpicelli, F.; Ferraro, A.; Leopoldo, M.; Lacivita, E.; Perrone-Capano, C.; Tempia, F.; Miniaci, M.C. The 5-ht7 receptor triggers cerebellar long-term synaptic depression via pkc-mapk. Neuropharmacology 2016, 101, 426–438.
  10. Volpicelli, F.; Speranza, L.; di Porzio, U.; Crispino, M.; Perrone-Capano, C. The serotonin receptor 7 and the structural plasticity of brain circuits. Front. Behav. Neurosci. 2014, 8, 318.
  11. Gocho, Y.; Sakai, A.; Yanagawa, Y.; Suzuki, H.; Saitow, F. Electrophysiological and pharmacological properties of gabaergic cells in the dorsal raphe nucleus. J. Physiol. Sci. 2013, 63, 147–154.
  12. Okada, M.; Fukuyama, K.; Okubo, R.; Shiroyama, T.; Ueda, Y. Lurasidone sub-chronically activates serotonergic transmission via desensitization of 5-ht1a and 5-ht7 receptors in dorsal raphe nucleus. Pharmaceuticals 2019, 12, 149.
  13. Nativio, P.; Zoratto, F.; Romano, E.; Lacivita, E.; Leopoldo, M.; Pascale, E.; Passarelli, F.; Laviola, G.; Adriani, W. Stimulation of 5-ht 7 receptor during adolescence determines its persistent upregulation in adult rat forebrain areas. Synapse 2015, 69, 533–542.
  14. Aubert, Y.; Allers, K.A.; Sommer, B.; de Kloet, E.R.; Abbott, D.H.; Datson, N.A. Brain regionspecific transcriptomic markers of serotonin1a receptor agonist action mediating sexual rejection and aggression in female marmoset monkeys. J. Sex. Med. 2013, 10, 1461–1475.
  15. Zareifopoulos, N.; Papatheodoropoulos, C. Effects of 5-ht-7 receptor ligands on memory and cognition. Neurobiol. Learn. Mem. 2016, 136, 204–209.
  16. Okubo, R.; Hasegawa, T.; Fukuyama, K.; Shiroyama, T.; Okada, M. Current limitations and candidate potential of 5-ht7 receptor antagonism in psychiatric pharmacotherapy. Front. Psychiatry 2021, 12, 623684.
  17. Bonaventure, P.; Nepomuceno, D.; Hein, L.; Sutcliffe, J.G.; Lovenberg, T.; Hedlund, P.B. Radioligand binding analysis of knockout mice reveals 5-hydroxytryptamine(7) receptor distribution and uncovers 8-hydroxy-2-(di-n-propylamino)tetralin interaction with alpha(2) adrenergic receptors. Neuroscience 2004, 124, 901–911.
  18. Fatima, S.; Shi, X.; Lin, Z.; Chen, G.Q.; Pan, X.H.; Wu, J.C.; Ho, J.W.; Lee, N.P.; Gao, H.; Zhang, G.; et al. 5-hydroxytryptamine promotes hepatocellular carcinoma proliferation by influencing beta-catenin. Mol. Oncol. 2016, 10, 195–212.
  19. Tzirogiannis, K.N.; Kourentzi, K.T.; Zyga, S.; Papalimneou, V.; Tsironi, M.; Grypioti, A.D.; Protopsaltis, I.; Panidis, D.; Panoutsopoulos, G.I. Effect of 5-ht7 receptor blockade on liver regeneration after 60–70% partial hepatectomy. BMC Gastroenterol. 2014, 14, 201.
  20. Svejda, B.; Kidd, M.; Timberlake, A.; Harry, K.; Kazberouk, A.; Schimmack, S.; Lawrence, B.; Pfragner, R.; Modlin, I.M. Serotonin and the 5-ht7 receptor: The link between hepatocytes, igf-1 and small intestinal neuroendocrine tumors. Cancer Sci. 2013, 104, 844–855.
  21. Gautam, J.; Banskota, S.; Regmi, S.C.; Ahn, S.; Jeon, Y.H.; Jeong, H.; Kim, S.J.; Nam, T.G.; Jeong, B.S.; Kim, J.A. Tryptophan hydroxylase 1 and 5-ht(7) receptor preferentially expressed in triple-negative breast cancer promote cancer progression through autocrine serotonin signaling. Mol. Cancer 2016, 15, 75.
  22. Hedlund, P.B.; Huitron-Resendiz, S.; Henriksen, S.J.; Sutcliffe, J.G. 5-ht7 receptor inhibition and inactivation induce antidepressantlike behavior and sleep pattern. Biol. Psychiatry 2005, 58, 831–837.
  23. Wesolowska, A.; Nikiforuk, A.; Stachowicz, K.; Tatarczynska, E. Effect of the selective 5-ht7 receptor antagonist sb 269970 in animal models of anxiety and depression. Neuropharmacology 2006, 51, 578–586.
  24. Schmidt, S.; Furini, C.; Zinn, C.; Cavalcante, L.; Ferreira, F.; Behling, J.; Myskiw, J.; Izquierdo, I. Modulation of the consolidation and reconsolidation of fear memory by three different serotonin receptors in hippocampus. Neurobiol. Learn. Mem. 2017, 142, 48–54.
  25. 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.
  26. Costa, L.; Spatuzza, M.; D’Antoni, S.; Bonaccorso, C.M.; Trovato, C.; Musumeci, S.A.; Leopoldo, M.; Lacivita, E.; Catania, M.V.; Ciranna, L. Activation of 5-ht7 serotonin receptors reverses metabotropic glutamate receptor-mediated synaptic plasticity in wild-type and fmr1 knockout mice, a model of fragile x syndrome. Biol. Psychiatry 2012, 72, 924–933.
  27. Ishibashi, T.; Horisawa, T.; Tokuda, K.; Ishiyama, T.; Ogasa, M.; Tagashira, R.; Matsumoto, K.; Nishikawa, H.; Ueda, Y.; Toma, S.; et al. Pharmacological profile of lurasidone, a novel antipsychotic agent with potent 5-hydroxytryptamine 7 (5-ht7) and 5-ht1a receptor activity. J. Pharmacol. Exp. Ther. 2010, 334, 171–181.
  28. Shapiro, D.A.; Renock, S.; Arrington, E.; Chiodo, L.A.; Liu, L.X.; Sibley, D.R.; Roth, B.L.; Mailman, R. Aripiprazole, a novel atypical antipsychotic drug with a unique and robust pharmacology. Neuropsychopharmacology 2003, 28, 1400–1411.
  29. Ghanizadeh, A.; Sahraeizadeh, A.; Berk, M. A head-to-head comparison of aripiprazole and risperidone for safety and treating autistic disorders, a randomized double blind clinical trial. Child Psychiatry Hum. Dev. 2014, 45, 185–192.
  30. Maeda, K.; Sugino, H.; Akazawa, H.; Amada, N.; Shimada, J.; Futamura, T.; Yamashita, H.; Ito, N.; McQuade, R.D.; Mork, A.; et al. Brexpiprazole i: In vitro and in vivo characterization of a novel serotonin-dopamine activity modulator. J. Pharmacol. Exp. Ther. 2014, 350, 589–604.
  31. Su, T.P.; Malhotra, A.K.; Hadd, K.; Breier, A.; Pickar, D. D2 dopamine receptor occupancy: A crossover comparison of risperidone with clozapine therapy in schizophrenic patients. Arch. Gen. Psychiatry 1997, 54, 972–973.
  32. Meltzer, H.Y. The mechanism of action of novel antipsychotic drugs. Schizophr. Bull. 1991, 17, 263–287.
  33. Fernandez, J.; Alonso, J.M.; Andres, J.I.; Cid, J.M.; Diaz, A.; Iturrino, L.; Gil, P.; Megens, A.; Sipido, V.K.; Trabanco, A.A. Discovery of new tetracyclic tetrahydrofuran derivatives as potential broad-spectrum psychotropic agents. J. Med. Chem. 2005, 48, 1709–1712.
  34. Elmaci, I.; Altinoz, M.A. Targeting the cellular schizophrenia. Likely employment of the antipsychotic agent pimozide in treatment of refractory cancers and glioblastoma. Crit. Rev. Oncol./Hematol. 2018, 128, 96–109.
  35. Lopez-Munoz, F.; Alamo, C. Active metabolites as antidepressant drugs: The role of norquetiapine in the mechanism of action of quetiapine in the treatment of mood disorders. Front. Psychiatry 2013, 4, 102.
  36. Smith, C.; Rahman, T.; Toohey, N.; Mazurkiewicz, J.; Herrick-Davis, K.; Teitler, M. Risperidone irreversibly binds to and inactivates the h5-ht7 serotonin receptor. Mol. Pharmacol. 2006, 70, 1264–1270.
  37. Schotte, A.; Janssen, P.F.; Gommeren, W.; Luyten, W.H.; Van Gompel, P.; Lesage, A.S.; De Loore, K.; Leysen, J.E. Risperidone compared with new and reference antipsychotic drugs: In vitro and in vivo receptor binding. Psychopharmacology 1996, 124, 57–73.
  38. Bang-Andersen, B.; Ruhland, T.; Jorgensen, M.; Smith, G.; Frederiksen, K.; Jensen, K.G.; Zhong, H.; Nielsen, S.M.; Hogg, S.; Mork, A.; et al. Discovery of 1-piperazine (lu aa21004): A novel multimodal compound for the treatment of major depressive disorder. J. Med. Chem. 2011, 54, 3206–3221.
  39. Okada, M.; Okubo, R.; Fukuyama, K. Vortioxetine subchronically activates serotonergic transmission via desensitization of serotonin 5-ht1a receptor with 5-ht3 receptor inhibition in rats. Int. J. Mol. Sci. 2019, 20, 6235.
  40. Okada, M.; Fukuyama, K.; Ueda, Y. Lurasidone inhibits nmda/glutamate antagonist-induced functional abnormality of thalamocortical glutamatergic transmission via 5-ht7 receptor blockade. Br. J. Pharmacol. 2019, 176, 4002–4018.
  41. Okada, M.; Matsumoto, R.; Yamamoto, Y.; Fukuyama, K. Effects of subchronic administrations of vortioxetine, lurasidone and escitalopram on thalamocortical glutamatergic transmission associated with serotonin 5-ht7 receptor. Int. J. Mol. Sci. 2021, 22, 1351.
  42. Fukuyama, K.; Motomura, E.; Shiroyama, T.; Okada, M. Impact of 5-ht7 receptor inverse agonism of lurasidone on monoaminergic tripartite synaptic transmission and pathophysiology of lower risk of weight gain. Biomed. Pharmacother. 2022, 148, 112750.
  43. Bymaster, F.P.; Calligaro, D.O.; Falcone, J.F.; Marsh, R.D.; Moore, N.A.; Tye, N.C.; Seeman, P.; Wong, D.J. Radioreceptor binding profile of the atypical antipsychotic olanzapine. Neuropsychopharmacology 1996, 14, 87–96.
  44. Belenky, M.A.; Pickard, G.E. Subcellular distribution of 5-ht1b and 5-ht7 receptors in the mouse suprachiasmatic nucleus. J. Comp. Neurol. 2001, 432, 371–388.
  45. Rusak, B.; Zucker, I. Neural regulation of circadian rhythms. Physiol. Rev. 1979, 59, 449–526.
  46. Shiroyama, T.; Fukuyama, K.; Okada, M. Distinct effects of escitalopram and vortioxetine on astroglial l-glutamate release associated with connexin43. Int. J. Mol. Sci. 2021, 22, 10013.
  47. Mahe, C.; Loetscher, E.; Feuerbach, D.; Muller, W.; Seiler, M.P.; Schoeffter, P. Differential inverse agonist efficacies of sb-258719, sb-258741 and sb-269970 at human recombinant serotonin 5-ht7 receptors. Eur. J. Pharmacol. 2004, 495, 97–102.
  48. Hagan, J.J.; Price, G.W.; Jeffrey, P.; Deeks, N.J.; Stean, T.; Piper, D.; Smith, M.I.; Upton, N.; Medhurst, A.D.; Middlemiss, D.N. Characterization of sb-269970-a, a selective 5-ht7 receptor antagonist. Br. J. Pharmacol. 2000, 130, 539–548.
  49. Winokur, A.; Gary, K.A.; Rodner, S.; Rae-Red, C.; Fernando, A.T.; Szuba, M.P. Depression, sleep physiology, and antidepressant drugs. Depress. Anxiety 2001, 14, 19–28.
  50. Guscott, M.; Bristow, L.J.; Hadingham, K.; Rosahl, T.W.; Beer, M.S.; Stanton, J.A.; Bromidge, F.; Owens, A.P.; Huscroft, I.; Myers, J.; et al. Genetic knockout and pharmacological blockade studies of the 5-ht7 receptor suggest therapeutic potential in depression. Neuropharmacology 2005, 48, 492–502.
  51. Bonaventure, P.; Kelly, L.; Aluisio, L.; Shelton, J.; Lord, B.; Galici, R.; Miller, K.; Atack, J.; Lovenberg, T.W.; Dugovic, C. Selective blockade of 5-hydroxytryptamine (5-ht)7 receptors enhances 5-ht transmission, antidepressant-like behavior, and rapid eye movement sleep suppression induced by citalopram in rodents. J. Pharmacol. Exp. Ther. 2007, 321, 690–698.
  52. Wesolowska, A.; Tatarczynska, E.; Nikiforuk, A.; Chojnacka-Wojcik, E. Enhancement of the anti-immobility action of antidepressants by a selective 5-ht7 receptor antagonist in the forced swimming test in mice. Eur. J. Pharmacol. 2007, 555, 43–47.
  53. Wesolowska, A.; Kowalska, M. Influence of serotonin 5-ht(7) receptor blockade on the behavioral and neurochemical effects of imipramine in rats. Pharmacol. Rep. PR 2008, 60, 464–474.
  54. Mnie-Filali, O.; Faure, C.; Lambas-Senas, L.; El Mansari, M.; Belblidia, H.; Gondard, E.; Etievant, A.; Scarna, H.; Didier, A.; Berod, A.; et al. Pharmacological blockade of 5-ht7 receptors as a putative fast acting antidepressant strategy. Neuropsychopharmacology 2011, 36, 1275–1288.
  55. Wesolowska, A.; Nikiforuk, A.; Stachowicz, K. Potential anxiolytic and antidepressant effects of the selective 5-ht7 receptor antagonist sb 269970 after intrahippocampal administration to rats. Eur. J. Pharmacol. 2006, 553, 185–190.
  56. Zhang, Q.J.; Du, C.X.; Tan, H.H.; Zhang, L.; Li, L.B.; Zhang, J.; Niu, X.L.; Liu, J. Activation and blockade of serotonin7 receptors in the prelimbic cortex regulate depressive-like behaviors in a 6-hydroxydopamine-induced parkinson’s disease rat model. Neuroscience 2015, 311, 45–55.
  57. Han, L.N.; Zhang, L.; Sun, Y.N.; Du, C.X.; Zhang, Y.M.; Wang, T.; Zhang, J.; Liu, J. Serotonin7 receptors in the lateral habenular nucleus regulate depressive-like behaviors in the hemiparkinsonian rats. Brain Res. 2016, 1644, 79–87.
  58. Yau, J.L.; Noble, J.; Seckl, J.R. Acute restraint stress increases 5-ht7 receptor mrna expression in the rat hippocampus. Neurosci. Lett. 2001, 309, 141–144.
  59. Roberts, A.J.; Krucker, T.; Levy, C.L.; Slanina, K.A.; Sutcliffe, J.G.; Hedlund, P.B. Mice lacking 5-ht receptors show specific impairments in contextual learning. Eur. J. Neurosci. 2004, 19, 1913–1922.
  60. Clemett, D.A.; Cockett, M.I.; Marsden, C.A.; Fone, K.C. Antisense oligonucleotide-induced reduction in 5-hydroxytryptamine7 receptors in the rat hypothalamus without alteration in exploratory behaviour or neuroendocrine function. J. Neurochem. 1998, 71, 1271–1279.
  61. Hedlund, P.B.; Sutcliffe, J.G. The 5-ht7 receptor influences stereotypic behavior in a model of obsessive-compulsive disorder. Neurosci. Lett. 2007, 414, 247–251.
  62. Semenova, S.; Geyer, M.A.; Sutcliffe, J.G.; Markou, A.; Hedlund, P.B. Inactivation of the 5-ht(7) receptor partially blocks phencyclidine-induced disruption of prepulse inhibition. Biol. Psychiatry 2008, 63, 98–105.
  63. Waters, K.A.; Stean, T.O.; Hammond, B.; Virley, D.J.; Upton, N.; Kew, J.N.; Hussain, I. Effects of the selective 5-ht(7) receptor antagonist sb-269970 in animal models of psychosis and cognition. Behav. Brain Res. 2012, 228, 211–218.
  64. Galici, R.; Boggs, J.D.; Miller, K.L.; Bonaventure, P.; Atack, J.R. Effects of sb-269970, a 5-ht7 receptor antagonist, in mouse models predictive of antipsychotic-like activity. Behav. Pharmacol. 2008, 19, 153–159.
  65. Nikiforuk, A.; Kos, T.; Fijal, K.; Holuj, M.; Rafa, D.; Popik, P. Effects of the selective 5-ht7 receptor antagonist sb-269970 and amisulpride on ketamine-induced schizophrenia-like deficits in rats. PLoS ONE 2013, 8, e66695.
  66. Pouzet, B. Sb-258741: A 5-ht7 receptor antagonist of potential clinical interest. CNS Drug Rev. 2002, 8, 90–100.
  67. Liy-Salmeron, G.; Meneses, A. Effects of 5-ht drugs in prefrontal cortex during memory formation and the ketamine amnesia-model. Hippocampus 2008, 18, 965–974.
  68. 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.
  69. 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.
  70. Nikiforuk, A.; Holuj, M.; Potasiewicz, A.; Popik, P. Effects of the selective 5-ht7 receptor antagonist sb-269970 on premature responding in the five-choice serial reaction time test in rats. Behav. Brain Res. 2015, 289, 149–156.
  71. Huhn, M.; Nikolakopoulou, A.; Schneider-Thoma, J.; Krause, M.; Samara, M.; Peter, N.; Arndt, T.; Backers, L.; Rothe, P.; Cipriani, A.; et al. Comparative efficacy and tolerability of 32 oral antipsychotics for the acute treatment of adults with multi-episode schizophrenia: A systematic review and network meta-analysis. Lancet 2019, 394, 939–951.
  72. Krause, M.; Zhu, Y.; Huhn, M.; Schneider-Thoma, J.; Bighelli, I.; Chaimani, A.; Leucht, S. Efficacy, acceptability, and tolerability of antipsychotics in children and adolescents with schizophrenia: A network meta-analysis. Eur. Neuropsychopharmacol. J. Eur. Coll. Neuropsychopharmacol. 2018, 28, 659–674.
  73. Kishi, T.; Ikuta, T.; Sakuma, K.; Okuya, M.; Iwata, N. Efficacy and safety of antipsychotic treatments for schizophrenia: A systematic review and network meta-analysis of randomized trials in japan. J. Psychiatr. Res. 2021, 138, 444–452.
  74. Phalguni, A.; McCool, R.; Wood, H.; Sanderson, A.; Rydevik, G.; Franklin, B.; James, D. Systematic literature review and network meta-analysis of lurasidone, brexpiprazole and cariprazine for schizophrenia. Int. Clin. Psychopharmacol. 2023, 38, 45–56.
  75. Ostacher, M.; Ng-Mak, D.; Patel, P.; Ntais, D.; Schlueter, M.; Loebel, A. Lurasidone compared to other atypical antipsychotic monotherapies for bipolar depression: A systematic review and network meta-analysis. World J. Biol. Psychiatry Off. J. World Fed. Soc. Biol. Psychiatry 2018, 19, 586–601.
  76. Bahji, A.; Ermacora, D.; Stephenson, C.; Hawken, E.R.; Vazquez, G. Comparative efficacy and tolerability of pharmacological treatments for the treatment of acute bipolar depression: A systematic review and network meta-analysis. J. Affect. Disord. 2020, 269, 154–184.
  77. Kishi, T.; Ikuta, T.; Matsuda, Y.; Sakuma, K.; Okuya, M.; Mishima, K.; Iwata, N. Mood stabilizers and/or antipsychotics for bipolar disorder in the maintenance phase: A systematic review and network meta-analysis of randomized controlled trials. Mol. Psychiatry 2021, 26, 4146–4157.
  78. Loebel, A.; Cucchiaro, J.; Silva, R.; Kroger, H.; Hsu, J.; Sarma, K.; Sachs, G. Lurasidone monotherapy in the treatment of bipolar i depression: A randomized, double-blind, placebo-controlled study. Am. J. Psychiatry 2014, 171, 160–168.
  79. Loebel, A.; Cucchiaro, J.; Silva, R.; Kroger, H.; Sarma, K.; Xu, J.; Calabrese, J.R. Lurasidone as adjunctive therapy with lithium or valproate for the treatment of bipolar i depression: A randomized, double-blind, placebo-controlled study. Am. J. Psychiatry 2014, 171, 169–177.
  80. Sajatovic, M.; Forester, B.P.; Tsai, J.; Kroger, H.; Pikalov, A.; Cucchiaro, J.; Loebel, A. Efficacy of lurasidone in adults aged 55 years and older with bipolar depression: Post hoc analysis of 2 double-blind, placebo-controlled studies. J. Clin. Psychiatry 2016, 77, e1324–e1331.
  81. Suppes, T.; Silva, R.; Cucchiaro, J.; Mao, Y.; Targum, S.; Streicher, C.; Pikalov, A.; Loebel, A. Lurasidone for the treatment of major depressive disorder with mixed features: A randomized, double-blind, placebo-controlled study. Am. J. Psychiatry 2016, 173, 400–407.
  82. Tsai, J.; Thase, M.E.; Mao, Y.; Ng-Mak, D.; Pikalov, A.; Loebel, A. Lurasidone for major depressive disorder with mixed features and anxiety: A post-hoc analysis of a randomized, placebo-controlled study. CNS Spectr. 2017, 22, 236–245.
  83. Vieta, E.; Florea, I.; Schmidt, S.N.; Areberg, J.; Ettrup, A. Intravenous vortioxetine to accelerate onset of effect in major depressive disorder: A 2-week, randomized, double-blind, placebo-controlled study. Int. Clin. Psychopharmacol. 2019, 34, 153–160.
  84. Heidmann, D.E.; Szot, P.; Kohen, R.; Hamblin, M.W. Function and distribution of three rat 5-hydroxytryptamine7 (5-ht7) receptor isoforms produced by alternative splicing. Neuropharmacology 1998, 37, 1621–1632.
  85. Heidmann, D.E.; Metcalf, M.A.; Kohen, R.; Hamblin, M.W. Four 5-hydroxytryptamine7 (5-ht7) receptor isoforms in human and rat produced by alternative splicing: Species differences due to altered intron-exon organization. J. Neurochem. 1997, 68, 1372–1381.
  86. Krobert, K.A.; Bach, T.; Syversveen, T.; Kvingedal, A.M.; Levy, F.O. The cloned human 5-ht7 receptor splice variants: A comparative characterization of their pharmacology, function and distribution. Naunyn Schmiedebergs Arch. Pharmacol. 2001, 363, 620–632.
  87. Andressen, K.W.; Manfra, O.; Brevik, C.H.; Ulsund, A.H.; Vanhoenacker, P.; Levy, F.O.; Krobert, K.A. The atypical antipsychotics clozapine and olanzapine promote down-regulation and display functional selectivity at human 5-ht7 receptors. Br. J. Pharmacol. 2015, 172, 3846–3860.
  88. Guthrie, C.R.; Murray, A.T.; Franklin, A.A.; Hamblin, M.W. Differential agonist-mediated internalization of the human 5-hydroxytryptamine 7 receptor isoforms. J. Pharmacol. Exp. Ther. 2005, 313, 1003–1010.
  89. Bijata, M.; Labus, J.; Guseva, D.; Stawarski, M.; Butzlaff, M.; Dzwonek, J.; Schneeberg, J.; Bohm, K.; Michaluk, P.; Rusakov, D.A.; et al. Synaptic remodeling depends on signaling between serotonin receptors and the extracellular matrix. Cell Rep. 2017, 19, 1767–1782.
  90. Kvachnina, E.; Liu, G.; Dityatev, A.; Renner, U.; Dumuis, A.; Richter, D.W.; Dityateva, G.; Schachner, M.; Voyno-Yasenetskaya, T.A.; Ponimaskin, E.G. 5-ht7 receptor is coupled to g alpha subunits of heterotrimeric g12-protein to regulate gene transcription and neuronal morphology. J. Neurosci. 2005, 25, 7821–7830.
  91. Baker, L.P.; Nielsen, M.D.; Impey, S.; Metcalf, M.A.; Poser, S.W.; Chan, G.; Obrietan, K.; Hamblin, M.W.; Storm, D.R. Stimulation of type 1 and type 8 ca2+/calmodulin-sensitive adenylyl cyclases by the gs-coupled 5-hydroxytryptamine subtype 5-ht7a receptor. J Biol Chem 1998, 273, 17469–17476.
  92. Grimes, M.T.; Powell, M.; Gutierrez, S.M.; Darby-King, A.; Harley, C.W.; McLean, J.H. Epac activation initiates associative odor preference memories in the rat pup. Learn. Mem. 2015, 22, 74–82.
  93. Lin, S.L.; Johnson-Farley, N.N.; Lubinsky, D.R.; Cowen, D.S. Coupling of neuronal 5-ht7 receptors to activation of extracellular-regulated kinase through a protein kinase a-independent pathway that can utilize epac. J. Neurochem. 2003, 87, 1076–1085.
  94. Speranza, L.; Chambery, A.; Di Domenico, M.; Crispino, M.; Severino, V.; Volpicelli, F.; Leopoldo, M.; Bellenchi, G.C.; di Porzio, U.; Perrone-Capano, C. The serotonin receptor 7 promotes neurite outgrowth via erk and cdk5 signaling pathways. Neuropharmacology 2013, 67, 155–167.
  95. Costa, L.; Sardone, L.M.; Bonaccorso, C.M.; D’Antoni, S.; Spatuzza, M.; Gulisano, W.; Tropea, M.R.; Puzzo, D.; Leopoldo, M.; Lacivita, E.; et al. Activation of serotonin 5-ht7 receptors modulates hippocampal synaptic plasticity by stimulation of adenylate cyclases and rescues learning and behavior in a mouse model of fragile x syndrome. Front. Mol. Neurosci. 2018, 11, 353.
  96. Okada, M.; Fukuyama, K.; Motomura, E. Dose-dependent biphasic action of quetiapine on ampk signalling via 5-ht7 receptor: Exploring pathophysiology of clinical and adverse effects of quetiapine. Int. J. Mol. Sci. 2022, 23, 9103.
  97. Fukuyama, K.; Motomura, E.; Okada, M. Brexpiprazole reduces 5-ht7 receptor function on astroglial transmission systems. Int. J. Mol. Sci. 2022, 23, 6571.
  98. Salminen, A.; Kaarniranta, K.; Kauppinen, A. Age-related changes in ampk activation: Role for ampk phosphatases and inhibitory phosphorylation by upstream signaling pathways. Ageing Res. Rev. 2016, 28, 15–26.
  99. Ponimaskin, E.; Voyno-Yasenetskaya, T.; Richter, D.W.; Schachner, M.; Dityatev, A. Morphogenic signaling in neurons via neurotransmitter receptors and small gtpases. Mol. Neurobiol. 2007, 35, 278–287.
  100. Wirth, A.; Holst, K.; Ponimaskin, E. How serotonin receptors regulate morphogenic signalling in neurons. Prog. Neurobiol. 2017, 151, 35–56.
  101. Speranza, L.; Giuliano, T.; Volpicelli, F.; De Stefano, M.E.; Lombardi, L.; Chambery, A.; Lacivita, E.; Leopoldo, M.; Bellenchi, G.C.; di Porzio, U.; et al. Activation of 5-ht7 receptor stimulates neurite elongation through mtor, cdc42 and actin filaments dynamics. Front. Behav. Neurosci. 2015, 9, 62.
  102. Crispino, M.; Volpicelli, F.; Perrone-Capano, C. Role of the serotonin receptor 7 in brain plasticity: From development to disease. Int. J. Mol. Sci. 2020, 21, 505.
  103. Ibata, K.; Sun, Q.; Turrigiano, G.G. Rapid synaptic scaling induced by changes in postsynaptic firing. Neuron 2008, 57, 819–826.
  104. Citri, A.; Malenka, R.C. Synaptic plasticity: Multiple forms, functions, and mechanisms. Neuropsychopharmacology 2008, 33, 18–41.
  105. Beaudet, G.; Jozet-Alves, C.; Asselot, R.; Schumann-Bard, P.; Freret, T.; Boulouard, M.; Paizanis, E. Deletion of the serotonin receptor type 7 disrupts the acquisition of allocentric but not egocentric navigation strategies in mice. Behav. Brain Res. 2017, 320, 179–185.
  106. Vasefi, M.S.; Yang, K.; Li, J.; Kruk, J.S.; Heikkila, J.J.; Jackson, M.F.; MacDonald, J.F.; Beazely, M.A. Acute 5-ht7 receptor activation increases nmda-evoked currents and differentially alters nmda receptor subunit phosphorylation and trafficking in hippocampal neurons. Mol. Brain 2013, 6, 24.
  107. Bacon, W.L.; Beck, S.G. 5-hydroxytryptamine(7) receptor activation decreases slow afterhyperpolarization amplitude in ca3 hippocampal pyramidal cells. J. Pharmacol. Exp. Ther. 2000, 294, 672–679.
  108. Tokarski, K.; Zahorodna, A.; Bobula, B.; Hess, G. 5-ht7 receptors increase the excitability of rat hippocampal ca1 pyramidal neurons. Brain Res 2003, 993, 230–234.
  109. Costa, L.; Trovato, C.; Musumeci, S.A.; Catania, M.V.; Ciranna, L. 5-ht(1a) and 5-ht(7) receptors differently modulate ampa receptor-mediated hippocampal synaptic transmission. Hippocampus 2012, 22, 790–801.
  110. Andreetta, F.; Carboni, L.; Grafton, G.; Jeggo, R.; Whyment, A.D.; van den Top, M.; Hoyer, D.; Spanswick, D.; Barnes, N.M. Hippocampal 5-ht7 receptors signal phosphorylation of the glua1 subunit to facilitate ampa receptor mediated-neurotransmission in vitro and in vivo. Br. J. Pharmacol. 2016, 173, 1438–1451.
  111. Leo, D.; Adriani, W.; Cavaliere, C.; Cirillo, G.; Marco, E.; Romano, E.; Di Porzio, U.; Papa, M.; Perrone-Capano, C.; Laviola, G. Methylphenidate to adolescent rats drives enduring changes of accumbal htr7 expression: Implications for impulsive behavior and neuronal morphology. Genes Brain Behav. 2009, 8, 356–368.
  112. Ansorge, M.S.; Morelli, E.; Gingrich, J.A. Inhibition of serotonin but not norepinephrine transport during development produces delayed, persistent perturbations of emotional behaviors in mice. J. Neurosci. 2008, 28, 199–207.
  113. Beaudet, G.; Bouet, V.; Jozet-Alves, C.; Schumann-Bard, P.; Dauphin, F.; Paizanis, E.; Boulouard, M.; Freret, T. Spatial memory deficit across aging: Current insights of the role of 5-ht7 receptors. Front. Behav. Neurosci. 2014, 8, 448.
  114. Rusheen, A.E.; Gee, T.A.; Jang, D.P.; Blaha, C.D.; Bennet, K.E.; Lee, K.H.; Heien, M.L.; Oh, Y. Evaluation of electrochemical methods for tonic dopamine detection in vivo. Trends Analyt. Chem. 2020, 132, 116049.
  115. Fukuyama, K.; Fukuzawa, M.; Shiroyama, T.; Okada, M. Pathomechanism of nocturnal paroxysmal dystonia in autosomal dominant sleep-related hypermotor epilepsy with s284l-mutant alpha4 subunit of nicotinic ach receptor. Biomed. Pharmacother. 2020, 126, 110070.
  116. Fukuyama, K.; Nakano, T.; Shiroyama, T.; Okada, M. Chronic administrations of guanfacine on mesocortical catecholaminergic and thalamocortical glutamatergic transmissions. Int. J. Mol. Sci. 2021, 22, 4122.
  117. Fukuyama, K.; Hasegawa, T.; Okada, M. Cystine/glutamate antiporter and aripiprazole compensate nmda antagonist-induced dysfunction of thalamocortical l-glutamatergic transmission. Int. J. Mol. Sci. 2018, 19, 3645.
  118. Fukuyama, K.; Kato, R.; Murata, M.; Shiroyama, T.; Okada, M. Clozapine normalizes a glutamatergic transmission abnormality induced by an impaired nmda receptor in the thalamocortical pathway via the activation of a group iii metabotropic glutamate receptor. Biomolecules 2019, 9, 234.
  119. Nakano, T.; Hasegawa, T.; Suzuki, D.; Motomura, E.; Okada, M. Amantadine combines astroglial system xc(-) activation with glutamate/nmda receptor inhibition. Biomolecules 2019, 9, 191.
  120. Okada, M.; Fukuyama, K.; Kawano, Y.; Shiroyama, T.; Ueda, Y. Memantine protects thalamocortical hyper-glutamatergic transmission induced by nmda receptor antagonism via activation of system xc(). Pharmacol. Res. Perspect. 2019, 7, e00457.
  121. Okada, M.; Fukuyama, K.; Nakano, T.; Ueda, Y. Pharmacological discrimination of effects of mk801 on thalamocortical, mesothalamic, and mesocortical transmissions. Biomolecules 2019, 9, 746.
  122. Fukuyama, K.; Fukuzawa, M.; Okada, M. Upregulated and hyperactivated thalamic connexin 43 plays important roles in pathomechanisms of cognitive impairment and seizure of autosomal dominant sleep-related hypermotor epilepsy with s284l-mutant alpha4 subunit of nicotinic ach receptor. Pharmaceuticals 2020, 13, 99.
  123. Fukuyama, K.; Fukuzawa, M.; Okubo, R.; Okada, M. Upregulated connexin 43 induced by loss-of-functional s284l-mutant alpha4 subunit of nicotinic ach receptor contributes to pathomechanisms of autosomal dominant sleep-related hypermotor epilepsy. Pharmaceuticals 2020, 13, 58.
  124. Fukuyama, K.; Fukuzawa, M.; Shiroyama, T.; Okada, M. Pathogenesis and pathophysiology of autosomal dominant sleep-related hypermotor epilepsy with s284l-mutant alpha4 subunit of nicotinic ach receptor. Br. J. Pharmacol. 2020, 177, 2143–2162.
  125. Okada, M.; Fukuyama, K.; Kawano, Y.; Shiroyama, T.; Suzuki, D.; Ueda, Y. Effects of acute and sub-chronic administrations of guanfacine on catecholaminergic transmissions in the orbitofrontal cortex. Neuropharmacology 2019, 156, 107547.
  126. Okada, M.; Fukuyama, K. Interaction between mesocortical and mesothalamic catecholaminergic transmissions associated with nmda receptor in the locus coeruleus. Biomolecules 2020, 10, 990.
  127. Hwang, K.; Shine, J.M.; Bruss, J.; Tranel, D.; Boes, A. Neuropsychological evidence of multi-domain network hubs in the human thalamus. eLife 2021, 10, e69480.
  128. Hwang, K.; Shine, J.M.; Cole, M.W.; Sorenson, E. Thalamocortical contributions to cognitive task activity. eLife 2022, 11, e81282.
  129. Reber, J.; Hwang, K.; Bowren, M.; Bruss, J.; Mukherjee, P.; Tranel, D.; Boes, A.D. Cognitive impairment after focal brain lesions is better predicted by damage to structural than functional network hubs. Proc. Natl. Acad. Sci. USA 2021, 118, e2018784118.
  130. Fukuyama, K.; Okada, M. High frequency oscillations play important roles in development of epileptogenesis/ictogenesis via activation of astroglial signallings. Biomed. Pharmacother. 2022, 149, 112846.
  131. Fernandez, L.M.J.; Luthi, A. Sleep spindles: Mechanisms and functions. Physiol. Rev. 2020, 100, 805–868.
  132. Fukuyama, K.; Okada, M. Brivaracetam and levetiracetam suppress astroglial l-glutamate release through hemichannel via inhibition of synaptic vesicle protein. Int. J. Mol. Sci. 2022, 23, 4473.
  133. Yang, M.; Logothetis, N.K.; Eschenko, O. Occurrence of hippocampal ripples is associated with activity suppression in the mediodorsal thalamic nucleus. J. Neurosci. 2019, 39, 434–444.
  134. Okada, M. Can rodent models elucidate the pathomechanisms of genetic epilepsy? Br. J. Pharmacol. 2022, 179, 1620–1639.
  135. Okada, M.; Kawano, Y.; Fukuyama, K.; Motomura, E.; Shiroyama, T. Candidate strategies for development of a rapid-acting antidepressant class without neuropsychiatric adverse effects: Prevention of ketamine-induced neuropsychiatric adverse reactions. Int. J. Mol. Sci. 2020, 21, 7951.
  136. Okada, M.; Fukuyama, K.; Shiroyama, T.; Murata, M. A working hypothesis regarding identical pathomechanisms between clinical efficacy and adverse reaction of clozapine via the activation of connexin43. Int. J. Mol. Sci. 2020, 21, 7019.
  137. Fu, C.; Armstrong, D.; Marsh, E.; Lieberman, D.; Motil, K.; Witt, R.; Standridge, S.; Nues, P.; Lane, J.; Dinkel, T.; et al. Consensus guidelines on managing rett syndrome across the lifespan. BMJ Paediatr. Open 2020, 4, e000717.
  138. Neul, J.L.; Fang, P.; Barrish, J.; Lane, J.; Caeg, E.B.; Smith, E.O.; Zoghbi, H.; Percy, A.; Glaze, D.G. Specific mutations in methyl-cpg-binding protein 2 confer different severity in rett syndrome. Neurology 2008, 70, 1313–1321.
  139. Kulikovskaja, L.; Sarajlija, A.; Savic-Pavicevic, D.; Dobricic, V.; Klein, C.; Westenberger, A. Wdr45 mutations may cause a mecp2 mutation-negative rett syndrome phenotype. Neurol. Genet. 2018, 4, e227.
  140. Collins, A.L.; Levenson, J.M.; Vilaythong, A.P.; Richman, R.; Armstrong, D.L.; Noebels, J.L.; David Sweatt, J.; Zoghbi, H.Y. Mild overexpression of mecp2 causes a progressive neurological disorder in mice. Hum. Mol. Genet. 2004, 13, 2679–2689.
  141. De Filippis, B.; Nativio, P.; Fabbri, A.; Ricceri, L.; Adriani, W.; Lacivita, E.; Leopoldo, M.; Passarelli, F.; Fuso, A.; Laviola, G. Pharmacological stimulation of the brain serotonin receptor 7 as a novel therapeutic approach for rett syndrome. Neuropsychopharmacology 2014, 39, 2506–2518.
  142. De Filippis, B.; Chiodi, V.; Adriani, W.; Lacivita, E.; Mallozzi, C.; Leopoldo, M.; Domenici, M.R.; Fuso, A.; Laviola, G. Long-lasting beneficial effects of central serotonin receptor 7 stimulation in female mice modeling rett syndrome. Front. Behav. Neurosci. 2015, 9, 86.
  143. Hull, M.; Berger, M.; Heneka, M. Disease-modifying therapies in alzheimer’s disease: How far have we come? Drugs 2006, 66, 2075–2093.
  144. Lalut, J.; Karila, D.; Dallemagne, P.; Rochais, C. Modulating 5-ht(4) and 5-ht(6) receptors in alzheimer’s disease treatment. Future Med. Chem. 2017, 9, 781–795.
  145. Hashemi-Firouzi, N.; Komaki, A.; Soleimani Asl, S.; Shahidi, S. The effects of the 5-ht7 receptor on hippocampal long-term potentiation and apoptosis in a rat model of alzheimer’s disease. Brain Res. Bull. 2017, 135, 85–91.
  146. Querfurth, H.W.; LaFerla, F.M. Alzheimer’s disease. N. Engl. J. Med. 2010, 362, 329–344.
  147. Villegas, A.Q.; Manzo, H.S.A.; Almada, M.O.V.; Modragon, C.B.; Guzman, R.G. Procognitive and neuroprotective effect of 5-ht7 agonist in an animal model by icv amyloid-b injection (p1.1-005). Neurology 2019, 92, P1.1-005.
  148. Miller-Thomas, M.M.; Sipe, A.L.; Benzinger, T.L.; McConathy, J.; Connolly, S.; Schwetye, K.E. Multimodality review of amyloid-related diseases of the central nervous system. Radiographics 2016, 36, 1147–1163.
  149. Meneses, A. Neural activity, memory, and dementias: Serotonergic markers. Behav. Pharmacol. 2017, 28, 132–141.
  150. Werner, F.M.; Covenas, 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, 22, 2064–2071.
  151. Bourson, A.; Kapps, V.; Zwingelstein, C.; Rudler, A.; Boess, F.G.; Sleight, A.J. Correlation between 5-ht7 receptor affinity and protection against sound-induced seizures in dba/2j mice. Naunyn Schmiedebergs Arch. Pharmacol. 1997, 356, 820–826.
  152. Graf, M.; Jakus, R.; Kantor, S.; Levay, G.; Bagdy, G. Selective 5-ht1a and 5-ht7 antagonists decrease epileptic activity in the wag/rij rat model of absence epilepsy. Neurosci. Lett. 2004, 359, 45–48.
  153. Yang, Z.; Liu, X.; Yin, Y.; Sun, S.; Deng, X. Involvement of 5-ht(7) receptors in the pathogenesis of temporal lobe epilepsy. Eur. J. Pharmacol. 2012, 685, 52–58.
  154. Pai, V.P.; Marshall, A.M.; Hernandez, L.L.; Buckley, A.R.; Horseman, N.D. Altered serotonin physiology in human breast cancers favors paradoxical growth and cell survival. Breast Cancer Res. 2009, 11, R81.
More
Information
Contributors MDPI registered users' name will be linked to their SciProfiles pages. To register with us, please refer to https://encyclopedia.pub/register : , ,
View Times: 355
Revisions: 2 times (View History)
Update Date: 24 Mar 2023
1000/1000
Video Production Service