Skip to main content
NIHPA Author Manuscripts logoLink to NIHPA Author Manuscripts
. Author manuscript; available in PMC: 2012 Feb 20.
Published in final edited form as: Curr Opin Pharmacol. 2011 Feb 20;11(1):75–80. doi: 10.1016/j.coph.2011.01.010

Allosteric modulation of the 5-HT3 receptor

Paul Andrew Davies 1
PMCID: PMC3072441  NIHMSID: NIHMS273361  PMID: 21342788

Abstract

5-Hydroxytryptamine type 3 (5-HT3) receptors are ligand-gated ion channels that play important roles in depression, anxiety, substance abuse, emesis, inflammatory pain, spinal nociception, gastrointestinal function, and cardiovascular reflexes. Probably the most studied modulators of 5-HT3 receptors are the high affinity competitive ‘setron’ antagonists typified by ondansetron. However, there exists a broad range of compounds that modulate the 5-HT3 receptor, not through the orthosteric site but by binding to allosteric sites. Most notable are therapeutic compounds ascribed to certain targets but that allosterically modulate 5-HT3 receptors at clinically relevant concentrations.

Keywords: anesthetics, alcohols, antidepressants, antipsychotis, cannabinoids

Introduction

The 5-HT3 receptor is a member of the Cys-loop ligand-gated ion channel (LGIC) superfamily which includes the nicotinic acetylcholine, γ-aminobutyric acid type A (GABAA), glycine and ZAC receptor ion channels [1]. Five genes encoding for individual subunits (5-HT3A-E) have been identified within the human genome [2*]. However, current knowledge of 5-HT3 receptor pharmacology is heavily biased towards information from human and rodent homomeric 5-HT3A receptors expressed in recombinant cells or native 5-HT3 receptors of unknown subunit composition expressed in rodent cells (rodents have genes only for 5-HT3A and 5-HT3B subunits).

Agonists and positive modulators of 5-HT3 receptors are not favorable for clinical use because of enhanced anxiety and pro-emetic effects. In contrast, antagonists are successfully used to treat chemotherapy-induced vomiting, post-operative nausea and vomiting (PONV), alcohol craving and irritable bowel syndrome with diarrhea. Modulators of 5-HT3 receptors have also been suggested to have therapeutic relevance for schizophrenia, anxiety, cognition and nociception [3].

Most members of the LGIC superfamily have several different subunits that can assemble to form multiple receptor subtypes. Each subunit structurally influences channel properties; hence heteromeric receptors provide diversity in pharmacological and biophysical properties within a receptor family. Allosteric modulators bind to a site distinct from the orthosteric site (the endogenous agonist binding site) but alter receptor conformations that influence the binding and/or functional properties of agonists binding to the orthosteric site. Compared to homomeric receptors, heteromeric receptors contain an increased number of potential allosteric sites for drug interaction.

This review examines clinically relevant allosteric modulators of 5-HT3 receptors, and focuses on compounds that modulate 5-HT3 receptors at physiological concentrations. The current list of compounds is based on work performed almost entirely on homomeric 5-HT3A receptors. With the existence of five genes encoding for 5-HT3 receptor subunits in the human genome, it is inevitable that the known pharmacology of the listed allosteric compounds will change when tested against novel 5-HT3 subunit combinations.

Alcohols and volatile anesthetics

Alcohols and volatile anesthetics can produce an assortment of behavioral effects. Numerous targets have been proposed to explain the effects of alcohols and anesthetics, including the 5-HT3 receptor. Activation of 5-HT3 receptors enhances the release of the neurotransmitters dopamine (DA) and γ-aminobutyric acid (GABA), which are believed to be the principal neurotransmitters for addiction and intoxication respectively. 5-HT3 receptor antagonists reduce the halothane-mediated inhibition of spinal sensory neuronal responses to noxious peripheral stimulation, indicating that 5-HT3 receptors are anesthetic targets for the reduction in nociception [4]. In addition, 5-HT3 receptor antagonists are used to prevent and treat PONV, which has a strong association with the use of halogenated volatile anesthetics [5], implicating a possible interaction with 5-HT3 receptors.

For human 5-HT3A receptors, currents evoked by low concentrations of 5-HT were enhanced by compounds of smaller molecular volumes (<120 Å3) such as butanol and halothane, whereas larger compounds (e.g. octanol, hexanol and sevoflurane) inhibited such currents [6]. The smaller compounds shifted the 5-HT concentration-response relationship to the left, reflecting an increase in the 5-HT3A receptors’ sensitivity to agonist. Compared with 5-HT3A receptors, the current amplitude enhancement by butanol and pentanol was significantly less in 5-HT3AB receptors with no shift in 5-HT EC50 values, yet the two receptor subtypes were equally sensitive to the inhibitory action of octanol [7]. Similarly, Hayrapetyan et al. observed that, when compared to rodent 5-HT3A receptors, 5-HT3AB receptors were much less sensitive to the enhancing action of ethanol [8]. The smaller anesthetics and alcohols enhance the 5-HT3A receptors’ channel gating efficacy without altering the agonist binding affinity. Incorporation of the 5-HT3B subunit reduces this enhancement of gating efficacy [910].

The increase in channel gating by alcohols strongly suggests that the open state of the channel has been stabilized. However, due to the low single-channel conductance of the 5-HT3A receptor the direct observation of single open channels was not possible until recently. Using a triple mutant receptor (5-HT3A(QDA)) that forms homomeric channels exhibiting a measurable single-channel conductance [1112] the stability of the open channel by ethanol was confirmed (Figure 1). By examining the different open states evoked by saturating 5-HT in the absence and presence of ethanol the prolonged open state appeared to come at the expense of the desensitized state [13**]. This prolongation of the open state of the channel at the expense of the desensitized state is consistent with the slowed desensitization effect of ethanol and trichloroethanol on macroscopic currents [1415].

Figure 1. Ethanol increases the number of open single-channel events.

Figure 1

Left, Cell-attached recordings of human 5-HT3A(QDA)-mediated currents evoked by 100 μM 5-HT in the absence (A) or presence (B) of 25 mM ethanol. Twenty second examples of single-channel activity with an expanded view of a burst (left panel). Dotted lines indicating closed and open states. Right, all-points histograms for the 20 s recordings of 5-HT3A(QDA)-mediated currents evoked by 100 μM 5-HT shown in left panel. Histograms were fitted by two Gaussians, percentages of events in the closed and open states are shown. Figure reproduced, with permission, from Feinberg-Zadek and Davies [13].

Intravenous anesthetics

In comparison to the potentiation of 5-HT-evoked currents by volatile anesthetics of low molecular volume, intravenous anesthetics have an inhibitory effect on 5-HT3 receptor channels. Various experimental approaches such as 5-HT3 receptor-mediated depolarizations of the rat isolated vagus nerve, currents recorded from oocytes and HEK293 cells expressing 5-HT3 receptors or rodent cell lines with native 5-HT3 receptors have been employed to study intravenous anesthetics modulation of 5-HT3 receptors. In all the experimental approaches, the IC50 values for barbiturate anesthetics, etomidate and propofol are significantly higher than the clinical anesthetic concentration [1617].

At clinically relevant concentrations, the dissociative anesthetic ketamine (approximately 10μM, [18]) has been reported to enhance 5-HT3 receptor-mediated currents in rabbit neurons [19]. However, an inhibition at higher concentrations was also reported. Conversely, no modulation of 5-HT3 receptor-evoked currents was observed with clinically relevant concentrations in neurons from guinea pig or from mouse 5-HT3A receptors stably expressed in HEK293 cells, suggesting a species-dependent effect [20].

Antidepressants and antipsychotics

The therapeutic endpoints of antidepressants are often stated to increase the levels of neurotransmitters by various mechanisms. However, there is increasing awareness that antidepressants associate with 5-HT3 receptors, resulting in an allosteric inhibition. Fan [21] showed in rat neurons that imipramine (tricyclic), fluoxetine (selective serotonin reuptake inhibitor, SSRI), phenelzine and iproniazid (monoamine oxidase inhibitors, MAOI) inhibit 5-HT3 receptor-mediated currents.

At low, clinically relevant concentrations fluoxetine, imipramine and doxepin (tricyclic) inhibit 5-HT3A receptors in both the open and closed conformational states with the block in the closed state resulting in a greater inhibition [2223*]. The major metabolite of fluoxetine - norfluoxetine - is a more potent blocker than fluoxetine [22]. Fluoxetine, imipramine, phenelzine and iproniazid also accelerate desensitization [2122].

Activation of presynaptic 5-HT3 receptors on dopaminergic neurons of the mesolimbic system enhances dopamine release. Accordingly, antagonists of 5-HT3 receptors may hold novel antipsychotic properties due to the dopamine hypothesis for schizophrenia. Current antipsychotic compounds have already been described to block 5-HT3 receptors at clinically relevant concentrations. For human 5-HT3A receptors expressed in HEK293 cells and endogenous receptors expressed in murine N1E-115 cells, clozapine blocked in a competitive manner but flupentixol, fluphenazine, haloperidol, levomepromazine, and thioridazine acted as allosteric inhibitors [24].

Antidepressants (desipramine, fluoxetine and reboxetine), antipsychotics (fluphenazine, haloperidol and clozapine) and 5-HT3 receptors have been shown to co-localize in lipid raft domains. This was initially thought to imply that lipid rafts were crucial for the antagonistic properties of these compounds [25]. However, further studies have shown that disruption of lipid raft integrity altered 5-HT3 receptor kinetics and current amplitude while block of 5-HT3 receptors by desipramine and fluoxetine remained unaltered [26].

Cannabinoids

Both natural (Δ9-THC and anandamide) and synthetic (WIN55,212-2, JWH-015 and CP55940) cannabinoids have been shown to efficiently suppress nausea and vomiting and to exhibit analgesic properties [27]. However, these cannabinoid compounds have limited therapeutic potential due to their psychotropic actions mediated by their agonistic profile at cannabinoid (CB) receptors. In addition, cannabinoids inhibit 5-HT3 receptors.

Anandamide exhibited a high potency for block of human 5-HT3 receptors compared to its Ki for human CB1 and CB2 receptors [28]. As a possible mechanism of action for anandamide-induced block of 5-HT3A receptors Xiong et al [29] suggested that anandamide accelerates receptor desensitization.

Recently the nonpsychotropic cannabinoid, cannabidiol, has been shown to be an allosteric inhibitor of mouse 5-HT3A receptors [30*]. Cannabidiol’s nonpsychotropic properties are explained by a very low affinity for the CB1 and CB2 receptors. However, cannabidiol still has antiemetic effects. These data demonstrate that part of the desired therapeutic endpoints of cannabinoids may be due to an allosteric inhibition of 5-HT3 receptors.

Other compounds

A number of other compounds have been shown to modulate 5-HT3 receptors. These include local anesthetics; lidocaine and bupivacaine which are both competitive and allosteric antagonists at the 5-HT3A receptor [31]. Steroids are non-competitive inhibitors of 5-HT3 receptors, but only at concentrations that exceed physiological concentrations [2*]. Morphine, hydromorphone, and fentanyl have been shown to cause a competitive block of 5-HT3A receptors [3233]. Methadone, an opioid often used to treat morphine addiction, also competitively inhibits human 5-HT3A receptors and increases desensitization. However, an insurmountable block was observed for heteromeric human 5-HT3AB receptors [34*].

In addition to the natural cannabinoid compounds mentioned above, other naturally occurring compounds are known to modulate 5-HT3 receptors [2*]. These naturally occurring compounds include quinine, extracts from ginger, ginseng and liquorice as well as α-thujone, a component of the volatile oils from wormwood, thyme, sage. However, it is difficult to directly compare the in vivo concentrations of the reputed therapeutic benefits of some “traditional medicines” with the in vitro concentrations.

Kinetic mechanisms of 5-HT3A receptor modulation

So how do the allosteric compounds modulate 5-HT-elicited current amplitudes and shift 5-HT concentration response curves? We have recently developed a kinetic model (Figure 2) [35] that defines agonist-evoked activation of 5-HT3A receptors. Agonist (A) binds to the closed, resting state of the receptor (R) and the agonist-bound receptor (AR) then transitions to an open channel state (AO). From the open state the receptor can desensitize (AD) or, if the agonist is removed, can close either through the open states (AnO) or the pre-open states (AnR). Positive modulators may increase the 5-HT3 receptors’ sensitivity to agonist by increasing agonist binding affinity (increasing k1/k2), increasing channel gating efficacy (increasing β/α), or by reducing the rate of desensitization (decrease kd+/kd-). Allosteric antagonists would bind to the receptor and cause opposite effects on these kinetic transitions.

Figure 2. Kinetic model of 5-HT3A receptor function.

Figure 2

The major states and transitions required to account for the actions of agonists on 5-HT3 receptors are shown in black. The minor transitions, which are shown in grey, are assumed to be possible, but negligible Values of rate constants (denoted above and below arrows) determined from macroscopic current experiments with rapid solution exchange are given in Solt et al. [40].

Polymorphisms of 5-HT3 receptors influence pharmacology

In recent years an increasing amount of literature has been published detailing the occurrence and influence of 5-HT3 subunit polymorphisms in a number of pathological states. Some polymorphisms alter channel kinetics [36], which may influence the binding and/or action of allosteric compounds such that they alter the effectiveness and side effects of some therapeutic compounds. Therapeutic responses to and side effects of the SSRI paroxetine are influenced by variants in the 5-HT3A subunit and 5-HT3B subunit respectively [3738]. A variant in the 5-HT3A subunit alters the clinical response to the atypical antipsychotic risperidone in schizophrenic patients [39]. Increased potency for clozapine block has been observed in the rare 5-HT3A(P391R) variant observed in at least one schizophrenic patient [40]. It has been proposed that variants in 5-HT3 subunits underlie treatment-resistant schizophrenia [4143]. The area of 5-HT3 receptor polymorphisms is a new and exciting area that may enhance our knowledge of certain pathologies and lead to novel therapies.

Future aspects

Most studies to date have been performed on homomeric 5-HT3A receptors, usually performed on expressed rodent subunits and at room temperature. Rodents have only two orthologs of the five genes that encode human 5-HT3 subunit genes. It has been noted that rodents do not make good models for gastrointestinal research [44*]. If rodents are not a good model for 5-HT3 receptor mechanisms in the “little brain” then perhaps the rodent is an unsatisfactory model for learning about 5-HT3 receptor physiology or potential 5-HT3 receptor-based CNS therapies. However, humanized mice will undoubtedly expand our knowledge of the complexities of the 5-HT3 receptor and its pharmacology. Our knowledge of heteromeric 5-HT3 receptors is limited; we have just begun to appreciate the complexity of human 5-HT3 receptors in vivo and their potential for pharmacological manipulation.

Acknowledgments

The author is supported by a grant from the National Institutes of Health National Institute of Alcoholism and Alcohol Abuse [Grant R21-AA017938].

Footnotes

Publisher's Disclaimer: This is a PDF file of an unedited manuscript that has been accepted for publication. As a service to our customers we are providing this early version of the manuscript. The manuscript will undergo copyediting, typesetting, and review of the resulting proof before it is published in its final citable form. Please note that during the production process errors may be discovered which could affect the content, and all legal disclaimers that apply to the journal pertain.

References

  • 1.Davies PA, Wang W, Hales TG, Kirkness EF. A novel class of ligand-gated ion channel is activated by Zn2+ J Biol Chem. 2003;278:712–717. doi: 10.1074/jbc.M208814200. [DOI] [PubMed] [Google Scholar]
  • *2.Walstab J, Rappold G, Niesler B. 5-HT(3) receptors: role in disease and target of drugs. Pharmacol Ther. 2010;128:146–169. doi: 10.1016/j.pharmthera.2010.07.001. This review is a comprehensive review on 5-HT3 receptors covering genetics, pathology and pharmacology. It contains an updated review of competitve antagonists. [DOI] [PubMed] [Google Scholar]
  • 3.Thompson AJ, Lummis SC. The 5-HT3 receptor as a therapeutic target. Expert Opin Ther Targets. 2007;11:527–540. doi: 10.1517/14728222.11.4.527. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 4.Koshizaki M, Kawamata M, Shimada SG, Saito Y, Collins JG. 5-HT3 receptors partially mediate halothane depression of spinal dorsal horn sensory neurons. Anesth Analg. 2003;96:1027–1031. doi: 10.1213/01.ANE.0000050769.34933.03. [DOI] [PubMed] [Google Scholar]
  • 5.Apfel CC, Kranke P, Katz MH, Goepfert C, Papenfuss T, Rauch S, Heineck R, Greim CA, Roewer N. Volatile anaesthetics may be the main cause of early but not delayed postoperative vomiting: a randomized controlled trial of factorial design. Br J Anaesth. 2002;88:659–668. doi: 10.1093/bja/88.5.659. [DOI] [PubMed] [Google Scholar]
  • 6.Stevens RJ, Rusch D, Davies PA, Raines DE. Molecular properties important for inhaled anesthetic action on human 5-HT3A receptors. Anesth Analg. 2005;100:1696–1703. doi: 10.1213/01.ANE.0000151720.36988.09. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 7.Stevens R, Rusch D, Solt K, Raines DE, Davies PA. Modulation of human 5-hydroxytryptamine type 3AB receptors by volatile anesthetics and n-alcohols. J Pharmacol Exp Ther. 2005;314:338–345. doi: 10.1124/jpet.105.085076. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 8.Hayrapetyan V, Jenschke M, Dillon GH, Machu TK. Co-expression of the 5-HT(3B) subunit with the 5-HT(3A) receptor reduces alcohol sensitivity. Brain Res Mol Brain Res. 2005;142:146–150. doi: 10.1016/j.molbrainres.2005.09.011. [DOI] [PubMed] [Google Scholar]
  • 9.Solt K, Stevens RJ, Davies PA, Raines DE. General anesthetic-induced channel gating enhancement of 5-hydroxytryptamine type 3 receptors depends on receptor subunit composition. J Pharmacol Exp Ther. 2005;315:771–776. doi: 10.1124/jpet.105.090621. [DOI] [PubMed] [Google Scholar]
  • 10.Rusch D, Musset B, Wulf H, Schuster A, Raines DE. Subunit-dependent modulation of the 5-hydroxytryptamine type 3 receptor open-close equilibrium by n-alcohols. J Pharmacol Exp Ther. 2007;321:1069–1074. doi: 10.1124/jpet.106.118752. [DOI] [PubMed] [Google Scholar]
  • 11.Kelley SP, Dunlop JI, Kirkness EF, Lambert JJ, Peters JA. A cytoplasmic region determines single-channel conductance in 5-HT3 receptors. Nature. 2003;424:321–324. doi: 10.1038/nature01788. [DOI] [PubMed] [Google Scholar]
  • 12.Hales TG, Dunlop JI, Deeb TZ, Carland JE, Kelley SP, Lambert JJ, Peters JA. Common determinants of single channel conductance within the large cytoplasmic loop of 5-hydroxytryptamine type 3 and alpha4beta2 nicotinic acetylcholine receptors. J Biol Chem. 2006;281:8062–8071. doi: 10.1074/jbc.M513222200. [DOI] [PubMed] [Google Scholar]
  • **13.Feinberg-Zadek PL, Davies PA. Ethanol stabilizes the open state of single 5-hydroxytryptamine(3A)(QDA) receptors. J Pharmacol Exp Ther. 2010;333:896–902. doi: 10.1124/jpet.109.164863. This study used a high conducting mutant 5-HT3A subunit that allowed for the study of ethanol modulation 5-HT3A receptors at a single channel level. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 14.Zhou Q, Verdoorn TA, Lovinger DM. Alcohols potentiate the function of 5-HT3 receptor-channels on NCB-20 neuroblastoma cells by favouring and stabilizing the open channel state. J Physiol. 1998;507:335–352. doi: 10.1111/j.1469-7793.1998.335bt.x. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 15.Hu XQ, Sun H, Peoples RW, Hong R, Zhang L. An interaction involving an arginine residue in the cytoplasmic domain of the 5-HT3A receptor contributes to receptor desensitization mechanism. J Biol Chem. 2006;281:21781–21788. doi: 10.1074/jbc.M600676200. [DOI] [PubMed] [Google Scholar]
  • 16.Barann M, Linden I, Witten S, Urban BW. Molecular actions of propofol on human 5-HT3A receptors: enhancement as well as inhibition by closely related phenol derivatives. Anesth Analg. 2008;106:846–857. doi: 10.1213/ane.0b013e318162ca7c. [DOI] [PubMed] [Google Scholar]
  • 17.Rusch D, Braun HA, Wulf H, Schuster A, Raines DE. Inhibition of human 5-HT(3A) and 5-HT(3AB) receptors by etomidate, propofol and pentobarbital. Eur J Pharmacol. 2007;573:60–64. doi: 10.1016/j.ejphar.2007.07.009. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 18.Grant IS, Nimmo WS, McNicol LR, Clements JA. Ketamine disposition in children and adults. Br J Anaesth. 1983;55:1107–1111. doi: 10.1093/bja/55.11.1107. [DOI] [PubMed] [Google Scholar]
  • 19.Peters JA, Malone HM, Lambert JJ. Ketamine potentiates 5-HT3 receptor-mediated currents in rabbit nodose ganglion neurones. Br J Pharmacol. 1991;103:1623–1625. doi: 10.1111/j.1476-5381.1991.tb09837.x. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 20.Yamakura T, Chavez-Noriega LE, Harris RA. Subunit-dependent inhibition of human neuronal nicotinic acetylcholine receptors and other ligand-gated ion channels by dissociative anesthetics ketamine and dizocilpine. Anesthesiology. 2000;92:1144–1153. doi: 10.1097/00000542-200004000-00033. [DOI] [PubMed] [Google Scholar]
  • 21.Fan P. Effects of antidepressants on the inward current mediated by 5-HT3 receptors in rat nodose ganglion neurones. Br J Pharmacol. 1994;112:741–744. doi: 10.1111/j.1476-5381.1994.tb13140.x. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 22.Choi JS, Choi BH, Ahn HS, Kim MJ, Rhie DJ, Yoon SH, Min DS, Jo YH, Kim MS, Sung KW, et al. Mechanism of block by fluoxetine of 5-hydroxytryptamine3 (5-HT3)-mediated currents in NCB-20 neuroblastoma cells. Biochem Pharmacol. 2003;66:2125–2132. doi: 10.1016/j.bcp.2003.08.012. [DOI] [PubMed] [Google Scholar]
  • *23.Gumilar F, Bouzat C. Tricyclic antidepressants inhibit homomeric Cys-loop receptors by acting at different conformational states. Eur J Pharmacol. 2008;584:30–39. doi: 10.1016/j.ejphar.2008.01.023. This study used the high conducting 5-HT3A subunit mutant to demonstrate that tricyclic antidepressants inhibit through both open and closed states. The study also examined the block at a single-channel level. [DOI] [PubMed] [Google Scholar]
  • 24.Rammes G, Eisensamer B, Ferrari U, Shapa M, Gimpl G, Gilling K, Parsons C, Riering K, Hapfelmeier G, Bondy B, et al. Antipsychotic drugs antagonize human serotonin type 3 receptor currents in a noncompetitive manner. Mol Psychiatry. 2004;9:846–858. 818. doi: 10.1038/sj.mp.4001490. [DOI] [PubMed] [Google Scholar]
  • 25.Eisensamer B, Uhr M, Meyr S, Gimpl G, Deiml T, Rammes G, Lambert JJ, Zieglgansberger W, Holsboer F, Rupprecht R. Antidepressants and antipsychotic drugs colocalize with 5-HT3 receptors in raft-like domains. J Neurosci. 2005;25:10198–10206. doi: 10.1523/JNEUROSCI.2460-05.2005. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 26.Nothdurfter C, Tanasic S, Di Benedetto B, Rammes G, Wagner EM, Kirmeier T, Ganal V, Kessler JS, Rein T, Holsboer F, et al. Impact of lipid raft integrity on 5-HT3 receptor function and its modulation by antidepressants. Neuropsychopharmacology. 2010;35:1510–1519. doi: 10.1038/npp.2010.20. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 27.Pertwee RG. Emerging strategies for exploiting cannabinoid receptor agonists as medicines. Br J Pharmacol. 2009;156:397–411. doi: 10.1111/j.1476-5381.2008.00048.x. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 28.Barann M, Molderings G, Bruss M, Bonisch H, Urban BW, Gothert M. Direct inhibition by cannabinoids of human 5-HT3A receptors: probable involvement of an allosteric modulatory site. Br J Pharmacol. 2002;137:589–596. doi: 10.1038/sj.bjp.0704829. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 29.Xiong W, Hosoi M, Koo BN, Zhang L. Anandamide inhibition of 5-HT3A receptors varies with receptor density and desensitization. Mol Pharmacol. 2008;73:314–322. doi: 10.1124/mol.107.039149. [DOI] [PubMed] [Google Scholar]
  • *30.Yang KH, Galadari S, Isaev D, Petroianu G, Shippenberg TS, Oz M. The nonpsychoactive cannabinoid cannabidiol inhibits 5-hydroxytryptamine3A receptor-mediated currents in Xenopus laevis oocytes. J Pharmacol Exp Ther. 2010;333:547–554. doi: 10.1124/jpet.109.162594. This study demonstrates that the non-psychotropic, analgesic and antiemetic cannabinoid, cannabidiol (CBD) is an allosteric inhibitor of 5-HT3 receptors. This raises the possibility that some of the therapeutic benefits of cannabinoids maybe through the 5-HT3 receptor and that allosteric modulators of 5-HT3 receptors show useful therapeutic potential. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 31.Ueta K, Suzuki T, Sugimoto M, Uchida I, Mashimo T. Local anesthetics have different mechanisms and sites of action at recombinant 5-HT3 receptors. Reg Anesth Pain Med. 2007;32:462–470. doi: 10.1016/j.rapm.2007.06.391. [DOI] [PubMed] [Google Scholar]
  • 32.Wittmann M, Peters I, Schaaf T, Wartenberg HC, Wirz S, Nadstawek J, Urban BW, Barann M. The effects of morphine on human 5-HT3A receptors. Anesth Analg. 2006;103:747–752. doi: 10.1213/01.ane.0000229706.84471.4d. [DOI] [PubMed] [Google Scholar]
  • 33.Wittmann M, Schaaf T, Peters I, Wirz S, Urban BW, Barann M. The effects of fentanyl-like opioids and hydromorphone on human 5-HT3A receptors. Anesth Analg. 2008;107:107–112. doi: 10.1213/ane.0b013e31817342c2. [DOI] [PubMed] [Google Scholar]
  • *34.Deeb TZ, Sharp D, Hales TG. Direct subunit-dependent multimodal 5-hydroxytryptamine3 receptor antagonism by methadone. Mol Pharmacol. 2009;75:908–917. doi: 10.1124/mol.108.053322. This study further demonstrates how subunit composition of the receptor can alter 5-HT3 receptor pharmacology. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 35.Solt K, Ruesch D, Forman SA, Davies PA, Raines DE. Differential effects of serotonin and dopamine on human 5-HT3A receptor kinetics: interpretation within an allosteric kinetic model. J Neurosci. 2007;27:13151–13160. doi: 10.1523/JNEUROSCI.3772-07.2007. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 36.Krzywkowski K, Davies PA, Feinberg-Zadek PL, Brauner-Osborne H, Jensen AA. High-frequency HTR3B variant associated with major depression dramatically augments the signaling of the human 5-HT3AB receptor. Proc Natl Acad Sci U S A. 2008;105:722–727. doi: 10.1073/pnas.0708454105. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 37.Kato M, Fukuda T, Wakeno M, Fukuda K, Okugawa G, Ikenaga Y, Yamashita M, Takekita Y, Nobuhara K, Azuma J, et al. Effects of the serotonin type 2A, 3A and 3B receptor and the serotonin transporter genes on paroxetine and fluvoxamine efficacy and adverse drug reactions in depressed Japanese patients. Neuropsychobiology. 2006;53:186–195. doi: 10.1159/000094727. [DOI] [PubMed] [Google Scholar]
  • 38.Sugai T, Suzuki Y, Sawamura K, Fukui N, Inoue Y, Someya T. The effect of 5-hydroxytryptamine 3A and 3B receptor genes on nausea induced by paroxetine. Pharmacogenomics J. 2006;6:351–356. doi: 10.1038/sj.tpj.6500382. [DOI] [PubMed] [Google Scholar]
  • 39.Gu B, Wang L, Zhang AP, Ma G, Zhao XZ, Li HF, Feng GY, He L, Xing QH. Association between a polymorphism of the HTR3A gene and therapeutic response to risperidone treatment in drug-naive Chinese schizophrenia patients. Pharmacogenet Genomics. 2008;18:721–727. doi: 10.1097/FPC.0b013e32830500e2. [DOI] [PubMed] [Google Scholar]
  • 40.Rammes G, Hosp C, Eisensamer B, Tanasic S, Nothdurfter C, Zieglgansberger W, Rupprecht R. Identification of a domain which affects kinetics and antagonistic potency of clozapine at 5-HT3 receptors. PLoS One. 2009;4:e6715. doi: 10.1371/journal.pone.0006715. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 41.Ji X, Takahashi N, Branko A, Ishihara R, Nagai T, Mouri A, Saito S, Maeno N, Inada T, Ozaki N. An association between serotonin receptor 3B gene (HTR3B) and treatment-resistant schizophrenia (TRS) in a Japanese population. Nagoya J Med Sci. 2008;70:11–17. [PubMed] [Google Scholar]
  • 42.Ji X, Takahashi N, Saito S, Ishihara R, Maeno N, Inada T, Ozaki N. Relationship between three serotonin receptor subtypes (HTR3A, HTR2A and HTR4) and treatment-resistant schizophrenia in the Japanese population. Neurosci Lett. 2008;435:95–98. doi: 10.1016/j.neulet.2008.01.083. [DOI] [PubMed] [Google Scholar]
  • 43.Meineke C, Tzvetkov MV, Bokelmann K, Oetjen E, Hirsch-Ernst K, Kaiser R, Brockmoller J. Functional characterization of a −100_-102delAAG deletion-insertion polymorphism in the promoter region of the HTR3B gene. Pharmacogenet Genomics. 2008;18:219–230. doi: 10.1097/FPC.0b013e3282f51092. [DOI] [PubMed] [Google Scholar]
  • *44.Holbrook JD, Gill CH, Zebda N, Spencer JP, Leyland R, Rance KH, Trinh H, Balmer G, Kelly FM, Yusaf SP, et al. Characterisation of 5-HT3C, 5-HT3D and 5-HT3E receptor subunits: evolution, distribution and function. J Neurochem. 2009;108:384–396. doi: 10.1111/j.1471-4159.2008.05775.x. Study examining the function of 5-HT3 receptors containing 5-HT3C, 5-HT3D and 5-HT3E subunits. Discusses the potential importance of these subunits to human physiology and the difficulty of using rodents to study 5-HT3 receptors due to the lack of genes encoding these subunits. [DOI] [PubMed] [Google Scholar]

RESOURCES