Skip to main content
NIHPA Author Manuscripts logoLink to NIHPA Author Manuscripts
. Author manuscript; available in PMC: 2019 Feb 21.
Published in final edited form as: Handb Exp Pharmacol. 2018;252:227–260. doi: 10.1007/164_2018_107

Serotonergic Psychedelics: Experimental Approaches for Assessing Mechanisms of Action

Clinton E Canal 1,
PMCID: PMC6136989  NIHMSID: NIHMS968438  PMID: 29532180

Abstract

Recent, well-controlled – albeit small-scale – clinical trials show that serotonergic psychedelics, including psilocybin and lysergic acid diethylamide, possess great promise for treating psychiatric disorders, including treatment-resistant depression. Additionally, fresh results from a deluge of clinical neuroimaging studies are unveiling the dynamic effects of serotonergic psychedelics on functional activity within, and connectivity across, discrete neural systems. These observations have led to testable hypotheses regarding neural processing mechanisms that contribute to psychedelic effects and therapeutic benefits. Despite these advances and a plethora of preclinical and clinical observations supporting a central role for brain serotonin 5-HT2A receptors in producing serotonergic psychedelic effects, lingering and new questions about mechanisms abound. These chiefly pertain to molecular neuropharmacology. This chapter is devoted to illuminating and discussing such questions in the context of preclinical experimental approaches for studying mechanisms of action of serotonergic psychedelics, classic and new.

Keywords: α Adrenergic, 5-HT2A, 5-HT2C, Cingulate cortex, Head-twitch, Ketanserin, Psychedelic mechanisms, Receptor binding, Receptor conformations, Receptor dimers, Receptor function, Serotonin, Signal transduction

1 Introduction

Humans have been reporting their experiences with psychoactive substances in exquisite detail for centuries, at least. In the mid 1800s, J. J. Moreau de Tours wrote about hashish intoxication (Moreau 1973). Sigmund Freud later scripted his personal relationship with cocaine (Freud and Byck 1975). Ann and Alexander Shulgin then gave the world PiHKAL (Shulgin and Shulgin 1991) and TiHKAL (Shulgin and Shulgin 1997), books describing mostly novel psychedelic drugs and their effects. Fast-forward to today, we see the practice of self-reporting experiences with psychoactive substances is rampant, organized, archived, and accessible (e.g., Erowid.org). The surge of information about new psychoactive substances reported on the internet paralleling the surge in online vendors selling such “research chemicals” has catalyzed their spread and use. Systematic research aimed to discover and document their unique mechanisms has, understandably, lagged behind.

Fortunately for researchers, most novel psychedelics still fit within familiar chemotype classes and have overlapping pharmacology with their classic predecessors – mainly 5-HT2(A, B, and C) receptor agonist activity in the case of serotonergic psychedelics that include lysergamides (e.g., LSD), tryptamines (e.g., psilocybin and DMT), and phenethylamines (e.g., mescaline and including phenylisopropylamines, e.g., DOB). Chemists have synthesized dozens of relatively obscure serotonergic psychedelics, which have appeared recently on the clandestine market. Examples include lysergamides like AL-LAD (Brandt et al. 2017b) and PARGY-LAD, tryptamines like 5-MeO-DALT (Cozzi and Daley 2016), and phenylethylamines like bk-2C-B, the β-ketone analog of 2C-B and 25C-NBOMe, another analog of 2C-B (Halberstadt 2017). Recent literature covers what is currently known regarding the physiological, psychological, and visual perceptual effects of serotonergic psychedelics, their neuropharmacology and effects on human brain functional connectivity, their use as potential medicines, their inherent risks, and the phenomenology of the psychedelic experience (Halberstadt 2017; Kometer and Vollenweider 2016; Liechti 2017; Nichols 2016; Nichols et al. 2017; Preller and Vollenweider 2016). This chapter covers preclinical research strategies used to elucidate common and divergent mechanisms of serotonergic psychedelics, classic and new.

2 5-HT2A Receptors: The End of the Beginning

For research probing mechanisms, it is clear we have reached the end of the beginning – a cadre of researchers agree that 5-HT2A receptor activation is necessary for most of the psychoactive effects of serotonergic psychedelics (Kometer et al. 2013; Kraehenmann et al. 2017; Nichols 2016; Preller et al. 2017; Vollenweider et al. 1998). To understand mechanisms, though, requires delineating atomic-level drug and receptor interactions and attendant consequences on signal transduction (Wacker et al. 2017) and cellular intrinsic excitability, subsequent short-term and long-term effects on electrochemical communication within and between micro- and macroneural circuits (Petri et al. 2014), as well as the interplay of neural circuits with the user’s personality, psychological and cognitive state, personal history and genetic background (“set”), and the external environment (“setting”) (Studerus et al. 2012).

Questions from these wide-ranging levels of analyses remain. At the highest level, self-reported subjective experiences, there appear to be differences in effects (beyond pharmacokinetics) across and even within classes of serotonergic psychedelics (Glennon 1992; Shulgin and Shulgin 1991, 1997). Although representatives from each chemotype class can induce similar, visual “form constants” of the lattice and tunnel types (elementary psychedelic patterns) (Kometer and Vollenweider 2016), users typically report differences in their “body load,” stimulant and entactogenic effects, the degree to which they produce “organic” or “synthetic” visual or aural hallucinations, and how deeply and clearly they affect emotional and cognitive states. For example, the psychedelic 5-HT2A agonist, DIPT, appears to produce distinct aural, hallucinatory effects (Blough et al. 2014; Rickli et al. 2016; Shulgin and Shulgin 1997), whereas DMT (and analogs including 5-MeO-DMT) is distinguished by its marked proclivity to induce complex hallucinations (Strassman 2001), such as visual hallucinations of things, entities, or events separated from consensus reality. If the effects of different serotonergic psychedelics can be distinguished reliably, then what, mechanistically, differentiates them? Are differences caused by unique, dynamic, active conformations of 5-HT2A receptors, or do some serotonergic psychedelics preferentially target distinct populations of 5-HT2A receptors, e.g., pre- or postsynaptic (Bécamel et al. 2017)? Alternatively, other receptor targets or 5-HT2A receptors functionally linked to other systems, for example, the endocannabinoid system (Best and Regehr 2008; Parrish and Nichols 2006), may contribute to psychedelic effects. Furthermore, unique 5-HT2 receptor homo- or heterodimers or oligomers could be the mechanistic target of serotonergic psychedelics.

3 Binding Events and Cellular Signal Transduction

3.1 Radioligand Receptor Binding

Radioligand receptor binding assays were integral in determining that 5-HT2 receptors were the primary targets of classic psychedelics, such as LSD and DOB (Glennon et al. 1984), and radiolabeled psychedelics applied in autoradiography of brain of slices determined the location of psychedelic receptor targets across neural systems in rodents (McKenna et al. 1987; McKenna and Saavedra 1987). Presently, researchers are developing selective 5-HT2A agonist radioligands to analyze 5-HT2A receptors in humans, using PET imaging techniques (Johansen et al. 2017), and a recent clinical study employing [11C]Cimbi-36, a selective 5-HT2 agonist, reports dense expression of 5-HT2 receptors across all cortical regions, but limited expression in subcortical regions (Beliveau et al. 2017).

Radioligand competition binding and saturation binding assays are used to quantify the affinity, or strength of interaction, of new compounds at receptors. In a competition binding assay, a compound with unknown affinity at the receptor of interest is added to a multi-well microplate at increasing concentrations across wells, typically at half-log units, together with a radiolabeled ligand, with known affinity at the receptor of interest, at one fixed concentration (e.g., its Kd at the receptor, the equilibrium dissociation constant of the ligand). Cell membranes expressing the receptor are then added to the wells, and the mixture is incubated for a period of time until equilibrium is achieved, i.e., the amount of free ligand and ligand bound to the receptor remains constant. The contents of the wells are then rapidly passed through a fiberglass filter mat that collects radioligand bound to receptor but permits free radioligand to pass through. Samples are then added to scintillation cocktail, and a scintillation counter detects the amount of radioactivity emitted from each sample over a fixed time (e.g., counts per minute). The affinity, Ki (equilibrium dissociation constant of a ligand determined in inhibition studies), of the unknown is then determined by fitting a nonlinear dose-response inhibition curve, calculating the concentration of the compound required to displace 50% of the radioligand from the receptor, followed by correction incorporating the radioligand’s affinity and concentration (Cheng and Prusoff 1973).

Saturation binding assays include a similar workflow, but the radionuclide is attached chemically to the compound with unknown affinity at the receptor of interest. The radioligand is then incubated, with tissue expressing the receptor, at increasing concentrations across wells. The affinity, Kd, of the radioligand, or the concentration required to occupy 50% of the receptors at equilibrium, is then calculated, as well as the saturating concentration, which can be used to calculate the density of receptor binding sites labeled by the radioligand, or Bmax. For details, refer to McKinney and Raddatz (2006). A saturation binding assay provides a more accurate affinity value than a single competition binding assay, due to the fact that a radioligand in a competition assay may selectively bind a subset of receptors existing in specific conformations – or may interact with unique receptor amino acids with which the unknown compound may not interact. In other words, Ki values at a specific receptor population may be different depending on the radioligand used, but Kd values remain constant.

Agonist affinities at G-protein-coupled receptors (GPCRs) are strongly affected by the state of G-proteins linked to them. Agonists stabilize active receptor conformations – receptors bound to guanine nucleotide-free Gα proteins – and then dissociate slowly from these active conformations, imparting high affinity at them (DeVree et al. 2016). Thus, competition binding assays in the presence or absence of non-hydrolyzable guanine nucleotides (e.g., GTPγS which blocks agonist high-affinity binding) can be used to determine the affinity of ligands at inactive or active receptor conformations. Moreover, since agonist ligands typically have a higher affinity at receptors labeled with agonists (high-affinity, KH) compared to antagonists (low-affinity, KL), comparisons of these affinities can be used as first-pass screens to test novel ligands for agonist activity. Also, functional efficacies of 5-HT2 ligands correlate strongly with their KL to KH ratios (Egan et al. 2000).

Radioligand binding assays can also be used to measure ligand—receptor association and dissociation rates (Sykes et al. 2010). Ligand—receptor kinetics data can inform efficacy, selectivity, and duration of action in vivo (de Witte et al. 2016). A ligand’s residence time at a receptor—the duration of time a ligand is bound to a receptor—may also be a critical factor for recruiting intracellular signaling molecules. Receptor-mediated activation of cellular cascades may be time-dependent; if an agonist ligand’s dissociation rate is too quick, a substantial proportion of a cellular cascade may remain inactive. Recently, scientists showed that LSD has a relatively long residence time at the 5-HT2A receptor (Wacker et al. 2017). Such studies may inform mechanistic differences between serotonergic psychedelics. The actions of DMT, for example, are very short-lived compared to LSD. It may be discovered that residence time dictates transduction signals that initiate unique cellular psychedelic cascades. LSD potently recruits β-arrestin2 to 5-HT2A receptors, but mutating lysine residue 229 to alanine to decrease LSD’s residence time strongly reduces β-arrestin2 recruitment to 5-HT2A (Wacker et al. 2017).

Classic psychedelics of the tryptamine and lysergamide chemotypes are not selective for 5-HT2 receptors. Psilocin, for example, has appreciable affinity (between 4 and 220 nM Ki) at human 5-HT1B, 5-HT1D, 5-HT1E, 5-HT5, 5-HT6, and 5-HT7 receptors, c.f. PDSP database (Roth et al. 2000). Novel serotonergic psychedelics, such as N-benzylphenethylamines, and putatively psychedelic N-benzylated-5-methoxytryptamines, possess high affinity at 5-HT2 receptors but also bind to other receptors. For example, 25I-NBOMe has significant affinity (<300 nM Ki) at µ-opioid, κ-opioid, and histamine H1 receptors (Nichols et al. 2008). Others have also observed activity of certain N-benzylphenethylamines at H1 (Ki as low as 80 nM), α1A- and α2A-adrenergic (Ki as low as ∼300 nM) receptors (Rickli et al. 2015b), rendering them somewhat similar to LSD, which is promiscuous (PDSP database). Relative to their 2C-x classic hallucinogen predecessors, these drugs have substantially greater selectivity for 5-HT2 over 5-HT1A receptors (Rickli et al. 2015b). Many N-benzylated-5-methoxytryptamines and N-benzylphenethylamines also possess significant affinity at 5-HT6, and N-3-iodobenzyl-5-methoxytryptamine also has significant activity at 5-HT7 receptors (Nichols et al. 2015). A recent report of 25CN-NBOH, however, shows that this compound is very selective at 5-HT2 receptors compared to a host of other receptors; the only relevant affinities (Ki ∼300 nM or less) noted are at human 5-HT6 and rat sigma-1 and sigma-2 (Jensen et al. 2017).

3.2 Receptor Functional Assays

Serotonergic psychedelics stimulate 5-HT2 receptor – Gαq signaling, as measured by activation of phospholipase C-β, causing increases in phosphoinositide hydrolysis, thereby stimulating inositol phosphate production and activation of protein kinase C (Jope et al. 1994; Sanders-Bush et al. 1988); this is the canonical 5-HT2 receptor pathway. Most experimenters focus on activation of Gα subunits, because of technical challenges measuring βγ activation from Gαq-coupled receptors (Kadamur and Ross 2013). Laborious techniques to assess ligand functional effects involving chromatography-based detection or anion exchange columns (Felder et al. 1990; Hide et al. 1989) have frequently been replaced by relatively simple, high-throughput, kit assays that employ highly selective, fluorophore-labeled monoclonal antibodies that bind signaling molecules (Canal et al. 2013b). With the advent of protocols and toolkits to measure ligand-stimulated 5-HT2-β-arrestin2 recruitment, researchers increasingly examine this event, especially with the objective to determine ligand bias (Kenakin 2016; Wacker et al. 2017; Wang et al. 2013).

Other signaling pathways activated by 5-HT2 receptors, such as Gα13-dependent or Gα13-independent phospholipase D pathways (Barclay et al. 2011; McGrew et al. 2002), remain difficult to measure. The workflow can include extraction steps and chromatography and is low-throughput (Walker et al. 2004). Accordingly, much less is known about how psychedelics affect these cellular signaling cascades. For similar reasons, the activity of psychedelics is not fully characterized at other members of the Gαq subclass or other signaling pathways that may be linked endogenously to 5-HT2 receptors, including Gαq-p63RhoGEF-RhoA, Gα11, Gα12, Gα14, Gα15, and Gα16 (Milligan and Kostenis 2006). 5-HT2 receptor activation stimulates calcium mobilization, but this response can be triggered by Gαq-dependent increases in inositol phosphates, i.e. IP3, that activate IP3 receptors (Ca2+ release channels) on endoplasmic reticulum, or by activation of Ca2+ permeable channels on cell membranes. There is much to be discovered about the ability of 5-HT2 receptors to modulate activity of distinct ion channels important for neurotransmission. It is incumbent to determine the effects of serotonergic psychedelics at noncanonical signal transduction pathways, because the 5-HT2A intracellular signals that generate psychedelic effects remain mysterious (Nichols 2016).

Although there are correlations between the psychedelic potencies of phenethylamines, their phenylisopropylamine counterparts, and 5-HT2A agonist efficacy to stimulate inositol phosphate production (Moya et al. 2007; Parrish et al. 2005), LSD, one of the most potent psychedelics, is a notoriously weak 5-HT2A agonist at this pathway (Berg et al. 1998; Egan et al. 1998a). Moreover, high-affinity agonist binding of several 5-HT2A ligands does not correlate with efficacy to stimulate 5-HT2A-mediated inositol phosphate production (Roth et al. 1997). Also, ligand efficacy at this pathway does not correlate with efficacy to substitute for LSD or DOM in drug discrimination tests (Rabin et al. 2002). Serotonergic psychedelics are most often observed to be partial agonists at the 5-HT2A inositol phosphate production pathway, relative to 5-HT. This extends across all classes of serotonergic psychedelics, classic and new, including N-benzylphenethylamines, DOB, 2C-B, novel tryptamines, psilocin, and LSD (Acuna-Castillo et al. 2002; Moya et al. 2007; Parrish et al. 2005; Rickli et al. 2015b, 2016).

Lorcaserin (Belviq®, a 5-HT2C-preferring agonist for obesity), however, efficaciously stimulates 5-HT2A-mediated inositol phosphate production in vitro (75% efficacy compared to 5-HT at human 5-HT2A receptors) (Thomsen et al. 2008), and at clinical doses, likely stimulates this 5-HT2A receptor pathway. From the FDA briefing document (NDA22529):

Assuming that distribution of lorcaserin in monkeys and humans is most comparable, brain levels of lorcaserin may reach 430 ng/ml or 1.7 µM from the clinical dose of 10mg bid. This concentration of lorcaserin would be expected to activate central 5HT2A and potentially [5HT]2B receptors, assuming that lorcaserin has access to receptor sites in the CNS.

Yet, at this dose, lorcaserin is not psychedelic. As a benzazapine, it is also structurally quite different from any of the serotonergic psychedelics – rehashing thoughts that chemotype, and by extension chemotype-dependent stabilization of special 5-HT2A psychedelic conformations, drives psychedelic effects. Other structurally unique ligands that activate 5-HT2A-Gαq signaling, including the piperazine mCPP, also do not elicit psychoactive effects like the serotonergic psychedelics. It has been argued that increased activity at 5-HT2C relative to 5-HT2A receptors, which mCPP and lorcaserin possess, attenuates psychedelic effects (Fantegrossi et al. 2010), but several observations disprove this postulation (Canal and Morgan 2012). For example, all serotonergic psychedelics are 5-HT2C agonists, and psychedelic effects do not abate by increasing dose, i.e., by increasing stimulation of 5-HT2C receptors.

The classic first-generation antipsychotic, chlorpromazine (Thorazine®), a potent antagonist at the 5-HT2A-Gαq-inositol phosphate pathway (Canton et al. 1994), does “not significantly influence the somatic and psychological disturbances” caused by LSD (Clark and Bliss 1957). Others reported that chlorpromazine attenuates, but does not fully block, the psychedelic effects of LSD (Isbell and Logan 1957) or DOM (Snyder et al. 1967) – chlorpromazine doses ranged from 50 to 200 mg (P.O.). Also peculiar is that the non-hallucinogenic 5-HT2A lysergamides, ergotamine and BOL-148, do not significantly alter the psychoactive effects of psilocybin or LSD, respectively, in humans (Clark and Bliss 1957; Pokorny et al. 2016). Though, ergotamine may not readily cross the blood-brain barrier (Verhoeff et al. 1993), and BOL-148 appears to elicit psychoactive effects in some people (Richards et al. 1958). Collectively, these data cast major doubt on the 5-HT2-Gαq-inositol phosphate pathway as a central mediator of psychedelic effects. Direct in vivo support for this conclusion emanates from a preclinical study that showed knockout of Gαq does not eliminate (but does attenuate) the 5-HT2A-dependent head-twitch response induced by the psychedelic phenylisopropylamine DOI (Garcia et al. 2007).

Knockout of β-arrestin2 in mice has no effect on the DOI-elicited head-twitch response (Schmid et al. 2008) and actually increases the head-twitch response elicited by 5-MeO-DMT (Schmid and Bohn 2010). Moreover, measurements of other signal transduction molecules in vitro, including 5-HT2A-Gαi/o-elicited arachidonic acid release (phospholipase A activation) do not reveal unique signaling properties of psychedelics, and like the phospholipase C-β-inositol phosphate pathway, many psychedelics are partial agonists relative to 5-HT (Kurrasch-Orbaugh et al. 2003a, b; Moya et al. 2007). Gonzalez-Maeso’s group has focused on alternative 5-HT2-Gαi/o signaling in vivo. They show that serotonergic psychedelics, but not lisuride (considered a non-hallucinogenic 5-HT2A agonist), alter gene expression through pertussis toxin-sensitive Gαi/o signaling (Gonzalez-Maeso et al. 2007). It should be noted, however, that gene expression changes peak after the induction of behavioral responses, thus, likely do not cause them (Gonzalez-Maeso et al. 2007). Nevertheless, in vitro studies show that pertussis toxin decreases DOI- and LSD-elicited inositol phosphate production and abolishes their potentiation of Erk1,2 phosphorylation but does not impact lisuride and ergotamine responses (Karaki et al. 2014).

These observations show that our understanding of serotonergic psychedelic mechanisms is unripe, and creative studies need to be conducted. For example, clozapine (Clozaril®), an inverse agonist at the 5-HT2A-Gαq-inositol phosphate pathway (Egan et al. 1998b) and arguably the most effective antipsychotic (Wenthur and Lindsley 2013) activates 5-HT2A-mediated AKT phosphorylation, an effect blocked by the selective 5-HT2A antagonist M100907 (Schmid et al. 2014). Also, like other 5-HT2A agonists, clozapine causes 5-HT2A receptor internalization, whereas ketanserin does not (Raote et al. 2013). Recent studies also revealed that psilocin is a 5-HT reuptake inhibitor, i.e., blocks the serotonin transporter, SERT (Blough et al. 2014; Rickli et al. 2016). Also, classic psychedelics can activate TAAR1 and sigma-1 receptors (Bunzow et al. 2001; Fontanilla et al. 2009; Simmler et al. 2016). Intriguingly, 2C-B, traditionally viewed as a selective 5-HT2 agonist, has an inhibitory potency at SERT similar to MDMA (Montgomery et al. 2007); similar effects were observed with DIPT (Rickli et al. 2016). Moreover, DMT causes serotonin efflux from SERT with efficacies similar to MDMA (Rickli et al. 2016).

Despite micromolar 5-HT1A affinities (Rickli et al. 2015b), N-benzylphenethylamines retain potent psychedelic effects. Also, benzofurans, such as 5-APB and 6-APB, are potent and efficacious 5-HT2B agonists but have very low potency at 5-HT2A receptors. They also stimulate efflux of DA and 5-HT and have activity at TAAR1 receptors (Iversen et al. 2013; Rickli et al. 2015a), but anecdotal reports note that psychedelic effects are relatively minor compared to classic psychedelics. These observations provide further credence that the 5-HT2A receptor, but not 5-HT1A, 5-HT2B, TAAR1, or monoamine transporters, initiates psychedelic effects. These and other proteins may modulate psychedelic effects. The 5-HT1A partial agonist (and β-adrenergic antagonist (Hoffmann et al. 2004)), pindolol, for example, strongly potentiates psychedelic effects of DMT (Strassman 2001). In conclusion, despite the central role of 5-HT2A receptors in producing psychedelic effects, we are still lurking in a fuzzy arena regarding mechanisms after the receptor binding event.

3.3 X-Ray Crystallography

Molecular modeling, molecular dynamics simulations, medicinal chemistry, and molecular pharmacology studies, employing point mutations of 5-HT2A receptor amino acids that alter ligand-receptor molecular interactions, help illustrate how serotonergic psychedelics interact with the 5-HT2A receptor (Braden and Nichols 2007; Braden et al. 2006; Chambers and Nichols 2002; Choudhary et al. 1995; Isberg et al. 2011; Perez-Aguilar et al. 2014). The new gold standard, however, for deciphering the precise fit of a ligand at a receptor and the conformation(s) of the receptor it stabilizes is to isolate crystals of the receptor with the ligand bound, and to develop atomic-level resolution (low ångström, i.e., <3.0 Å) crystallographic images of them. Numerous, resolved GPCR crystal structures with agonists, antagonists, or inverse agonists bound have been reported recently (Hua et al. 2016, 2017; Thal et al. 2016; Wang et al. 2017) and are poised to quickly evolve the structure-based drug discovery process (Ranganathan et al. 2017).

The 5-HT2B crystal structure with LSD bound revealed how a classic psychedelic precisely interacts with a 5-HT2 receptor and delivered a putative snapshot of a psychedelic receptor conformation (Wacker et al. 2017). LSD binds in the orthosteric pocket of 5-HT2B, which is characterized by many hydrophobic side chains from residues in transmembranes III, V, VI, and VII; recent mutagenesis studies and a resolved 5-HT2C crystal structure confirm that this pocket is also quite similar in the 5-HT2C receptor (Canal et al. 2011; Cordova-Sintjago et al. 2014; Liu et al. 2017; Peng et al. 2018). The basic nitrogen of the ergoline system forms a salt bridge with aspartic acid residue D135 in transmembrane III – this critical interaction is conversed across aminergic GPCRs (Katritch et al. 2013). LSD’s ergoline system has aromatic interactions with F340 and F341 in transmembrane VI, and its indole nitrogen hydrogen bonds with G221 in transmembrane V. LSD binds differently than ergoline, as ergoline’s indole nitrogen forms a distinct hydrogen bond with T140 in 5-HT2B’s transmembrane VII (Wang et al. 2013), a bond not seen with LSD-5-HT2B. Mutating the homologous residue in 5-HT2C to alanine significantly reduces 5-HT’s affinity and agonist potency to stimulate 5-HT2C-phosphoinositide hydrolysis (Liu et al. 2017).

LSD also interacts with the previously described extended binding pocket of 5-HT2B (Wang et al. 2013); specifically, its ethyl groups interact with residues W131 and L132 in transmembrane III, and L362 in transmembrane VII. These interactions may be key to LSD’s psychedelic effects, as molecular modeling and ligand docking at 5-HT2A appear to show that they persist for LSD but are lost with LSA, which lacks the diethylamide moiety of LSD (Wacker et al. 2017). (Though, L132 in 5-HT2B is I132 in 5-HT2A.) TiHKAL (Shulgin and Shulgin 1997) describes an LSA self-report from Albert Hoffman (who discovered LSD):

An i.m. administration of a 500 microgram dose led to a tired, dreamy state with an inability to maintain clear thoughts. After a short period of sleep, the effects were gone and normal baseline was recovered within five hours. Other observers have confirmed this clouding of consciousness leading to sleep.

This report illustrates that LSA is substantially less psychoactive than LSD. Since it is distinct from LSD only in its lack of the diethylamide moiety, the hydrophobic interactions between the diethyl structures and 5-HT2A may be central to LSD’s psychedelic effects via 5-HT2A. LSA is as efficacious as LSD at activating 5-HT2A- Gαq-mediated calcium flux and 5-HT2A-mediated β-arrestin2 recruitment, but 1–2 orders of magnitude less potent (Wacker et al. 2017).

The LSD-5-HT2B structure has similar conformational features as active GPCRs, but with a bias towards a β-arrestin2 state; this was subsequently confirmed in functional assays that show LSD has a potency bias towards β-arrestin2 versus Gαq signaling (Wacker et al. 2013, 2017). These data may suggest that 5-HT2A- β-arrestin2 signaling contributes significantly to psychedelic effects. However, knockout of β-arrestin2 does not reduce the 5-HT2A-mediated head-twitch response caused by two psychedelics, DOI and 5-MeO-DMT (Schmid and Bohn 2010; Schmid et al. 2008). Resolution of 5-HT2A crystal structures with serotonergic psychedelics from the phenethylamine and tryptamine classes may reveal commonalities regarding ligand-receptor interactions and receptor conformations that may ignite the psychedelic experience (Nichols 2017). However, crystal structures (∼3 Å) of the β2-adrenergic receptor with an antagonists or inverse agonists bound did not reveal robust conformational differences, suggesting ligands with different functional properties may alter receptor dynamics more so than receptor structure (Wacker et al. 2010). Because ligand-receptor signaling is a dynamic, spatial-temporal process (Grundmann and Kostenis 2017), advanced molecular dynamics (Saleh et al. 2017) describing ligand–receptor–G-protein-binding sequences may be needed to reveal the subtleties of psychedelics acting at 5-HT2A receptors.

4 Preclinical Animal Models

4.1 Head-Twitch Response

The psychedelic-induced head-twitch response in rodents was first reported in 1956 (Keller and Umbreit 1956; Winter and Flataker 1956), and validation of the assay was provided in 1967 (Corne and Pickering 1967). Since then, numerous groups have shown that serotonergic psychedelics elicit the behavior via a 5-HT2A mechanism. Tested psychedelics include LSD, psilocybin, psilocin, DMT, mescaline, 5-MeO-DMT, 5-MeO-DIPT, DPT, 2C-T-7, DOM, DOB, DOI, 2C–I, and several new phenethylamines, tryptamines, and lysergamides (Brandt et al. 2016, 2017a, b; Canal and Morgan 2012; Corne and Pickering 1967; Fantegrossi et al. 2005, 2006, 2008; Halberstadt and Geyer 2013, 2014, 2017; Halberstadt et al. 2011; Moya et al. 2007; Nichols et al. 2015).

Mice display a head-twitch response – observed as rapid, lateral rotations of the head (Halberstadt and Geyer 2013) – commencing within a few minutes after peripheral administration of a psychedelic, and with phenylisopropylamines, the response peaks in about 10 min and persists for at least 2 h (Canal and Morgan 2012). 5-HT2A antagonists block the head-twitch in mice, rats, and the least shrew (Canal and Morgan 2012; Darmani et al. 1994; Halberstadt and Geyer 2017; Schreiber et al. 1995), whether administered before or after induction of the head-twitch response, as observed in C57BL/6J mice (Canal et al. 2013a). Serotonergic psychedelics do not elicit a head-twitch response in 5-HT2A knockout mice, but restoration of 5-HT2A receptors to cortical neurons restores the ability of psychedelics to elicit the response (Gonzalez-Maeso et al. 2007).

What has made the head-twitch response particularly attractive for studying serotonergic psychedelic mechanisms is that lisuride does not produce it in mice (Gonzalez-Maeso et al. 2007; Halberstadt and Geyer 2013). Lisuride does, however, elicit a head-twitch response in the least shrew (Darmani et al. 1994), which appears a particularly sensitive species; (±)-DOI, 0.63 mg/kg, elicits an average of 263 head-twitches in 30 min, whereas lisuride, 1.25 mg/kg, elicits an average of 49 head-twitches in the same time period (Darmani et al. 1994). As a comparison, adult, male C57BL/6J mice exhibit about 30–40 head-twitches in a 10-min period after (±)-DOI, 1 mg/kg (Canal and Morgan 2012; Halberstadt and Geyer 2013). Furthermore, lisuride is a low-efficacy 5-HT2A agonist (Berg et al. 1998) and appears to be distinguished from other 5-HT2A agonists, especially, by its weak potency and efficacy at stimulating intracellular calcium mobilization; for example, its efficacy relative to 5-HT is ∼49%, whereas LSD’s efficacy is ∼85% (Cussac et al. 2008). A recent study reports a significant correlation between the potencies of phenethylamines and tryptamines to activate 5-HT2A-mediated calcium mobilization and their potencies to elicit the head-twitch response (Nichols et al. 2015). Calcium mobilization can be independent of the phospholipase C-β-IP3 receptor pathway, and this signaling pathway does not appear to control all psychoactive effects, as noted above. Thus, these results provide an intriguing possibility that IP3 receptor-independent calcium mobilization may uniquely contribute to psychedelic effects.

Nevertheless, there are clear false positives in the head-twitch assay. The 5-HT releaser d-fenfluramine is non-hallucinogenic but elicits the head-twitch in mice (Darmani 1998) – fenfluramine does, however, displace the 5-HT2A agonist radioligand [11C]Cimbi-36 from binding sites in primate brains (Yang et al. 2017), suggesting that the head-twitch may be sensitive to drugs that indirectly stimulate 5-HT2A receptors. False negatives also show that it has questionable translational validity. To illustrate, anecdotal reports note that cannabis – psychoactive due to THC partial agonist activity at cannabinoid 1 (CB1) receptors – potentiates serotonergic psychedelic effects in humans. However, numerous compounds that stimulate CB1 receptors, including THC, eliminate the head-twitch elicited by DOI, whereas the CB1 inverse agonist, SR 141716A (Rimonibant®), can elicit the head-twitch (Ceci et al. 2015; Darmani 2001; Darmani et al. 2003; Darmani and Pandya 2000; Egashira et al. 2004, 2011).

The psychedelic-elicited head-twitch response can be modulated, albeit mostly suppressed, by a number of compounds that target receptors other than 5-HT2A, including 5-HT2C, 5-HT1A, glutamate NMDA, AMPA and mGluR2, α-adrenergic, and dopamine D2 receptors, and others; regardless of the modulatory effect, selective 5-HT2A blockade abolishes the head-twitch (Canal and Morgan 2012). Thus, the 5-HT2A receptor appears to functionally interact with numerous neurotransmitter systems. Some mice including C57BL/6J mice naturally exhibit the head-twitch at low levels. For example, we have observed scores of adult, male, drug-naïve C57BL/6J mice, and each exhibits ∼2–5 robust head-twitches in 10 min (Canal et al. 2013a); others report similar observations (Halberstadt and Geyer 2013). Interestingly, it too is blocked by selective 5-HT2A antagonism (Canal et al. 2013a), corroborating the conclusion that the 5-HT2A receptor, regardless of whether it is activated by a psychedelic or by endogenous mechanisms, mediates the head-twitch response.

Many novel lysergamides including 1P–LSD, LSZ, and AL-LAD elicit a dose-dependent head-twitch in C57BL/6J mice (Brandt et al. 2016, 2017b). LSM-775 does not produce a head-twitch in C57BL/6J mice unless they are pretreated with a 5-HT1A antagonist (Brandt et al. 2017a). LSM-775 also appears to lack psychedelic effects in humans, which is peculiar, as other psychedelics are potent and efficacious 5-HT1A agonists, including LSD (Pauwels et al. 1993), and as noted above, the 5-HT1A partial agonist, pindolol, potentiates the psychedelic effects of DMT (Strassman 2001). Moreover, the potent 5-HT1A agonist (±)-8-OH-DPAT enhances the stimulus effects of DOM (Glennon 1991). However, 5-HT1A agonists including both enantiomers of 8-OH-DPAT potently block the head-twitch response elicited by some, but not all, serotonergic psychedelics (Canal et al. 2015; Dursun and Handley 1993; Goodwin and Green 1985), and the 5-HT1A antagonist/dopamine D2/D3 agonist S(-)-UH-301 alone can elicit a head-twitch response in mice (Darmani and Reeves 1996).

4.2 Drug Discrimination

The two-lever, appetitive, drug discrimination task is an authoritative, preclinical tool for determining psychedelic drug mechanisms in vivo. A food-motivated animal is trained in an operant task, under one of several reinforcement schedules, to press one lever for a food reward when it is under the influence of a training drug, and the other lever when it is not. Once the animal clearly shows it can discriminate or recognize the effects of the drug by successfully pressing the correct lever on repeated trials, e.g., with accuracy ≥80% (typically requiring several weeks of training), it can then be treated with test drugs to observe whether they substitute (partially to fully) for the training drug, or when co-administered with the training drug, suppress (partially to fully) its discriminative stimulus effects. For example, if a test drug causes animals to make ≥80% of their responses on the lever associated with the training drug, investigators infer that the two compounds produce similar subjective or stimulus effects. Conversely, if a test drug causes animals to make ≤20% of their responses on the lever associated with the training drug, investigators infer that the test drug does not produce subjective effects that are like the training drug. Even if the test drug has discriminative effects, if they are unlike the training drug, animals typically default to responding by pressing the vehicle-associated lever. If a test drug causes animals to split their responses between the levers, then investigators infer that it has effects somewhat similar to the training drug (Glennon and Young 2011). Like the head-twitch procedure, drug discrimination can provide information regarding drug pharmacokinetics and pharmacodynamics (e.g., drug onset, duration, potency, and mechanism of action), and drug discrimination has high predictive validity, i.e., it translates well to human subjects. Importantly, the drug discrimination procedure distinguishes psychoactive drugs from various classes, and germane here, primates trained using two-choice drug discrimination unmistakably distinguish different types of hallucinogens, e.g., κ-opioid agonist hallucinogens from NMDA antagonist hallucinogens and from serotonergic psychedelics (Butelman et al. 2010). For a detailed study of the drug discrimination procedure, including its utility and arguments regarding its superiority relative to other behavioral assays, see Glennon and Young (2011).

Early drug discrimination studies with rodents that showed 5-HT2A antagonists reduce the stimulus effects of diverse serotonergic psychedelics (Glennon 1992; Glennon et al. 1983) provided commanding evidence that 5-HT2A receptor activation is their unifying and common mechanism. This evidence was corroborated by studies employing DOx psychedelics, which are 5-HT2 selective agonists, as test drugs that substitute for LSD (Fiorella et al. 1995). Other studies confirmed that selective blockade of 5-HT2A receptors, i.e., by M100907 (Kehne et al. 1996; Palfreyman et al. 1993), occludes the discriminative stimulus effects of some serotonergic psychedelics in rats and primates (Li et al. 2007, 2009a; May et al. 2009; Schreiber et al. 1994).

Recent studies employing drug discrimination in rats show that novel psychedelics including 25I-, 25B-, and 25C-NBOMe, and 5-MeO-DALT fully substitute for DOM; interestingly, the NBOMe drugs also substitute for MDMA, but 5-MeO-DALT does not (Gatch et al. 2017). This study and others illustrate the utility of drug discrimination assays to differentiate unique effects of different serotonergic psychedelics. The selective 5-HT2C antagonist SB242084 (1 mg/kg) blocks ∼70% of DIPT lever responding but does not affect the DMT discriminative stimulus (Carbonaro et al. 2015). Intriguingly, also from this study, M100907 does not completely block the discriminative stimulus effects of DIPT but does completely block DMT’s effects. Also, 2C-T-7 only partially substitutes for psilocybin and LSD (∼40% and ∼75%, respectively) in rats at a dose (1 mg/kg) that appears to be maximal for eliciting the head-twitch response in mice (Fantegrossi et al. 2005; Winter et al. 2007). Also from these studies, psilocybin at 1 mg/kg partially (∼50%) substitutes for LSD, and M100907 completely blocks the substitution. M100907 (0.5 mg/kg) also completely blocks 2C-T-7’s stimulus effects. M100907 (0.2 mg/kg) partially (∼40%) blocks psilocybin’s stimulus effects, whereas (±)-8-OH-DPAT mostly (∼80%) blocks them; the effect of (±)-8-OH-DPAT is not blocked by the selective 5-HT1A antagonist, WAY-100635 (0.2 mg/kg), nor does this compound on its own (up to 0.6 mg/kg) alter the stimulus effects of psilocybin. In humans, however, the non-hallucinogenic 5-HT1A receptor partial agonist buspirone reduces psilocybin-induced simple and complex hallucinations (Pokorny et al. 2016). Clearly, different mechanisms contribute to the stimulus effects of different serotonergic psychedelics, though, again, like the head-twitch response, 5-HT2A receptors appear to control a significant portion of the effects, with some exceptions. This translates well to humans, as the psychedelic effects of both psilocybin and LSD are blocked by a fairly selective 5-HT2A antagonist, ketanserin – though see Sect. 3.6.

Similar to the head-twitch model, compounds targeting receptors other than 5-HT2A modulate the discriminative stimulus effects of serotonergic psychedelics, and false positives, false negatives, and misunderstood results have emerged (Benneyworth et al. 2005; Reissig et al. 2005; Winter 2009). For example, lisuride substitutes for a number of serotonergic psychedelics in the two-lever drug discrimination paradigm; however, this can be overcome by training animals to discriminate two training drugs and vehicle. Thus, when animals are trained to discriminate lisuride, LSD, and vehicle, lisuride does not substitute for LSD (Appel et al. 2004). Regarding false negative responses, LSD, DOM, and DOI substitute for fenfluramine (Glennon 1991; McCreary et al. 2003). DOI, at 1 mg/kg, engenders rats to respond to the (±)-fenfluramine (2 mg/kg)-associated lever ∼73% of the time, and this effect is completely blocked by the 5-HT2B/5-HT2C inverse agonist, SB206553 (1 mg/kg); interestingly, M100907 also dose-dependently attenuates (but does not fully block) the effect (McCreary et al. 2003). In C57BL/6J mice, SB206553 suppresses the DOI-elicited head-twitch response (Canal et al. 2010, 2013a), suggesting that 5-HT2B or 5-HT2C receptors may contribute to DOI’s effects. Often overlooked, M100907 has relevant affinity at mouse and human 5-HT2C receptors (Ki ∼40–100 nM (Canal et al. 2013a); PDSP certified), where it acts as a 5-HT2C inverse agonist with potency and efficacy similar to the well-appreciated 5-HT2C inverse agonist, clozapine (Herrick-Davis et al. 2000; Kehne et al. 1996; Navailles et al. 2006; Rauser et al. 2001).

MDMA nearly (∼80%) substitutes for DMT and similarly nearly (∼80%) substitutes for methamphetamine in two-lever drug discrimination assays (Gatch et al. 2009). mGluR2 activation, which suppresses DOB- and DOI-elicited head-twitches, fails to alter discriminative stimulus effects of DIPT, DMT, and DOB in mice (Benneyworth et al. 2008; Carbonaro et al. 2015; Griebel et al. 2016). However, the prolonged training regimen and multidosing of serotonergic psychedelics that drug discrimination requires may cause functional changes in mGluR2 receptors that mask effects of mGluR2 activation (Benneyworth et al. 2008). Interestingly, the 5-HT3 antagonist/5-HT4 agonist zacopride potently and efficaciously reduces the discriminative stimulus properties of DOM (and MDMA) (Glennon et al. 1992). Finally, unique serotonergic psychedelics may produce different stimulus properties depending on their training dose or depending on when the training drug is administered prior to engaging in the associative learning task. For example, LSD’s stimulus effects are 5-HT2A mediated 30 min after its administration but appear to be dopamine D2 receptor mediated 90 min after administration (Marona-Lewicka et al. 2005). Interpretation of results from drug discrimination studies should consider different targets engaged by training drugs administered at different doses and at different times.

5 Measuring Localized and System Effects in the Brain

Recent, clinical neuroimaging studies have revealed serotonergic psychedelic effects that may explain not only neural perturbations that underlie visual hallucinations, e.g., decreases in alpha oscillations in visual cortex (Carhart-Harris et al. 2016; Kometer et al. 2013; Kometer and Vollenweider 2016), but also psychotherapeutic benefits of serotonergic psychedelics. One hypothesis, supported by recent observations, is that psychedelics have entropic effects on cortical activity, razing entrenched functional connectivity while sprouting new patterns of connectivity (Carhart-Harris et al. 2014; Petri et al. 2014; Tagliazucchi et al. 2014, 2016); for detailed discussions of this evolving topic, refer to Atasoy et al. (2017), Carhart-Harris et al. (2017), and Viol et al. (2017). Studies show that brains from subjects with treatment-resistant depression exhibit hyperactivity (entrenchment) within certain circuits, including the subcallosal cingulate white matter, which has been subsequently targeted by deep-brain stimulation (Mayberg et al. 1997; Riva-Posse et al. 2017). Psilocybin helps relieve treatment-resistant depression (Carhart-Harris et al. 2017), and intriguingly, psilocybin and LSD alter activity within the cingulate cortex and functional connectivity between the cingulate cortex and other brain systems (Carhart-Harris et al. 2012, 2016). These neural perturbations correlate with dissolution of ego boundaries (loss of the self), suggesting the possibility that this psychedelic phenomenon may, in certain individuals, help relieve psychiatric distress (c.f. Griffiths et al. 2016; Ross et al. 2016; Vollenweider 2001).

Preclinical strategies can provide additional spatial and temporal precision and a reductionist understanding of mechanisms which span genetic, molecular, cellular, and neural systems. Approaches to measure localized effects include direct brain injections, via drug infusions through stereotaxically implanted cannulae as well as systemic injections followed by measurements of changes in neurochemicals in discrete neural systems, e.g., via in vivo microdialysis combined with high-performance liquid chromatography and/or liquid chromatography-mass spectrometry or voltammetry detection (Bucher and Wightman 2015; Kennedy 2013). Measurements of electrophysiological effects on distinct brain cell types in distinct brain regions, e.g., via multichannel recordings from precisely implanted electrodes or from brain slice preparations (Du et al. 2017), can provide information about 5-HT2 effects on cell and network excitability, neurotransmission, and neuroplasticity. Invasive approaches also allow researchers to measure, for example, psychedelic-induced changes in DNA methylation (epigenetics), RNA transcription (gene expression), protein synthesis, and phosphorylation (posttranslational modifications). Serotonergic psychedelics alter the expression of genes that contribute to synaptic plasticity, providing physiological evidence that these compounds cause persistent changes in the brain that may underlie their therapeutic effects (Martin and Nichols 2017). For example, 24-h treatment of 45-day-old, brain organoids with 5-MeO-DMT alters the expression of proteins involved in memory consolidation, cytoskeletal organization, and inflammation, e.g., CaMKII, ephrin-B2, and NF-κB, respectively (Dakic et al. 2017).

Invasive techniques also permit analyses of interactions between receptor targets and other proteins. 5-HT2A and 5-HT2C receptors directly bind to synaptic scaffolding proteins, such as PSD-95 (Becamel et al. 2004; Xia et al. 2003). Also, 5-HT2A and glutamate mGluR2 receptors interact closely, and their interaction may be a key mechanism underlying psychedelic effects (Benneyworth et al. 2007; Delille et al. 2012; Gonzalez-Maeso et al. 2008; Lee et al. 2014; Moreno et al. 2011). Similarly, analyzing gene expression patterns allows researchers to observe which genes the brain expresses in regions where receptor targets of serotonergic psychedelics are expressed. Using the Allen Brain Atlas online resource, one can see that the gene coding for 5-HT2A receptors, HTR2A, is expressed in cortical brain regions that overlap very closely with expression of the gene coding for one of the subunits of glutamate NMDA receptors, NR2A, which the psychedelic dissociative ketamine targets. This connection fits well with results from PET (18fluorodeoxyglucose) imaging studies that show psilocybin and ketamine produce similar prefrontal cortex-limbic activation patterns in humans (Vollenweider 2001).

5.1 Serotonergic Psychedelics Impact on Neurotransmission

The effects of serotonergic psychedelics on neurotransmission have only been resolved partially. After systemic injections in rodents, serotonergic psychedelics increase glutamate in the cortex and also the ventral tegmentum area (Muschamp et al. 2004; Pehek et al. 2006; Scruggs et al. 2003). They also increase dopamine release in the cortex, but not the nucleus accumbens or the striatum (Di Matteo et al. 2000; Gudelsky et al. 1994; Pehek and Hernan 2015; Pehek et al. 2001, 2006). DOI increases acetylcholine release in the prefrontal cortex and the hippocampus (Nair and Gudelsky 2004; Zhelyazkova-Savova et al. 1997, 1999); also Nair and Gudelsky (2004) show that mescaline increases acetylcholine release in the prefrontal cortex, but not the hippocampus. DOI decreases norepinephrine release in the hippocampus (Done and Sharp 1992), though this study was performed in anesthetized rats. Finally, DOI increases GABA release in the prefrontal cortex (Abi-Saab et al. 1999). Most of the neurochemical effects of compounds in the aforementioned studies were blocked by 5-HT2 antagonists.

Early studies showed that serotonergic psychedelics increase brain 5-HT levels (Freedman and Giarman 1962; Giarman and Freedman 1965), but few studies since have systematically dissected this effect. One study shows that peripherally administered DOI has no effect on 5-HT release in the prefrontal cortex (Gobert and Millan 1999), yet others show that DOI significantly decreases 5-HT release there, in anesthetized rats (Martin-Ruiz et al. 2001; Wright et al. 1990). Other studies show that direct prefrontal cortex application of DOI increases 5-HT release there (Amargos-Bosch et al. 2004; Bortolozzi et al. 2003; Martin-Ruiz et al. 2001) or has no effect (Wright et al. 1990). Still another shows that DOI decreases 5-HT release from cortical slices while concordantly increasing GABA release; the increase in GABA release caused by DOI is also observed using cortical synaptosome preparations (Luparini et al. 2004). Finally, direct striatal DOI application increases 5-HT release there (Abellan et al. 2000).

A few recent studies have employed electrophysiological approaches to examine effects on neurotransmission. The 5-HT2A agonist and putative psychedelic, TCB-2, inhibits pyramidal neurons in layer 6 of the prefrontal cortex (Tian et al. 2016); an earlier study showed that 5-HT2 receptor activation stimulates (presumably) GABA interneurons in layer 2/3 of piriform cortex (Gellman and Aghajanian 1994). 5-HT2 receptors also increase activity of layer 5 GABAergic interneurons and glutamatergic pyramidal neurons (Aghajanian and Marek 1997; Spindle and Thomas 2014; Weber and Andrade 2010; Zhang and Arsenault 2005). These results align with the robust expression of 5-HT2 receptors on both GABAergic and glutamatergic neurons in the cortex (Puig et al. 2010; Willins et al. 1997). Finally, a recent study shows that low-dose LSD inhibits dorsal raphe nuclei 5-HT firing, and high-dose LSD additionally decreases firing of ventral tegmentum area dopamine neurons; the former effects are blocked by haloperidol and M100907, and latter effects are blocked by haloperidol, WAY-100635, and the TAAR1 antagonist, EPPTB (De Gregorio et al. 2016).

5.2 Neural Systems Underlying Serotonergic Psychedelic Effects

Elaine Sanders-Bush’s group combined brain microinfusions with drug discrimination to directly test the contribution of the anterior cingulate cortex to the discriminative stimulus properties of LSD; they found that local infusions of LSD substitute for systemically administered LSD, and that local infusions of M100907 block LSD’s discriminative stimulus properties (Gresch et al. 2007). Similarly, DOI directly infused into the anterior cingulate cortex elicits a head-twitch response, which is blocked by systemic injections of M100907 (Willins and Meltzer 1997). Despite these major findings, few other studies have directly assessed the involvement of other neural systems in serotonergic psychedelic effects (Halberstadt and Geyer 2017). Most information is based on correlation analyses from clinical trials, but these reports support preclinical observations. PET imaging with [18F]altanserin, a 5-HT2A antagonist radioligand, showed that psilocybin’s receptor occupancy in the anterior cingulate and medial prefrontal cortices correlates with psychedelic intensity (Quednow et al. 2010). Functional changes in the brain caused by serotonergic psychedelics include activity increases and decreases as well as modulation of interactions within and across cortical regions, between the thalamus and cortex, hippocampus, amygdala, and cortex, and across regions typically not functionally associated, typifying entropic effects of serotonergic psychedelics – some of these changes significantly correlate with distinct psychedelic effects (c.f. Carhart-Harris et al. 2016; Mueller et al. 2017; Palhano-Fontes et al. 2015; Petri et al. 2014; Tagliazucchi et al. 2016).

6 On Ketanserin, Conformations, and Dimerization

Reports from clinical trials conclude that the psychedelic effects of psilocybin and LSD are mediated by 5-HT2A receptors, because they are blocked by ketanserin (40 mg, P.O.), typically viewed as a selective 5-HT2A antagonist (Kometer et al. 2012; Kraehenmann et al. 2017; Preller et al. 2017; Quednow et al. 2012). Haloperidol, typically viewed as a selective dopamine D2 antagonist, is much less effective than ketanserin at blocking psilocybin’s effects, but risperidone, an antipsychotic with combined D2/5-HT2 activity, is as effective as ketanserin (Vollenweider et al. 1998).

Ketanserin, however, at <2 nM concentration labels a site(s) distinct from 5-HT2A receptors in several species, including humans, and in several neural systems, notably the striatum, substantia nigra, and raphe nuclei; in rats, this site appears to control the release of the dopamine metabolite DOPAC from dopamine nerve terminals (Leysen et al. 1987; Lopez-Gimenez et al. 1998; Pazos et al. 1987). Recently, Glennon commented on ketanserin’s off-target effects (Glennon 2017):

The lack of ketanserin’s selectivity for 5-HT2 receptors over some other receptors, notably histamine receptors and certain adrenoceptors, was a drawback when brain homogenates were being employed as the receptor source.

Importantly, M100907, viewed as one of the most selective, commercially available 5-HT2A antagonists, only effects ∼50% of ketanserin-appropriate lever responding in rats trained to discriminate ketanserin from saline; prazosin, an α1-adrenergic receptor antagonist, combined with M100907 causes full substitution (Li et al. 2009b). These observations suggest that ketanserin blocks α1-adrenergic receptors in vivo, which produces a subjectively recognizable effect. Importantly, α1-adrenergic receptors co-localize with 5-HT2A receptors in the prefrontal cortex (Santana et al. 2013), suggesting they may functionally interact in vivo. In addition to α1-adrenergic receptors, ketanserin also has relevant (off-target) affinity at human H1, 5-HT1D, 5-HT1F, and 5-HT2C receptors (Boess and Martin 1994; Bonhaus et al. 1995; Domenech et al. 1997; Ghoneim et al. 2006), and 5-HT2C receptors also co-localize with 5-HT2A receptors in certain parts of the cortex (Santana and Artigas 2017).

When considering haloperidol’s inefficacy at blocking psychedelic effects of psilocybin in humans and DOI’s discriminative stimulus properties in rats (Schreiber et al. 1994; Vollenweider et al. 1998), it should be noted that haloperidol has no activity at 5-HT2C receptors but possesses relevant affinity at 5-HT2A receptors (Herrick-Davis et al. 2000; Kroeze et al. 2003; Leysen et al. 1993; Rauser et al. 2001; Richelson and Souder 2000). Conversely, ketanserin and risperidone are efficacious 5-HT2C inverse agonists that also have high affinity at 5-HT2A (Hartman and Northup 1996; Herrick-Davis et al. 2000; Richelson and Souder 2000). Moreover, in competition binding assays, risperidone and ketanserin recognize two 5-HT2A receptor sites (defined by two slopes in the displacement curves) labeled with [3H](±)-DOB, whereas haloperidol recognizes only one site. In functional assays, each drug antagonizes 5-HT2A-stimulated inositol phosphate production and arachidonic acid release, but risperidone and ketanserin antagonize the latter in a biphasic manner similar to their binding characteristics (Brea et al. 2009). The authors suggest that the pharmacological differences are due to differential recognition of 5-HT2A receptor homodimers – that risperidone and ketanserin bind 5-HT2A homodimers, but haloperidol does not. It is, therefore, intriguing to consider that haloperidol may be ineffective at blocking psilocybin’s psychedelic effects, because it may not block putative 5-HT2A homodimers activated by psilocybin, whereas ketanserin and risperidone may. Risperidone potently blocks 5-HT2 mediated activation of (presumably) GABAergic interneurons in piriform cortex, whereas haloperidol, up to 10 µM, only weakly blocks these effects (Gellman and Aghajanian 1994). Alternatively, since haloperidol has negligible affinity at 5-HT2C receptors, by extension, it may not be able to bind 5-HT2A-5-HT2C heterodimers, which were recently observed in vitro and in brains (Moutkine et al. 2017). Co-activation of 5-HT2A and 5-HT2C receptors has been postulated as a mechanism underlying serotonergic psychedelic effects (Burris et al. 1991; Canal et al. 2010).

Finally, a couple of recent clinical trials suggest that 5-HT2A receptor activation may not be necessary or sufficient to produce all of the psychoactive effects of serotonergic psychedelics. Ketanserin (40 mg, P.O.) does not entirely block the effects of an ayahuasca brew (containing DMT, harmine, harmaline, and THH); notably, it does not block “modifications in time perception,” feeling “high,” or “visions” (Valle et al. 2016). This report suggests that there is a mechanism other than 5-HT2A at large, and the authors propose to further investigate sigma-1 receptor activation (Valle et al. 2016). Alternatively, ketanserin may not block unique 5-HT2 receptor ensembles and/or conformations stabilized by ayahuasca. [125I]DOI autoradiography studies show that 5-HT2A and 5-HT2C receptors exist in multiple conformations across neural systems in the rat brain, and ketanserin and M100907 have different affinities at them (Lopez-Gimenez et al. 2013). Collectively, then, data suggest that different 5-HT2 antagonists may selectively block unique 5-HT2 receptor homodimer or heterodimer ensembles and/or receptor conformations – it remains unclear which are the mechanistic targets of psychedelics. Clearly, more serotonergic psychedelics research aimed at elucidating their mechanisms needs to be conducted.

7 Conclusions

Recent experiments have begun to unveil neuropsychological mechanisms of serotonergic psychedelics, and clinical studies with ketanserin support a central role for serotonin 5-HT2A receptors in producing psychedelic effects. There remains fertile ground, however, for much more discovery. Unknown are the cellular signal transduction conduits of 5-HT2 receptor activation, the 5-HT2 receptor – protein interactions, and the neural circuits and neurochemical processes within those circuits, that produce distinct psychedelic effects, e.g., “oceanic boundlessness” or “visionary restructuralization” (Dittrich 1998). Also mysterious are the mechanisms underlying unique psychoactive effects engendered by unique psychedelics. Indeed, it is unclear whether all serotonergic psychedelics can induce psychedelic effects matching those of LSD – experiencing oneness with the universe or an all-encompassing unity, transcending time and space, tapping into the unconscious or experiencing archetypes, and an ethereal, positive, overwhelming luminescent, mental state filled with awe and profound philosophical, spiritual, or religious meaning that is ineffable with words (Pahnke et al. 1970; Pahnke and Richards 1966). Future discoveries will weave together reductionist and emergentist points of view to construct lucid neurobiological pictures illuminating how serotonergic psychedelics work.

Acknowledgments

This work was supported by grants from the National Institute on Drug Abuse (R21DA040907) and the Department of Defense (W81XWH-17-1-0329). I express my sincere gratitude to Dr. Nader Moniri and Austen Casey for critically reading and providing feedback on an early draft of this manuscript.

Abbreviations

1P-LSD

1-Propionyl-lysergic acid diethylamide

25C-NBOMe

N-(2-Methoxybenzyl)-2,5-dimethoxy-4-bromophenethylamine

25CN-NBOH

N-(2-Hydroxybenzyl)-2,5-dimethoxy-4-cyanophenylethylamine

25I-NBOMe

N-(2-Methoxybenzyl)-2,5-dimethoxy-4-iodophenethylamine

2C-B

4-Bromo-2,5-dimethoxyphenethylamine

2C-I

4-Iodo-2,5-dimethoxyphenethylamine

2C-T-7

2,5-Dimethoxy-4-propylthiophenethylamine

5-APB

5-(2-Aminopropyl)benzofuran

5-HT

5-Hydroxytryptamine (serotonin)

5-MeO-DALT

5-Methoxy-N,N-diallyltryptamine

5-MeO-DIPT

5-Methoxy-N,N-diisopropyltryptamine

5-MeO-DMT

5-Methoxy-N,N-dimethyltryptamine

6-APB

6-(2-Aminopropyl)benzofuran

AL-LAD

N6-allyl-6-norlysergic acid diethylamide

AMPA

α-Amino-3-hydroxy-5-methyl-4-isoxazolepropionic acid

bk-2C-B

β-Keto-2,5-dimethoxy-4-bromophenethylamine

BOL-148

2-Bromo-lysergic acid diethylamide

CB1

Cannabinoid 1 receptor

DA

Dopamine

DIPT

N,N-Diisopropyltryptamine

DMT

N,N-Dimethyltryptamine

DOB

2,5-Dimethoxy-4-bromoamphetamine

DOI

2,5-Dimethoxy-4-iodoamphetamine

DOM

2,5-Dimethoxy-4-methylamphetamine

DOPAC

3,4-Dihydroxyphenylacetic acid

DPT

N,N-Dipropyltryptamine

IP3

Inositol 1,4,5-trisphosphate

LSA

Lysergamide

LSD

Lysergic acid diethylamide

LSM-775

Lysergic acid morpholide

LSZ

Lysergic acid 2,4-dimethylazetidide

mCPP

meta-Chlorophenylpiperazine

MDMA

3,4-Methylenedioxymethamphetamine

mGluR2

Metabotropic glutamate receptor 2

NMDA

N-Methyl-D-aspartate

PARGY-LAD

N6-Propynyl-6-norlysergic acid diethylamide

PET

Positron emission tomography

SERT

Serotonin transporter

TAAR1

Trace amine-associated receptor 1

TCB-2

1-(3-Bromo-2,5-dimethoxybicyclo[4.2.0]octa-1,3,5-trien-7-yl) methanamine

THC

Δ9-Tetrahydrocannabinol

THH

Tetrahydroharmine

References

  1. Abellan MT, Martin-Ruiz R, Artigas F. Local modulation of the 5-HT release in the dorsal striatum of the rat: an in vivo microdialysis study. Eur Neuropsychopharmacol. 2000;10:455–462. doi: 10.1016/s0924-977x(00)00118-8. [DOI] [PubMed] [Google Scholar]
  2. Abi-Saab WM, Bubser M, Roth RH, Deutch AY. 5-HT2 receptor regulation of extracellular GABA levels in the prefrontal cortex. Neuropsychopharmacology. 1999;20:92–96. doi: 10.1016/S0893-133X(98)00046-3. [DOI] [PubMed] [Google Scholar]
  3. Acuna-Castillo C, Villalobos C, Moya PR, Saez P, Cassels BK, Huidobro-Toro JP. Differences in potency and efficacy of a series of phenylisopropylamine/phenylethylamine pairs at 5-HT(2A) and 5-HT(2C) receptors. Br J Pharmacol. 2002;136:510–519. doi: 10.1038/sj.bjp.0704747. [DOI] [PMC free article] [PubMed] [Google Scholar]
  4. Aghajanian GK, Marek GJ. Serotonin induces excitatory postsynaptic potentials in apical dendrites of neocortical pyramidal cells. Neuropharmacology. 1997;36:589–599. doi: 10.1016/s0028-3908(97)00051-8. [DOI] [PubMed] [Google Scholar]
  5. Amargos-Bosch M, Bortolozzi A, Puig MV, Serrats J, Adell A, Celada P, Toth M, Mengod G, Artigas F. Co-expression and in vivo interaction of serotonin1A and serotonin2A receptors in pyramidal neurons of prefrontal cortex. Cereb Cortex. 2004;14:281–299. doi: 10.1093/cercor/bhg128. [DOI] [PubMed] [Google Scholar]
  6. Appel JB, West WB, Buggy J. LSD, 5-HT (serotonin), and the evolution of a behavioral assay. Neurosci Biobehav Rev. 2004;27:693–701. doi: 10.1016/j.neubiorev.2003.11.012. [DOI] [PubMed] [Google Scholar]
  7. Atasoy S, Roseman L, Kaelen M, Kringelbach ML, Deco G, Carhart-Harris RL. Connectome-harmonic decomposition of human brain activity reveals dynamical repertoire re-organization under LSD. Sci Rep. 2017;7:17661. doi: 10.1038/s41598-017-17546-0. [DOI] [PMC free article] [PubMed] [Google Scholar]
  8. Barclay Z, Dickson L, Robertson DN, Johnson MS, Holland PJ, Rosie R, Sun L, Fleetwood-Walker S, Lutz EM, Mitchell R. 5-HT2A receptor signalling through phospholipase D1 associated with its C-terminal tail. Biochem J. 2011;436:651–660. doi: 10.1042/BJ20101844. [DOI] [PubMed] [Google Scholar]
  9. Bécamel C, Gavarini S, Chanrion B, Alonso G, Galeotti N, Dumuis A, Bockaert J, Marin P. The serotonin 5-HT2A and 5-HT2C receptors interact with specific sets of PDZ proteins. J Biol Chem. 2004;279:20257–20266. doi: 10.1074/jbc.M312106200. [DOI] [PubMed] [Google Scholar]
  10. Bécamel C, Berthoux C, Barre A, Marin P. Growing evidence for heterogeneous synaptic localization of 5-HT2A receptors. ACS Chem Neurosci. 2017;8:897–899. doi: 10.1021/acschemneuro.6b00409. [DOI] [PubMed] [Google Scholar]
  11. Beliveau V, Ganz M, Feng L, Ozenne B, Hojgaard L, Fisher PM, Svarer C, Greve DN, Knudsen GM. A high-resolution in vivo atlas of the human brain’s serotonin system. J Neurosci. 2017;37:120–128. doi: 10.1523/JNEUROSCI.2830-16.2016. [DOI] [PMC free article] [PubMed] [Google Scholar]
  12. Benneyworth MA, Smith RL, Barrett RJ, Sanders-Bush E. Complex discriminative stimulus properties of (+)lysergic acid diethylamide (LSD) in C57Bl/6J mice. Psychopharmacology. 2005;179:854–862. doi: 10.1007/s00213-004-2108-z. [DOI] [PubMed] [Google Scholar]
  13. Benneyworth MA, Xiang Z, Smith RL, Garcia EE, Conn PJ, Sanders-Bush E. A selective positive allosteric modulator of metabotropic glutamate receptor subtype 2 blocks a hallucinogenic drug model of psychosis. Mol Pharmacol. 2007;72:477–484. doi: 10.1124/mol.107.035170. [DOI] [PubMed] [Google Scholar]
  14. Benneyworth MA, Smith RL, Sanders-Bush E. Chronic phenethylamine hallucinogen treatment alters behavioral sensitivity to a metabotropic glutamate 2/3 receptor agonist. Neuropsychopharmacology. 2008;33:2206–2216. doi: 10.1038/sj.npp.1301600. [DOI] [PubMed] [Google Scholar]
  15. Berg KA, Maayani S, Goldfarb J, Scaramellini C, Leff P, Clarke WP. Effector pathway-dependent relative efficacy at serotonin type 2A and 2C receptors: evidence for agonist-directed trafficking of receptor stimulus. Mol Pharmacol. 1998;54:94–104. [PubMed] [Google Scholar]
  16. Best AR, Regehr WG. Serotonin evokes endocannabinoid release and retrogradely suppresses excitatory synapses. J Neurosci. 2008;28:6508–6515. doi: 10.1523/JNEUROSCI.0678-08.2008. [DOI] [PMC free article] [PubMed] [Google Scholar]
  17. Blough BE, Landavazo A, Decker AM, Partilla JS, Baumann MH, Rothman RB. Interaction of psychoactive tryptamines with biogenic amine transporters and serotonin receptor subtypes. Psychopharmacology. 2014;231:4135–4144. doi: 10.1007/s00213-014-3557-7. [DOI] [PMC free article] [PubMed] [Google Scholar]
  18. Boess FG, Martin IL. Molecular biology of 5-HT receptors. Neuropharmacology. 1994;33:275–317. doi: 10.1016/0028-3908(94)90059-0. [DOI] [PubMed] [Google Scholar]
  19. Bonhaus DW, Bach C, DeSouza A, Salazar FH, Matsuoka BD, Zuppan P, Chan HW, Eglen RM. The pharmacology and distribution of human 5-hydroxytryptamine2B (5-HT2B) receptor gene products: comparison with 5-HT2A and 5-HT2C receptors. Br J Pharmacol. 1995;115:622–628. doi: 10.1111/j.1476-5381.1995.tb14977.x. [DOI] [PMC free article] [PubMed] [Google Scholar]
  20. Bortolozzi A, Amargos-Bosch M, Adell A, Diaz-Mataix L, Serrats J, Pons S, Artigas F. In vivo modulation of 5-hydroxytryptamine release in mouse prefrontal cortex by local 5-HT(2A) receptors: effect of antipsychotic drugs. Eur J Neurosci. 2003;18:1235–1246. doi: 10.1046/j.1460-9568.2003.02829.x. [DOI] [PubMed] [Google Scholar]
  21. Braden MR, Nichols DE. Assessment of the roles of serines 5.43(239) and 5.46(242) for binding and potency of agonist ligands at the human serotonin 5-HT2A receptor. Mol Pharmacol. 2007;72:1200–1209. doi: 10.1124/mol.107.039255. [DOI] [PubMed] [Google Scholar]
  22. Braden MR, Parrish JC, Naylor JC, Nichols DE. Molecular interaction of serotonin 5-HT2A receptor residues Phe339(6.51) and Phe340(6.52) with superpotent N-benzyl phenethylamine agonists. Mol Pharmacol. 2006;70:1956–1964. doi: 10.1124/mol.106.028720. [DOI] [PubMed] [Google Scholar]
  23. Brandt SD, Kavanagh PV, Westphal F, Stratford A, Elliott SP, Hoang K, Wallach J, Halberstadt AL. Return of the lysergamides. Part I: Analytical and behavioural characterization of 1-propionyl-d-lysergic acid diethylamide (1P–LSD) Drug Test Anal. 2016;8:891–902. doi: 10.1002/dta.1884. [DOI] [PMC free article] [PubMed] [Google Scholar]
  24. Brandt SD, Kavanagh PV, Twamley B, Westphal F, Elliott SP, Wallach J, Stratford A, Klein LM, McCorvy JD, Nichols DE, Halberstadt AL. Return of the lysergamides. Part IV: Analytical and pharmacological characterization of lysergic acid morpholide (LSM-775) Drug Test Anal. 2017a doi: 10.1002/dta.2222. [DOI] [PMC free article] [PubMed]
  25. Brandt SD, Kavanagh PV, Westphal F, Elliott SP, Wallach J, Colestock T, Burrow TE, Chapman SJ, Stratford A, Nichols DE, Halberstadt AL. Return of the lysergamides. Part II: Analytical and behavioural characterization of N6 -allyl-6-norlysergic acid diethylamide (AL-LAD) and (2’S,4’S)-lysergic acid 2,4-dimethylazetidide (LSZ) Drug Test Anal. 2017b;9:38–50. doi: 10.1002/dta.1985. [DOI] [PMC free article] [PubMed] [Google Scholar]
  26. Brea J, Castro M, Giraldo J, Lopez-Gimenez JF, Padin JF, Quintian F, Cadavid MI, Vilaro MT, Mengod G, Berg KA, Clarke WP, Vilardaga JP, Milligan G, Loza MI. Evidence for distinct antagonist-revealed functional states of 5-hydroxytryptamine(2A) receptor homodimers. Mol Pharmacol. 2009;75:1380–1391. doi: 10.1124/mol.108.054395. [DOI] [PubMed] [Google Scholar]
  27. Bucher ES, Wightman RM. Electrochemical analysis of neurotransmitters. Annu Rev Anal Chem (Palo Alto, Calif) 2015;8:239–261. doi: 10.1146/annurev-anchem-071114-040426. [DOI] [PMC free article] [PubMed] [Google Scholar]
  28. Bunzow JR, Sonders MS, Arttamangkul S, Harrison LM, Zhang G, Quigley DI, Darland T, Suchland KL, Pasumamula S, Kennedy JL, Olson SB, Magenis RE, Amara SG, Grandy DK. Amphetamine, 3,4-methylenedioxymethamphetamine, lysergic acid diethylamide, and metabolites of the catecholamine neurotransmitters are agonists of a rat trace amine receptor. Mol Pharmacol. 2001;60:1181–1188. doi: 10.1124/mol.60.6.1181. [DOI] [PubMed] [Google Scholar]
  29. Burris KD, Breeding M, Sanders-Bush E. (+)Lysergic acid diethylamide, but not its nonhallucinogenic congeners, is a potent serotonin 5HT1C receptor agonist. J Pharmacol Exp Ther. 1991;258:891–896. [PubMed] [Google Scholar]
  30. Butelman ER, Rus S, Prisinzano TE, Kreek MJ. The discriminative effects of the kappa-opioid hallucinogen salvinorin A in nonhuman primates: dissociation from classic hallucinogen effects. Psychopharmacology. 2010;210:253–262. doi: 10.1007/s00213-009-1771-5. [DOI] [PMC free article] [PubMed] [Google Scholar]
  31. Canal CE, Morgan D. Head-twitch response in rodents induced by the hallucinogen 2,5-dimethoxy-4-iodoamphetamine: a comprehensive history, a re-evaluation of mechanisms, and its utility as a model. Drug Test Anal. 2012;4:556–576. doi: 10.1002/dta.1333. [DOI] [PMC free article] [PubMed] [Google Scholar]
  32. Canal CE, Olaghere da Silva UB, Gresch PJ, Watt EE, Sanders-Bush E, Airey DC. The serotonin 2C receptor potently modulates the head-twitch response in mice induced by a phenethylamine hallucinogen. Psychopharmacology. 2010;209:163–174. doi: 10.1007/s00213-010-1784-0. [DOI] [PMC free article] [PubMed] [Google Scholar]
  33. Canal CE, Cordova-Sintjago TC, Villa NY, Fang LJ, Booth RG. Drug discovery targeting human 5-HT(2C) receptors: residues S3.36 and Y7.43 impact ligand-binding pocket structure via hydrogen bond formation. Eur J Pharmacol. 2011;673:1–12. doi: 10.1016/j.ejphar.2011.10.006. [DOI] [PMC free article] [PubMed] [Google Scholar]
  34. Canal CE, Booth RG, Morgan D. Support for 5-HT2C receptor functional selectivity in vivo utilizing structurally diverse, selective 5-HT2C receptor ligands and the 2,5-dimethoxy-4-iodoamphetamine elicited head-twitch response model. Neuropharmacology. 2013a;70:112–121. doi: 10.1016/j.neuropharm.2013.01.007. [DOI] [PMC free article] [PubMed] [Google Scholar]
  35. Canal CE, Cordova-Sintjago T, Liu Y, Kim MS, Morgan D, Booth RG. Molecular pharmacology and ligand docking studies reveal a single amino acid difference between mouse and human serotonin 5-HT2A receptors that impacts behavioral translation of novel 4-phenyl-2-dimethylaminotetralin ligands. J Pharmacol Exp Ther. 2013b;347:705–716. doi: 10.1124/jpet.113.208637. [DOI] [PMC free article] [PubMed] [Google Scholar]
  36. Canal CE, Felsing DE, Liu Y, Zhu W, Wood JT, Perry CK, Vemula R, Booth RG. An orally active phenylaminotetralin-chemotype serotonin 5-HT7 and 5-HT1A receptor partial agonist that corrects motor stereotypy in mouse models. ACS Chem Neurosci. 2015;6:1259–1270. doi: 10.1021/acschemneuro.5b00099. [DOI] [PubMed] [Google Scholar]
  37. Canton H, Verriele L, Millan MJ. Competitive antagonism of serotonin (5-HT)2C and 5-HT2A receptor-mediated phosphoinositide (PI) turnover by clozapine in the rat: a comparison to other antipsychotics. Neurosci Lett. 1994;181:65–68. doi: 10.1016/0304-3940(94)90561-4. [DOI] [PubMed] [Google Scholar]
  38. Carbonaro TM, Eshleman AJ, Forster MJ, Cheng K, Rice KC, Gatch MB. The role of 5-HT2A, 5-HT 2C and mGlu2 receptors in the behavioral effects of tryptamine hallucinogens N,N-dimethyltryptamine and N,N-diisopropyltryptamine in rats and mice. Psychopharmacology. 2015;232:275–284. doi: 10.1007/s00213-014-3658-3. [DOI] [PMC free article] [PubMed] [Google Scholar]
  39. Carhart-Harris RL, Erritzoe D, Williams T, Stone JM, Reed LJ, Colasanti A, Tyacke RJ, Leech R, Malizia AL, Murphy K, Hobden P, Evans J, Feilding A, Wise RG, Nutt DJ. Neural correlates of the psychedelic state as determined by fMRI studies with psilocybin. Proc Natl Acad Sci. 2012;109:2138–2143. doi: 10.1073/pnas.1119598109. [DOI] [PMC free article] [PubMed] [Google Scholar]
  40. Carhart-Harris RL, Leech R, Hellyer PJ, Shanahan M, Feilding A, Tagliazucchi E, Chialvo DR, Nutt D. The entropic brain: a theory of conscious states informed by neuroimaging research with psychedelic drugs. Front Hum Neurosci. 2014;8:20. doi: 10.3389/fnhum.2014.00020. [DOI] [PMC free article] [PubMed] [Google Scholar]
  41. Carhart-Harris RL, Muthukumaraswamy S, Roseman L, Kaelen M, Droog W, Murphy K, Tagliazucchi E, Schenberg EE, Nest T, Orban C, Leech R, Williams LT, Williams TM, Bolstridge M, Sessa B, McGonigle J, Sereno MI, Nichols D, Hellyer PJ, Hobden P, Evans J, Singh KD, Wise RG, Curran HV, Feilding A, Nutt DJ. Neural correlates of the LSD experience revealed by multimodal neuroimaging. Proc Natl Acad Sci U S A. 2016;113:4853–4858. doi: 10.1073/pnas.1518377113. [DOI] [PMC free article] [PubMed] [Google Scholar]
  42. Carhart-Harris RL, Roseman L, Bolstridge M, Demetriou L, Pannekoek JN, Wall MB, Tanner M, Kaelen M, McGonigle J, Murphy K, Leech R, Curran HV, Nutt DJ. Psilocybin for treatment-resistant depression: fMRI-measured brain mechanisms. Sci Rep. 2017;7:13187. doi: 10.1038/s41598-017-13282-7. [DOI] [PMC free article] [PubMed] [Google Scholar]
  43. Ceci C, Proietti Onori M, Macri S, Laviola G. Interaction between the endocannabinoid and serotonergic system in the exhibition of head twitch response in four mouse strains. Neurotox Res. 2015;27:275–283. doi: 10.1007/s12640-014-9510-z. [DOI] [PubMed] [Google Scholar]
  44. Chambers JJ, Nichols DE. A homology-based model of the human 5-HT2A receptor derived from an in silico activated G-protein coupled receptor. J Comput Aided Mol Des. 2002;16:511–520. doi: 10.1023/a:1021275430021. [DOI] [PubMed] [Google Scholar]
  45. Cheng Y, Prusoff WH. Relationship between the inhibition constant (K1) and the concentration of inhibitor which causes 50% inhibition (I50) of an enzymatic reaction. Biochem Pharmacol. 1973;22:3099–3108. doi: 10.1016/0006-2952(73)90196-2. [DOI] [PubMed] [Google Scholar]
  46. Choudhary MS, Sachs N, Uluer A, Glennon RA, Westkaemper RB, Roth BL. Differential ergoline and ergopeptine binding to 5-hydroxytryptamine2A receptors: ergolines require an aromatic residue at position 340 for high affinity binding. Mol Pharmacol. 1995;47:450–457. [PubMed] [Google Scholar]
  47. Clark LD, Bliss EL. Psychopharmacological studies of lysergic acid diethylamide (LSD-25) intoxication; effects of premedication with BOL-128 (2-bromo-d-lysergic acid diethylamide), mescaline, atropine, amobarbital, and chlorpromazine. AMA Arch Neurol Psychiatry. 1957;78:653–655. doi: 10.1001/archneurpsyc.1957.02330420113020. [DOI] [PubMed] [Google Scholar]
  48. Cordova-Sintjago T, Villa N, Fang L, Booth RG. Aromatic interactions impact ligand binding and function at serotonin 5-HT2C G protein-coupled receptors: receptor homology modeling, ligand docking, and molecular dynamics results validated by experimental studies. Mol Phys. 2014;112:398–407. doi: 10.1080/00268976.2013.833656. [DOI] [PMC free article] [PubMed] [Google Scholar]
  49. Corne SJ, Pickering RW. A possible correlation between drug-induced hallucinations in man and a behavioural response in mice. Psychopharmacologia. 1967;11:65–78. doi: 10.1007/BF00401509. [DOI] [PubMed] [Google Scholar]
  50. Cozzi NV, Daley PF. Receptor binding profiles and quantitative structure-affinity relationships of some 5-substituted-N,N-diallyltryptamines. Bioorg Med Chem Lett. 2016;26:959–964. doi: 10.1016/j.bmcl.2015.12.053. [DOI] [PubMed] [Google Scholar]
  51. Cussac D, Boutet-Robinet E, Ailhaud MC, Newman-Tancredi A, Martel JC, Danty N, Rauly-Lestienne I. Agonist-directed trafficking of signalling at serotonin 5-HT2A, 5-HT2B and 5-HT2C-VSV receptors mediated Gq/11 activation and calcium mobilisation in CHO cells. Eur J Pharmacol. 2008;594:32–38. doi: 10.1016/j.ejphar.2008.07.040. [DOI] [PubMed] [Google Scholar]
  52. Dakic V, Minardi Nascimento J, Costa Sartore R, Maciel RM, de Araujo DB, Ribeiro S, Martins-de-Souza D, Rehen SK. Short term changes in the proteome of human cerebral organoids induced by 5-MeO-DMT. Sci Rep. 2017;7:12863. doi: 10.1038/s41598-017-12779-5. [DOI] [PMC free article] [PubMed] [Google Scholar]
  53. Darmani NA. Cocaine and selective monoamine uptake blockers (sertraline, nisoxetine, and GBR 12935) prevent the d-fenfluramine-induced head-twitch response in mice. Pharmacol Biochem Behav. 1998;60:83–90. doi: 10.1016/s0091-3057(97)00548-0. [DOI] [PubMed] [Google Scholar]
  54. Darmani NA. Cannabinoids of diverse structure inhibit two DOI-induced 5-HT (2A) receptor-mediated behaviors in mice. Pharmacol Biochem Behav. 2001;68:311–317. doi: 10.1016/s0091-3057(00)00477-9. [DOI] [PubMed] [Google Scholar]
  55. Darmani NA, Pandya DK. Involvement of other neurotransmitters in behaviors induced by the cannabinoid CB1 receptor antagonist SR 141716A in naive mice. J Neural Transm (Vienna) 2000;107:931–945. doi: 10.1007/s007020070043. [DOI] [PubMed] [Google Scholar]
  56. Darmani NA, Reeves SL. The mechanism by which the selective 5-HT1A receptor antagonist S-(-) UH 301 produces head-twitches in mice. Pharmacol Biochem Behav. 1996;55:1–10. doi: 10.1016/0091-3057(96)00072-x. [DOI] [PubMed] [Google Scholar]
  57. Darmani NA, Mock OB, Towns LC, Gerdes CF. The head-twitch response in the least shrew (Cryptotis parva) is a 5-HT2- and not a 5-HT1C-mediated phenomenon. Pharmacol Biochem Behav. 1994;48:383–396. doi: 10.1016/0091-3057(94)90542-8. [DOI] [PubMed] [Google Scholar]
  58. Darmani NA, Janoyan JJ, Kumar N, Crim JL. Behaviorally active doses of the CB1 receptor antagonist SR 141716A increase brain serotonin and dopamine levels and turnover. Pharmacol Biochem Behav. 2003;75:777–787. doi: 10.1016/s0091-3057(03)00150-3. [DOI] [PubMed] [Google Scholar]
  59. De Gregorio D, Posa L, Ochoa-Sanchez R, McLaughlin R, Maione S, Comai S, Gobbi G. The hallucinogen d-lysergic diethylamide (LSD) decreases dopamine firing activity through 5-HT1A, D2 and TAAR1 receptors. Pharmacol Res. 2016;113:81–91. doi: 10.1016/j.phrs.2016.08.022. [DOI] [PubMed] [Google Scholar]
  60. de Witte WE, Danhof M, van der Graaf PH, de Lange EC. In vivo target residence time and kinetic selectivity: the association rate constant as determinant. Trends Pharmacol Sci. 2016;37:831–842. doi: 10.1016/j.tips.2016.06.008. [DOI] [PubMed] [Google Scholar]
  61. Delille HK, Becker JM, Burkhardt S, Bleher B, Terstappen GC, Schmidt M, Meyer AH, Unger L, Marek GJ, Mezler M. Heterocomplex formation of 5-HT2A-mGlu2 and its relevance for cellular signaling cascades. Neuropharmacology. 2012;62:2184–2191. doi: 10.1016/j.neuropharm.2012.01.010. [DOI] [PubMed] [Google Scholar]
  62. DeVree BT, Mahoney JP, Vélez-Ruiz GA, Rasmussen SGF, Kuszak AJ, Edwald E, Fung J-J, Manglik A, Masureel M, Du Y, Matt RA, Pardon E, Steyaert J, Kobilka BK, Sunahara RK. Allosteric coupling from G protein to the agonist-binding pocket in GPCRs. Nature. 2016;535:182. doi: 10.1038/nature18324. [DOI] [PMC free article] [PubMed] [Google Scholar]
  63. Di Matteo V, Di Giovanni G, Di Mascio M, Esposito E. Biochemical and electrophysiologi-cal evidence that RO 60-0175 inhibits mesolimbic dopaminergic function through serotonin (2C) receptors. Brain Res. 2000;865:85–90. doi: 10.1016/s0006-8993(00)02246-0. [DOI] [PubMed] [Google Scholar]
  64. Dittrich A. The standardized psychometric assessment of altered states of consciousness (ASCs) in humans. Pharmacopsychiatry. 1998;31(Suppl 2):80–84. doi: 10.1055/s-2007-979351. [DOI] [PubMed] [Google Scholar]
  65. Domenech T, Beleta J, Palacios JM. Characterization of human serotonin 1D and 1B receptors using [3H]-GR-125743, a novel radiolabelled serotonin 5HT1D/1B receptor antagonist. Naunyn Schmiedeberg’s Arch Pharmacol. 1997;356:328–334. doi: 10.1007/pl00005058. [DOI] [PubMed] [Google Scholar]
  66. Done CJ, Sharp T. Evidence that 5-HT2 receptor activation decreases noradrenaline release in rat hippocampus in vivo. Br J Pharmacol. 1992;107:240–245. doi: 10.1111/j.1476-5381.1992.tb14493.x. [DOI] [PMC free article] [PubMed] [Google Scholar]
  67. Du C, Collins W, Cantley W, Sood D, Kaplan DL. Tutorials for electrophysiological recordings in neuronal tissue engineering. ACS Biomater Sci Eng. 2017;3:2235–2246. doi: 10.1021/acsbiomaterials.7b00318. [DOI] [PubMed] [Google Scholar]
  68. Dursun SM, Handley SL. The effects of alpha 2-adrenoceptor antagonists on the inhibition of 1-(2,5-dimethoxy-4-iodophenyl)-2-aminopropane (DOI)-induced head shakes by 5-HT1A receptor agonists in the mouse. Br J Pharmacol. 1993;109:1046–1052. doi: 10.1111/j.1476-5381.1993.tb13727.x. [DOI] [PMC free article] [PubMed] [Google Scholar]
  69. Egan CT, Herrick-Davis K, Miller K, Glennon RA, Teitler M. Agonist activity of LSD and lisuride at cloned 5HT2A and 5HT2C receptors. Psychopharmacology. 1998a;136:409–414. doi: 10.1007/s002130050585. [DOI] [PubMed] [Google Scholar]
  70. Egan CT, Herrick-Davis K, Teitler M. Creation of a constitutively activated state of the 5-hydroxytryptamine2A receptor by site-directed mutagenesis: inverse agonist activity of anti-psychotic drugs. J Pharmacol Exp Ther. 1998b;286:85–90. [PubMed] [Google Scholar]
  71. Egan C, Grinde E, Dupre A, Roth BL, Hake M, Teitler M, Herrick-Davis K. Agonist high and low affinity state ratios predict drug intrinsic activity and a revised ternary complex mechanism at serotonin 5-HT(2A) and 5-HT(2C) receptors. Synapse. 2000;35:144–150. doi: 10.1002/(SICI)1098-2396(200002)35:2<144::AID-SYN7>3.0.CO;2-K. [DOI] [PubMed] [Google Scholar]
  72. Egashira N, Mishima K, Uchida T, Hasebe N, Nagai H, Mizuki A, Iwasaki K, Ishii H, Nishimura R, Shoyama Y, Fujiwara M. Anandamide inhibits the DOI-induced head-twitch response in mice. Psychopharmacology. 2004;171:382–389. doi: 10.1007/s00213-003-1611-y. [DOI] [PubMed] [Google Scholar]
  73. Egashira N, Shirakawa A, Okuno R, Mishima K, Iwasaki K, Oishi R, Fujiwara M. Role of endocannabinoid and glutamatergic systems in DOI-induced head-twitch response in mice. Pharmacol Biochem Behav. 2011;99:52–58. doi: 10.1016/j.pbb.2011.04.003. [DOI] [PubMed] [Google Scholar]
  74. Fantegrossi WE, Harrington AW, Eckler JR, Arshad S, Rabin RA, Winter JC, Coop A, Rice KC, Woods JH. Hallucinogen-like actions of 2,5-dimethoxy-4-(n)-propylthiophenethylamine (2C-T-7) in mice and rats. Psychopharmacology. 2005;181:496–503. doi: 10.1007/s00213-005-0009-4. [DOI] [PubMed] [Google Scholar]
  75. Fantegrossi WE, Harrington AW, Kiessel CL, Eckler JR, Rabin RA, Winter JC, Coop A, Rice KC, Woods JH. Hallucinogen-like actions of 5-methoxy-N,N-diisopropyltryptamine in mice and rats. Pharmacol Biochem Behav. 2006;83:122–129. doi: 10.1016/j.pbb.2005.12.015. [DOI] [PubMed] [Google Scholar]
  76. Fantegrossi WE, Reissig CJ, Katz EB, Yarosh HL, Rice KC, Winter JC. Hallucinogen-like effects of N,N-dipropyltryptamine (DPT): possible mediation by serotonin 5-HT1A and 5-HT2A receptors in rodents. Pharmacol Biochem Behav. 2008;88:358–365. doi: 10.1016/j.pbb.2007.09.007. [DOI] [PMC free article] [PubMed] [Google Scholar]
  77. Fantegrossi WE, Simoneau J, Cohen MS, Zimmerman SM, Henson CM, Rice KC, Woods JH. Interaction of 5-HT2A and 5-HT2C receptors in R(-)-2,5-dimethoxy-4-iodoamphetamine-elicited head twitch behavior in mice. J Pharmacol Exp Ther. 2010;335:728–734. doi: 10.1124/jpet.110.172247. [DOI] [PMC free article] [PubMed] [Google Scholar]
  78. Felder CC, Kanterman RY, Ma AL, Axelrod J. Serotonin stimulates phospholipase A2 and the release of arachidonic acid in hippocampal neurons by a type 2 serotonin receptor that is independent of inositolphospholipid hydrolysis. Proc Natl Acad Sci U S A. 1990;87:2187–2191. doi: 10.1073/pnas.87.6.2187. [DOI] [PMC free article] [PubMed] [Google Scholar]
  79. Fiorella D, Rabin RA, Winter JC. The role of the 5-HT2A and 5-HT2C receptors in the stimulus effects of hallucinogenic drugs. I: antagonist correlation analysis. Psychopharmacology. 1995;121:347–356. doi: 10.1007/BF02246074. [DOI] [PubMed] [Google Scholar]
  80. Fontanilla D, Johannessen M, Hajipour AR, Cozzi NV, Jackson MB, Ruoho AE. The hallucinogen N,N-dimethyltryptamine (DMT) is an endogenous sigma-1 receptor regulator. Science. 2009;323:934–937. doi: 10.1126/science.1166127. [DOI] [PMC free article] [PubMed] [Google Scholar]
  81. Freedman DX, Giarman NJ. LSD-25 and the status and level of brain serotonin. Ann N Y Acad Sci. 1962;96:98–107. doi: 10.1111/j.1749-6632.1962.tb50105.x. [DOI] [PubMed] [Google Scholar]
  82. Freud S, Byck R. Cocaine papers. Stonehill; New York: 1975. [Google Scholar]
  83. Garcia EE, Smith RL, Sanders-Bush E. Role of G(q) protein in behavioral effects of the hallucinogenic drug 1-(2,5-dimethoxy-4-iodophenyl)-2-aminopropane. Neuropharmacology. 2007;52:1671–1677. doi: 10.1016/j.neuropharm.2007.03.013. [DOI] [PMC free article] [PubMed] [Google Scholar]
  84. Gatch MB, Rutledge MA, Carbonaro T, Forster MJ. Comparison of the discriminative stimulus effects of dimethyltryptamine with different classes of psychoactive compounds in rats. Psychopharmacology. 2009;204:715–724. doi: 10.1007/s00213-009-1501-z. [DOI] [PMC free article] [PubMed] [Google Scholar]
  85. Gatch MB, Dolan SB, Forster MJ. Locomotor and discriminative stimulus effects of four novel hallucinogens in rodents. Behav Pharmacol. 2017;28:375–385. doi: 10.1097/FBP.0000000000000309. [DOI] [PMC free article] [PubMed] [Google Scholar]
  86. Gellman RL, Aghajanian GK. Serotonin2 receptor-mediated excitation of interneurons in piriform cortex: antagonism by atypical antipsychotic drugs. Neuroscience. 1994;58:515–525. doi: 10.1016/0306-4522(94)90077-9. [DOI] [PubMed] [Google Scholar]
  87. Ghoneim OM, Legere JA, Golbraikh A, Tropsha A, Booth RG. Novel ligands for the human histamine H1 receptor: synthesis, pharmacology, and comparative molecular field analysis studies of 2-dimethylamino-5-(6)-phenyl-1,2,3,4-tetrahydronaphthalenes. Bioorg Med Chem. 2006;14:6640–6658. doi: 10.1016/j.bmc.2006.05.077. [DOI] [PubMed] [Google Scholar]
  88. Giarman NJ, Freedman DX. Biochemical aspects of the actions of psychotomimetic drugs. Pharmacol Rev. 1965;17:1–25. [PubMed] [Google Scholar]
  89. Glennon RA. Discriminative stimulus properties of hallucinogens and related designer drugs. NIDA Res Monogr. 1991;116:25–44. [PubMed] [Google Scholar]
  90. Glennon RA. Animal models for assessing hallucinogenic agents. In: Boulton AA, Baker GB, Wu PH, editors. Animal models of drug addiction. Humana Press; Totowa: 1992. pp. 345–381. [Google Scholar]
  91. Glennon RA. The 2014 Philip S. Portoghese Medicinal Chemistry Lectureship: the “Phenylalkylaminome” with a focus on selected drugs of abuse. J Med Chem. 2017;60:2605–2628. doi: 10.1021/acs.jmedchem.7b00085. [DOI] [PMC free article] [PubMed] [Google Scholar]
  92. Glennon RA, Young R. Drug discrimination: applications to medicinal chemistry and drug studies. Wiley; Hoboken: 2011. [Google Scholar]
  93. Glennon RA, Young R, Rosecrans JA. Antagonism of the effects of the hallucinogen DOM and the purported 5-HT agonist quipazine by 5-HT2 antagonists. Eur J Pharmacol. 1983;91:189–196. doi: 10.1016/0014-2999(83)90464-8. [DOI] [PubMed] [Google Scholar]
  94. Glennon RA, Titeler M, McKenney JD. Evidence for 5-HT2 involvement in the mechanism of action of hallucinogenic agents. Life Sci. 1984;35:2505–2511. doi: 10.1016/0024-3205(84)90436-3. [DOI] [PubMed] [Google Scholar]
  95. Glennon RA, Higgs R, Young R, Issa H. Further studies on N-methyl-1 (3,4-methylenedioxyphenyl)-2-aminopropane as a discriminative stimulus: antagonism by 5-hydroxytryptamine3 antagonists. Pharmacol Biochem Behav. 1992;43:1099–1106. doi: 10.1016/0091-3057(92)90488-2. [DOI] [PubMed] [Google Scholar]
  96. Gobert A, Millan MJ. Serotonin (5-HT)2A receptor activation enhances dialysate levels of dopamine and noradrenaline, but not 5-HT, in the frontal cortex of freely-moving rats. Neuro-pharmacology. 1999;38:315–317. doi: 10.1016/s0028-3908(98)00188-9. [DOI] [PubMed] [Google Scholar]
  97. Gonzalez-Maeso J, Weisstaub NV, Zhou M, Chan P, Ivic L, Ang R, Lira A, Bradley-Moore M, Ge Y, Zhou Q, Sealfon SC, Gingrich JA. Hallucinogens recruit specific cortical 5-HT (2A) receptor-mediated signaling pathways to affect behavior. Neuron. 2007;53:439–452. doi: 10.1016/j.neuron.2007.01.008. [DOI] [PubMed] [Google Scholar]
  98. Gonzalez-Maeso J, Ang RL, Yuen T, Chan P, Weisstaub NV, Lopez-Gimenez JF, Zhou M, Okawa Y, Callado LF, Milligan G, Gingrich JA, Filizola M, Meana JJ, Sealfon SC. Identification of a serotonin/glutamate receptor complex implicated in psychosis. Nature. 2008;452:93–97. doi: 10.1038/nature06612. [DOI] [PMC free article] [PubMed] [Google Scholar]
  99. Goodwin GM, Green AR. A behavioural and biochemical study in mice and rats of putative selective agonists and antagonists for 5-HT1 and 5-HT2 receptors. Br J Pharmacol. 1985;84:743–753. doi: 10.1111/j.1476-5381.1985.tb16157.x. [DOI] [PMC free article] [PubMed] [Google Scholar]
  100. Gresch PJ, Barrett RJ, Sanders-Bush E, Smith RL. 5-Hydroxytryptamine (serotonin)2A receptors in rat anterior cingulate cortex mediate the discriminative stimulus properties of d-lysergic acid diethylamide. J Pharmacol Exp Ther. 2007;320:662–669. doi: 10.1124/jpet.106.112946. [DOI] [PubMed] [Google Scholar]
  101. Griebel G, Pichat P, Boulay D, Naimoli V, Potestio L, Featherstone R, Sahni S, Defex H, Desvignes C, Slowinski F, Vige X, Bergis OE, Sher R, Kosley R, Kongsamut S, Black MD, Varty GB. The mGluR2 positive allosteric modulator, SAR218645, improves memory and attention deficits in translational models of cognitive symptoms associated with schizophrenia. Sci Rep. 2016;6:35320. doi: 10.1038/srep35320. [DOI] [PMC free article] [PubMed] [Google Scholar]
  102. Griffiths RR, Johnson MW, Carducci MA, Umbricht A, Richards WA, Richards BD, Cosimano MP, Klinedinst MA. Psilocybin produces substantial and sustained decreases in depression and anxiety in patients with life-threatening cancer: a randomized double-blind trial. J Psychopharmacol. 2016;30:1181–1197. doi: 10.1177/0269881116675513. [DOI] [PMC free article] [PubMed] [Google Scholar]
  103. Grundmann M, Kostenis E. Temporal bias: time-encoded dynamic GPCR signaling. Trends Pharmacol Sci. 2017;38:1110–1124. doi: 10.1016/j.tips.2017.09.004. [DOI] [PubMed] [Google Scholar]
  104. Gudelsky GA, Yamamoto BK, Nash JF. Potentiation of 3,4-methylenedioxymethamphetamine-induced dopamine release and serotonin neurotoxicity by 5-HT2 receptor agonists. Eur J Pharmacol. 1994;264:325–330. doi: 10.1016/0014-2999(94)90669-6. [DOI] [PubMed] [Google Scholar]
  105. Halberstadt AL. Pharmacology and toxicology of N-benzylphenethylamine (“NBOMe”) hallucinogens. Curr Top Behav Neurosci. 2017;32:283–311. doi: 10.1007/7854_2016_64. [DOI] [PubMed] [Google Scholar]
  106. Halberstadt AL, Geyer MA. Characterization of the head-twitch response induced by hallucinogens in mice: detection of the behavior based on the dynamics of head movement. Psychopharmacology. 2013;227:727–739. doi: 10.1007/s00213-013-3006-z. [DOI] [PMC free article] [PubMed] [Google Scholar]
  107. Halberstadt AL, Geyer MA. Effects of the hallucinogen 2,5-dimethoxy-4-iodophenethylamine (2C–I) and superpotent N-benzyl derivatives on the head twitch response. Neuropharmacology. 2014;77:200–207. doi: 10.1016/j.neuropharm.2013.08.025. [DOI] [PMC free article] [PubMed] [Google Scholar]
  108. Halberstadt AL, Geyer MA. Effect of hallucinogens on unconditioned behavior. Curr Top Behav Neurosci. 2017 doi: 10.1007/7854_2016_466. [DOI] [PMC free article] [PubMed]
  109. Halberstadt AL, Koedood L, Powell SB, Geyer MA. Differential contributions of serotonin receptors to the behavioral effects of indoleamine hallucinogens in mice. J Psychopharmacol. 2011;25:1548–1561. doi: 10.1177/0269881110388326. [DOI] [PMC free article] [PubMed] [Google Scholar]
  110. Hartman JL, Northup JK. Functional reconstitution in situ of 5-hydroxytryptamine2c (5HT2c) receptors with alphaq and inverse agonism of 5HT2c receptor antagonists. J Biol Chem. 1996;271:22591–22597. doi: 10.1074/jbc.271.37.22591. [DOI] [PubMed] [Google Scholar]
  111. Herrick-Davis K, Grinde E, Teitler M. Inverse agonist activity of atypical antipsychotic drugs at human 5-hydroxytryptamine2C receptors. J Pharmacol Exp Ther. 2000;295:226–232. [PubMed] [Google Scholar]
  112. Hide I, Kato T, Yamawaki S. In vivo determination of 5-hydroxytryptamine receptor-stimulated phosphoinositide turnover in rat brain. J Neurochem. 1989;53:556–560. doi: 10.1111/j.1471-4159.1989.tb07369.x. [DOI] [PubMed] [Google Scholar]
  113. Hoffmann C, Leitz MR, Oberdorf-Maass S, Lohse MJ, Klotz K-N. Comparative pharmacology of human β-adrenergic receptor subtypes – characterization of stably transfected receptors in CHO cells. Naunyn Schmiedeberg’s Arch Pharmacol. 2004;369:151–159. doi: 10.1007/s00210-003-0860-y. [DOI] [PubMed] [Google Scholar]
  114. Hua T, Vemuri K, Pu M, Qu L, Han GW, Wu Y, Zhao S, Shui W, Li S, Korde A, Laprairie RB, Stahl EL, Ho JH, Zvonok N, Zhou H, Kufareva I, Wu B, Zhao Q, Hanson MA, Bohn LM, Makriyannis A, Stevens RC, Liu ZJ. Crystal structure of the human cannabinoid receptor CB1. Cell. 2016;167(750–762):e14. doi: 10.1016/j.cell.2016.10.004. [DOI] [PMC free article] [PubMed] [Google Scholar]
  115. Hua T, Vemuri K, Nikas SP, Laprairie RB, Wu Y, Qu L, Pu M, Korde A, Jiang S, Ho JH, Han GW, Ding K, Li X, Liu H, Hanson MA, Zhao S, Bohn LM, Makriyannis A, Stevens RC, Liu ZJ. Crystal structures of agonist-bound human cannabinoid receptor CB1. Nature. 2017;547:468–471. doi: 10.1038/nature23272. [DOI] [PMC free article] [PubMed] [Google Scholar]
  116. Isbell H, Logan CR. Studies on the diethylamide of lysergic acid (LSD-25). II. Effects of chlorpromazine, azacyclonol, and reserpine on the intensity of the LSD-reaction. A M A Arch Neurol Psychiatry. 1957;77:350–358. doi: 10.1001/archneurpsyc.1957.02330340026002. [DOI] [PubMed] [Google Scholar]
  117. Isberg V, Balle T, Sander T, Jorgensen FS, Gloriam DE. G protein- and agonist-bound serotonin 5-HT2A receptor model activated by steered molecular dynamics simulations. J Chem Inf Model. 2011;51:315–325. doi: 10.1021/ci100402f. [DOI] [PubMed] [Google Scholar]
  118. Iversen L, Gibbons S, Treble R, Setola V, Huang X-P, Roth BL. Neurochemical profiles of some novel psychoactive substances. Eur J Pharmacol. 2013;700:147–151. doi: 10.1016/j.ejphar.2012.12.006. [DOI] [PMC free article] [PubMed] [Google Scholar]
  119. Jensen AA, McCorvy JD, Leth-Petersen S, Bundgaard C, Liebscher G, Kenakin TP, Brauner-Osborne H, Kehler J, Kristensen JL. Detailed characterization of the in vitro pharmacological and pharmacokinetic properties of N-(2-hydroxybenzyl)-2,5-dimethoxy-4-cyanophenylethylamine (25CN-NBOH), a highly selective and brain-penetrant 5-HT2A receptor agonist. J Pharmacol Exp Ther. 2017;361:441–453. doi: 10.1124/jpet.117.239905. [DOI] [PubMed] [Google Scholar]
  120. Johansen A, Hansen HD, Svarer C, Lehel S, Leth-Petersen S, Kristensen JL, Gillings N, Knudsen GM. The importance of small polar radiometabolites in molecular neuroimaging: a PET study with [(11)C]Cimbi-36 labeled in two positions. J Cereb Blood Flow Metab. 2017 doi: 10.1177/0271678X17746179. [DOI] [PMC free article] [PubMed]
  121. Jope RS, Song L, Powers R. [3H]PtdIns hydrolysis in postmortem human brain membranes is mediated by the G-proteins Gq/11 and phospholipase C-beta. Biochem J. 1994;304(Pt 2):655–659. doi: 10.1042/bj3040655. [DOI] [PMC free article] [PubMed] [Google Scholar]
  122. Kadamur G, Ross EM. Mammalian phospholipase C. Annu Rev Physiol. 2013;75:127–154. doi: 10.1146/annurev-physiol-030212-183750. [DOI] [PubMed] [Google Scholar]
  123. Karaki S, Becamel C, Murat S, Mannoury la Cour C, Millan MJ, Prezeau L, Bockaert J, Marin P, Vandermoere F. Quantitative phosphoproteomics unravels biased phosphorylation of serotonin 2A receptor at Ser280 by hallucinogenic versus nonhallucinogenic agonists. Mol Cell Proteomics. 2014;13:1273–1285. doi: 10.1074/mcp.M113.036558. [DOI] [PMC free article] [PubMed] [Google Scholar]
  124. Katritch V, Cherezov V, Stevens RC. Structure-function of the G protein-coupled receptor superfamily. Annu Rev Pharmacol Toxicol. 2013;53:531–556. doi: 10.1146/annurev-pharmtox-032112-135923. [DOI] [PMC free article] [PubMed] [Google Scholar]
  125. Kehne JH, Baron BM, Carr AA, Chaney SF, Elands J, Feldman DJ, Frank RA, van Giersbergen PL, McCloskey TC, Johnson MP, McCarty DR, Poirot M, Senyah Y, Siegel BW, Widmaier C. Preclinical characterization of the potential of the putative atypical antipsychotic MDL 100,907 as a potent 5-HT2A antagonist with a favorable CNS safety profile. J Pharmacol Exp Ther. 1996;277:968–981. [PubMed] [Google Scholar]
  126. Keller DL, Umbreit WW. Permanent alteration of behavior in mice by chemical and psychological means. Science. 1956;124:723–724. doi: 10.1126/science.124.3225.723. [DOI] [PubMed] [Google Scholar]
  127. Kenakin T. Measurement of receptor signaling bias. Curr Protoc Pharmacol. 2016;74:2–15. doi: 10.1002/cpph.11. [DOI] [PubMed] [Google Scholar]
  128. Kennedy RT. Emerging trends in in vivo neurochemical monitoring by microdialysis. Curr Opin Chem Biol. 2013;17:860–867. doi: 10.1016/j.cbpa.2013.06.012. [DOI] [PMC free article] [PubMed] [Google Scholar]
  129. Kometer M, Vollenweider FX. Serotonergic hallucinogen-induced visual perceptual alterations. Curr Top Behav Neurosci. 2016 doi: 10.1007/7854_2016_461. [DOI] [PubMed]
  130. Kometer M, Schmidt A, Bachmann R, Studerus E, Seifritz E, Vollenweider FX. Psilocybin biases facial recognition, goal-directed behavior, and mood state toward positive relative to negative emotions through different serotonergic subreceptors. Biol Psychiatry. 2012;72:898–906. doi: 10.1016/j.biopsych.2012.04.005. [DOI] [PubMed] [Google Scholar]
  131. Kometer M, Schmidt A, Jancke L, Vollenweider FX. Activation of serotonin 2A receptors underlies the psilocybin-induced effects on alpha oscillations, N170 visual-evoked potentials, and visual hallucinations. J Neurosci. 2013;33:10544–10551. doi: 10.1523/JNEUROSCI.3007-12.2013. [DOI] [PMC free article] [PubMed] [Google Scholar]
  132. Kraehenmann R, Pokorny D, Vollenweider L, Preller KH, Pokorny T, Seifritz E, Vollenweider FX. Dreamlike effects of LSD on waking imagery in humans depend on serotonin 2A receptor activation. Psychopharmacology. 2017;234:2031–2046. doi: 10.1007/s00213-017-4610-0. [DOI] [PubMed] [Google Scholar]
  133. Kroeze WK, Hufeisen SJ, Popadak BA, Renock SM, Steinberg S, Ernsberger P, Jayathilake K, Meltzer HY, Roth BL. H1-histamine receptor affinity predicts short-term weight gain for typical and atypical antipsychotic drugs. Neuropsychopharmacology. 2003;28:519–526. doi: 10.1038/sj.npp.1300027. [DOI] [PubMed] [Google Scholar]
  134. Kurrasch-Orbaugh DM, Parrish JC, Watts VJ, Nichols DE. A complex signaling cascade links the serotonin2A receptor to phospholipase A2 activation: the involvement of MAP kinases. J Neurochem. 2003a;86:980–991. doi: 10.1046/j.1471-4159.2003.01921.x. [DOI] [PubMed] [Google Scholar]
  135. Kurrasch-Orbaugh DM, Watts VJ, Barker EL, Nichols DE. Serotonin 5-hydroxytryptamine 2A receptor-coupled phospholipase C and phospholipase A2 signaling pathways have different receptor reserves. J Pharmacol Exp Ther. 2003b;304:229–237. doi: 10.1124/jpet.102.042184. [DOI] [PubMed] [Google Scholar]
  136. Lee MY, Chiang CC, Chiu HY, Chan MH, Chen HH. N-acetylcysteine modulates hallucinogenic 5-HT(2A) receptor agonist-mediated responses: behavioral, molecular, and electro-physiological studies. Neuropharmacology. 2014;81:215–223. doi: 10.1016/j.neuropharm.2014.02.006. [DOI] [PubMed] [Google Scholar]
  137. Leysen JE, Eens A, Gommeren W, Van Gompel P, Wynants J, Janssen PA. Non-serotonergic [3H]ketanserin binding sites in striatal membranes are associated with a dopac release system on dopaminergic nerve endings. Eur J Pharmacol. 1987;134:373–375. doi: 10.1016/0014-2999(87)90373-6. [DOI] [PubMed] [Google Scholar]
  138. Leysen JE, Janssen PM, Schotte A, Luyten WH, Megens AA. Interaction of antipsychotic drugs with neurotransmitter receptor sites in vitro and in vivo in relation to pharmacological and clinical effects: role of 5HT2 receptors. Psychopharmacology. 1993;112:S40–S54. doi: 10.1007/BF02245006. [DOI] [PubMed] [Google Scholar]
  139. Li JX, Rice KC, France CP. Behavioral effects of dipropyltryptamine in rats: evidence for 5-HT1A and 5-HT2A agonist activity. Behav Pharmacol. 2007;18:283–288. doi: 10.1097/FBP.0b013e3281f19ca0. [DOI] [PubMed] [Google Scholar]
  140. Li JX, Rice KC, France CP. Discriminative stimulus effects of 1-(2,5-dimethoxy-4-methylphenyl)-2-aminopropane in rhesus monkeys: antagonism and apparent pA2 analyses. J Pharmacol Exp Ther. 2009a;328:976–981. doi: 10.1124/jpet.108.145458. [DOI] [PMC free article] [PubMed] [Google Scholar]
  141. Li JX, Unzeitig A, Javors MA, Rice KC, Koek W, France CP. Discriminative stimulus effects of 1-(2,5-dimethoxy-4-methylphenyl)-2-aminopropane (DOM), ketanserin, and (R)-(+)-{alpha}-(2,3-dimethoxyphenyl)-1-[2-(4-fluorophenyl)ethyl]-4-pipidinemetha nol (MDL100907) in rats. J Pharmacol Exp Ther. 2009b;331:671–679. doi: 10.1124/jpet.109.157560. [DOI] [PMC free article] [PubMed] [Google Scholar]
  142. Liechti ME. Modern clinical research on LSD. Neuropsychopharmacology. 2017;42:2114–2127. doi: 10.1038/npp.2017.86. [DOI] [PMC free article] [PubMed] [Google Scholar]
  143. Liu Y, Canal CE, Cordova-Sintjago TC, Zhu W, Booth RG. Mutagenesis analysis reveals distinct amino acids of the human serotonin 5-HT2C receptor underlying the pharmacology of distinct ligands. ACS Chem Neurosci. 2017;8:28–39. doi: 10.1021/acschemneuro.6b00124. [DOI] [PubMed] [Google Scholar]
  144. Lopez-Gimenez JF, Vilaro MT, Palacios JM, Mengod G. [3H]MDL 100,907 labels 5-HT2A serotonin receptors selectively in primate brain. Neuropharmacology. 1998;37:1147–1158. doi: 10.1016/s0028-3908(98)00102-6. [DOI] [PubMed] [Google Scholar]
  145. Lopez-Gimenez JF, Vilaro MT, Palacios JM, Mengod G. Multiple conformations of 5-HT2A and 5-HT 2C receptors in rat brain: an autoradiographic study with [125I](+/−)DOI. Exp Brain Res. 2013;230:395–406. doi: 10.1007/s00221-013-3636-8. [DOI] [PubMed] [Google Scholar]
  146. Luparini MR, Garrone B, Pazzagli M, Pinza M, Pepeu G. A cortical GABA-5HT interaction in the mechanism of action of the antidepressant trazodone. Prog Neuro-Psychopharmacol Biol Psychiatry. 2004;28:1117–1127. doi: 10.1016/j.pnpbp.2004.05.046. [DOI] [PubMed] [Google Scholar]
  147. Marona-Lewicka D, Thisted RA, Nichols DE. Distinct temporal phases in the behavioral pharmacology of LSD: dopamine D2 receptor-mediated effects in the rat and implications for psychosis. Psychopharmacology. 2005;180:427–435. doi: 10.1007/s00213-005-2183-9. [DOI] [PubMed] [Google Scholar]
  148. Martin DA, Nichols CD. The effects of hallucinogens on gene expression. Curr Top Behav Neurosci. 2017 doi: 10.1007/7854_2017_479. [DOI] [PubMed]
  149. Martin-Ruiz R, Puig MV, Celada P, Shapiro DA, Roth BL, Mengod G, Artigas F. Control of serotonergic function in medial prefrontal cortex by serotonin-2A receptors through a glutamate-dependent mechanism. J Neurosci. 2001;21:9856–9866. doi: 10.1523/JNEUROSCI.21-24-09856.2001. [DOI] [PMC free article] [PubMed] [Google Scholar]
  150. May JA, Sharif NA, Chen HH, Liao JC, Kelly CR, Glennon RA, Young R, Li JX, Rice KC, France CP. Pharmacological properties and discriminative stimulus effects of a novel and selective 5-HT2 receptor agonist AL-38022A [(S)-2-(8,9-dihydro-7H-pyrano[2,3-g]indazol-1-yl)-1-methylethylamine] Pharmacol Biochem Behav. 2009;91:307–314. doi: 10.1016/j.pbb.2008.07.015. [DOI] [PMC free article] [PubMed] [Google Scholar]
  151. Mayberg HS, Brannan SK, Mahurin RK, Jerabek PA, Brickman JS, Tekell JL, Silva JA, McGinnis S, Glass TG, Martin CC, Fox PT. Cingulate function in depression: a potential predictor of treatment response. Neuroreport. 1997;8:1057–1061. doi: 10.1097/00001756-199703030-00048. [DOI] [PubMed] [Google Scholar]
  152. McCreary AC, Filip M, Cunningham KA. Discriminative stimulus properties of (+/−)-fenfluramine: the role of 5-HT2 receptor subtypes. Behav Neurosci. 2003;117:212–221. doi: 10.1037/0735-7044.117.2.212. [DOI] [PubMed] [Google Scholar]
  153. McGrew L, Chang MS, Sanders-Bush E. Phospholipase D activation by endogenous 5-hydroxytryptamine 2C receptors is mediated by Galpha13 and pertussis toxin-insensitive Gbetagamma subunits. Mol Pharmacol. 2002;62:1339–1343. doi: 10.1124/mol.62.6.1339. [DOI] [PubMed] [Google Scholar]
  154. McKenna DJ, Saavedra JM. Autoradiography of LSD and 2,5-dimethoxyphenylisopropylamine psychotomimetics demonstrates regional, specific cross-displacement in the rat brain. Eur J Pharmacol. 1987;142:313–315. doi: 10.1016/0014-2999(87)90121-x. [DOI] [PubMed] [Google Scholar]
  155. McKenna DJ, Mathis CA, Shulgin AT, Sargent T3rd, Saavedra JM. Autoradiographic localization of binding sites for 125I-DOI, a new psychotomimetic radioligand, in the rat brain. Eur J Pharmacol. 1987;137:289–290. doi: 10.1016/0014-2999(87)90239-1. [DOI] [PubMed] [Google Scholar]
  156. McKinney M, Raddatz R. Practical aspects of radioligand binding. Curr Protoc Pharmacol. 2006;1:1.3. doi: 10.1002/0471141755.ph0103s33. [DOI] [PubMed] [Google Scholar]
  157. Milligan G, Kostenis E. Heterotrimeric G-proteins: a short history. Br J Pharmacol. 2006;147(Suppl 1):S46–S55. doi: 10.1038/sj.bjp.0706405. [DOI] [PMC free article] [PubMed] [Google Scholar]
  158. Montgomery T, Buon C, Eibauer S, Guiry PJ, Keenan AK, McBean GJ. Comparative potencies of 3,4-methylenedioxymethamphetamine (MDMA) analogues as inhibitors of [(3)H]noradrenaline and [(3)H]5-HT transport in mammalian cell lines. Br J Pharmacol. 2007;152:1121–1130. doi: 10.1038/sj.bjp.0707473. [DOI] [PMC free article] [PubMed] [Google Scholar]
  159. Moreau JJ. Hashish and mental illness. Raven Press; New York: 1973. [Google Scholar]
  160. Moreno JL, Holloway T, Albizu L, Sealfon SC, Gonzalez-Maeso J. Metabotropic glutamate mGlu2 receptor is necessary for the pharmacological and behavioral effects induced by hallucinogenic 5-HT2A receptor agonists. Neurosci Lett. 2011;493:76–79. doi: 10.1016/j.neulet.2011.01.046. [DOI] [PMC free article] [PubMed] [Google Scholar]
  161. Moutkine I, Quentin E, Guiard BP, Maroteaux L, Doly S. Heterodimers of serotonin receptor subtypes 2 are driven by 5-HT2C protomers. J Biol Chem. 2017;292:6352–6368. doi: 10.1074/jbc.M117.779041. [DOI] [PMC free article] [PubMed] [Google Scholar]
  162. Moya PR, Berg KA, Gutierrez-Hernandez MA, Saez-Briones P, Reyes-Parada M, Cassels BK, Clarke WP. Functional selectivity of hallucinogenic phenethylamine and phenylisopropylamine derivatives at human 5-hydroxytryptamine (5-HT)2A and 5-HT2C receptors. J Pharmacol Exp Ther. 2007;321:1054–1061. doi: 10.1124/jpet.106.117507. [DOI] [PubMed] [Google Scholar]
  163. Mueller F, Lenz C, Dolder PC, Harder S, Schmid Y, Lang UE, Liechti ME, Borgwardt S. Acute effects of LSD on amygdala activity during processing of fearful stimuli in healthy subjects. Transl Psychiatry. 2017;7:e1084. doi: 10.1038/tp.2017.54. [DOI] [PMC free article] [PubMed] [Google Scholar]
  164. Muschamp JW, Regina MJ, Hull EM, Winter JC, Rabin RA. Lysergic acid diethylamide and [-]-2,5-dimethoxy-4-methylamphetamine increase extracellular glutamate in rat prefrontal cortex. Brain Res. 2004;1023:134–140. doi: 10.1016/j.brainres.2004.07.044. [DOI] [PubMed] [Google Scholar]
  165. Nair SG, Gudelsky GA. Activation of 5-HT2 receptors enhances the release of acetylcholine in the prefrontal cortex and hippocampus of the rat. Synapse. 2004;53:202–207. doi: 10.1002/syn.20054. [DOI] [PubMed] [Google Scholar]
  166. Navailles S, De Deurwaerdere P, Spampinato U. Clozapine and haloperidol differentially alter the constitutive activity of central serotonin2C receptors in vivo. Biol Psychiatry. 2006;59:568–575. doi: 10.1016/j.biopsych.2005.07.035. [DOI] [PubMed] [Google Scholar]
  167. Nichols DE. Psychedelics. Pharmacol Rev. 2016;68:264–355. doi: 10.1124/pr.115.011478. [DOI] [PMC free article] [PubMed] [Google Scholar]
  168. Nichols DE. Chemistry and structure-activity relationships of psychedelics. Curr Top Behav Neurosci. 2017 doi: 10.1007/7854_2017_475. [DOI] [PubMed]
  169. Nichols DE, Frescas SP, Chemel BR, Rehder KS, Zhong D, Lewin AH. High specific activity tritium-labeled N-(2-methoxybenzyl)-2,5-dimethoxy-4-iodophenethylamine (INBMeO): a high-affinity 5-HT2A receptor-selective agonist radioligand. Bioorg Med Chem. 2008;16:6116–6123. doi: 10.1016/j.bmc.2008.04.050. [DOI] [PMC free article] [PubMed] [Google Scholar]
  170. Nichols DE, Sassano MF, Halberstadt AL, Klein LM, Brandt SD, Elliott SP, Fiedler WJ. N-Benzyl-5-methoxytryptamines as potent serotonin 5-HT2 receptor family agonists and comparison with a series of phenethylamine analogues. ACS Chem Neurosci. 2015;6:1165–1175. doi: 10.1021/cn500292d. [DOI] [PMC free article] [PubMed] [Google Scholar]
  171. Nichols DE, Johnson MW, Nichols CD. Psychedelics as medicines: an emerging new paradigm. Clin Pharmacol Ther. 2017;101:209–219. doi: 10.1002/cpt.557. [DOI] [PubMed] [Google Scholar]
  172. Pahnke WN, Richards WA. Implications of LSD and experimental mysticism. J Relig Health. 1966;5:175–208. doi: 10.1007/BF01532646. [DOI] [PubMed] [Google Scholar]
  173. Pahnke WN, Kurland AA, Unger S, Savage C, Grof S. The experimental use of psychedelic (LSD) psychotherapy. JAMA. 1970;212:1856–1863. [PubMed] [Google Scholar]
  174. Palfreyman MG, Schmidt CJ, Sorensen SM, Dudley MW, Kehne JH, Moser P, Gittos MW, Carr AA. Electrophysiological, biochemical and behavioral evidence for 5-HT2 and 5-HT3 mediated control of dopaminergic function. Psychopharmacology. 1993;112:S60–S67. doi: 10.1007/BF02245008. [DOI] [PubMed] [Google Scholar]
  175. Palhano-Fontes F, Andrade KC, Tofoli LF, Santos AC, Crippa JA, Hallak JE, Ribeiro S, de Araujo DB. The psychedelic state induced by ayahuasca modulates the activity and connectivity of the default mode network. PLoS One. 2015;10:e0118143. doi: 10.1371/journal.pone.0118143. [DOI] [PMC free article] [PubMed] [Google Scholar]
  176. Parrish JC, Nichols DE. Serotonin 5-HT(2A) receptor activation induces 2-arachidonoylglycerol release through a phospholipase c-dependent mechanism. J Neurochem. 2006;99:1164–1175. doi: 10.1111/j.1471-4159.2006.04173.x. [DOI] [PubMed] [Google Scholar]
  177. Parrish JC, Braden MR, Gundy E, Nichols DE. Differential phospholipase C activation by phenylalkylamine serotonin 5-HT 2A receptor agonists. J Neurochem. 2005;95:1575–1584. doi: 10.1111/j.1471-4159.2005.03477.x. [DOI] [PubMed] [Google Scholar]
  178. Pauwels PJ, Van Gompel P, Leysen JE. Activity of serotonin (5-HT) receptor agonists, partial agonists and antagonists at cloned human 5-HT1A receptors that are negatively coupled to adenylate cyclase in permanently transfected HeLa cells. Biochem Pharmacol. 1993;45:375–383. doi: 10.1016/0006-2952(93)90073-6. [DOI] [PubMed] [Google Scholar]
  179. Pazos A, Probst A, Palacios JM. Serotonin receptors in the human brain – IV. Autoradiographic mapping of serotonin-2 receptors. Neuroscience. 1987;21:123–139. doi: 10.1016/0306-4522(87)90327-7. [DOI] [PubMed] [Google Scholar]
  180. Pehek EA, Hernan AE. Stimulation of glutamate receptors in the ventral tegmental area is necessary for serotonin-2 receptor-induced increases in mesocortical dopamine release. Neuro-science. 2015;290:159–164. doi: 10.1016/j.neuroscience.2015.01.029. [DOI] [PMC free article] [PubMed] [Google Scholar]
  181. Pehek EA, McFarlane HG, Maguschak K, Price B, Pluto CP. M100,907, a selective 5-HT2A antagonist, attenuates dopamine release in the rat medial prefrontal cortex. Brain Res. 2001;888:51–59. doi: 10.1016/s0006-8993(00)03004-3. [DOI] [PubMed] [Google Scholar]
  182. Pehek EA, Nocjar C, Roth BL, Byrd TA, Mabrouk OS. Evidence for the preferential involvement of 5-HT2A serotonin receptors in stress- and drug-induced dopamine release in the rat medial prefrontal cortex. Neuropsychopharmacology. 2006;31:265–277. doi: 10.1038/sj.npp.1300819. [DOI] [PubMed] [Google Scholar]
  183. Peng Y, McCorvy JD, Harpsøe K, Lansu K, Yuan S, Popov P, Qu L, Pu M, Che T, Nikolajsen LF, Huang X-P, Wu Y, Shen L, Bjørn-Yoshimoto WE, Ding K, Wacker D, Han GW, Cheng J, Katritch V, Jensen AA, Hanson MA, Zhao S, Gloriam DE, Roth BL, Stevens RC, Liu Z-J. 5-HT2C receptor structures reveal the structural basis of GPCR polypharmacology. Cell. 2018;172(4):719–730. doi: 10.1016/j.cell.2018.01.001. e14. [DOI] [PMC free article] [PubMed] [Google Scholar]
  184. Perez-Aguilar JM, Shan J, LeVine MV, Khelashvili G, Weinstein H. A functional selectivity mechanism at the serotonin-2A GPCR involves ligand-dependent conformations of intracellular loop 2. J Am Chem Soc. 2014;136:16044–16054. doi: 10.1021/ja508394x. [DOI] [PMC free article] [PubMed] [Google Scholar]
  185. Petri G, Expert P, Turkheimer F, Carhart-Harris R, Nutt D, Hellyer PJ, Vaccarino F. Homological scaffolds of brain functional networks. J R Soc Interface. 2014;11:20140873. doi: 10.1098/rsif.2014.0873. [DOI] [PMC free article] [PubMed] [Google Scholar]
  186. Pokorny T, Preller KH, Kraehenmann R, Vollenweider FX. Modulatory effect of the 5-HT1A agonist buspirone and the mixed non-hallucinogenic 5-HT1A/2A agonist ergotamine on psilocybin-induced psychedelic experience. Eur Neuropsychopharmacol. 2016;26:756–766. doi: 10.1016/j.euroneuro.2016.01.005. [DOI] [PubMed] [Google Scholar]
  187. Preller KH, Vollenweider FX. Phenomenology, structure, and dynamic of psychedelic states. Curr Top Behav Neurosci. 2016 doi: 10.1007/7854_2016_459. [DOI] [PubMed]
  188. Preller KH, Herdener M, Pokorny T, Planzer A, Kraehenmann R, Stampfli P, Liechti ME, Seifritz E, Vollenweider FX. The fabric of meaning and subjective effects in LSD-induced states depend on serotonin 2A receptor activation. Curr Biol. 2017;27:451–457. doi: 10.1016/j.cub.2016.12.030. [DOI] [PubMed] [Google Scholar]
  189. Puig MV, Watakabe A, Ushimaru M, Yamamori T, Kawaguchi Y. Serotonin modulates fast-spiking interneuron and synchronous activity in the rat prefrontal cortex through 5-HT1A and 5-HT2A receptors. J Neurosci. 2010;30:2211–2222. doi: 10.1523/JNEUROSCI.3335-09.2010. [DOI] [PMC free article] [PubMed] [Google Scholar]
  190. Quednow BB, Geyer MA, Halberstadt AL. Serotonin and schizophrenia. In: Muller CP, Jacobs B, editors. Handbook of the behavioral neurobiology of serotonin. Academic Press; London: 2010. pp. 585–620. [Google Scholar]
  191. Quednow BB, Kometer M, Geyer MA, Vollenweider FX. Psilocybin-induced deficits in automatic and controlled inhibition are attenuated by ketanserin in healthy human volunteers. Neuropsychopharmacology. 2012;37:630–640. doi: 10.1038/npp.2011.228. [DOI] [PMC free article] [PubMed] [Google Scholar]
  192. Rabin RA, Regina M, Doat M, Winter JC. 5-HT2A receptor-stimulated phosphoinositide hydrolysis in the stimulus effects of hallucinogens. Pharmacol Biochem Behav. 2002;72:29–37. doi: 10.1016/s0091-3057(01)00720-1. [DOI] [PubMed] [Google Scholar]
  193. Ranganathan A, Rodríguez D, Carlsson J. Structure-based discovery of GPCR ligands from crystal structures and homology models. Springer; Berlin, Heidelberg: 2017. pp. 1–35. [Google Scholar]
  194. Raote I, Bhattacharyya S, Panicker MM. Functional selectivity in serotonin receptor 2A (5-HT2A) endocytosis, recycling, and phosphorylation. Mol Pharmacol. 2013;83:42–50. doi: 10.1124/mol.112.078626. [DOI] [PubMed] [Google Scholar]
  195. Rauser L, Savage JE, Meltzer HY, Roth BL. Inverse agonist actions of typical and atypical antipsychotic drugs at the human 5-hydroxytryptamine(2C) receptor. J Pharmacol Exp Ther. 2001;299:83–89. [PubMed] [Google Scholar]
  196. Reissig CJ, Eckler JR, Rabin RA, Winter JC. The 5-HT1A receptor and the stimulus effects of LSD in the rat. Psychopharmacology. 2005;182:197–204. doi: 10.1007/s00213-005-0068-6. [DOI] [PMC free article] [PubMed] [Google Scholar]
  197. Richards N, Chapman LF, Goodell H, Wolff HG. LSD-like delirium following ingestion of a small amount of its brom analog (BOL-148) Ann Intern Med. 1958;48:1078–1082. doi: 10.7326/0003-4819-48-5-1078. [DOI] [PubMed] [Google Scholar]
  198. Richelson E, Souder T. Binding of antipsychotic drugs to human brain receptors focus on newer generation compounds. Life Sci. 2000;68:29–39. doi: 10.1016/s0024-3205(00)00911-5. [DOI] [PubMed] [Google Scholar]
  199. Rickli A, Kopf S, Hoener MC, Liechti ME. Pharmacological profile of novel psychoactive benzofurans. Br J Pharmacol. 2015a;172:3412–3425. doi: 10.1111/bph.13128. [DOI] [PMC free article] [PubMed] [Google Scholar]
  200. Rickli A, Luethi D, Reinisch J, Buchy D, Hoener MC, Liechti ME. Receptor interaction profiles of novel N-2-methoxybenzyl (NBOMe) derivatives of 2,5-dimethoxy-substituted phenethylamines (2C drugs) Neuropharmacology. 2015b;99:546–553. doi: 10.1016/j.neuropharm.2015.08.034. [DOI] [PubMed] [Google Scholar]
  201. Rickli A, Moning OD, Hoener MC, Liechti ME. Receptor interaction profiles of novel psychoactive tryptamines compared with classic hallucinogens. Eur Neuropsychopharmacol. 2016;26:1327–1337. doi: 10.1016/j.euroneuro.2016.05.001. [DOI] [PubMed] [Google Scholar]
  202. Riva-Posse P, Choi KS, Holtzheimer PE, Crowell AL, Garlow SJ, Rajendra JK, McIntyre CC, Gross RE, Mayberg HS. A connectomic approach for subcallosal cingulate deep brain stimulation surgery: prospective targeting in treatment-resistant depression. Mol Psychiatry. 2017 doi: 10.1038/mp.2017.59. [DOI] [PMC free article] [PubMed]
  203. Ross S, Bossis A, Guss J, Agin-Liebes G, Malone T, Cohen B, Mennenga SE, Belser A, Kalliontzi K, Babb J, Su Z, Corby P, Schmidt BL. Rapid and sustained symptom reduction following psilocybin treatment for anxiety and depression in patients with life-threatening cancer: a randomized controlled trial. J Psychopharmacol. 2016;30:1165–1180. doi: 10.1177/0269881116675512. [DOI] [PMC free article] [PubMed] [Google Scholar]
  204. Roth BL, Choudhary MS, Khan N, Uluer AZ. High-affinity agonist binding is not sufficient for agonist efficacy at 5-hydroxytryptamine2A receptors: evidence in favor of a modified ternary complex model. J Pharmacol Exp Ther. 1997;280:576–583. [PubMed] [Google Scholar]
  205. Roth BL, Lopez E, Patel S, Kroeze WK. The multiplicity of serotonin receptors: uselessly diverse molecules or an embarrassment of riches? Neuroscientist. 2000;6:252–262. [Google Scholar]
  206. Saleh N, Ibrahim P, Saladino G, Gervasio FL, Clark T. An efficient metadynamics-based protocol to model the binding affinity and the transition state ensemble of G-protein-coupled receptor ligands. J Chem Inf Model. 2017;57:1210–1217. doi: 10.1021/acs.jcim.6b00772. [DOI] [PubMed] [Google Scholar]
  207. Sanders-Bush E, Burris KD, Knoth K. Lysergic acid diethylamide and 2,5-dimethoxy-4-methylamphetamine are partial agonists at serotonin receptors linked to phosphoinositide hydrolysis. J Pharmacol Exp Ther. 1988;246:924–928. [PubMed] [Google Scholar]
  208. Santana N, Artigas F. Expression of serotonin2c receptors in pyramidal and GABAergic neurons of rat prefrontal cortex: a comparison with striatum. Cereb Cortex. 2017;27:3125–3139. doi: 10.1093/cercor/bhw148. [DOI] [PubMed] [Google Scholar]
  209. Santana N, Mengod G, Artigas F. Expression of alpha(1)-adrenergic receptors in rat prefrontal cortex: cellular co-localization with 5-HT(2A) receptors. Int J Neuropsychopharmacol. 2013;16:1139–1151. doi: 10.1017/S1461145712001083. [DOI] [PubMed] [Google Scholar]
  210. Schmid CL, Bohn LM. Serotonin, but not N-methyltryptamines, activates the serotonin 2A receptor via a ss-arrestin2/Src/Akt signaling complex in vivo. J Neurosci. 2010;30:13513–13524. doi: 10.1523/JNEUROSCI.1665-10.2010. [DOI] [PMC free article] [PubMed] [Google Scholar]
  211. Schmid CL, Raehal KM, Bohn LM. Agonist-directed signaling of the serotonin 2A receptor depends on beta-arrestin-2 interactions in vivo. Proc Natl Acad Sci U S A. 2008;105:1079–1084. doi: 10.1073/pnas.0708862105. [DOI] [PMC free article] [PubMed] [Google Scholar]
  212. Schmid CL, Streicher JM, Meltzer HY, Bohn LM. Clozapine acts as an agonist at serotonin 2A receptors to counter MK-801-induced behaviors through a betaarrestin2-independent activation of Akt. Neuropsychopharmacology. 2014;39:1902–1913. doi: 10.1038/npp.2014.38. [DOI] [PMC free article] [PubMed] [Google Scholar]
  213. Schreiber R, Brocco M, Millan MJ. Blockade of the discriminative stimulus effects of DOI by MDL 100,907 and the ‘atypical’ antipsychotics, clozapine and risperidone. Eur J Pharmacol. 1994;264:99–102. doi: 10.1016/0014-2999(94)90643-2. [DOI] [PubMed] [Google Scholar]
  214. Schreiber R, Brocco M, Audinot V, Gobert A, Veiga S, Millan MJ. (1-(2,5-dimethoxy-4 iodophenyl)-2-aminopropane)-induced head-twitches in the rat are mediated by 5-hydroxytryptamine (5-HT) 2A receptors: modulation by novel 5-HT2A/2C antagonists, D1 antagonists and 5-HT1A agonists. J Pharmacol Exp Ther. 1995;273:101–112. [PubMed] [Google Scholar]
  215. Scruggs JL, Schmidt D, Deutch AY. The hallucinogen 1-[2,5-dimethoxy-4-iodophenyl]-2-aminopropane (DOI) increases cortical extracellular glutamate levels in rats. Neurosci Lett. 2003;346:137–140. doi: 10.1016/s0304-3940(03)00547-0. [DOI] [PubMed] [Google Scholar]
  216. Shulgin A, Shulgin A. Pihkal: a chemical love story. Transform Press; Berkeley: 1991. [Google Scholar]
  217. Shulgin A, Shulgin A. Tihkal: the continuation. Transform Press; Berkeley: 1997. [Google Scholar]
  218. Simmler LD, Buchy D, Chaboz S, Hoener MC, Liechti ME. In vitro characterization of psychoactive substances at rat, mouse, and human trace amine-associated receptor 1. J Pharmacol Exp Ther. 2016;357:134–144. doi: 10.1124/jpet.115.229765. [DOI] [PubMed] [Google Scholar]
  219. Snyder SH, Faillace L, Hollister L. 2,5-Dimethoxy-4-methyl-amphetamine (STP): a new hallucinogenic drug. Science. 1967;158:669–670. doi: 10.1126/science.158.3801.669. [DOI] [PubMed] [Google Scholar]
  220. Spindle MS, Thomas MP. Activation of 5-HT2A receptors by TCB-2 induces recurrent oscillatory burst discharge in layer 5 pyramidal neurons of the mPFC in vitro. Physiol Rep. 2014;2:e12003. doi: 10.14814/phy2.12003. [DOI] [PMC free article] [PubMed] [Google Scholar]
  221. Strassman R. DMT: the spirit molecule: a doctor’s revolutionary research into the biology of near-death and mystical experiences. Park Street Press; Rochester: 2001. [Google Scholar]
  222. Studerus E, Gamma A, Kometer M, Vollenweider FX. Prediction of psilocybin response in healthy volunteers. PLoS One. 2012;7:e30800. doi: 10.1371/journal.pone.0030800. [DOI] [PMC free article] [PubMed] [Google Scholar]
  223. Sykes DA, Dowling MR, Charlton SJ. Measuring receptor target coverage: a radioligand competition binding protocol for assessing the association and dissociation rates of unlabeled compounds. Curr Protoc Pharmacol. 2010;9:14. doi: 10.1002/0471141755.ph0914s50. [DOI] [PubMed] [Google Scholar]
  224. Tagliazucchi E, Carhart-Harris R, Leech R, Nutt D, Chialvo DR. Enhanced repertoire of brain dynamical states during the psychedelic experience. Hum Brain Mapp. 2014;35:5442–5456. doi: 10.1002/hbm.22562. [DOI] [PMC free article] [PubMed] [Google Scholar]
  225. Tagliazucchi E, Roseman L, Kaelen M, Orban C, Muthukumaraswamy SD, Murphy K, Laufs H, Leech R, McGonigle J, Crossley N, Bullmore E, Williams T, Bolstridge M, Feilding A, Nutt DJ, Carhart-Harris R. Increased global functional connectivity correlates with LSD-induced ego dissolution. Curr Biol. 2016;26:1043–1050. doi: 10.1016/j.cub.2016.02.010. [DOI] [PubMed] [Google Scholar]
  226. Thal DM, Sun B, Feng D, Nawaratne V, Leach K, Felder CC, Bures MG, Evans DA, Weis WI, Bachhawat P, Kobilka TS, Sexton PM, Kobilka BK, Christopoulos A. Crystal structures of the M1 and M4 muscarinic acetylcholine receptors. Nature. 2016;531:335–340. doi: 10.1038/nature17188. [DOI] [PMC free article] [PubMed] [Google Scholar]
  227. Thomsen WJ, Grottick AJ, Menzaghi F, Reyes-Saldana H, Espitia S, Yuskin D, Whelan K, Martin M, Morgan M, Chen W, Al-Shamma H, Smith B, Chalmers D, Behan D. Lorcaserin, a novel selective human 5-hydroxytryptamine2C agonist: in vitro and in vivo pharmacological characterization. J Pharmacol Exp Ther. 2008;325:577–587. doi: 10.1124/jpet.107.133348. [DOI] [PubMed] [Google Scholar]
  228. Tian MK, Schmidt EF, Lambe EK. Serotonergic suppression of mouse prefrontal circuits implicated in task attention. eNeuro. 2016;3 doi: 10.1523/ENEURO.0269-16.2016. [DOI] [PMC free article] [PubMed] [Google Scholar]
  229. Valle M, Maqueda AE, Rabella M, Rodriguez-Pujadas A, Antonijoan RM, Romero S, Alonso JF, Mananas MA, Barker S, Friedlander P, Feilding A, Riba J. Inhibition of alpha oscillations through serotonin-2A receptor activation underlies the visual effects of ayahuasca in humans. Eur Neuropsychopharmacol. 2016;26:1161–1175. doi: 10.1016/j.euroneuro.2016.03.012. [DOI] [PubMed] [Google Scholar]
  230. Verhoeff NP, Visser WH, Ferrari MD, Saxena PR, van Royen EA. Dopamine D2-receptor imaging with 123I-iodobenzamide SPECT in migraine patients abusing ergotamine: does ergotamine cross the blood brain barrier? Cephalalgia. 1993;13:325–329. doi: 10.1046/j.1468-2982.1993.1305325.x. [DOI] [PubMed] [Google Scholar]
  231. Viol A, Palhano-Fontes F, Onias H, de Araujo DB, Viswanathan GM. Shannon entropy of brain functional complex networks under the influence of the psychedelic Ayahuasca. Sci Rep. 2017;7:7388. doi: 10.1038/s41598-017-06854-0. [DOI] [PMC free article] [PubMed] [Google Scholar]
  232. Vollenweider FX. Brain mechanisms of hallucinogens and entactogens. Dialogues Clin Neurosci. 2001;3:265–279. doi: 10.31887/DCNS.2001.3.4/fxvollenweider. [DOI] [PMC free article] [PubMed] [Google Scholar]
  233. Vollenweider FX, Vollenweider-Scherpenhuyzen MF, Babler A, Vogel H, Hell D. Psilocybin induces schizophrenia-like psychosis in humans via a serotonin-2 agonist action. Neuroreport. 1998;9:3897–3902. doi: 10.1097/00001756-199812010-00024. [DOI] [PubMed] [Google Scholar]
  234. Wacker D, Fenalti G, Brown MA, Katritch V, Abagyan R, Cherezov V, Stevens RC. Conserved binding mode of human β2 adrenergic receptor inverse agonists and antagonist revealed by X-ray crystallography. J Am Chem Soc. 2010;132:11443–11445. doi: 10.1021/ja105108q. [DOI] [PMC free article] [PubMed] [Google Scholar]
  235. Wacker D, Wang C, Katritch V, Han GW, Huang XP, Vardy E, McCorvy JD, Jiang Y, Chu M, Siu FY, Liu W, Xu HE, Cherezov V, Roth BL, Stevens RC. Structural features for functional selectivity at serotonin receptors. Science. 2013;340:615–619. doi: 10.1126/science.1232808. [DOI] [PMC free article] [PubMed] [Google Scholar]
  236. Wacker D, Wang S, McCorvy JD, Betz RM, Venkatakrishnan AJ, Levit A, Lansu K, Schools ZL, Che T, Nichols DE, Shoichet BK, Dror RO, Roth BL. Crystal structure of an LSD-bound human serotonin receptor. Cell. 2017;168:377–389. doi: 10.1016/j.cell.2016.12.033. e12. [DOI] [PMC free article] [PubMed] [Google Scholar]
  237. Walker SJ, Brown HA Molecular Cellular Biology CUINYUSA. Measurement of G protein-coupled receptor-stimulated phospholipase D activity in intact cells. Methods Mol Biol. 2004;237:89–97. doi: 10.1385/1-59259-430-1:89. [DOI] [PubMed] [Google Scholar]
  238. Wang C, Jiang Y, Ma J, Wu H, Wacker D, Katritch V, Han GW, Liu W, Huang XP, Vardy E, McCorvy JD, Gao X, Zhou XE, Melcher K, Zhang C, Bai F, Yang H, Yang L, Jiang H, Roth BL, Cherezov V, Stevens RC, Xu HE. Structural basis for molecular recognition at serotonin receptors. Science. 2013;340:610–614. doi: 10.1126/science.1232807. [DOI] [PMC free article] [PubMed] [Google Scholar]
  239. Wang S, Wacker D, Levit A, Che T, Betz RM, McCorvy JD, Venkatakrishnan AJ, Huang XP, Dror RO, Shoichet BK, Roth BL. D4 dopamine receptor high-resolution structures enable the discovery of selective agonists. Science. 2017;358:381–386. doi: 10.1126/science.aan5468. [DOI] [PMC free article] [PubMed] [Google Scholar]
  240. Weber ET, Andrade R. Htr2a gene and 5-HT(2A) receptor expression in the cerebral cortex studied using genetically modified mice. Front Neurosci. 2010;4:36. doi: 10.3389/fnins.2010.00036. [DOI] [PMC free article] [PubMed] [Google Scholar]
  241. Wenthur CJ, Lindsley CW. Classics in chemical neuroscience: clozapine. ACS Chem Neurosci. 2013;4:1018–1025. doi: 10.1021/cn400121z. [DOI] [PMC free article] [PubMed] [Google Scholar]
  242. Willins DL, Meltzer HY. Direct injection of 5-HT2A receptor agonists into the medial prefrontal cortex produces a head-twitch response in rats. J Pharmacol Exp Ther. 1997;282:699–706. [PubMed] [Google Scholar]
  243. Willins DL, Deutch AY, Roth BL. Serotonin 5-HT2A receptors are expressed on pyramidal cells and interneurons in the rat cortex. Synapse. 1997;27:79–82. doi: 10.1002/(SICI)1098-2396(199709)27:1<79::AID-SYN8>3.0.CO;2-A. [DOI] [PubMed] [Google Scholar]
  244. Winter JC. Hallucinogens as discriminative stimuli in animals: LSD, phenethylamines, and tryptamines. Psychopharmacology. 2009;203:251–263. doi: 10.1007/s00213-008-1356-8. [DOI] [PubMed] [Google Scholar]
  245. Winter CA, Flataker L. Effects of lysergic acid diethylamide upon performance of trained rats. Proc Soc Exp Biol Med. 1956;92:285–289. doi: 10.3181/00379727-92-22453. [DOI] [PubMed] [Google Scholar]
  246. Winter JC, Rice KC, Amorosi DJ, Rabin RA. Psilocybin-induced stimulus control in the rat. Pharmacol Biochem Behav. 2007;87:472–480. doi: 10.1016/j.pbb.2007.06.003. [DOI] [PMC free article] [PubMed] [Google Scholar]
  247. Wright IK, Garratt JC, Marsden CA. Effects of a selective 5-HT2 agonist, DOI, on 5-HT neuronal firing in the dorsal raphe nucleus and 5-HT release and metabolism in the frontal cortex. Br J Pharmacol. 1990;99:221–222. doi: 10.1111/j.1476-5381.1990.tb14683.x. [DOI] [PMC free article] [PubMed] [Google Scholar]
  248. Xia Z, Gray JA, Compton-Toth BA, Roth BL. A direct interaction of PSD-95 with 5-HT2A serotonin receptors regulates receptor trafficking and signal transduction. J Biol Chem. 2003;278:21901–21908. doi: 10.1074/jbc.M301905200. [DOI] [PubMed] [Google Scholar]
  249. Yang KC, Stepanov V, Martinsson S, Ettrup A, Takano A, Knudsen GM, Halldin C, Farde L, Finnema SJ. Fenfluramine reduces [11C]Cimbi-36 binding to the 5-HT2A receptor in the nonhuman primate brain. Int J Neuropsychopharmacol. 2017;20:683–691. doi: 10.1093/ijnp/pyx051. [DOI] [PMC free article] [PubMed] [Google Scholar]
  250. Zhang ZW, Arsenault D. Gain modulation by serotonin in pyramidal neurones of the rat prefrontal cortex. J Physiol. 2005;566:379–394. doi: 10.1113/jphysiol.2005.086066. [DOI] [PMC free article] [PubMed] [Google Scholar]
  251. Zhelyazkova-Savova M, Giovannini MG, Pepeu G. Increase of cortical acetylcholine release after systemic administration of chlorophenylpiperazine in the rat: an in vivo microdialysis study. Neurosci Lett. 1997;236:151–154. doi: 10.1016/s0304-3940(97)00785-4. [DOI] [PubMed] [Google Scholar]
  252. Zhelyazkova-Savova M, Giovannini MG, Pepeu G. Systemic chlorophenylpiperazine increases acetylcholine release from rat hippocampus-implication of 5-HT2C receptors. Pharmacol Res. 1999;40:165–170. doi: 10.1006/phrs.1999.0473. [DOI] [PubMed] [Google Scholar]

RESOURCES