Next Article in Journal
Integrated Analysis of Metabolome and Transcriptome Revealed Different Regulatory Networks of Metabolic Flux in Tea Plants [Camellia sinensis (L.) O. Kuntze] with Varied Leaf Colors
Previous Article in Journal
Molecular Dynamics Investigations of Human DNA-Topoisomerase I Interacting with Novel Dewar Valence Photo-Adducts: Insights into Inhibitory Activity
Previous Article in Special Issue
The Novel Atypical Antipsychotic Lurasidone Affects Cytochrome P450 Expression in the Liver and Peripheral Blood Lymphocytes
 
 
Font Type:
Arial Georgia Verdana
Font Size:
Aa Aa Aa
Line Spacing:
Column Width:
Background:
Review

Molecular and Medical Aspects of Psychedelics

by
Adam Wojtas
and
Krystyna Gołembiowska
*
Unit II, Department of Pharmacology, Maj Institute of Pharmacology Polish Academy of Sciences, 12 Smętna Street, 31-343 Kraków, Poland
*
Author to whom correspondence should be addressed.
Int. J. Mol. Sci. 2024, 25(1), 241; https://doi.org/10.3390/ijms25010241
Submission received: 17 November 2023 / Revised: 17 December 2023 / Accepted: 20 December 2023 / Published: 23 December 2023
(This article belongs to the Special Issue Molecular Advances in Psychiatric Therapies)

Abstract

:
Psychedelics belong to the oldest psychoactive drugs. They arouse recent interest due to their therapeutic applications in the treatment of major depressive disorder, substance use disorder, end-of-life anxiety,= and anxiety symptoms, and obsessive–compulsive disorder. In this review, the current state of preclinical research on the mechanism of action, neurotoxicity, and behavioral impact of psychedelics is summarized. The effect of selective 5-HT2A receptor agonists, 25I- and 25B-NBOMe, after acute and repeated administration is characterized and compared with the effects of a less selective drug, psilocybin. The data show a significant effect of NBOMes on glutamatergic, dopaminergic, serotonergic, and cholinergic neurotransmission in the frontal cortex, striatum, and nucleus accumbens. The increases in extracellular levels of neurotransmitters were not dose-dependent, which most likely resulted from the stimulation of the 5-HT2A receptor and subsequent activation of the 5-HT2C receptors. This effect was also observed in the wet dog shake test and locomotor activity. Chronic administration of NBOMes elicited rapid development of tolerance, genotoxicity, and activation of microglia. Acute treatment with psilocybin affected monoaminergic and aminoacidic neurotransmitters in the frontal cortex, nucleus accumbens, and hippocampus but not in the amygdala. Psilocybin exhibited anxiolytic properties resulting from intensification of GABAergic neurotransmission. The data indicate that NBOMes as selective 5-HT2A agonists exert a significant effect on neurotransmission and behavior of rats while also inducing oxidative DNA damage. In contrast to NBOMes, the effects induced by psilocybin suggest a broader therapeutic index of this drug.

1. Introduction

Serotonergic hallucinogens might be the oldest psychoactive substances used by humanity. These substances, known for inducing profound changes in perception, mood, and cognitive processes, are often associated with therapeutic, religious, and recreational applications [1]. It is due to those properties that Humphrey Osmond coined the widely-used name “psychedelic”, originating from the Greek words psukhḗ, “mind”, and delos, “to reveal”. The modern era of psychedelic research began in the 19th century with the discovery of mescaline, the active ingredient of hallucinogenic Peyote cacti [2]. The next chapter opened in 1938 with synthesis and 5 years later the discovery of the psychoactive properties of lysergic acid diethylamide (LSD) [3]. Due to its extreme potency, LSD quickly became the most intensely researched psychedelic compound, with more than 1000 published articles by the end of the 1960s [4]. It was studied as a possible aid in psychotherapy, or otherwise explored as a potential treatment for substance abuse disorders, anxiety, and mood disorders [5]. Unfortunately, alongside medical use, recreational use of these substances swept across the globe, resulting in the passage of the “Controlled Substances Act “ in 1970 where psychedelics were classified as “drugs with no currently accepted medical use and a high potential for abuse”. These circumstances made it difficult to continue research concerning psychedelic drugs and nearly all studies (with only a few exceptions) came to an abrupt end, followed by several decades of hiatus in psychedelic research. However, in recent years, clinical trials have been conducted and several psychedelic drugs are indications for the treatment of major depressive disorder and treatment-resistant depression, substance use disorder, end-of-life anxiety, cancer-related depression and/or anxiety symptoms, and obsessive–compulsive disorder [6].
In this work, we review the current state of preclinical knowledge about the mechanism of action, receptor targets, and toxicity of psychedelics at the level of neurotransmission and behavioral response. Specifically, the NBOMe class and psilocybin are chosen in this review as these substances show different pharmacological properties and distinct medical potential.

2. Classification of Psychedelics

All psychedelic compounds can be divided by their structure into two main categories: phenylalkylamines, e.g., mescaline (3,4,5-trimethoxyphenethylamine) or DOI (2,5-dimethoxy-4-iodoamphetamine) [7] and indoleamines, e.g., DMT (N,N-dimethyltryptamine) or LSD. Both of them bear a resemblance to endogenous compounds—either phenethylamine or serotonin. While the former bind mainly to the 5-HT2 receptor family [8,9], the latter demonstrate an affinity for several types of receptors and nearly all 5-HT receptors [10].
The only hallucinogenic phenethylamine that exists in nature is mescaline, the active compound of Peyote and Echinopsis cacti [11], while the biggest group being the “2C” series act primarily as stimulants, though in higher doses they exert hallucinogenic effects [12]. Hallucinogenic amphetamines cannot be found in nature and have to be obtained through synthesis. Their best-known representatives are the 4-substituted-2,5-dimethoxyamphetamines, with the most important being 2,5-dimethoxy-4-iodoamphetamine (DOI), 2,5-dimethoxy-4-methylamphetamine (DOM), and 2,5-dimethoxy-4-bromoamphetamine (DOB) and (DOC). Due to their high affinity (nanomolar or even subnanomolar) for the 5-HT2A receptor, they have been used as radioligands to map the distribution of the aforementioned receptor in the brain [13]. A new class of serotonergic hallucinogens, N-(2-methoxybenzyl)-2,5-dimethoxy-4-substituted phenethylamines, in short NBOMes, became very popular as recreational drugs [4]. Many compounds belonging to this group have been synthesized, differing in substituents on the phenyl ring at position 4. The addition of the N-2-methoxybenzyl group significantly increased the affinity for the 5-HT2A serotonin receptor subtype (Table 1). Quite like phenethylamines, indoleamines can also be further differentiated into two subclasses: simple tryptamines like DMT or psilocin and ergolines (lysergamides), which are tryptamine derivatives with more rigid conformation; they primarily consist of LSD and its derivatives. Tryptamines are quite often encountered in nature. DMT can be found in Ayahuasca, a brew made from the leaves of Psychotria viridis; bufotenine (5-OH-DMT) is secreted by glands of Bufo alvarius, an American species of toad; and the most common forms include psilocybin and its primary active metabolite psilocin, the compounds originating from Psilocybe fungi, which can be found all around the globe [1,14]. Tryptamines show affinity for a large number of 5-HT receptors. This includes 5-HT1A with the affinity for this receptor being sometimes almost as high as for 5-HT1B, 5-HT1D, 5-HT1E, 5-HT2A, 5-HT2B, 5-HT2C, 5-HT5A, 5-HT6, and 5-HT7 (Table 1) [10]. In high concentrations, they also bind to α-adrenergic receptors, dopaminergic receptors, and serotonin transporters (SERT) [10]. Nevertheless, these compounds induce their hallucinogenic effects via activation of the 5-HT2A receptor. Finally, ergolines are derivatives of alkaloids secreted by the ergot fungi. Due to their tetracyclic structure, they happen to be more rigid than the usual “simple” tryptamines [15]. Their history is inseparably bound with their most famous representative—LSD. It is, up to this day, one of the most potent psychedelics [11,16]. Similarly to tryptamines, ergolines exhibit high affinity for 5-HT1A, 5-HT1B, 5-HT1D, 5-HT1E, 5-HT2A, 5-HT2B, 5-HT2C, 5-HT5A, 5-HT5B, 5-HT6, and 5-HT7 receptors (Table 1) [17]. What is more unique to them is that they also bind to the dopaminergic D1 and D2 receptors and adrenergic α1 and α2 receptors [17,18,19].

3. The Mechanism of Action of Psychedelics

3.1. 5-HT2A Receptor as a Primary Target for Psychedelics

Hallucinogens exert their psychoactive effects by acting as agonists for the cortical 5-HT2A receptor [13,22,23,24]. The 5-HT2A receptor belongs to the G-protein-coupled receptor (GPCR) family. It is coupled with the Gq/11 protein, and its activation leads to phosphoinositide hydrolysis resulting in the formation of diacylglycerol and inositol triphosphate, which leads to the mobilization of intracellular calcium and subsequent membrane depolarization [4]. Furthermore, the intensity of the psychedelic experience in humans is correlated with the occupancy of the 5-HT2A receptor, mainly in the prefrontal cortex (PFC) [25]. This activation of 5-HT2A receptors in the PFC launches a downstream cascade of changes in connectivity and alterations in blood flow across multiple regions of the brain, e.g., cingulate cortex, inferior parietal lobule, lateral temporal cortex, hippocampus (HP), thalamus, amygdala, and claustrum [26,27,28] involved in cognition, emotional processing, sensory perception, or even self-recognition and theory of mind processes [4,29]. What is interesting is that even though psychedelics activate 5-HT2A receptors on glutamatergic pyramidal neurons in the brain, this typically does not induce depolarization or generation of action potentials. Instead, there is an increase in glutamate release from depolarized neurons, resulting in recurrent activity [30]. Notably, Beique et al. [31] identified a subset of large neurons in the deep cortical layers that exhibited significant sensitivity to 5-HT. These neurons exhibited substantial membrane depolarizations, leading to spiking activity. Based on these findings, Martin and Nichols [32] isolated a specific subset of psychedelic-activated neurons from rat brains. They revealed that psychedelics directly stimulated only a minor proportion of 5-HT2A receptor-expressing excitatory neurons, especially in crucial brain areas, such as the PFC and claustrum. In particular, the psychedelic-responsive neurons exhibit elevated gene expression for the 5-HT2A receptor, which likely underlies their heightened sensitivity to psychedelics compared with other neurons. The authors postulate that this distinct neuron group acts as a “trigger population”. The activation of these neurons subsequently recruits other cell types including subpopulations of somatostatin or parvalbumin inhibitory GABAergic interneurons or astrocytes [32].

3.2. The Impact of 5-HT2A Receptor Activation on Behavior

The 5-HT2A receptor is a key player in inducing the psychedelic experience in humans and its activation is considered as a proxy for hallucinogenic effect in animal models [4]. The head twitch response/wet dog shake (HTR/WDS) test is based on this mechanism, exhibiting significant construct validity. The assay quantifies rapid, rhythmic head movements observed in rodents post-administration of psychedelic 5-HT2A receptor agonists [33]. While some false positives have been identified, such as fenfluramine, p-chloroamphetamine, and 5-hydroxytryptophan, the test predominantly exhibits specificity for 5-HT2A receptor agonists [15]. Furthermore, the HTR assay seems to be highly sensitive to 5-HT2A receptor agonists known to produce psychedelic effects in humans, proving its predictive validity. This is evidenced by the fact that the non-psychedelic 5-HT2A receptor agonist, namely lisuride, does not evoke the head twitch phenomenon [33]. Unfortunately, the face validity of the assay is poor, as humans do not exhibit head-twitching behavior after administration of psychoactive drugs.

4. NBOMes

N-2-methoxybenzyl substitution of 2-C compounds [11] drastically enhanced the binding to the 5-HT2A receptor, which resulted in the synthesis of a number of NBOMe agents differing in substituents on the phenyl ring at position 4. The examples of this group (shown in Table 2) are called in short 25I-NBOMe, 25B-NBOMe, and 25C-NBOMe and have in their molecule a substituent of halogens: iodine, bromine, or chlorine [34].

4.1. The Effects of NBOMes on Behavior Observed in Rodents

A study conducted on C57BL/6J mice demonstrated that 25I-NBOMe, administered at doses of 0.1, 0.3, and 1 mg/kg, was 14 times more potent than its “parent” compound, 2C-I, in inducing HTRs, and that this effect was fully reversible using volinserin, a selective 5-HT2A receptor antagonist [36]. Escalating doses of 25B-NBOMe (0.1–10 mg/kg) caused a hormetic response in the number of WDS episodes in Wistar Han rats [37], and this effect was replicated in C57BL/6J mice and was reversed by the 5-HT2A receptor antagonist, ketanserin [38].
A study by Gatch et al. [39] showed that 25I-, 25B-, and 25C-NBOMe dose-dependently reduced animal locomotor activity. When administered subcutaneously, 25I-NBOMe (0.03–3 mg/kg) produced a bell-shaped effect on C57BL/6J mouse activity [40]. In line with this finding, the researchers have also shown that low (0.001–1 mg/kg) and moderate (0.1–1 mg/kg) doses of 25I-NBOMe increase mobility, while a high dose (10 mg/kg) causes a sharp decline in the spontaneous motor activity of male ICR mice [41]. Similarly, 25B-NBOMe inhibited the motor activity of Wistar Han rats measured in an open field (0.3–3 mg/kg) [37]. This phenomenon is similar to the effects of another 5-HT2A receptor agonist, DOI, which modifies locomotor activity via a 5-HT2A/5HT2C-dependent mechanism. Low and moderate doses primarily activate the 5-HT2A receptors located on pyramidal neurons, leading to neuronal excitation and stimulation of locomotor behavior. As the plasma levels of the drug increase, it leads to the activation of the 5-HT2C receptors located on GABAergic interneurons, which results in the suppression of locomotor activity [42].
Some studies suggest the potentially addictive properties of the NBOMes. 25I-NBOMe (0.3 mg/kg) induced place preference in the conditioned place preference (CPP) test and increased vocalization frequency, in a similar way to methamphetamine, in male C57BL/6J mice. On the other hand, those effects were not replicated in self-administration experiments in Sprague Dawley rats (0.03 mg/kg/infusion) since this study reported weak addictive properties compared with place preference studies [43]. 25B-NBOMe at a dose of 1 mg/kg induced place preference in the CPP test in male C57BL/6J mice. Moreover, it produced statistically significant but weaker than those reported for methamphetamine responses in the self-administration procedure at doses of 0.03–0.3 mg/kg/infusion [38]. 25N-NBOMe increased place preference in male C57BL/6J mice at a dose of 3 mg/kg and also produced self-administration in Sprague Dawley rats (0.1 mg/kg/infusion) [44]. 25D-NBOMe (1 mg/kg) induced CPP and was readily self-administered (0.03 mg/kg/infusion) in rats [45].
The mechanism behind those behavioral symptoms seems to originate from a dopaminergic mechanism. 25B-NBOMe [37], 25I-NBOMe [46], 25N-NBOMe [44], and 25D-NBOMe [45] increased extracellular dopamine (DA) levels in the striatum (STR) and nucleus accumbens (NAC), the structures involved in the reward system. Furthermore, the administration of NBOMe compounds affected the protein levels of DA receptors, a hallmark of abuse potential. 25B-NBOMe elevated D1 receptor expression and decreased D2 receptor expression in the mouse NAC. 25N-NBOMe reduced the expression of D2 receptors both in NAC and dorsal STR. It also decreased the expression of DA transporter (DAT) in the NAC, while increasing its phosphorylation in the NAC and dorsal STR. Furthermore, the drug significantly reduced the expression of tyrosine hydroxylase (TH) in the NAC. 25D-NBOMe increased the expression of the dopamine receptor D1 while decreasing the expression of the D2 receptor, and downregulated DAT [45]. Taken together, these changes in dopaminergic activity, while not as potent as those induced by methamphetamine, may originate from a similar mechanism. The excessive DA levels in NAC would stimulate the D2 autoreceptors, which are involved in the regulation of DA release. Overstimulation of D2 receptors would then lead to their downregulation and reduction in the activity of DAT and TH [47].

4.2. The Effects of NBOMes Observed in Humans

NBOMes became available to drug users when they first appeared on the illicit drug market in 2010 [48,49]. Similarly to LSD, they are most often sold on blotter paper; sometimes, they are even sold as LSD. They are administered either nasally or orally (either by swallowing or sublingually) in small, sub-milligram doses [48]. The duration of action varies for each route of administration [36]. The effects of ingestion are usually an outcome of the activation of serotoninergic and adrenergic pathways and may include severe visual and auditory hallucinations, agitation, aggressiveness, long-lasting seizures, tachycardia, sweating, hypertension and hyperthermia, and psychotic/paranoid behavior [12,50,51,52]. Based on recent knowledge, the risk of NBOMes toxicity excludes these 5-HT2 receptor agonists from the treatment of a range of psychiatric disorders.

4.3. The Effects Exerted on Neurotransmitter Release by a Single Exposure to NBOMes

While there is scientific consensus that the primary mechanism behind the psychedelic effect of NBOMes is related to the increase in glutamatergic neurotransmission in the PFC [4,53,54,55], the effects exerted on other neurotransmitters are not so consistent and vary from drug to drug. The NBOMe class is reported to be extremely potent [55,56,57,58] in elevating the extracellular levels of DA, 5-HT, and glutamate after acute administration of 25I-NBOMe, and numerous WDS episodes were also seen.
The effect of administration of 25B-NBOMe is consistent with these findings as an increase in cortical extracellular glutamate levels was observed [37], and this effect was more robust than the one exerted by 25I-NBOMe [55]. Furthermore, 25B-NBOMe induced a similar number of WDS to 25I-NBOMe but with a three times smaller dose (0.3 mg/kg) [37]. Altogether, these findings confirm the stronger affinity and potency of 25B-NBOMe at the 5HT2A receptor, reported in vitro by Rickli et al. [20]. Like 25I-NBOMe [55], 25B-NBOMe significantly increased the extracellular 5-HT levels [37]. This phenomenon may be the reason for the serotonin syndrome reported in humans after ingestion of 25B-NBOMe [51]. Furthermore, it increased the volume of dopaminergic neurotransmission akin to 25I-NBOMe. Those changes observed in NAC and STR suggest that it might exhibit reinforcing properties [37].
The dose–response curve observed in microdialysis studies had an inverted “U” shape, most likely resulting from the activation of the different subtypes of 5-HT receptors. The progressive elevation of extracellular levels of neurotransmitters except acetylcholine (ACh), starting from the lowest dose (0.1 mg/kg) and reaching the plateau at 0.3 mg/kg, is a result of the activation of 5-HT2A receptors located on pyramidal glutamatergic cells [53,55,59,60]. As the plasma levels of the drug increase, subsequent activation of the 5-HT2C receptor located on cortical GABAergic interneurons occurs, leading to an increase in GABAergic neurotransmission and attenuation of the observed effect (Figure 1).
Both local [61] and systemic [62] administration of 5-HT2A/2C receptor agonists were reported to induce a dose-dependent increase in extracellular levels of GABA. This effect was further replicated when assessing hallucinogenic potency, with the WDS response curve which acquired a similar shape reaching the peak at the dose of 0.3 mg/kg [37], similar to 25I-NBOMe [55] and HTR in mice [38]. Furthermore, 25B-NBOMe reduced locomotor activity and induced anxiety in animals; these effects are also akin to those induced by 25I-NBOMe [46] and other psychedelics [42]. Surprisingly, while being more active at the 5-HT2A receptor than 25I-NBOME and inducing a greater increase in extracellular levels of neurotransmitters, 25B-NBOMe induced a very weak genotoxic effect evidenced by oxidative DNA damage in the nuclei from rat PFC and HP when compared with the control group [37] or when comparing it with the effect of 25I-NBOMe [63].
As reported by Quirion et al. [64], the 5-HT2A receptors are located on cortical cholinergic terminals, suggesting their role in the stimulation of ACh release. Nair and Gudelsky [65] reported that activation of the 5-HT2A receptor by DOI or mescaline stimulated ACh release in the PFC, and this effect was abolished by a 5-HT2A/B/C antagonist. In contrast to these results, the study by Wojtas et al. [37] observed a decrease, no changes, or increase in extracellular levels of cortical ACh after the administration of 25B-NBOMe, and the effect varied depending on the dose. These results are hard to explain due to a lack of data concerning the effects of psychedelics on cholinergic neurotransmission.
These results indicate that while being a powerful psychedelic capable of inducing significant enhancement of neurotransmission, which results in changes in animal behavior similar to those induced by other psychedelics, NBOMes might not be as toxic as other representatives of its class.

4.4. The Consequences of Repeated Exposure to NBOMes

As mentioned earlier, 25B-NBOMe elevates extracellular levels of DA in NAC and STR, which are elements of the reward system. Unlike other psychedelics, the NBOMes have been reported to exhibit rewarding properties, inducing CPP in mice and self-administration in rats [44,45]. Most importantly, a study by Custodio et al. [38] revealed that 25B-NBOMe also exhibited these properties in CPP and self-administration, and the effects were neutralized when using the D1 or D2 receptor antagonist, clearly suggesting the involvement of the dopaminergic system. Furthermore, chronic administration of 25B-NBOMe induced changes in the expression of the D1 and D2 receptors, namely upregulation of the former and downregulation of the latter, which is a common phenomenon induced by addictive compounds [38].
As stated before, frequent exposure to psychedelic drugs leads to rapid induction of tolerance [66,67], originating from the downregulation of the 5-HT2A receptor. This was also reported for 25I-NBOMe in rats [46] and 25B-NBOMe in mice [38]. Repeated administration of 25B-NBOMe completely suppressed its effect on cortical glutamatergic and dopaminergic neurotransmission and significantly reduced the effect it induced on extracellular serotonin levels [68]. This effect was more robust than the one elicited by 25I-NBOMe [46]. The influence of tolerance on the WDS response was similar to 25I-NBOMe, starting with a rapid decline in the number of WDS episodes on the second day of treatment. This effect can be explained by the downregulation of the 5-HT2A receptor, which diminishes the enhancement of glutamatergic neurotransmission in the PFC, an essential event needed for HTR/WDS to occur [4]. The observed decrease in locomotor activity after chronic exposure to 25B-NBOMe in the open field test may also be a result of tolerance. With the 5-HT2A receptor downregulation, the 5-HT2C receptor activation plays a greater role in modulating animal behavior, thereby leading to suppression of locomotor activity, as suggested by Halberstadt et al. [42].
The potency of 25B-NBOMe-induced increases in extracellular levels of the examined neurotransmitters was also reduced in NAC and STR. It is important to notice that in the NAC, the observed tolerance was much smaller [68]. Importantly, after a 7-day treatment, the compound still increased extracellular levels of dopamine, which corresponds with its effect on DA receptors in NAC reported in mice [38] and increased Δ-fosB expression. Taken together, these findings suggest that 25B-NBOMe might exhibit addictive properties like other representatives of its family. Nevertheless, it is worth noticing that these effects, while significant, are much smaller than the addictive properties of other classes of drugs of abuse, e.g., psychostimulants [38].
While acute administration of 25B-NBOMe resulted only in a minor genotoxic effect [37], it produced significant DNA damage in both PFC and HP after chronic treatment [68], in a similar way to 25I-NBOMe [64]. Repeated administration of 25B-NBOMe increased basal levels of glutamate, DA, and 5-HT, suggesting the appearance of maladaptive changes in neurotransmission. These increases might lead to genotoxicity directly when elevated glutamate levels overstimulate ionotropic glutamatergic receptors, resulting in DNA damage [69], and indirectly, when an excess of monoamines leads to disturbances in their metabolism, thus increasing the production of free radicals [69,70]. Moreover, phenethylamines may directly produce oxidative stress, which was shown by Xu et al. [71] in in vitro studies. Nevertheless, the observed genotoxic effect did not translate into permanent loss of brain tissue, measured as the volume of cortical and hippocampal regions [68]. Alongside the previous study, these results suggest that although it induces some DNA damage, the toxicity of 25-NBOMe cannot be explained by this phenomenon and is most likely a result of serotonin syndrome [72].

5. Psilocybin

5.1. History of Psilocybin

Psilocybin-containing mushrooms belong to a diverse group within the Basidiomycota fungi, comprising over 200 species across genera, such as Psilocybe, Gymnopilus, and Panaeolus, with the most prevalent genus being Psilocybe, with about 144 species [73]. Psychedelic mushrooms have been consumed and revered as religious objects since prehistoric times by many cultures, with evidence of such practices found in Europe, Africa, and most notably, Mesoamerica [74,75,76].
The modern history of psilocybin starts with Albert Hofmann, the chemist who discovered the psychedelic properties of LSD. In 1957, he received a sample of Psilocybe Mexicana mushrooms from which he extracted two crystalline compounds. Through self-experimentation, he confirmed their psychoactive effects, and by 1958, he had identified these compounds as psilocybin and psilocin, which he synthesized in 1958 [77].

5.2. Pharmacology of Psilocybin

After oral intake, psilocybin is quickly dephosphorylated in the stomach or by alkaline phosphatase in the intestine, kidney, and potentially blood, forming psilocin, which easily penetrates the blood–brain barrier [78,79,80]. Studies in rodents confirm the almost complete conversion of psilocybin to psilocin before entering the bloodstream [81]. Both psilocybin and psilocin have similar psychotropic effects on humans at equimolar concentrations. Thus, psilocybin acts as a prodrug, with psilocin being the active agent responsible for its in vivo effects [82]. Moreover, inhibition of alkaline phosphatase completely suppresses the psychoactive effects of psilocybin administration [83], further supporting the hypothesis that psilocin is the primary psychoactive agent in hallucinogenic mushrooms [84].
As psilocin is structurally akin to the neurotransmitter serotonin, it undergoes a similar metabolism. It is further degraded in the liver by monoamine oxidase (MAO) or aldehyde dehydrogenase, leading to various intermediates and end products [85,86]. This is why MAO inhibitors are sometimes taken by users in an attempt to intensify psilocin’s hallucinogenic effects. Furthermore, psilocin itself may competitively inhibit MAO, which may lead to elevated brain 5-HT levels and a decrease in 5-hydroxyindoleacetic acid (5-HIAA) levels [87].
Like all psychedelics, psilocin has a strong affinity for 5-HT receptors in the brain, primarily acting as an agonist at the 5-HT2A, 5-HT2C, and 5-HT1A receptors. The psychedelic effects of psilocin are largely nullified by ketanserin, a 5-HT2A receptor antagonist, indicating the central role of the 5-HT2A receptor in its subjective effects [88,89,90]. The role of 5-HT1A receptors in the psychoactive effects of psilocin is yet to be examined. Furthermore, psilocin interacts with other serotonin receptors, including 5-HT1B, 5-HT1D, 5-HT2B, 5-HT5A, 5-HT6, and 5-HT7 [84].
The subjective and behavioral effects of psilocybin are influenced by interactions with non-serotonergic receptors. Research suggests that psilocybin indirectly elevates DA levels in the striatum, which is associated with depersonalization and euphoria [91]. Nonetheless, the dopaminergic system plays only a partial role in the psychological effects of psilocybin, as haloperidol, a nonselective DA receptor antagonist, reduces only about 30% of these symptoms [90]. Contrary to LSD, which binds to D2 receptors, psilocybin exhibits no affinity for dopamine D2 receptors [92,93]. While there is evidence suggesting that classical psychedelics can elevate dopaminergic transmission in human striatal regions, they do not significantly activate the NAC in positron emission tomography (PET) imaging studies. This observation aligns with the absence of data linking classical psychedelics to substance use disorder [90,91,94,95].

5.3. The Effects of Psilocybin Observed in Animal Studies

Both psilocybin and psilocin have been observed to evoke HTR and WDS in mice and rats, respectively [96,97], and the effect was absent in 5-HT2A receptor knockout mice [15]. Psilocybin fully substituted for psilocin, DOM, and LSD [98], and these effects were abolished by coadministration of volinserin, further confirming that the primary effects of psilocybin in HTR are mediated via the 5-HT2A receptor [98]. Psilocin dose-dependently (1.25–5.0 mg/kg) reduced locomotor activity in rats [10] and mice [98]. What is interesting is that the treatment with the selective 5-HT1A receptor antagonist, WAY-100635, completely reversed the effects of psilocin on the locomotor activity of mice. Furthermore, deletion of the 5-HT2A receptor gene did not affect the response induced by psilocin [42]. These findings suggest that the effect of psilocybin/psilocin on locomotor behavior may be primarily mediated via the 5-HT1A receptor and not the 5-HT2A receptor.
To date, there are no animal studies suggesting that psilocybin exhibits addictive properties. Fantegrossi et al. [99] reported no significant differences between psilocybin and saline in self-administration in rhesus monkeys. A microdialysis study by Sakashita et al. [100] demonstrated that psilocin elevated extracellular levels of DA in NAC in a slight but significant way but did not affect them in the ventral tegmental area (VTA), which suggests rather low reinforcing properties. Furthermore, psilocin inhibited methamphetamine-induced conditioned place preference formation during the acquisition phase [101]. It also reduced intracranial self-stimulation [102], and this effect was only partially reduced by the administration of volinserin, suggesting that other receptors, besides the 5-HT2A receptor, may also contribute to this effect [103]. These experiments are consistent with the general view that classic psychedelics lack reinforcing properties [4] and suggest a possible use of psilocybin in the treatment of addictive disorders.
Acute administration of psilocin has been reported to either increase anxiety, measured as the center avoidance in mice [97], or exert no effect in this paradigm [104]. The anxiogenic effect was also reported in rats, but it diminished with repeated exposure to the experimental arena, suggesting that the anxiety is a result of drug-enhanced neophobia [15]. A recent study by Hibicke et al. [105] demonstrated the anxiolytic properties of psilocybin if the animals were repeatedly exposed to the novel environment. Anxiety was not observed in rats 24 h after psilocybin administration either in the open field or light/dark box (L/D) paradigm [62]. Furthermore, psilocybin/psilocin facilitated fear extinction in rats [106,107].
Psilocybin tested acutely in Flinders Sensitive Line rats [108] and 24 h after administration in naive controls [62] did not exert an antidepressant effect in the forced swim test (FST) but demonstrated a significant antidepressant effect 5 weeks after its administration [105]. Furthermore, it attenuated learned helplessness in mice 24 h after the drug administration [109] and reversed anhedonia in chronically stressed mice [110], while not affecting the immobility time measured in the FST. These findings support the thesis that (a) psilocybin/psilocin may exhibit antidepressant properties and (b) FST might not be suitable for evaluating the antidepressant qualities of psychedelics.

5.4. The Effects of Psilocybin Observed in Humans

Psilocybin has demonstrated minimal toxicity in animals, with an LD50 in rodents being 2000 to 3000 times the standard human dose on a mg/kg basis [84]. When assessing the acute toxicity, safety, and addictive potential of various psychoactive substances, psilocybin is consistently ranked at the lower harm end while, in comparison, opioids, notably heroin, are at the higher harm end [111,112,113]. Physiologically, psilocybin poses minimal risk to humans, showing no association with major organ damage, carcinogenicity, teratogenicity, enduring neuropsychological deficits, or overdose fatalities [76].
Oral psilocybin doses range from 0.045–0.429 mg/kg, with the psychedelic effect observed with oral doses > 15 mg [114] or plasma psilocin levels of 4–6 ng/mL [80]. Doses exceeding 25 mg orally are considered high but not dangerous [76]. After oral intake, onset of action is 20–40 min, peaking at 60–90 min with effects lasting 4–6 h, and complete cessation by 24 h [90,114].
Psilocybin administration results in dose-dependent effects: low doses produce drowsiness and amplify pre-existing moods [114]; medium doses instigate a controllable altered state of consciousness [82]; and high doses generate intense psychedelic experiences, characterized by altered perception, dream-like states, illusions, hallucinations, synesthesia, and alterations in the perception of time and space [95,114,115].
According to the Altered States of Consciousness scale (ASC) scale [116], when juxtaposed with ketamine, psilocybin exhibits pronounced visual hallucinatory effects, whereas ketamine more markedly disrupts physical integrity [93,117,118]. Pharmacological interventions have provided insights into the mechanism of action of psilocybin. Both ketanserin (5-HT2A/C antagonist) and risperidone (mixed 5-HT2A/C and D2 antagonist) have been observed to normalize psilocybin-induced alterations. Conversely, haloperidol, a D2 receptor antagonist, only modulates euphoria, derealization, and depersonalization but fails to influence visual hallucinations [90].

5.5. The Effects of Psilocybin as a Fast-Acting Antidepressant

While possessing distinct pharmacological targets, namely, the 5-HT2A receptor for psychedelics and the NMDA receptor for ketamine, those drugs exhibit some overlapping properties. Either stimulation of cortical 5-HT2A receptors located on pyramidal cells by psychedelics or inhibition of cortical GABAergic interneurons by ketamine is supposed to result in a “glutamate surge”, which is a sudden elevation of cortical glutamate levels [119,120]. Subsequently, this phenomenon leads to an increase in neuroplasticity, producing an antidepressant effect [121,122]. The molecular mechanism underlying the neuroplastic effects of psychedelics involves 5-HT2A receptor signaling initiated by increased Ca2+ influx in the dendritic compartment that drives the release of neurotrophic factors (e.g., brain-derived neurotrophic factor, BDNF), which act on tropomyosin receptor kinase B (TrkB) receptors to stimulate mammalian target of rapamycin (mTOR) signaling crucial to synapse formation [109,119,123]. Overall, in this model alterations in glutamate release contribute to neuroplasticity. However, in addition to being agonists of the 5-HT2A receptor, psychedelics are also agonists of the 5-HT2C and 5-HT1A receptors [4]. 5-HT1A and 5-HT2A receptors are highly expressed and often co-expressed in PFC pyramidal neurons and GABAergic interneurons. Almost 60% of cells in the PFC contain mRNA for 5-HT1A and 5-HT2A receptors, with co-localization of up to 80% [124,125,126]. Thus, the cellular response is determined by the summation of 5-HT1A receptor-induced inhibition and 5-HT2A receptor-induced excitation. The 5-HT1A receptor signaling at GABAergic interneurons may inhibit this inhibitory control of pyramidal cells leading to their disinhibition. In this way, psilocybin modulates local and downstream effects. Psilocybin could also activate pyramidal neurons via 5-HT2C receptors expressed in layer V neurons of the PFC [127]. However, the depolarizing action of 5-HT in layer V pyramidal neurons of the PFC does not seem to depend on 5-HT2C receptor activation since it was not blocked by the selective antagonist SB 242084 [128]. According to the “bipartite” model proposed by Carhart-Harris and Nutt [129], 5-HT1A and 5-HT2A receptors play a crucial role in psilocybin-induced alterations in brain activity (Figure 2).
On the global level, the administration of psychedelics [130,131] reduces the activity of the default mode network (DMN), which is hyperactivated in depressive disorders. As stated by Vollenweider and Kometer [93], this translates into partially overlapping subjective experiences produced by psilocybin.

5.6. The Effect of Psilocybin Exerted on Cortical and Thalamic Neurotransmitters, Receptors, and Behavior

As reported in other works regarding psychedelics [37,53,55], psilocybin elevated the extracellular glutamate levels in the PFC, providing further evidence for the “glutamate surge” hypothesis [82]. Furthermore, it elevated cortical levels of DA only at a smaller dose, with the high dose being ineffective. This finding is in accordance with the work of Sakashita et al. [100], who reported a slight but significant decrease in cortical DA after administration of a similar dose (10 mg/kg) of psilocin. The authors explain this fact by the activation of 5-HT2C receptors, which are reported to decrease DA activity in the PFC [132]. Psilocybin dose-dependently raised the extracellular levels of 5-HT in the PFC [62], which was observed previously for psilocin by Sakashita et al. [100]. This effect might be mediated via the 5-HT2A receptor, as local administration of 5-HT2A agonist (DOI) into the PFC elevated extracellular levels of cortical 5-HT and this effect was neutralized with 5-HT2A receptor antagonist volinserin [133]. Furthermore, psilocybin dose-dependently increased GABA in the PFC [62]. What is important to note is that this also happens in humans after the administration of psilocybin, as stated by Mason et al. [134]. This could be explained by either the activation of the 5-HT2C [124] or 5-HT2A [4,135] receptor. Both are located on GABAergic interneurons in the PFC, and psilocin exhibits similar affinities for them [10].
As reported by Rodriguez et al. [136], 5-HT2A receptors exhibit high expression in the reticular nucleus (RN) of the thalamus, which is composed mainly of GABAergic interneurons. Vollenweider and Geyer [118] proposed that RN was responsible for filtering the thalamic input into the cortex and that psychedelics enhanced its GABAergic activity, disrupting the negative feedback. The observations of Wojtas et al. [62] seem to support this hypothesis, as a dose-dependent increase in GABAergic neurotransmission was reported while any significant effect on glutamatergic neurotransmission was not observed (Figure 3).
Due to the release of glutamate induced by the administration of psilocybin, the expression of GluN2A and GluN2B subunits of the NMDA receptor and GluA1 and GluA2 subunits of the AMPA receptor in the PFC 24 h after administration of psilocybin was examined. No significant effect was observed, except for the increase in expression of the GluN2A subunit when using the higher (10 mg/kg) dose of psilocybin [62]. These results are hard to explain, and perhaps it would be useful to replicate this experiment with different time points to evaluate if changes might happen earlier/later after the drug was administered.
While it is generally acclaimed that classical psychedelics are physiologically safe to use, a significant increase in glutamatergic activity in the PFC after high doses of psilocybin was observed, which may result in genotoxicity. To examine this, the oxidative DNA damage was assessed with the Comet Assay [62]. The 10 mg/kg dose of psilocybin significantly harmed the DNA in both the PFC and HP, suggesting caution when using high doses of this drug.
To investigate the long-lasting effect of psilocybin administration on rats’ behavior, behavioral tests were conducted 24 h after drug administration to ensure that the injected drug would leave the system. No effect on the locomotor activity was observed [62] when measured in the open field test and only a slight drop in the distance traveled was seen in the L/D box test after administration of 2 mg/kg of psilocybin. This suggests that after 24 h, the drug is washed out of the body and nearly no long-lasting effects are present, supporting the hypothesis about its safety. Since it did not produce an antidepressant effect in the forced swim test (FST), it is worth noticing that this assay may not be suitable for testing fast-acting antidepressant drugs [108].

5.7. The Limbic Response to Psilocybin

Alongside the PFC, the limbic system also exhibits maladaptive changes in depressive disorders [137]. Both the HP and amygdala undergo hypotrophy during depression, while NAC exhibits dysfunctional hypertrophy [137,138,139]. Recent studies have reported that the administration of ketamine is able to reverse those changes in all aforementioned structures [139,140], and these effects are correlated with antidepressant effect. The data concerning the effect of psilocybin on the limbic system and limbic-related behavior are scarce.
As reported by Sakashita et al. [100] and Wojtas et al. [141], psilocin dose-dependently increased extracellular DA levels in the NAC. This effect can be explained by the stimulation of 5-HT2A receptors, as local administration of 5-HT2A agonists enhances DA release in the NAC [142,143], while administration of 5-HT2A antagonists leads to inhibition of DA release in the NAC [143]. Interestingly, while Sakashita and colleagues [100] observed a slight but significant and dose-independent drop in the extracellular level of 5-HT in the NAC after administration of psilocin, Wojtas et al. [141] reported a dose-dependent increase. This effect might be explained by the stimulation of pyramidal cells, which send their projections to the raphe nuclei, and subsequent stimulation of serotonergic neurons projecting to the NAC [144].
Intriguingly, psilocybin decreased accumbal glutamate levels [141]. Previous studies by Wojtas et al. [37] with a more selective 5-HT2A agonist, 25B-NBOMe, reported increases in glutamatergic activity in the NAC. This suggests the involvement of other receptor subtypes, as psilocin is a more promiscuous drug, but the 5-HT1A receptors located on glutamatergic cells projecting into the NAC are the most probable target [145]. The observed increase in the extracellular level of GABA might result from the stimulation of 5-HT2A receptors located on GABAergic interneurons [146].
The opposing effect of lower and higher doses of psilocybin on extracellular levels of glutamate in the HP is hard to explain; the dose-dependent increase in GABA excludes the involvement of GABAergic interneurons [141]. Furthermore, psilocybin increased extracellular levels of ACh in the HP, with the lower dose being more potent [141]. This might be a result of subsequent activation of the 5-HT2A receptor, which stimulates ACh release [65], and then the 5-HT1B receptor, which inhibits ACh release in the rat HP [147]. Perhaps the inhibition of glutamatergic activity by the lower dose of psilocybin results from the elevated levels of ACh acting at M4 muscarinic receptors located on hippocampal pyramidal cells [148].
Psilocybin did not affect extracellular levels of glutamate and GABA in the amygdala [141]. 5-HT2A receptors are highly expressed on pyramidal cells and both parvalbumin and somatostatin GABAergic interneurons in the amygdala, which suggests that the effect of their activation may be mutually suppressive [149].
Despite the fact that psilocin is a 5-HT2A receptor agonist and it elevated the extracellular levels of 5-HT in the NAC, it did not affect the expression of the 5-HT2A receptor. This may be due to its low density in the NAC [150]. Changes were observed for the D2 receptor when using a higher dose of psilocybin [141]. This may be a result of an interplay between dopaminergic and serotonergic neurotransmission, as the administration of haloperidol attenuates the psychotomimetic effects induced by psilocybin administration [82].
Both 5-HT2A and 5-HT1A receptors show high-density expression in the HP [149,151]. The observed decrease in 5-HT1A receptor density might be a result of their stimulation either by psilocybin or the release of serotonin, but there is a lack of data regarding the latter [141]. The decrease in 5-HT2A receptor expression induced by the lower dose of psilocybin [141] may be a result of rapid downregulation, while the increase caused by the higher dose might be a result of an increase in synapto- and neurogenesis [123].
Psilocybin exhibited a dose-dependent anxiolytic effect measured as a decrease in center avoidance in the open field test both 1 and 24 h after the drug administration, and this effect might result from intensification of GABAergic neurotransmission observed in limbic structures [141].

5.8. The Therapeutic Potential of Psychedelics

Besides being a non-toxic and well-tolerated drug, over the last decade, psilocybin has been thoroughly examined as a potential fast-acting antidepressant drug. Carhart-Harris et al. [152] reported that acute administration of psilocybin produced significant effects in 67% of patients with treatment-resistant depression in the first week after the treatment, with 47% of treated individuals staying in remission for 3-month and 6-month periods [153]. A single dose of psilocybin was associated with a reduction in depressive symptoms without serious adverse events in a randomized clinical study of Raison et al. [154]. A study by Gukasyan et al. [155] reported long-lasting antidepressant effects of psilocybin administration, with significant response to long treatment maintained in 75% of participants and the remission rate was 58% at the 12-month follow-up. The drug also withstands comparison with classical antidepressant treatment, as double administration of psilocybin (separated by a 3-week interval) was as efficient as a 6-week chronic treatment with escitalopram [156,157].
Besides depression, numerous clinical studies have shown the therapeutic potential of psychedelics in treating disorders including addiction to alcohol [94,158] and nicotine [159,160,161], anorexia nervosa [162], distress and anxiety concerning death [163,164,165,166], obsessive–compulsive disorder [167], and post-traumatic stress disorder (PTSD) [168]. Psilocybin significantly increased abstinence and decreased craving up to 36 weeks after administration in alcohol-dependent participants [158] and elicited alterations in their relationship with alcohol [94]. Administration of moderate (20 mg/70 kg) and high (30 mg/70 kg) doses of psilocybin was a potentially efficacious adjunct to current smoking cessation treatment and promoted long-term smoking abstinence [159,160,161]. Classical psychedelics including psilocybin alleviated symptoms of anorexia nervosa that relate to serotonergic signaling and cognitive inflexibility [162]. Administration of low (1 or 3 mg/70 kg), moderate (0.2 mg/kg), or high (22 or 30 mg/kg) doses of psilocybin produced immediate, substantial, and sustained improvements in anxiety and depression in patients with life-threatening cancer at 1 to 6 months after treatment [163,164,165]. A similar reduction in anxiety was demonstrated in LSD-assisted psychotherapy [166]. Psilocybin in single doses ranging from sub-hallucinogenic (100 µg/kg), medium (200 µg/kg), and high (300 µg/kg) relieved symptoms of obsessive–compulsive disorder in a controlled clinical environment [167]. Classical psychedelics such as psilocybin and LSD showed significant potential for treating PTSD. However, their unpredictable psychological effects might not make them the best candidates compared with MDMA [168].
Serotonergic hallucinogens, acting on 5-HT2A receptors, induce perceptual and behavioral alterations possibly related to psychotic symptoms. It is suggested that the psychotomimetic properties of psychedelics are mediated by an alteration in thalamocortical activity, which is the neurobiological basis of schizophrenia [169,170]. Given the enhancing effect of psychedelics on neural plasticity and their effects on inflammatory processes, it is possible that psychedelics could have a role in treating cortical atrophy and cell loss in schizophrenia and the negative symptoms associated with this illness. It is thus possible that microdosing, in which psilocybin is taken in one-tenth of the typical psychedelic dose, has no psychosis-inducing effects while it may have positive effects on mood, may alleviate depression and anxiety, and improve cognitive function [171].

6. Conclusions and Future Questions

The presented data support the hypothesis about a greater risk associated with the use of Novel Psychoactive Substances (i.e., NBOMes) in comparison with naturally occurring psychedelics. Therefore, the potential efficacy of these substances for medical treatment is ambiguous or low in comparison with psilocybin. The reviewed studies allow for the characterization of the effects of selective 5-HT2A/C receptor agonists on excitatory neurotransmitter systems and establish how those changes may translate into behavior and damage inflicted upon the central nervous system.
A similar approach was then applied to a compound with a broader receptor profile (psilocybin), uncovering its subtler effects resulting from the interplay of 5-HT1A and 5-HT2A receptors, with possible beneficial influence (i.e., lasting anxiolytic effect).
Mechanistically, psychedelics induce changes in neuronal structure that activate BDNF/TrkB/mTOR signaling crucial to synapse formation by stimulating 5-HT2A receptors [122,123]. The localization of 5-HT2A receptors highly expressed on layer V pyramidal neurons of the PFC could explain why psychedelics do not promote plasticity in the mesolimbic pathway and are not addictive. However, it is important to consider the potential risks associated with excessive release of glutamate leading to excitotoxicity and cell atrophy. Another question that needs to be resolved is whether the subjective effects of psychedelics are a critical component of their therapeutic mechanism. The development of non-hallucinogenic compounds such as analogs of 5-MeO-DMT, 6-MeO-isoDMT or tabernanthalog (TBG), which exhibit reduced hallucinogenic potential while retaining psychoplastogenic potency, may greatly improve the accessibility of therapy with psychedelics [172]. The recent discovery of linking intracellular 5-HT2A receptors with the induction of plasticity by molecules having lipophilic properties can lead to new drug discovery [173].

Author Contributions

Conceptualization, A.W. and K.G.; writing, review, and editing, A.W. and K.G. All authors have read and agreed to the published version of the manuscript.

Funding

This research was funded by the National Science Centre grant nos. 2020/37/B/NZ7/03753 and 2016/21/B/NZ7/01131 and statutory funds of the Maj Institute of Pharmacology, Polish Academy of Sciences.

Institutional Review Board Statement

Not applicable.

Informed Consent Statement

Not applicable.

Data Availability Statement

Data sharing is not applicable to this article. No new data were created or analyzed in this study.

Acknowledgments

The authors thank Agnieszka Bysiek and Izabela Szpręgiel for their help in editing this article.

Conflicts of Interest

The authors declare no conflict of interest.

References

  1. Schultes, R.E.; Hofmann, A.; Rätsch, C. Plants of the Gods: Their Sacred, Healing, and Hallucinogenic Powers. Heal. Art. Press. 1998, 1–208. [Google Scholar] [CrossRef]
  2. Nichols, D.E.; Walter, H. The History of Psychedelics in Psychiatry. Pharmacopsychiatry 2021, 54, 151–166. [Google Scholar] [CrossRef] [PubMed]
  3. Hofmann, A. How LSD originated. J. Psychedelic Drugs 1979, 11, 53–60. [Google Scholar] [CrossRef] [PubMed]
  4. Nichols, D.E. Psychedelics. Pharmacol. Rev. 2016, 68, 264–355. [Google Scholar] [CrossRef] [PubMed]
  5. Liechti, M.E. Modern Clinical Research on LSD. Neuropsychopharmacology 2017, 42, 2114–2127. [Google Scholar] [CrossRef] [PubMed]
  6. Seybert, C.; Cotovio, G.; Madeira, L.; Ricou, M.; Pires, A.M.; Oliviera-Maia, A.J. Psychedelic treatments for mental health conditions pose challenges for informed consent. Nat. Med. Comment. 2023, 29, 2167–2170. [Google Scholar] [CrossRef] [PubMed]
  7. Kwan, A.C.; Olson, D.E.; Preller, K.H.; Roth, B.L. The neural basis of psychedelic action. Nat. Neurosci. 2022, 25, 1407–1419. [Google Scholar] [CrossRef]
  8. Pierce, P.A.; Peroutka, S.J. Hallucinogenic drug interactions with neurotransmitter receptor binding sites in human cortex. Psychopharmacology 1989, 97, 118–122. [Google Scholar] [CrossRef]
  9. Titeler, M.; Lyon, R.A.; Glennon, R.A. Radioligand binding evidence implicates the brain 5-HT2 receptor as a site of action for LSD and phenylisopropylamine hallucinogens. Psychopharmacology 1988, 94, 213–216. [Google Scholar] [CrossRef]
  10. Halberstadt, A.L.; Geyer, M.A. Multiple receptors contribute to the behavioral effects of indoleamine hallucinogens. Neuropharmacology 2011, 61, 364–381. [Google Scholar] [CrossRef]
  11. Nichols, D.E. Structure-activity relationships of serotonin 5-HT2A agonists. Wiley Interdiscip. Rev. Membr. Transp. Signal. 2012, 1, 559–579. [Google Scholar] [CrossRef]
  12. Hill, S.L.; Thomas, S.H. Clinical toxicology of newer recreational drugs. Clin. Toxicol. 2011, 49, 705–719. [Google Scholar] [CrossRef] [PubMed]
  13. Glennon, R.A.; Titeler, M.; McKenney, J.D. Evidence for 5-HT2 involvement in the mechanism of action of hallucinogenic agents. Life Sci. 1984, 35, 2505–2511. [Google Scholar] [CrossRef] [PubMed]
  14. Araújo, A.M.; Carvalho, F.; Bastos, M.D.L.; Guedes de Pinho, P.; Carvalho, M. The hallucinogenic world of tryptamines: An updated review. Arch. Toxicol. 2015, 89, 1151–1173. [Google Scholar] [CrossRef] [PubMed]
  15. Halberstadt, A.L.; Geyer, M.A. Effect of Hallucinogens on Unconditioned Behavior. Curr. Top. Behav. Neurosci. 2018, 36, 159–199. [Google Scholar] [CrossRef] [PubMed]
  16. Nichols, D.E. Hallucinogens. Pharmacol. Ther. 2004, 101, 131–181. [Google Scholar] [CrossRef] [PubMed]
  17. Ray, T.S. Psychedelics and the human receptorome. PLoS ONE 2010, 5, e9019. [Google Scholar] [CrossRef]
  18. Giacomelli, S.; Palmery, M.; Romanelli, L.; Cheng, C.Y.; Silvestrini, B. Lysergic acid diethylamide (LSD) is a partial agonist of D2 dopaminergic receptors and it potentiates dopamine-mediated prolactin secretion in lactotrophs in vitro. Life Sci. 1998, 63, 215–222. [Google Scholar] [CrossRef]
  19. Watts, V.J.; Lawler, C.P.; Fox, D.R.; Neve, K.A.; Nichols, D.E.; Mailman, R.B. LSD and structural analogs: Pharmacological evaluation at D1 dopamine receptors. Psychopharmacology 1995, 118, 401–409. [Google Scholar] [CrossRef]
  20. Rickli, A.; Luethi, D.; Reinisch, J.; Buchy, D.; Hoener, M.C.; Liechti, M.E. Receptor interaction profiles of novel N-2-methoxybenzyl (NBOMe) derivatives of 2,5-dimethoxy-substituted phenethylamines (2C drugs). Neuropharmacology 2015, 99, 546–553. [Google Scholar] [CrossRef]
  21. Rickli, A.; Moning, O.D.; Hoener, M.C.; Liechti, M.E. Receptor interaction profiles of novel psychoactive tryptamines compared with classic hallucinogens. Eur. Neuropsychopharm. 2016, 26, 1327–1337. [Google Scholar] [CrossRef] [PubMed]
  22. Aghajanian, G.K.; Marek, G.J. Serotonin induces excitatory postsynaptic potentials in apical dendrites of neocortical pyramidal cells. Neuropharmacology 1997, 36, 589–599. [Google Scholar] [CrossRef] [PubMed]
  23. Sipes, T.E.; Geyer, M.A. DOI disruption of prepulse inhibition of startle in the rat is mediated by 5-HT(2A) and not by 5-HT(2C) receptors. Behav. Pharmacol. 1995, 6, 839–842. [Google Scholar] [CrossRef] [PubMed]
  24. Wing, L.L.; Tapson, G.S.; Geyer, M.A. 5HT-2 mediation of acute behavioral effects of hallucinogens in rats. Psychopharmacology 1990, 100, 417–425. [Google Scholar] [CrossRef] [PubMed]
  25. Madsen, M.K.; Fisher, P.M.; Burmester, D.; Dyssegaard, A.; Stenbæk, D.S.; Kristiansen, S.; Johansen, S.S.; Lehel, S.; Linnet, K.; Svarer, C.; et al. Correction: Psychedelic effects of psilocybin correlate with serotonin 2A receptor occupancy and plasma psilocin levels. Neuropsychopharmacology 2019, 44, 1336–1337. [Google Scholar] [CrossRef] [PubMed]
  26. Vollenweider, F.X.; Smallridge, J.W. Classic Psychedelic Drugs: Update on Biological Mechanisms. Pharmacopsychiatry 2022, 55, 121–138. [Google Scholar] [CrossRef]
  27. Barrett, F.S.; Krimmel, S.R.; Griffiths, R.R.; Seminowicz, D.A.; Mathur, B.N. Psilocybin acutely alters the functional connectivity of the claustrum with brain networks that support perception, memory, and attention. Neuroimage 2020, 218, 116980. [Google Scholar] [CrossRef]
  28. Kraehenmann, R.; Prellerm, K.H.; Scheidegger, M.; Pokorny, T.; Bosch, O.G.; Seifritz, E.; Vollenweider, F.X. Psilocybin-Induced Decrease in Amygdala Reactivity Correlates with Enhanced Positive Mood in Healthy Volunteers. Biol. Psychiatry 2015, 78, 572–581. [Google Scholar] [CrossRef]
  29. Carhart-Harris, R.L.; Leech, R.; Hellyer, P.J.; Shanahan, M.; Feilding, A.; Tagliazucchi, E.; Chialvo, D.R.; Nutt, D. The entropic brain: A theory of conscious states informed by neuroimaging research with psychedelic drugs. Front. Hum. Neurosci. 2014, 8, 20. [Google Scholar] [CrossRef]
  30. Aghajanian, G.K.; Marek, G.J. Serotonin model of schizophrenia: Emerging role of glutamate mechanisms. Brain Res. Brain Res. Rev. 2000, 31, 302–312. [Google Scholar] [CrossRef]
  31. Béïque, J.C.; Imad, M.; Mladenovic, L.; Gingrich, J.A.; Andrade, R. Mechanism of the 5-hydroxytryptamine 2A receptor-mediated facilitation of synaptic activity in prefrontal cortex. Proc. Natl. Acad. Sci. USA 2007, 104, 9870–9875. [Google Scholar] [CrossRef] [PubMed]
  32. Martin, D.A.; Nichols, C.D. Psychedelics Recruit Multiple Cellular Types and Produce Complex Transcriptional Responses Within the Brain. EBioMedicine 2016, 11, 262–277. [Google Scholar] [CrossRef] [PubMed]
  33. Halberstadt, A.L.; Geyer, M.A. 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. [Google Scholar] [CrossRef] [PubMed]
  34. Braden, M.R.; Parrishm, J.C.; Naylor, J.C.; Nichols, D.E. 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. [Google Scholar] [CrossRef] [PubMed]
  35. Eshleman, A.J.; Wolfrum, K.M.; Reed, J.F.; Kim, S.O.; Johnson, R.A.; Janowsky, A. Neurochemical pharmacology of psychoactive substituted N-benzylphenethylamines: High potency agonists at 5-HT2A receptors. Biochem. Pharmacol. 2018, 158, 27–34. [Google Scholar] [CrossRef] [PubMed]
  36. Halberstadt, A.L.; Geyer, M.A. 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. [Google Scholar] [CrossRef] [PubMed]
  37. Wojtas, A.; Herian, M.; Skawski, M.; Sobocińska, M.; González-Marín, A.; Noworyta-Sokołowska, K.; Gołembiowska, K. Neurochemical and Behavioral Effects of a New Hallucinogenic Compound 25B-NBOMe in Rats. Neurotox. Res. 2021, 39, 305–326. [Google Scholar] [CrossRef]
  38. Custodio, R.J.P.; Sayson, L.V.; Botanas, C.J.; Abiero, A.; You, K.Y.; Kim, M.; Lee, H.J.; Yoo, S.Y.; Lee, K.W.; Lee, Y.S.; et al. 25B-NBOMe, a novel N-2-methoxybenzyl-phenethylamine (NBOMe) derivative, may induce rewarding and reinforcing effects via a dopaminergic mechanism: Evidence of abuse potential. Addict. Biol. 2020, 25, e12850. [Google Scholar] [CrossRef]
  39. Gatch, M.B.; Dolan, S.B.; Forster, M.J. Locomotor and discriminative stimulus effects of four novel hallucinogens in rodents. Behav. Pharmacol. 2017, 28, 375–385. [Google Scholar] [CrossRef]
  40. Halberstadt, A.L. Pharmacology and Toxicology of N-Benzylphenethylamine (“NBOMe”) Hallucinogens. Curr. Top. Behav. Neurosci. 2017, 32, 283–311. [Google Scholar] [CrossRef]
  41. Tirri, M.; Bilel, S.; Arfè, R.; Corli, G.; Marchetti, B.; Bernardi, T.; Boccuto, F.; Serpelloni, G.; Botrè, F.; De-Giorgio, F.; et al. Effect of -NBOMe Compounds on Sensorimotor, Motor, and Prepulse Inhibition Responses in Mice in Comparison with the 2C Analogs and Lysergic Acid Diethylamide: From Preclinical Evidence to Forensic Implication in Driving Under the Influence of Drugs. Front. Psychiatry 2022, 13, 875722. [Google Scholar] [CrossRef] [PubMed]
  42. Halberstadt, A.L.; van der Heijden, I.; Ruderman, M.A.; Risbrough, V.B.; Gingrich, J.A.; Geyer, M.A.; Powell, S.B. 5-HT(2A) and 5-HT(2C) receptors exert opposing effects on locomotor activity in mice. Neuropsychopharmacology 2009, 34, 1958–1967. [Google Scholar] [CrossRef] [PubMed]
  43. Jeon, S.Y.; Kim, Y.H.; Kim, S.J.; Suh, S.K.; Cha, H.J. Abuse potential of 2-(4-iodo-2, 5-dimethoxyphenyl)N-(2-methoxybenzyl)ethanamine (25INBOMe); in vivo and ex vivo approaches. Neurochem. Int. 2019, 125, 74–81. [Google Scholar] [CrossRef] [PubMed]
  44. Seo, J.Y.; Hur, K.H.; Ko, Y.H.; Kim, K.; Lee, B.R.; Kim, Y.J.; Kim, S.K.; Kim, S.E.; Lee, Y.S.; Kim, H.C.; et al. A novel designer drug, 25N-NBOMe, exhibits abuse potential via the dopaminergic system in rodents. Brain Res. Bull. 2019, 152, 19–26. [Google Scholar] [CrossRef] [PubMed]
  45. Lee, J.G.; Hur, K.H.; Hwang, S.B.; Lee, S.; Lee, S.Y.; Jang, C.G. Designer Drug, 25D-NBOMe, Has Reinforcing and Rewarding Effects through Change of a Dopaminergic Neurochemical System. ACS Chem. Neurosci. 2023, 14, 2658–2666. [Google Scholar] [CrossRef]
  46. Herian, M.; Skawski, M.; Wojtas, A.; Sobocińska, M.K.; Noworyta, K.; Gołembiowska, K. Tolerance to neurochemical and behavioral effects of the hallucinogen 25I-NBOMe. Psychopharmacology 2021, 238, 2349–2364. [Google Scholar] [CrossRef]
  47. Ford, C.P. The role of D2-autoreceptors in regulating dopamine neuron activity and transmission. Neuroscience 2014, 282, 13–22. [Google Scholar] [CrossRef]
  48. Poklis, J.L.; Rasom, S.A.; Alfordm, K.N.; Poklism, A.; Peacem, M.R. Analysis of 25I-NBOMe, 25B-NBOMe, 25C-NBOMe and Other Dimethoxyphenyl-N-[(2-Methoxyphenyl) Methyl]Ethanamine Derivatives on Blotter Paper. J. Anal. Toxicol. 2015, 39, 617–623. [Google Scholar] [CrossRef]
  49. Zuba, D.; Sekuła, K.; Buczek, A. 25C-NBOMe—New potent hallucinogenic substance identified on the drug market. Forensic Sci. Int. 2013, 227, 7–14. [Google Scholar] [CrossRef]
  50. Al-Imam, A.; AbdulMajeed, B.A. NBOMe Compounds: Systematic Review and Data Crunching of the Surface Web. Glob. J. Health Sci. 2017, 9, 126. [Google Scholar] [CrossRef]
  51. Gee, P.; Schep, L.J.; Jensen, B.P.; Moore, G.; Barrington, S. Case series: Toxicity from 25B-NBOMe—A cluster of N-bomb cases. Clin. Toxicol. 2016, 54, 141–146. [Google Scholar] [CrossRef] [PubMed]
  52. Nikolaou, P.; Papoutsis, I.; Stefanidou, M.; Spiliopoulou, C.; Athanaselis, S. 2C-I-NBOMe, an “N-bomb” that kills with “Smiles”. Toxicological and legislative aspects. Drug Chem. Toxicol. 2015, 38, 113–119. [Google Scholar] [CrossRef] [PubMed]
  53. Muschamp, J.W.; Regina, M.J.; Hull, E.M.; Winter, J.C.; Rabin, R.A. Lysergic acid diethylamide and [-]-2,5-dimethoxy-4-methylamphetamine increase extracellular glutamate in rat prefrontal cortex. Brain Res. 2004, 1023, 134–140. [Google Scholar] [CrossRef] [PubMed]
  54. Noworyta-Sokołowska, K.; Kamińska, K.; Kreiner, G.; Rogóż, Z.; Gołembiowska, K. Neurotoxic Effects of 5-MeO-DIPT: A Psychoactive Tryptamine Derivative in Rats. Neurotox. Res. 2016, 30, 606–619. [Google Scholar] [CrossRef] [PubMed]
  55. Herian, M.; Wojtas, A.; Kamińska, K.; Świt, P.; Wach, A.; Gołembiowska, K. Hallucinogen-Like Action of the Novel Designer Drug 25I-NBOMe and Its Effect on Cortical Neurotransmitters in Rats. Neurotox. Res. 2019, 36, 91–100. [Google Scholar] [CrossRef] [PubMed]
  56. Poulie, C.B.M.; Jensen, A.A.; Halberstadt, A.L.; Kristensen, J.L. DARK Classics in Chemical Neuroscience: NBOMes. ACS Chem. Neurosci. 2020, 11, 3860–3869. [Google Scholar] [CrossRef] [PubMed]
  57. Zawilska, J.B.; Kacela, M.; Adamowicz, P. NBOMes-Highly Potent and Toxic Alternatives of LSD. Front. Neurosci. 2020, 14, 78. [Google Scholar] [CrossRef]
  58. Miliano, C.; Marti, M.; Pintori, N.; Castelli, M.P.; Tirri, M.; Arfè, R.; De Luca, M.A. Neurochemical and Behavioral Profiling in Male and Female Rats of the Psychedelic Agent 25I-NBOMe. Front. Pharmacol. 2019, 10, 1406. [Google Scholar] [CrossRef]
  59. Miner, L.A.; Backstrom, J.R.; Sanders-Bush, E.; Sesack, S.R. Ultrastructural localization of serotonin2A receptors in the middle layers of the rat prelimbic prefrontal cortex. Neuroscience 2003, 116, 107–117. [Google Scholar] [CrossRef]
  60. Scruggs, J.L.; Schmidt, D.; Deutch, A.Y. The hallucinogen 1-[2,5-dimethoxy-4-iodophenyl]-2-aminopropane (DOI) increases cortical extracellular glutamate levels in rats. Neurosci. Lett. 2003, 346, 137–140. [Google Scholar] [CrossRef]
  61. Abi-Saab, W.M.; Bubser, M.; Roth, R.H.; Deutch, A.Y. 5-HT2 receptor regulation of extracellular GABA levels in the prefrontal cortex. Neuropsychopharmacology 1999, 20, 92–96. [Google Scholar] [CrossRef] [PubMed]
  62. Wojtas, A.; Bysiek, A.; Wawrzczak-Bargiela, A.; Szych, Z.; Majcher-Maślanka, I.; Herian, M.; Maćkowiak, M.; Gołembiowska, K. Effect of Psilocybin and Ketamine on Brain Neurotransmitters, Glutamate Receptors, DNA and Rat Behavior. Int. J. Mol. Sci. 2022, 23, 6713. [Google Scholar] [CrossRef] [PubMed]
  63. Herian, M.; Wojtas, A.; Maćkowiak, M.; Wawrzczak-Bargiela, A.; Solarz, A.; Bysiek, A.; Madej, K.; Gołembiowska, K. Neurotoxicological profile of the hallucinogenic compound 25I-NBOMe. Sci. Rep. 2022, 12, 2939. [Google Scholar] [CrossRef] [PubMed]
  64. Quirion, R.; Richard, J.; Dam, T.V. Evidence for the existence of serotonin type-2 receptors on cholinergic terminals in rat cortex. Brain Res. 1985, 333, 345–349. [Google Scholar] [CrossRef] [PubMed]
  65. Nair, S.G.; Gudelsky, G.A. Activation of 5-HT2 receptors enhances the release of acetylcholine in the prefrontal cortex and hippocampus of the rat. Synapse 2004, 53, 202–207. [Google Scholar] [CrossRef]
  66. Abramson, H.; Jarvik, M.; Govin, M.; Hirsch, M. Lysergic acid diethylamide (LSD-25) antagonists. II. Development of tolerance in man to LSD-25 by prior administration of MLD-41 (1-methyl-d-lysergic acid diethylamide). AMA Arch. Neurol. Psychiatry 1958, 79, 201–207. [Google Scholar] [CrossRef]
  67. Angrist, B.; Rotrosen, J.; Gershon, S. Assessment of tolerance to the hallucinogenic effects of DOM. Psychopharmacologia 1974, 36, 203–207. [Google Scholar] [CrossRef]
  68. Wojtas, A.; Herian, M.; Maćkowiak, M.; Solarz, A.; Wawrzczak-Bargiela, A.; Bysiek, A.; Noworyta, K.; Gołembiowska, K. Hallucinogenic activity, neurotransmitters release, anxiolytic and neurotoxic effects in Rat’s brain following repeated administration of novel psychoactive compound 25B-NBOMe. Neuropharmacology 2023, 240, 109713. [Google Scholar] [CrossRef]
  69. Halliwell, B. Oxidative stress and neurodegeneration: Where are we now? J. Neurochem. 2006, 97, 1634–1658. [Google Scholar] [CrossRef]
  70. Wrona, M.Z.; Dryhurst, G. Oxidation of serotonin by superoxide radical: Implications to neurodegenerative brain disorders. Chem. Res. Toxicol. 1998, 11, 639–650. [Google Scholar] [CrossRef]
  71. Xu, P.; Qiu, Q.; Li, H.; Yan, S.; Yang, M.; Naman, C.B.; Wang, Y.; Zhou, W.; Shen, H.; Cui, W. 25C-NBOMe, a Novel Designer Psychedelic, Induces Neurotoxicity 50 Times More Potent Than Methamphetamine In Vitro. Neurotox. Res. 2019, 35, 993–998. [Google Scholar] [CrossRef] [PubMed]
  72. Kawahara, G.; Maeda, H.; Kikura-Hanajiri, R.; Yoshida, K.I.; Hayashi, Y.K. The psychoactive drug 25B-NBOMe recapitulates rhabdomyolysis in zebrafish larvae. Forensic Toxicol. 2017, 35, 369–375. [Google Scholar] [CrossRef] [PubMed]
  73. Guzmán, G.; Jacobs, J.Q.; Ramírez-Guillén, F.; Murrieta, D.; Gándara, E. The taxonomy of Psilocybe fagicola-complex. J. Microbiol. 2005, 43, 158–165. [Google Scholar] [PubMed]
  74. Wasson, R.G. Secret of “Divine Mushrooms”. Life . 13 May 1957, pp. 100–120. Available online: https://www.cuttersguide.com/pdf/Periodical-Publications/life-by-time-inc-published-may-13-1957.pdf (accessed on 16 November 2023).
  75. Carod-Artal, F.J. Hallucinogenic drugs in pre-Columbian Mesoamerican cultures. Neurologia 2015, 30, 42–49. [Google Scholar] [CrossRef] [PubMed]
  76. Johnson, M.W.; Griffiths, R.R.; Hendricks, P.S.; Henningfield, J.E. The abuse potential of medical psilocybin according to the 8 factors of the Controlled Substances Act. Neuropharmacology 2018, 142, 143–166. [Google Scholar] [CrossRef] [PubMed]
  77. Hofmann, A.; Frey, A.; Ott, H.; Petrzilka, T.; Troxler, F. Elucidation of the structure and the synthesis of psilocybin. Experientia 1958, 14, 397–399. [Google Scholar] [CrossRef] [PubMed]
  78. Horita, A.; Weber, L.J. Dephosphorylation of psilocybin to psilocin by alkaline phosphatase. Proc. Soc. Exp. Biol. Med. 1961, 106, 32–34. [Google Scholar] [CrossRef]
  79. Horita, A.; Weber, L.J. The enzymic dephosphorylation and oxidation of psilocybin and psilocin by mammalian tissue homogenates. Biochem. Pharmacol. 1961, 7, 47–54. [Google Scholar] [CrossRef]
  80. Hasler, F.; Bourquin, D.; Brenneisen, R.; Bär, T.; Vollenweider, F.X. Determination of psilocin and 4-hydroxyindole-3-acetic acid in plasma by HPLC-ECD and pharmacokinetic profiles of oral and intravenous psilocybin in man. Pharm. Acta Helv. 1997, 72, 175–184. [Google Scholar] [CrossRef]
  81. Eivindvik, K.; Rasmussen, K.E.; Sund, R.B. Handling of psilocybin and psilocin by everted sacs of rat jejunum and colon. Acta Pharm. Nord. 1989, 1, 295–302. [Google Scholar]
  82. Passie, T.; Seifert, J.; Schneider, U.; Emrich, H.M. The pharmacology of psilocybin. Addict. Biol. 2002, 7, 357–364. [Google Scholar] [CrossRef] [PubMed]
  83. Horita, A. Some biochemical studies on psilocybin and psilogin. J. Neuropsychiatry 1963, 4, 270–273. [Google Scholar]
  84. Tylš, F.; Páleníček, T.; Horáček, J. Psilocybin—Summary of knowledge and new perspectives. Eur. Neuropsychopharmacol. 2014, 24, 342–356. [Google Scholar] [CrossRef] [PubMed]
  85. Kalberer, F.; Kreis, W.; Rutschmann, J. The fate of psilocin in the rat. Biochem. Pharmacol. 1962, 11, 261–269. [Google Scholar] [CrossRef] [PubMed]
  86. Lindenblatt, H.; Krämer, E.; Holzmann-Erens, P.; Gouzoulis-Mayfrank, E.; Kovar, K.A. Quantitation of psilocin in human plasma by high-performance liquid chromatography and electrochemical detection: Comparison of liquid-liquid extraction with automated on-line solid-phase extraction. J. Chromatogr. B Biomed. Sci. Appl. 1998, 709, 255–263. [Google Scholar] [CrossRef] [PubMed]
  87. Dinis-Oliveira, R.J. Metabolism of psilocybin and psilocin: Clinical and forensic toxicological relevance. Drug Metab. Rev. 2017, 49, 84–91. [Google Scholar] [CrossRef] [PubMed]
  88. Preller, K.H.; Pokorny, T.; Hock, A.; Kraehenmann, R.; Stämpfli, P.; Seifritz, E.; Scheidegger, M.; Vollenweider, F.X. Effects of serotonin 2A/1A receptor stimulation on social exclusion processing. Proc. Natl. Acad. Sci. USA 2016, 113, 5119–5124. [Google Scholar] [CrossRef]
  89. Preller, K.H.; Herdener, M.; Pokorny, T.; Planzer, A.; Kraehenmann, R.; Stämpfli, P.; Liechti, M.E.; Seifritz, E.; Vollenweider, F.X. The Fabric of Meaning and Subjective Effects in LSD-Induced States Depend on Serotonin 2A Receptor Activation. Curr. Biol. 2017, 27, 451–457. [Google Scholar] [CrossRef]
  90. Vollenweider, F.X.; Vollenweider-Scherpenhuyzen, M.F.; Bäbler, A.; Vogel, H.; Hell, D. Psilocybin induces schizophrenia-like psychosis in humans via a serotonin-2 agonist action. Neuroreport 1998, 9, 3897–3902. [Google Scholar] [CrossRef]
  91. Vollenweider, F.X.; Vontobel, P.; Hell, D.; Leenders, K.L. 5-HT modulation of dopamine release in basal ganglia in psilocybin-induced psychosis in man—A PET study with [11C]raclopride. Neuropsychopharmacology 1999, 20, 424–433. [Google Scholar] [CrossRef]
  92. Marona-Lewicka, D.; Thisted, R.A.; Nichols, D.E. 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. [Google Scholar] [CrossRef] [PubMed]
  93. Vollenweider, F.X.; Kometer, M. The neurobiology of psychedelic drugs: Implications for the treatment of mood disorders. Nat. Rev. Neurosci. 2010, 11, 642–651. [Google Scholar] [CrossRef] [PubMed]
  94. Bogenschutz, M.P.; Podrebarac, S.K.; Duane, J.H.; Amegadzie, S.S.; Malone, T.C.; Owens, L.T.; Ross, S.; Mennenga, S.E. Clinical Interpretations of Patient Experience in a Trial of Psilocybin-Assisted Psychotherapy for Alcohol Use Disorder. Front. Pharmacol. 2018, 9, 100. [Google Scholar] [CrossRef] [PubMed]
  95. Geyer, M.A.; Vollenweider, F.X. Serotonin research: Contributions to understanding psychoses. Trends Pharmacol. Sci. 2008, 29, 445–453. [Google Scholar] [CrossRef]
  96. Corne, S.J.; Pickering, R.W. A possible correlation between drug-induced hallucinations in man and a behavioural response in mice. Psychopharmacologia 1967, 11, 65–78. [Google Scholar] [CrossRef]
  97. Halberstadt, A.L.; Koedood, L.; Powell, S.B.; Geyer, M.A. Differential contributions of serotonin receptors to the behavioral effects of indoleamine hallucinogens in mice. J. Psychopharmacol. 2011, 25, 1548–1561. [Google Scholar] [CrossRef]
  98. Winter, J.C.; Rice, K.C.; Amorosi, D.J.; Rabin, R.A. Psilocybin-induced stimulus control in the rat. Pharmacol. Biochem. Behav. 2007, 87, 472–480. [Google Scholar] [CrossRef]
  99. Fantegrossi, W.E.; Murnane, A.C.; Reissig, J.C. The behavioral pharmacology of hallucinogens. Behav. Biol. 2008, 75, 17–33. [Google Scholar] [CrossRef]
  100. Sakashita, Y.; Abe, K.; Katagiri, N.; Kambe, T.; Saitoh, T.; Utsunomiya, I.; Horiguchi, Y.; Taguchi, K. Effect of psilocin on extracellular dopamine and serotonin levels in the mesoaccumbens and mesocortical pathway in awake rats. Biol. Pharm. Bull. 2015, 38, 134–138. [Google Scholar] [CrossRef]
  101. Wang, J.; Liang, M.; Shang, Q.; Qian, H.; An, R.; Liu, H.; Shao, G.; Li, T.; Liu, X. Psilocin suppresses methamphetamine-induced hyperlocomotion and acquisition of conditioned place preference via D2R-mediated ERK signaling. CNS Neurosci. Ther. 2023, 29, 831–841. [Google Scholar] [CrossRef]
  102. Sakloth, F.; Leggett, E.; Moerke, M.J.; Townsend, E.A.; Banks, M.L.; Negus, S.S. Effects of acute and repeated treatment with serotonin 5-HT2A receptor agonist hallucinogens on intracranial self-stimulation in rats. Exp. Clin. Psychopharmacol. 2019, 27, 215–226. [Google Scholar] [CrossRef] [PubMed]
  103. Jaster, A.M.; Elder, H.; Marsh, S.A.; de la Fuente Revenga, M.; Negus, S.S.; González-Maeso, J. Effects of the 5-HT2A receptor antagonist volinanserin on head-twitch response and intracranial self-stimulation depression induced by different structural classes of psychedelics in rodents. Psychopharmacology 2022, 239, 1665–1677. [Google Scholar] [CrossRef] [PubMed]
  104. Chen, Y.; Liu, J.; Yao, Y.; Yan, H.; Su, R. Rearing behaviour in the mouse behavioural pattern monitor distinguishes the effects of psychedelics from those of lisuride and TBG. Front. Pharmacol. 2023, 14, 1021729. [Google Scholar] [CrossRef] [PubMed]
  105. Hibicke, M.; Landry, A.N.; Kramer, H.M.; Talman, Z.K.; Nichols, C.D. Psychedelics, but Not Ketamine, Produce Persistent Antidepressant-like Effects in a Rodent Experimental System for the Study of Depression. ACS Chem. Neurosci. 2020, 11, 864–871. [Google Scholar] [CrossRef]
  106. Catlow, B.J.; Song, S.; Paredes, D.A.; Kirstein, C.L.; Sanchez-Ramos, J. Effects of psilocybin on hippocampal neurogenesis and extinction of trace fear conditioning. Exp. Brain Res. 2013, 228, 481–491. [Google Scholar] [CrossRef] [PubMed]
  107. Hagsäter, S.M.; Pettersson, R.; Pettersson, C.; Atanasovski, D.; Näslund, J.; Eriksson, E. A Complex Impact of Systemically Administered 5-HT2A Receptor Ligands on Conditioned Fear. Int. J. Neuropsychopharmacol. 2021, 24, 749–757. [Google Scholar] [CrossRef] [PubMed]
  108. Jefsen, O.; Højgaard, K.; Christiansen, S.L.; Elfving, B.; Nutt, D.J.; Wegener, G.; Müller, H.K. Psilocybin lacks antidepressant-like effect in the Flinders Sensitive Line rat. Acta Neuropsychiatry 2019, 31, 213–219. [Google Scholar] [CrossRef]
  109. Shao, L.X.; Liao, C.; Gregg, I.; Davoudian, P.A.; Savalia, N.K.; Delagarza, K.; Kwan, A.C. Psilocybin induces rapid and persistent growth of dendritic spines in frontal cortex in vivo. Neuron 2021, 109, 2535–2544. [Google Scholar] [CrossRef]
  110. Hesselgrave, N.; Troppoli, T.A.; Wulff, A.B.; Cole, A.B.; Thompson, S.M. Harnessing psilocybin: Antidepressant-like behavioral and synaptic actions of psilocybin are independent of 5-HT2R activation in mice. Proc. Natl. Acad. Sci. USA 2021, 118, e2022489118. [Google Scholar] [CrossRef]
  111. Gable, R.S. Comparison of acute lethal toxicity of commonly abused psychoactive substances. Addiction 2004, 99, 686–696. [Google Scholar] [CrossRef]
  112. Morgan, C.J.; Noronha, L.A.; Muetzelfeldt, M.; Feilding, A.; Curran, H.V. Harms and benefits associated with psychoactive drugs: Findings of an international survey of active drug users. J. Psychopharmacol. 2013, 27, 497–506. [Google Scholar] [CrossRef] [PubMed]
  113. Nutt, D.; King, L.A.; Saulsbury, W.; Blakemore, C. Development of a rational scale to assess the harm of drugs of potential misuse. Lancet 2007, 369, 1047–1053. [Google Scholar] [CrossRef] [PubMed]
  114. Hasler, F.; Grimberg, U.; Benz, M.A.; Huber, T.; Vollenweider, F.X. Acute psychological and physiological effects of psilocybin in healthy humans: A double-blind, placebo-controlled dose-effect study. Psychopharmacology 2004, 172, 145–156. [Google Scholar] [CrossRef] [PubMed]
  115. Hollister, L.E. Clinical, biochemical and psychologic effects of psilocybin. Arch. Int. Pharmacodyn. Ther. 1961, 130, 42–52. [Google Scholar] [PubMed]
  116. Dittrich, A. The standardized psychometric assessment of altered states of consciousness (ASCs) in humans. Pharmacopsychiatry 1998, 31 (Suppl. 2), 80–84. [Google Scholar] [CrossRef] [PubMed]
  117. Studerus, E.; Kometer, M.; Hasler, F.; Vollenweider, F.X. Acute, subacute and long-term subjective effects of psilocybin in healthy humans: A pooled analysis of experimental studies. J. Psychopharmacol. 2011, 25, 1434–1452. [Google Scholar] [CrossRef] [PubMed]
  118. Vollenweider, F.X.; Geyer, M.A. A systems model of altered consciousness: Integrating natural and drug-induced psychoses. Brain Res. Bull. 2001, 56, 495–507. [Google Scholar] [CrossRef]
  119. Savalia, N.K.; Shao, L.X.; Kwan, A.C. A Dendrite-Focused Framework for Understanding the Actions of Ketamine and Psychedelics. Trends Neurosci. 2021, 44, 260–275. [Google Scholar] [CrossRef]
  120. Borsellino, P.; Krider, R.I.; Chea, D.; Grinnell, R.; Vida, T.A. Ketamine and the Disinhibition Hypothesis: Neurotrophic Factor-Mediated Treatment of Depression. Pharmaceuticals 2023, 16, 742. [Google Scholar] [CrossRef]
  121. De Gregorio, D.; Enns, J.P.; Nuñez, N.A.; Posa, L.; Gobbi, G. d-Lysergic acid diethylamide, psilocybin, and other classic hallucinogens: Mechanism of action and potential therapeutic applications in mood disorders. Prog. Brain Res. 2018, 242, 69–96. [Google Scholar] [CrossRef]
  122. Duman, R.S.; Aghajanian, G.K.; Sanacora, G.; Krystal, J.H. Synaptic plasticity and depression: New insights from stress and rapid-acting antidepressants. Nat. Med. 2016, 22, 238–249. [Google Scholar] [CrossRef] [PubMed]
  123. Ly, C.; Greb, A.C.; Cameron, L.P.; Wong, J.M.; Barragan, E.V.; Wilson, P.C.; Burbach, K.F.; Soltanzadeh Zarandi, S.; Sood, A.; Paddy, M.R.; et al. Psychedelics Promote Structural and Functional Neural Plasticity. Cell Rep. 2018, 23, 3170–3182. [Google Scholar] [CrossRef] [PubMed]
  124. 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. [Google Scholar] [CrossRef] [PubMed]
  125. Amargós-Bosch, M.; Bortolozzi, A.; Puig, M.V.; Serrats, J.; Adel, A.; Celada, P.; Totch, 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. [Google Scholar] [CrossRef] [PubMed]
  126. Santana, N. Expression of serotonin1A and serotonin2A receptors in pyramidal and GABAergic neurons of the rat prefrontal cortex. Cereb. Cortex 2004, 14, 1100–1109. [Google Scholar] [CrossRef]
  127. Sheldon, P.W.; Aghajanian, G.K. Excitatory responses to serotonin (5-HT) in neurons of the rat piriform cortex: Evidence for mediation by 5-HT1C receptors in pyramidal cells and 5-HT2 receptors in interneurons. Synapse 1991, 9, 208–218. [Google Scholar] [CrossRef]
  128. Béïque, J.C.; Campbell, B.; Perring, P.; Hamblin, M.V.; Walker, P.; Mladenovic, L.; Andrade, R. Serotonergic regulation of membrane potential in developing rat prefrontal cortex: Coordinated expression of 5-hydroxytryptamine (5-HT)1A, 5-HT2A, and 5-HT7 receptors. J. Neurosci. 2004, 24, 4807–4817. [Google Scholar] [CrossRef]
  129. Carhart-Harris, R.L.; Nutt, D.J. Serotonin and brain function: A tale of two receptors. J. Psychopharmacol. 2017, 31, 1091–1120. [Google Scholar] [CrossRef]
  130. Carhart-Harris, R.L.; Erritzoe, D.; Williams, T.; Stone, J.M.; Reed, L.J.; Colasanti, A.; Tyacke, R.J.; Leech, R.; Malizia, A.L.; Murphy, K.; et al. Neural correlates of the psychedelic state as determined by fMRI studies with psilocybin. Proc. Natl. Acad. Sci. USA 2012, 109, 2138–2143. [Google Scholar] [CrossRef]
  131. Lebedev, A.V.; Lövdén, M.; Rosenthal, G.; Feilding, A.; Nutt, D.J.; Carhart-Harris, R.L. Finding the self by losing the self: Neural correlates of ego-dissolution under psilocybin. Hum. Brain Mapp. 2015, 36, 3137–3153. [Google Scholar] [CrossRef]
  132. Millan, M.J.; Gobert, A.; Lejeune, F.; Dekeyne, A.; Newman-Tancredi, A.; Pasteau, V.; Rivet, J.M.; Cussac, D. The novel melatonin agonist agomelatine (S20098) is an antagonist at 5-hydroxytryptamine2C receptors, blockade of which enhances the activity of frontocortical dopaminergic and adrenergic pathways. J. Pharmacol. Exp. Ther. 2003, 306, 954–964. [Google Scholar] [CrossRef] [PubMed]
  133. Frånberg, O.; Marcus, M.M.; Svensson, T.H. Involvement of 5-HT2A receptor and α2-adrenoceptor blockade in the asenapine-induced elevation of prefrontal cortical monoamine outflow. Synapse 2012, 66, 650–660. [Google Scholar] [CrossRef] [PubMed]
  134. Mason, N.L.; Kuypers, K.P.C.; Müller, F.; Reckweg, J.; Tse, D.H.Y.; Toennes, S.W.; Hutten, N.R.P.W.; Jansen, J.F.A.; Stiers, P.; Feilding, A.; et al. Me, myself, bye: Regional alterations in glutamate and the experience of ego dissolution with psilocybin. Neuropsychopharmacology 2020, 45, 2003–2011. [Google Scholar] [CrossRef] [PubMed]
  135. Zhou, F.M.; Hablitz, J.J. Activation of serotonin receptors modulates synaptic transmission in rat cerebral cortex. J. Neurophysiol. 1999, 82, 2989–2999. [Google Scholar] [CrossRef]
  136. Rodrigues, N.B.; McIntyre, R.S.; Lipsitz, O.; Cha, D.S.; Lee, Y.; Gill, H.; Majeed, A.; Phan, L.; Nasri, F.; Ho, R.; et al. Changes in symptoms of anhedonia in adults with major depressive or bipolar disorder receiving IV ketamine: Results from the Canadian Rapid Treatment Center of Excellence. J. Affect. Disord. 2020, 276, 570–575. [Google Scholar] [CrossRef] [PubMed]
  137. Pandya, M.; Altinay, M.; Malone, D.A., Jr.; Anand, A. Where in the brain is depression? Curr. Psychiatry Rep. 2012, 14, 634–642. [Google Scholar] [CrossRef] [PubMed]
  138. Sheline, Y.I.; Gado, M.H.; Price, J.L. Amygdala core nuclei volumes are decreased in recurrent major depression. Neuroreport 1998, 9, 2023–2028. [Google Scholar] [CrossRef]
  139. Abdallah, C.G.; Jackowski, A.; Salas, R.; Gupta, S.; Sato, J.R.; Mao, X.; Coplan, J.D.; Shungu, D.C.; Mathew, S.J. The Nucleus Accumbens and Ketamine Treatment in Major Depressive Disorder. Neuropsychopharmacology 2017, 42, 1739–1746. [Google Scholar] [CrossRef]
  140. Zhou, Y.L.; Wu, F.C.; Wang, C.Y.; Zheng, W.; Lan, X.F.; Deng, X.R.; Ning, Y.P. Relationship between hippocampal volume and inflammatory markers following six infusions of ketamine in major depressive disorder. J. Affect. Disord. 2020, 276, 608–615. [Google Scholar] [CrossRef]
  141. Wojtas, A.; Bysiek, A.; Wawrzczak-Bargiela, A.; Maćkowiak, M.; Gołembiowska, K. Limbic system response to psilocybin and ketamine administration in rats: A neurochemical and behavioral study. Int. J. Mol. Sci. 2024, 25, 100. [Google Scholar] [CrossRef]
  142. Guan, X.M.; McBride, W.J. Serotonin microinfusion into the ventral tegmental area increases accumbens dopamine release. Brain Res. Bull. 1989, 23, 541–547. [Google Scholar] [CrossRef] [PubMed]
  143. Parsons, L.H.; Justice, J.B., Jr. Serotonin and dopamine sensitization in the nucleus accumbens, ventral tegmental area, and dorsal raphe nucleus following repeated cocaine administration. J. Neurochem. 1993, 61, 1611–1619. [Google Scholar] [CrossRef] [PubMed]
  144. De Deurwaerdere, P.; Di Giovanni, G. Serotonergic modulation of the activity of mesencephalic dopaminergic systems: Therapeutic implications. Prog. Neurobiol. 2017, 151, 175–236. [Google Scholar] [CrossRef] [PubMed]
  145. Celada, P.; Puig, M.V.; Artigas, F. Serotonin modulation of cortical neurons and networks. Front. Integr. Neurosci. 2013, 7, 25. [Google Scholar] [CrossRef] [PubMed]
  146. Cornea-Hébert, V.; Riad, M.; Wu, C.; Singh, S.K.; Descarries, L. Cellular and subcellular distribution of the serotonin 5-HT2A receptor in the central nervous system of adult rat. J. Comp. Neurol. 1999, 409, 187–209. [Google Scholar] [CrossRef]
  147. Izumi, J.; Washizuka, M.; Miura, N.; Hiraga, Y.; Ikeda, Y. Hippocampal serotonin 5-HT1A receptor enhances acetylcholine release in conscious rats. J. Neurochem. 1994, 62, 1804–1808. [Google Scholar] [CrossRef]
  148. Dasari, S.; Gulledge, A.T. M1 and M4 receptors modulate hippocampal pyramidal neurons. J. Neurophysiol. 2011, 105, 779–792. [Google Scholar] [CrossRef]
  149. Bombardi, C.; Di Giovanni, G. Functional anatomy of 5-HT2A receptors in the amygdala and hippocampal complex: Relevance to memory functions. Exp. Brain Res. 2013, 230, 427–439. [Google Scholar] [CrossRef]
  150. Pompeiano, M.; Palacios, J.M.; Mengod, G. Distribution of the serotonin 5-HT2 receptor family mRNAs: Comparison between 5-HT2A and 5-HT2C receptors. Brain Res. Mol. Brain Res. 1994, 23, 163–178. [Google Scholar] [CrossRef]
  151. Palchaudhuri, M.; Flügge, G. 5-HT1A receptor expression in pyramidal neurons of cortical and limbic brain regions. Cell Tissue Res. 2005, 321, 159–172. [Google Scholar] [CrossRef]
  152. Carhart-Harris, R.L.; Bolstridge, M.; Rucker, J.; Day, C.M.; Erritzoe, D.; Kaelen, M.; Bloomfield, M.; Rickard, J.A.; Forbes, B.; Feilding, A.; et al. Psilocybin with psychological support for treatment-resistant depression: An open-label feasibility study. Lancet Psychiatry 2016, 3, 619–627. [Google Scholar] [CrossRef] [PubMed]
  153. Carhart-Harris, R.L.; Bolstridge, M.; Day, C.M.J.; Rucker, J.; Watts, R.; Erritzoe, D.E.; Kaelen, M.; Giribaldi, B.; Bloomfield, M.; Pilling, S.; et al. Psilocybin with psychological support for treatment-resistant depression: Six-month follow-up. Psychopharmacology 2018, 235, 399–408. [Google Scholar] [CrossRef] [PubMed]
  154. Raison, C.L.; Sanacora, G.; Woolley, J.; Heinzerling, K.; Dunlop, B.W.; Brown, R.T.; Kakar, R.; Hassman, M.; Trivedi, R.P.; Robison, R.; et al. Single-dose psilocybin treatment for major depressive disorder. A randomized clinical trial. JAMA 2023, 330, 843–853. [Google Scholar] [CrossRef] [PubMed]
  155. Gukasyan, N.; Davis, A.K.; Barrett, F.S.; Cosimano, M.P.; Sepeda, N.D.; Johnson, M.W.; Griffiths, R.R. Efficacy and safety of psilocybin-assisted treatment for major depressive disorder: Prospective 12-month follow-up. J. Psychopharmacol. 2022, 36, 151–158. [Google Scholar] [CrossRef] [PubMed]
  156. Carhart-Harris, R.; Giribaldi, B.; Watts, R.; Baker-Jones, M.; Murphy-Beiner, A.; Murphy, R.; Martell, J.; Blemings, A.; Erritzoe, D.; Nutt, D.J. Trial of Psilocybin versus Escitalopram for Depression. N. Engl. J. Med. 2021, 384, 1402–1411. [Google Scholar] [CrossRef] [PubMed]
  157. Barba, T.; Buehler, S.; Kettner, H.; Radu, C.; Cunha, B.G.; Nutt, D.J.; Erritzoe, D.; Roseman, L.; Carhart-Harris, R. Effects of psilocybin versus escitalopram on rumination and thought suppression in depression. BJPsych Open 2022, 8, e163. [Google Scholar] [CrossRef] [PubMed]
  158. Bogenschutz, M.P.; Forcehimes, A.; Pommy, J.A.; Wilcox, C.E.; Barbosa, P.C.R.; Strassman, R.J. Psilocybin-assisted treatment for alcohol dependence: A proof-of-concept study. J. Psychopharmacol. 2015, 29, 289–299. [Google Scholar] [CrossRef]
  159. Johnson, M.W.; Garcia-Romeu, A.; Cosimano, M.P.; Griffiths, R.R. Pilot study of the 5-HT2AR agonist psilocybin in the treatment of tobacco addiction. J. Psychopharmacol. 2014, 28, 983–992. [Google Scholar] [CrossRef]
  160. Johnson, M.W.; Garcia-Romeu, A.; Griffiths, R.R. Long-term follow-up of psilocybin-facilitated smoking cessation. Am. J. Drug Alcohol Abus. 2017, 43, 55–60. [Google Scholar] [CrossRef]
  161. Garcia-Romeu, A.; Griffiths, R.R.; Johnson, M.W. Psilocybin-occasioned mystical experiences in the treatment of tobacco addiction. Curr. Drug Abus. Rev. 2014, 7, 157–164. [Google Scholar] [CrossRef]
  162. Foldi, C.; Liknaitzky, P.; Williams, M.; Oldfield, B.J. Rethinking therapeutic strategies for anorexia nervosa, insights from psychedelic medicine and animal models. Front. Neurosci. 2020, 14, 43. [Google Scholar] [CrossRef] [PubMed]
  163. Griffits, R.R.; Johnson, M.W.; Carducci, M.A.; Umbricht, A.; Richards, W.A.; Richards, B.D.; Cosimano, M.P.; Klinedinst, M.A. 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. [Google Scholar] [CrossRef] [PubMed]
  164. Ross, S.; Bossis, A.; Guss, J.; Agin-Liebes, G.; Malone, T.; Cohen, B.; Mennenga, S.E.; Belser, A.; Kalliontzi, K.; Babb, J.; et al. 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. [Google Scholar] [CrossRef] [PubMed]
  165. Grob, C.S.; Danforth, A.L.; Chopra, G.S.; Hagerty, M.; McKay, C.R.; Halberstadt, A.L.; Greer, G.R. Pilot study of psilocybin treatment for anxiety in patients with advanced-stage cancer. Arch. Gen. Psychiatry 2011, 68, 71–78. [Google Scholar] [CrossRef]
  166. Gasser, P.; Holstein, D.; Michel, Y.; Doblin, R.; Yazar-Klosinski, B.; Passie, T.; Brenneisen, R. Safety and efficacy of lysergic acid diethylamide-assisted psychotherapy for anxiety associated with life-threatening diseases. J. Nerv. Ment. Dis. 2014, 202, 513–520. [Google Scholar] [CrossRef]
  167. Moreno, F.A.; Wiegand, C.B.; Taitano, E.K.; Delgado, P.L. Safety, tolerability, and efficacy of psilocybin in 9 patients with obsessive-compulsive disorder. J. Clin. Psychiatry 2006, 67, 1735–1740. [Google Scholar] [CrossRef]
  168. Krediet, E.; Bostoen, T.; Breeksema, J.; Van Schagen, A.; Passie, T.; Vermetten, E. Reviewing the potential of psychedelics for treatment of PTSD. Int. J. Neuropsychopharmacol. 2020, 23, 385–400. [Google Scholar] [CrossRef]
  169. González-Maeso, J.; Sealfon, S.C. Psychedelics and schizophrenia. Trends Neurosci. 2009, 32, 225–232. [Google Scholar] [CrossRef]
  170. Celada, P.; Lladó-Pelfort, L.; Santana, N.; Kargieman, L.; Troyano-Rodriguez, E.; Riga, M.S.; Artigas, F. Disruption of thalamocortical activity in schizophrenia models: Relevance to antipsychotic drug action. Int. J. Neuropsychopharmacol. 2013, 16, 2145–2163. [Google Scholar] [CrossRef]
  171. Mahmood, D.; Alenzi, S.K.; Anwar, M.J.; Azam, F.; Qureshi, K.A.; Jaremko, M. New paradigms of old psychedelics in schizophrenia. Pharmaceuticals 2022, 15, 640. [Google Scholar] [CrossRef]
  172. Olson, D.E. The Subjective Effects of Psychedelics May Not Be Necessary for Their Enduring Therapeutic Effects. ACS Pharmacol. Transl. Sci. 2020, 4, 563–567. [Google Scholar] [CrossRef] [PubMed]
  173. Vargas, M.V.; Dunlap, L.E.; Dong, C.; Carter, S.J.; Tombari, R.J.; Jami, S.A.; Cameron, L.P.; Patel, S.D.; Hennessey, J.J.; Saeger, H.N.; et al. Psychedelics promote neuroplasticity through the activation of intracellular 5-HT2A receptors. Science 2023, 379, 700–706. [Google Scholar] [CrossRef] [PubMed]
Figure 1. The location of the 5-HT2A and 5HT2C receptors in the cortical neurons.
Figure 1. The location of the 5-HT2A and 5HT2C receptors in the cortical neurons.
Ijms 25 00241 g001
Figure 2. “Bipartite” model according to which 5-HT1A and 5-HT2A receptors play a crucial role in psilocybin-induced alterations in brain activity [129].
Figure 2. “Bipartite” model according to which 5-HT1A and 5-HT2A receptors play a crucial role in psilocybin-induced alterations in brain activity [129].
Ijms 25 00241 g002
Figure 3. Filtering of sensory input into the cortex by the thalamus via 5-HT2A receptors [118].
Figure 3. Filtering of sensory input into the cortex by the thalamus via 5-HT2A receptors [118].
Ijms 25 00241 g003
Table 1. Serotonin receptor interactions with some phenethylamine and indoleamine psychedelics (according to [20,21]).
Table 1. Serotonin receptor interactions with some phenethylamine and indoleamine psychedelics (according to [20,21]).
Compound/Receptor SubtypeKi ± SD (nM)
5-HT1A5-HT2A5-HT2B5-HT2C
Phenethylamines
Mescaline4600 ± 4006300 ± 1800>200017,000 ± 2000
25I-NBOMe1800 ± 3000.6 ± 0.2130 ± 804.6 ± 2
25B-NBOMe3600 ± 3000.5 ± 010 ± 106.2 ± 2.2
Indoleamines
Psilocin123 ± 2049 ± 10>20,00094 ± 9
DMT75 ± 20237 ± 4034 ± 32424 ± 150
LSD3 ± 0.54 ± 112,000 ± 40015 ± 3
Table 2. Abbreviations and chemical names of some NBOMe compounds [35].
Table 2. Abbreviations and chemical names of some NBOMe compounds [35].
AbbreviationChemical Name
25B-NBOMe2-(4-bromo-2,5-dimethoxyphenyl)-N-(2-methoxybenzyl)ethanamine
25C-NBOMe2-(4-chloro-2,5-dimethoxyphenyl)-N-(2-methoxybenzyl)ethanamine
25D-NBOMe2-(2,5-dimethoxy-4-methylphenyl)-N-(2-methoxybenzyl)ethanamine
25I-NBOMe2-(4-iodo-2,5-dimethoxyphenyl)-N-(2-methoxybenzyl)ethanamine
25N-NBOMe2-(2,5-dimethoxy-4-nitrophenyl)-N-(2-methoxybenzyl)ethanamine
Disclaimer/Publisher’s Note: The statements, opinions and data contained in all publications are solely those of the individual author(s) and contributor(s) and not of MDPI and/or the editor(s). MDPI and/or the editor(s) disclaim responsibility for any injury to people or property resulting from any ideas, methods, instructions or products referred to in the content.

Share and Cite

MDPI and ACS Style

Wojtas, A.; Gołembiowska, K. Molecular and Medical Aspects of Psychedelics. Int. J. Mol. Sci. 2024, 25, 241. https://doi.org/10.3390/ijms25010241

AMA Style

Wojtas A, Gołembiowska K. Molecular and Medical Aspects of Psychedelics. International Journal of Molecular Sciences. 2024; 25(1):241. https://doi.org/10.3390/ijms25010241

Chicago/Turabian Style

Wojtas, Adam, and Krystyna Gołembiowska. 2024. "Molecular and Medical Aspects of Psychedelics" International Journal of Molecular Sciences 25, no. 1: 241. https://doi.org/10.3390/ijms25010241

Note that from the first issue of 2016, this journal uses article numbers instead of page numbers. See further details here.

Article Metrics

Back to TopTop