Skip to main content
NIHPA Author Manuscripts logoLink to NIHPA Author Manuscripts
. Author manuscript; available in PMC: 2025 Feb 12.
Published in final edited form as: Annu Rev Physiol. 2023 Nov 6;86:27–47. doi: 10.1146/annurev-physiol-042022-020923

The effects of psychedelics on neuronal physiology

Cassandra J Hatzipantelis 1,2,3, David E Olson 1,2,3,4,*
PMCID: PMC10922499  NIHMSID: NIHMS1955211  PMID: 37931171

Abstract

Psychedelics are quite unique among drugs that impact the central nervous system, as a single administration of a psychedelic can both rapidly alter subjective experience in profound ways and produce sustained effects on circuits relevant to mood, fear, reward, and cognitive flexibility. These remarkable properties are a direct result of psychedelics interacting with several key neuroreceptors distributed across the brain. Stimulation of these receptors activates a variety of signaling cascades that ultimately culminate in changes in neuronal structure and function. Here, we describe the effects of psychedelics on neuronal physiology, highlighting their acute effects on serotonergic and glutamatergic neurotransmission as well as their long-lasting effects on structural and functional neuroplasticity in the cortex. We propose that the neurobiological changes leading to the acute and sustained effects of psychedelics might be distinct, which could provide opportunities for engineering compounds with optimized safety and efficacy profiles.

Keywords: psychedelic, hallucinogen, psilocybin, LSD, DMT, 5-HT2A receptor, neuroplasticity

INTRODUCTION

Classic serotonergic psychedelics are defined as psychoactive small molecules that produce acute effects on perception, mood, and cognition by activating serotonin 2A receptors (5-HT2ARs) (1). Psychedelic drugs have long been used by humans recreationally, therapeutically, and as part of cultural and spiritual traditions (2). This class of compounds includes molecules from tryptamine, phenethylamine, and ergoline chemical families such as psilocin, 2,5-dimethoxy-4-iodoamphetamince (DOI), and lysergic acid diethylamide (LSD) (3) (Figure 1A). While the acute effects of psychedelics on subjective experience are well known, these substances also produce long-lasting changes in neural circuits relevant to depression and other neuropsychiatric diseases (4). Recent clinical trials have revealed that psychedelics may possess both rapid and sustained therapeutic effects for patients with mood, anxiety, and substance-use disorders (59).

Figure 1.

Figure 1.

Polypharmacology of psychedelics. (a) Structures of several psychedelic drugs with tryptamine, phenethylamine, and ergoline frameworks highlighted in blue, red, and purple, respectively. Panel adapted with permission from Reference 3; copyright © 2022, Springer Nature America, Inc. (b) Radioligand binding profiles for prototypical members of the phenethylamine (DOI), tryptamine (DMT), and ergoline (LSD) families. Red boxes indicate Ki values <1 μM, pink boxes indicate Ki values 1–10 μM, and white boxes indicate Ki values >10 μM using either agonist or antagonist radioligands. Dark gray boxes indicate data that are not available. Data obtained from the Psychoactive Drug Screening Program Ki Database with the exception of TAAR1 data. Data for TAAR1 were taken from Reference 59, with red boxes indicating >50% activation of rat TAAR1 following a 1 μM treatment, (c) Radioligand binding to 5-HT2A receptors across the human brain. Panel adapted with permission from Reference 27. (d) Expression of 5-HT2A receptors in the mouse brain. Panel adapted with permission from Reference 25; copyright © 2010 Weber and Andrade. LSD, lysergic acid diethylamide; DMT, N,N-Dimethyltryptamine; DOI, 2,5-dimethoxy-4-iodoamphetamince; SERT, serotonin transporter; TAAR1, trace amine-associated receptor 1; AC, anterior cingulate cortex; Aud, auditory cortex; Ctx, cortex; Ent, entorhinal cortex; Hipp, hippocampus; Ins, insular cortex; M1, primary motor cortex; Orb, orbital cortex; PL, prelimbic cortex; Rh, rhinal cortex; S1, primary somatosensory cortex; S2, secondary somatosensory cortex; Str, striatum.

Because of their intense mind-altering effects and promising therapeutic potential, there has been a dramatic resurgence of research into the neuropsychopharmacological effects of psychedelics. Currently, it is unclear if psychological or neurobiological effects mediate the therapeutic properties of psychedelics (for an overview of the current debate with examples, see the following viewpoints; 10, 11).However, it is clear that psychedelics have profound effects on neuronal physiology due to their activation of specific receptors located within various circuits across the brain. These molecular-, cellular-, and circuit-level effects may directly contribute to the therapeutic properties of psychedelics, and at the very least, result in the profound subjective experiences characteristic of this class of compounds. Here, we review preclinical literature demonstrating that psychedelics regulate major neuronal physiological processes such as neurotransmission, neuroplasticity, and cell survival. By integrating what is known about psychedelic drug action at the molecular-, cellular-, and circuit- levels, we aim to stimulate new hypotheses about how these compounds may modulate key neuronal functions both acutely and in the long-term.

THE PHARMACOLOGY OF PYSCHEDELICS

The three major structural classes of psychedelics each possess unique polypharmacological profiles (Figure 1B). Ergolines are the least selective class of psychedelics with high affinities for serotonin, dopamine, histamine, adrenergic, and sigma receptors. In general, tryptamines have high affinity for most serotonin receptors, while phenethylamines tend to be more selective for the 5-HT2 family. A common feature of all classic psychedelics is that they potently activate 5-HT2Rs. Rodent drug discrimination studies have revealed a strong correlation with 5-HT2R binding affinity (12), and administration of the 5-HT2R antagonist ketanserin has been shown to block subjective, cognitive, and mood-related effects of psychedelics in humans (1319).

Of the three 5-HT2R subtypes, the 5-HT2AR has been found to be critically important for many of the effects induced by psychedelics. Drug discrimination studies revealed that the affinities of 5-HT2R antagonists for 5-HT2ARs, rather than 5-HT2CRs, correlated better with their abilities to block the effects of LSD (20). Moreover, occupancy of 5-HT2ARs correlates well with the intensity of subjective experiences induced by psychedelics (21). Human hallucinogenic potencies correlate exceptionally well with potencies in the mouse head-twitch response (HTR) assay, and 5-HT2AR knockout (KO) (22), but not 5-HT2C KO (23), completely abrogates psychedelic-induced HTR.

Some evidence suggests that cortical 5-HT2ARs play a key role in the HTR (22), but given that 5-HT2ARs are expressed widely throughout the brain, it has been challenging to determine which brain regions are critical for the many effects of psychedelics. A variety of studies in both humans and rodents have established particularly high densities of 5-HT2ARs in layer 5 of the cortex and in the claustrum (2427) (Figure 1C and D), however key differences between species do exist. In humans, expression of 5-HT2ARs is higher in the prefrontal and visual cortices rather than the motor and somatosensory cortices (Figure 1C), while rodents exhibit a more pronounced anterior to posterior expression gradient (Figure 1D).

The 5-HT2AR is a G protein coupled receptor (GPCR) that typically activates Gq signaling which leads to a cascade of events including activation of phospholipase C, hydrolysis of phosphatidylinositol-4,5-bisphosphate into inositol trisphosphate and diacylglycerol, and release of Ca2+ stores from the endoplasmic reticulum, ultimately activating CAMKII, protein kinase C, and MAPK pathways. Psychedelics have been primarily shown to activate these pathways in non-neuronal cells heterologously expressing 5-HT2ARs (2830), though these pathways are operative in neurons as well (31). Psychedelics increase inositol monophosphate accumulation in cortical cultures (32) and unpublished work suggests that psilocin can increase the firing rates of 5-HT2AR-expressing cortical neurons in a Gq-dependent manner (33). However, activation of 5-HT2ARs and canonical downstream signaling does not necessarily lead to hallucinogenic effects, as several non-hallucinogenic agonists of the 5-HT2AR have been discovered including lisuride, 6-fluoro-N,N-diethyltryptamine, tabernanthalog, ariadne, and IHCH-7079, among others (3441). Structural biology studies suggest that hallucinogenic and non-hallucinogenic agonists may induce distinct confirmations of the 5-HT2AR, leading to their disparate effects (35). Moreover, a novel biosensor that couples 5-HT2AR conformation to a fluorescence signal is capable of differentiating between hallucinogenic and non-hallucinogenic ligands (42).

Currently, very little is known about how biased agonism at the 5-HT2AR might impact the effects of psychedelics. Psychedelic-related behaviors induced by LSD are dependent on β-arrestin 2 (43), while those induced by DOI are not (44). Moreover, DOI can still produce a head-twitch response in Gq KO mice (45). Other forms of functional selectivity might be operative including location bias (32), and the activation of distinct heterodimeric complexes. The 5-HT2AR is known to form complexes with a variety of GPCRs that may alter is function, including metabotropic glutamate, dopamine, cannabinoid, and serotonin receptors (4649). Importantly, psychedelics have been shown to impact neuronal function in a manner reliant on such heterodimers (5052).

While the evidence for the 5-HT2AR in the subjective effects of psychedelics is substantial, to the best of our knowledge, no psychedelic has been described that completely lacks 5-HT2BR or 5-HT2CR agonism, leaving open the question of how these receptors might contribute to the various effects of psychedelics. A role for 5-HT2BRs in the psychoactive effects of psychedelics has been largely dismissed owing to low 5-HT2BR expression in the central nervous system compared to 5-HT2ARs and 5-HT2CRs (53). However, 5-HT2BRs have been shown to play a key role in compound-induced release of serotonin (54), and thus, have the potential to modulate the effects of psychedelics. Expression of 5-HT2CRs is much higher in the central nervous system (53), and they are likely to play major roles in the actions of psychedelics. Moreover, 5-HT2CRs are post-transcriptionally edited leading to several functionally distinct isoforms found in various brain regions (55, 56).

Outside of the 5-HT2 family of receptors, psychedelics target a wide variety of other neuroreceptors including 5-HT1Rs, trace amine-associated receptors (TAARs), and sigma-1 receptors (Figure 1B), among others. Activation of 5-HT1A autoreceptors on neurons of the raphe nucleus by LSD decreases their firing rate (57). Furthermore, 5-HT1BR and 5-HT1DR agonism could explain the propensity for psychedelics to induce vasoconstriction given that 5-HT1B and 5-HT1D receptors are known targets for the antimigraine triptan drugs (58), and triptans are structurally related to tryptamine-based psychedelics like DMT. Some psychedelics, like DMT, act as agonists at TAARs (59) and sigma-1 receptors (60). While DMT has been hypothesized to be an endogenous ligand for the sigma-1 receptor, it’s binding affinity for that target (>1 μM) is unremarkable (60). Recent work suggests that psychedelics may directly bind to the receptor tyrosine kinase TrkB, serving as positive allosteric modulators of brain-derived neurotrophic factor (BDNF) signaling(61).

EFFECTS OF PSYCHEDELICS ON GENE AND PROTEIN EXPRESSION

Gene microarray experiments have demonstrated that a single dose of LSD only regulates a very small number of genes in the brain, but many of them have been implicated in plasticity processes (62). Indeed, many psychedelics have relatively modest effects on gene expression despite their long-lasting effects on neuronal functions. Psychedelics have consistently been shown to increase the expression of immediate early genes like c-Fos, arc, egr-1, and egr-2, and these effects are absent in 5-HT2AR KO mice or following pretreatment with a 5-HT2R antagonist (22, 6368). Recent whole-brain mapping of c-Fos following an acute dose of psilocybin showed dramatic increases in c-Fos induction in the prefrontal cortex (PFC), claustrum, amygdala, thalamus, lateral habenula, and hippocampus, with decreased c-Fos in the raphe nuclei (69). The degree of c-Fos induction was best correlated with mRNA transcript levels of N-methyl-D-aspartate (NMDA) receptor subunits and the 5-HT2AR. Interestingly, brain-wide expression of c-Fos following psilocybin administration shared many similarities to that of the dissociative anesthetic ketamine.

Both hallucinogenic and non-hallucinogenic agonists of the 5-HT2AR can increase expression of various immediate early genes in the somatosensory cortex in a 5-HT2AR-dependent manner, albeit with unique transcriptional fingerprints (63). Unlike LSD, lisuride does not upregulate egr-1 or egr-2 expression (22). Both compounds increase c-Fos expression, but the magnitude of response appears to be greater for LSD (22). While LSD induces c-Fos in the PFC, hippocampus, and midbrain of rats, it only increases arc expression in the PFC (62), suggesting that particular immediate early gene-related pathways might be circuit-specific.

While psychedelic-induced c-Fos expression is 5-HT2AR-dependent (22, 64, 68), upregulation of this immediate early gene occurs in cells not expressing 5-HT2ARs (65). One mechanistic explanation is that BDNF or glutamate release may be critical mediators of psychedelic-induced changes in cortical microcircuit activity. Indeed, DOI dose-dependently increases cortical bdnf mRNA while decreasing hippocampal bdnf mRNA in a 5-HT2AR-dependent manner (70), and increases in cortical arc mRNA levels following DOI administration are abolished with the inducible KO of BDNF (71). Induction of bdnf expression in the cortex and claustrum is also blocked by mGlu2/3 receptor agonism and potentiated in the mPFC by mGlu2/3 receptor antagonism (72). Furthermore, a mGlu2/3 receptor agonist and an AMPA receptor antagonist abrogated DOI-induced c-Fos expression in the mPFC (73, 74), implicating glutamate signaling in psychedelic-induced IEG expression. Interestingly, different signal transduction pathways downstream of 5-HT2AR activation might lead to distinct gene expression responses. In cultured cortical neurons, arc, bdnf and egr-2 were upregulated following DOI administration in both a MAPK- and CAMKII-dependent manner, whereas DOI-induced upregulation of c-Fos and egr-1 were specifically dependent on CAMKII and MAPK signaling, respectively (75).

In addition to IEGs and bdnf, psychedelics also increase the expression of key pre- and postsynaptic proteins. In pigs, a single dose of psilocybin led to long-lasting increases in the presynaptic protein SV2A in both the PFC and hippocampus (76). In the PFC of rodents, psilocybin also had a marked effect on postsynaptic structures, increasing the expression of Psd95 (77) and dose-dependently increasing the synthesis of NMDA receptor subunits (78). Moreover, proteomic analysis of human cerebral organoids following treatment with 5-MeO-DMT or LSD revealed increased expression of proteins involved in structural and functional neuroplasticity (79, 80).

Psychedelics can also produce long-lasting changes in the neuronal epigenome that may explain the enduring effects of psychedelics. A single systemic administration of DOI increased the enhancer level of genes associated with structural and functional neuroplasticity persisting at least 7 days post-administration (81). Repeated (7-day) administration of LSD significantly increased the methylation level of CpG sites of genes related to neuronal development, growth and morphology, as well as neuronal death and survival mechanisms (82). In the same study, LSD primarily upregulated the synthesis of proteins involved in synaptic plasticity and microtubule polymerization (essential for neuronal structure), while downregulating the synthesis of proteins that regulate receptor-mediated endocytosis.

EFFECTS OF PSYCHEDELICS ON STRUCTURAL NEUROPLASTICITY

Psychedelic drugs have recently been classified as psychoplastogens—small molecules that produce therapeutic effects by rapidly promoting structural and functional neuroplasticity (83). In cortical cultures, psychedelics have been shown to increase the size of dendritic growth cones, promote the growth of new neurites, increase dendritic spine density, and alter spine morphology (8489) (Figure 2A). Moreover, only short stimulation periods are required to induce long-lasting changes in cortical neuron growth (90). Psychedelic-induced spine growth is likely associated with synaptogenesis given that colocalization of pre- and postsynaptic markers is also increased following treatment (84).

Figure 2.

Figure 2.

Psychedelics promote structural neuroplasticity. (a) DOI, DMT and LSD promote dendritogenesis in vitro. Dendrite tracings and Sholl plots are shown. Panel adapted from Reference 84, reprinted with permission from Elsevier, (b) 5-MeO-DMT increases dendritic spine density in the prefrontal cortex of wild type, but not 5-HT2AR knockout mice. Panel adapted with permission from Reference 32; copyright © 2023, The American Association for the Advancement of Science, (c) Psilocybin produces long-lasting changes in dendritic spine density in vivo. Panel adapted with permission from Reference 93; copyright © 2021, Alex C. Kwan. (d) DOI, and the non-hallucinogenic 5-HT2AR agonist TBG, increase dendritic spine formation in the primary sensory cortex as measured by 2-photon imaging in vivo. Blue and red arrowheads indicate newly formed and eliminated spines, respectively. White arrowheads indicate filopodia. Panel adapted with permission from Reference 34; copyright © 2020, David E. Olson, under exclusive license to Springer Nature Limited.

The first in vivo evidence for psychedelic-induced neuroplasticity was obtained from Golgi-Cox staining 24 h after a single systemic administration of DMT to rats, where treatment with DMT increased spine density in the PFC to a comparable extent as ketamine (84). Subsequently, a single dose DOI and 5-MeO-DMT were shown to increase dendritic spine density in the PFC of mice 24 h after administration (Figure 2B) (32, 81). Similar results following DOI treatment were observed in the somatosensory cortex using two-photon in vivo imaging (34). Repeated (7-day) systemic administration of LSD increased dendritic spine density in the PFC of wild type mice as well as animals exposed to 15 days of chronic restraint stress (91). However, chronic intermittent dosing of DMT over several weeks resulted in spine retraction in female, but not male rats (92), suggesting that overstimulation of cortical neuron growth pathways may lead to homeostatic changes curtailing growth.

Longitudinal in vivo imaging of cortical neurons following a single dose of psilocybin revealed significant increases in dendritic spine density in the cortex that persisted for at least a month (Figure 2C) (93). Interestingly, the magnitude of the effect was greater in female mice. Similar long-lasting effects have been observed following a single dose of 5-MeO-DMT (94). Two-photon imaging studies using psilocybin, DOI, and 5-MeO-DMT have all demonstrated that psychedelics increase the rate of spine formation but have little to no effect on the rate of spine elimination (34, 93, 94).

The effects of psychedelics on structural plasticity are dependent on 5-HT2AR signaling, as 5-HT2R antagonists block psychedelic-induced increases in dendritic branching and dendritic spine density in culture (84, 88). In vivo, 5-MeO-DMT and DOI promote robust spine growth in wild type mice, but these effects are not observed in 5-HT2AR KO animals (32, 81). While 5-HT2ARs exist on the plasma membrane, a large population is localized to intracellular compartments within cortical neurons. This intracellular pool plays a critical role in psychedelic-induced neuroplasticity, and location bias can potentially explain why polar, membrane-impermeable 5-HT2AR agonists like serotonin cannot promote cortical neuron growth (32).

While the mechanism of psychedelic-induced neuroplasticity has not been fully elucidated, several clues are beginning to emerge (95). In addition to 5-HT2AR activation, the psychoplastogenic effects of psychedelics also require TrkB, mammalian target of rapamycin (mTOR), and α-amino-3-hydroxy-5-methyl-4-isoxazolepropionic acid (AMPA) receptor signaling (84, 90). Proteomics experiments demonstrated that LSD treatment upregulated the mTOR pathway in human brain organoids (80), and mTOR activation in excitatory pyramidal cells of the cortex is essential for repeated (7-day) LSD administration to produce prosocial effects (96). Given that mTOR is the downstream effector of the canonical signaling pathway initiated by TrkB activation, these data again implicate BDNF-TrkB signaling in the effect of psychedelics on structural neuroplasticity. Intriguingly, a recent report found that LSD and psilocin might serve as positive allosteric modulators of BDNF signaling by directly binding to TrkB (61).

While the psychoplastogenic effects of psychedelics require 5-HT2AR activation, they do not appear to be directly coupled to hallucinations. Evidence continues to mount suggesting that non-hallucinogenic agonists of the 5-HT2AR may have therapeutic potential (3436, 39, 40, 97). Some of these compounds have been shown to promote cortical neuron growth in a 5-HT2AR-dependent manner (34, 36, 38). The non-hallucinogenic 5-HT2AR agonist tabernanthalog was shown to increase neuritogenesis and dendritic spine formation in cultured cortical neurons, and two-photon in vivo imaging revealed that its psychoplastogenic properties were comparable to that of the hallucinogen DOI (34) (Figure 2D). Follow-up studies revealed that a single dose of tabernanthalog rescued cortical pyramidal neuron spine loss following unpredictable mild stress. Over 30% of TBG-induced spines were formed near sites where stress had induced spine loss, and ~30% of tabernanthalog-induced spines survived for 12 days (98). The effects of tabernanthalog on spine growth are in sharp contrast to those of the traditional antidepressant fluoxetine, as a single dose of fluoxetine did not produce any measurable changes in spine density and did not rescue stress-induced behavioral deficits (98). Like tabernanthalog, the non-hallucinogenic 5-HT2AR agonist lisuride has been shown to promote dendritic spine growth following an insult that resulted in spine retraction (97). As cortical atrophy is a hallmark of many neuropsychiatric diseases (99), the long-term effects of psychedelics and non-hallucinogenic psychoplastogens on cortical neuron growth could have important implications for the treatment of numerous conditions.

EFFECTS OF PSYCHEDELICS ON NEURONAL SURVIVAL AND NEUROGENESIS

Psychedelics have been consistently shown to regulate the expression and synthesis of cell survival-related genes and proteins in the cortex, including serum glucocorticoid kinase, Iκβ-α, and NFκβ (62, 77, 100). While DOI significantly increased Iκβ-α mRNA in the somatosensory cortex in a 5-HT2A-dependent manner (63), LSD-induced changes in serum glucocorticoid kinase and Iκβ-α expression were unaffected by 5-HT2A receptor antagonism (100); suggesting that different psychedelics may regulate cell survival via distinct mechanisms.

Prolonged (6-day) exposure to DOI prevented kainate- and H2O2-induced excitotoxic and oxidative insults, respectively, in cultured cortical neurons by promoting mitochondrial biogenesis and reducing ROS levels (101). Interestingly, serotonin and DOI shared a common mechanism for driving neuroprotective effects, involving 5-HT2A receptor-dependent activation of sirtuin 1, a master regulator of mitochondrial function. These data imply a physiological role of 5-HT2AR activation in regulating neuronal mitochondria-driven bioenergetics and offer a potential mechanism by which some psychedelics can promote cell survival.

Importantly, DMT can bind to the sigma-1 receptor (60), a receptor localized to the endoplasmic reticulum-mitochondria interface where it ensures delivery of IP3-dependent Ca2+ ions released from intracellular stores to mitochondria to regulate cell survival processes (102). The sigma-1 receptor is also known to functionally modulate BDNF expression and secretion (102), and transactivate TrkB receptors (103, 104). BDNF promotes the survival of neurons produced via neurogenesis (105), and BDNF-TrkB-Akt signaling is an established neuroprotective and pro-survival signaling cascade (106). It is therefore possible that, at least some psychedelics regulate cell survival mechanisms via sigma-1 receptor activation. Indeed, DMT prevented human iPSC-derived cortical neuronal cell death in response to severe hypoxic stress and promoted cell survival following focal ischemia in mice, in a manner dependent on sigma-1 receptor expression (107, 108).

With respect to neurogenesis, the effects of psychedelics have been mixed. Intracerebroventricular administration of 5-Meo-DMT induced neurogenesis in the dentate gyrus just 15 min after injection, and these newborn neurons exhibited increased dendritic arbor complexity when compared to those not stimulated by 5-MeO-DMT (109). Conversely, acute administration of DOI and LSD had no effect on hippocampal neurogenesis (110), while psilocybin and 25I-NBOMe decreased hippocampal neurogenesis (111). Interestingly, 5-HT2R blockade by ketanserin decreased hippocampal progenitor cell turnover following acute treatment, while chronic treatment increases the turnover (110). Repeated (4-day) systemic injection of 5-Meo-DMT induced hippocampal neurogenesis that was blocked by a sigma-1 receptor antagonists, but not by 5-HT2AR or 5-HT1AR antagonists (112).

Clearly sigma-1 and 5-HT2A receptors play important roles in the effects of psychedelics on neuronal survival and neurogenesis, however whether these mechanisms are mutually exclusive or inherently linked to each other is unknown. 5-HT2AR-mediated Gq signaling may play a role in Ca2+ release from intracellular stores that the sigma-1 receptor then funnels to mitochondria to regulate bioenergetics and facilitate cell survival mechanisms, for example.

EFFECTS OF PSYCHEDELICS ON NEURONAL ACTIVITY

Psychedelics have profound acute and long-lasting functional effects on a variety of neuronal populations. Stimulation of rat cortical slices with DOI was shown to enhance the occurrence of late excitatory postsynaptic currents (EPSCs) in layer 5 pyramidal neurons in a 5-HT2A-dependent manner (113), an effect that was blocked by NR2B-selective antagonists (114). Furthermore, DOI increased the amplitude of evoked excitatory postsynaptic potentials (EPSPs) in layer 5 pyramidal neurons (115). Bath application of DOI also increases the frequency of spontaneous EPSPs in layer 5 pyramidal neurons, and this effect is suppressed by mGlu2/3 agonism (116). Enhanced excitatory neurotransmission in cortical slices has also been observed following treatment with DOB and was blocked with 5-HT2R antagonists (117). In vivo, 5-MeO-DMT increases cortical pyramidal neuron firing rate and burst activity following systemic administration (118).

In addition to excitatory neurons in the cortex, psychedelics also acutely impact the function of inhibitory and monoaminergic neurons. The 5-HT2AR is expressed in both glutamatergic and γ-aminobutyric acid (GABA)-ergic neurons in the cortex (119, 120). In slices from the piriform cortex, both DOI and LSD increased the firing rate of interneurons, and the effect was blocked by a 5-HT2R antagonist (121). In the locus coeruleus of rats, mescaline, DOM, and LSD decreased and increased the spontaneous and evoked activity of neurons, respectively (122). These effects are blocked by 5-HT2R antagonists and appear to be mediated by locus coeruleus afferents as iontophoretic application onto cell bodies in the locus coeruleus did not produce the same effects.

In the raphe, psychedelics can dramatically suppress the firing of serotonergic neurons, but these effects are only observed following treatment with psychedelics that have substantial 5-HT1AR activity (123125). The 5-HT1AR might also play a role in the effects of LSD on the firing rate of neurons of the ventral tegmental area (VTA), though the mechanism is more complex. Dose-response studies demonstrated that LSD decreased the firing rate of dopaminergic neurons in the VTA via a mechanism involving activation of 5-HT1A, D2, and TAAR1 receptors (57).

A unique property of psychedelics is their ability to produce changes in neuronal function that last long after the compounds have been cleared from the body. Systemic administration of DMT to rats increased both the amplitude and frequency of spontaneous EPSCs in cortical pyramidal neurons measured by ex vivo electrophysiology 24 h after dosing (84). Similar results have been observed following administration of 5-MeO-DMT to wild type mice, and these changes in functional plasticity were absent in 5-HT2AR KO mice (32) (Figure 3A). Similarly, psilocybin increased the frequency of miniature EPSCs in layer 5 pyramidal neurons 24 h following systemic administration (93). In general, effects on EPSC frequency seem quite robust, while effects on EPSC amplitude are more modest (32, 84, 93).

Figure 3.

Figure 3.

Psychedelics impact neuronal activity, (a) 5-MeO-DMT increases cortical EPSCs in wild type, but not 5-HT2AR knockout mice. Panel adapted with permission from Reference 32; copyright © 2023, The American Association for the Advancement of Science. (b) Psilocybin increases hippocampal slice EPSPs. Panel adapted with permission from Reference 126; copyright © 2021, National Academy of Sciences. 5-MeO, 5-methoxy-N,N-dimethyltryptamine; APV, 2-Aminophosphonovaleric acid; DNQX, 6,7-dinitroquinoxaline-2,3-dione; EPSP, excitatory postsynaptic potentials; KO, knockout; sEPSC, spontaneous excitatory postsynaptic currents; VEH, vehicle.

In addition to their long-lasting effects on EPSCs, psychedelics have also been found to have additional effects on cortical neuron function. A single systemic administration of DOI significantly enhanced cortical long-term potentiation in slices collected 24 h post-drug administration (81), while repeated (7-day) LSD administration increased the burst activity of PFC pyramidal neurons, and potentiated the excitatory response of DOI and the AMPA receptor agonist, quisqualate (96). Importantly, the effects of repeated dosing of LSD were dependent on mTOR activation. Outside of the cortex, systemic administration of psilocybin to mice that had been exposed to chronic stress led to strengthening of intrahippocampal synapses measured 3 days later (126) (Figure 3B). Currently, it is unclear if hippocampal plasticity induced by psychedelics is a result of direct action on hippocampal cells or an indirect effect from hippocampal afferents.

EFFECTS OF PSYCHEDELICS ON NEUROTRANSMITTER / NEUROMODULATOR RELEASE

Microdialysis studies have indicated that a number of psychedelics increase glutamate release in the PFC via 5-HT2AR activation including LSD, DOM, DOI, 25I-NBOMe, and psilocybin (78, 127129). Moreover, glutamate release appears to be a direct effect of cortical neuron stimulation as intracortical administration of psychedelics reproduces the glutamate release observed following systemic administration (128). Changes in excitatory neurotransmission following psilocybin administration are reflected in enhanced cortical glucose metabolism as measured in humans using [18F]fluorodeoxyglucose positron emission tomography (130, 131). Psychedelic-induced neuroplasticity has been hypothesized to involve glutamatergic transmission in the cortex; however, it is unclear if a large glutamate burst is necessary to induce cortical structural plasticity, and it is unknown if non-hallucinogenic psychoplastogens increase glutamate levels in the cortex to a comparable extent as psychedelics (95).

Psilocybin and DOI may regulate cortical microcircuits by activating 5-HT2AR expressing GABAergic interneurons and increasing GABA release in the PFC (78, 132). Psilocybin appears to regulate long-range cortical-subcortical circuits, by rapidly increasing GABA release in the thalamus and increasing GABA levels in the PFC after 140 min (78). The delayed release of cortical GABA may indicate that psilocybin impacts plasticity mechanisms that then facilitate delayed changes in neuronal circuit activity. Furthermore, ayahuasca ingestion by rats increased 5-HT and GABA release in the hippocampus, and increased 5-HT, GABA, and noradrenaline release in the amygdala (133).

Systemic administration of psilocybin in rats rapidly increased dopamine release in the PFC (78), suggesting that psilocin regulates a known top-down neurocircuit involving 5-HT2AR-expressing pyramidal neurons that project to and activate mesocortical dopaminergic VTA neurons. Indeed, activation of 5-HT2ARs in the PFC by DOI increases mesocortical dopaminergic signaling (134), and 25I-NBOMe dose-dependently increases dopamine in the frontal cortex (129). DOI and psilocin also activate mesolimbic dopaminergic VTA neurons, increasing dopamine release in the nucleus accumbens (135, 136). While increased mesolimbic dopamine is typically associated with the addictive potential of drugs, LSD induces much higher c-Fos expression in the nucleus accumbens when compared to highly addictive drugs cocaine and morphine (137), suggesting that distinct mechanisms may regulate addictive liability.

CIRCUIT-LEVEL EFFECTS OF PSYCHEDELICS

While 5-HT2ARs are expressed broadly throughout the brain, layer 5 of the cortex and the claustrum are the two brain regions with the highest densities (25). Given the extensive role the cortex plays in sensory processing and top-down control of various subcortical regions also involved in sensory processing, 5-HT2AR-dependent changes in cortical function have been hypothesized to mediate the subjective effects that constitute the classic ‘psychedelic experience.’ Though it is much harder to study due to its anatomy, the claustrum is likely to also play a key role in the effects of psychedelics. This enigmatic region was famously hypothesized by Crick and Koch to be the “seat of consciousness,” (138) due to its vast interconnectivity with the cortex and subcortical structures involved in sensory processing (139, 140). In humans, the claustrum is the most interconnected brain structure per volume (141), supporting a role for the claustrum in binding sensory information from across the cortex to form a unified conscious experience.

Given 5-HT2AR expression patterns, it is unsurprising that psychedelics can dramatically impact network activity across the cortex. Treatment with DOI, or 5-MeO-DMT decreased the amplitude of low frequency (0.3–4 Hz) oscillations in the cortex, and the effects were blocked by 5-HT2AR antagonists (118, 142). The two drugs appear to have opposite effects on high frequency oscillations in the cortex, with DOI and 5-MeO-DMT reducing and increasing gamma power in rodents, respectively (143, 144). Psilocybin also decreased power of low frequency bands in the cortex of rats, while potentially increasing gamma power (145). Another study found that DOI reduced local field potential power across a wide frequency range (146). Unlike DOI, 25C-NBOMe seems to increase the power of high frequency oscillations (120–150 Hz) in the cortex of rats, an effect blocked by a 5-HT2R antagonist (147). Electroencephalography studies in freely moving rats has revealed that psilocin, LSD, mescaline, and DOB—compounds representing the tryptamine, ergoline, and phenethylamine classes of psychedelics—all decreased spectral power in the 1–40 Hz frequency range (148).

Disruption of cortical oscillations might be directly related to the effects of psychedelics on cortical neuron excitability. Several studies have observed that psychedelics induce bidirectional modulation of cortical neuron function with approximately 1/3 of the cells in the cortex being excited, 1/3 being inhibited, and 1/3 being unaffected (118, 142). Interestingly, excitatory neurons in V1 of the mouse cortex with low and high firing rates were enhanced or suppressed, respectively, following DOI exposure (146). Similar observations have been made in V1 of cats and non-human primates (149, 150). The effect is likely determined by whether or not a neuron expresses 5-HT2ARs, as recent unpublished work using a tdTomato reporter for 5-HT2AR expression has shown that psilocin directly activates 5-HT2AR-expressing layer 5 pyramidal neurons in the cortex (33). Importantly, ketamine also appears to suppress cortical neurons that are normally active and activate those that are typically inactive (151). Thus, this switch in cortical neuron activity may represent a general feature of hallucinogenic drugs.

CONCLUSION AND UNANSWERED QUESTIONS

Psychedelics produce profound effects on nervous system function by activating 5-HT2ARs as well as several other targets (Figure 4). While the acute effects of these drugs on perception have long been appreciated, their sustained effects are now being investigated to explain how they can produce long-lasting therapeutic responses after only a single administration. Many questions remain about the mechanisms of psychedelics including why biological sex seems to impact both their acute and sustained effects. For example, female mice appear to be more sensitive to psychedelics in both the HTR assay (38) and in measures of structural neuroplasticity (93). It is unclear if sex hormones modulate responses to psychedelics or if differences in receptor expression patterns across the brain underlie the distinctions observed between males and females.

Figure 4.

Figure 4.

Schematic overview of the putative molecular mechanisms of psychedelic drug action as well as the molecular-, cellular-, and circuit-level effects of psychedelics on neuronal physiology.

Perhaps the most important unanswered questions about the mechanisms of psychedelics are related to which circuits mediate the various behavioral effects associated with this class of small molecules. Currently, we do not know which circuits are involved in hallucinogenic behaviors such as the HTR and which mediate long-lasting beneficial responses relevant to treating depression, post-traumatic stress disorder, and substance use disorder. It is quite possible that these circuits are distinct, and modern optogenetic, chemogenetic, and gene knockout tools should enable their elucidation. If the circuits mediating the hallucinogenic and therapeutic effects of psychedelics are distinct, it should be possible to engineer targeted therapeutics with improved efficacy and safety profiles.

ACKNOWLEDGEMENTS

This work was supported by funds from the National Institutes of Health (NIH) (R01GM128997 and R01DA056365).

Footnotes

DISCLOSURE

DEO is a co-founder of Delix Therapeutics, Inc., serves as the Chief Innovation Officer and Head of the Scientific Advisory Board, and has sponsored research agreements with Delix Therapeutics. Delix was not involved in the conceptualization, design, decision to publish, or preparation of this manuscript.

REFERENCES

  • 1.Nichols DE, Nichols CD, Hendricks PS. 2022. Proposed Consensus Statement on Defining Psychedelic Drugs. Psychedelic Medicine. 1(1), 12–13 [Google Scholar]
  • 2.Nichols DE. 2016. Psychedelics. Pharmacol Rev. 68(2):264–355 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 3.Kwan AC, Olson DE, Preller KH, Roth BL. 2022. The neural basis of psychedelic action. Nat Neurosci. 25(11):1407–19 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 4.Doss MK, Považan M, Rosenberg MD, Sepeda ND, Davis AK, et al. 2021. Psilocybin therapy increases cognitive and neural flexibility in patients with major depressive disorder. Transl Psychiatry. 11(1):1–10 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 5.Johnson MW, Garcia-Romeu A, Griffiths RR. 2017. Long-term Follow-up of Psilocybin-facilitated Smoking Cessation. Am J Drug Alcohol Abuse. 43(1):55–60 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 6.Bogenschutz MP, Forcehimes AA, Pommy JA, Wilcox CE, Barbosa P, Strassman RJ. 2015. Psilocybin-assisted treatment for alcohol dependence: A proof-of-concept study. J Psychopharmacol. 29(3):289–99 [DOI] [PubMed] [Google Scholar]
  • 7.Griffiths RR, Johnson MW, Carducci MA, Umbricht A, Richards WA, et al. 2016. Psilocybin produces substantial and sustained decreases in depression and anxiety in patients with life-threatening cancer: A randomized double-blind trial. J Psychopharmacol. 30(12):1181–97 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 8.Davis AK, Barrett FS, May DG, Cosimano MP, Sepeda ND, et al. 2021. Effects of Psilocybin-Assisted Therapy on Major Depressive Disorder: A Randomized Clinical Trial. JAMA Psychiatry. 78(5):481–89 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 9.Gukasyan N, Davis AK, Barrett FS, Cosimano MP, Sepeda ND, et al. 2022. Efficacy and safety of psilocybin-assisted treatment for major depressive disorder: Prospective 12-month follow-up. J Psychopharmacol. 36(2):151–58 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 10.Olson DE. 2021. The Subjective Effects of Psychedelics May Not Be Necessary for Their Enduring Therapeutic Effects. ACS Pharmacol. Transl. Sci 4(2):563–67 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 11.Yaden DB, Griffiths RR. 2021. The Subjective Effects of Psychedelics Are Necessary for Their Enduring Therapeutic Effects. ACS Pharmacol. Transl. Sci 4(2):568–72 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 12.Glennon RA, Titeler M, McKenney JD. 1984. Evidence for 5-HT2 involvement in the mechanism of action of hallucinogenic agents. Life Sciences. 35(25):2505–11 [DOI] [PubMed] [Google Scholar]
  • 13.Becker AM, Klaiber A, Holze F, Istampoulouoglou I, Duthaler U, et al. 2022. Ketanserin Reverses the Acute Response to LSD in a Randomized, Double-Blind, Placebo-Controlled, Crossover Study in Healthy Participants. International Journal of Neuropsychopharmacology. 26(2):97–106 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 14.Holze F, Vizeli P, Ley L, Müller F, Dolder P, et al. 2021. Acute dose-dependent effects of lysergic acid diethylamide in a double-blind placebo-controlled study in healthy subjects. Neuropsychopharmacology. 46(3):537–44 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 15.Preller KH, Herdener M, Pokorny T, Planzer A, Kraehenmann R, et al. 2017. The Fabric of Meaning and Subjective Effects in LSD-Induced States Depend on Serotonin 2A Receptor Activation. Current Biology. 27(3):451–57 [DOI] [PubMed] [Google Scholar]
  • 16.Valle M, Maqueda AE, Rabella M, Rodríguez-Pujadas A, Antonijoan RM, et al. 2016. Inhibition of alpha oscillations through serotonin-2A receptor activation underlies the visual effects of ayahuasca in humans. European Neuropsychopharmacology. 26(7):1161–75 [DOI] [PubMed] [Google Scholar]
  • 17.Kometer M, Schmidt A, Bachmann R, Studerus E, Seifritz E, Vollenweider FX. 2012. Psilocybin Biases Facial Recognition, Goal-Directed Behavior, and Mood State Toward Positive Relative to Negative Emotions Through Different Serotonergic Subreceptors. Biological Psychiatry. 72(11):898–906 [DOI] [PubMed] [Google Scholar]
  • 18.Quednow BB, Kometer M, Geyer MA, Vollenweider FX. 2012. Psilocybin-Induced Deficits in Automatic and Controlled Inhibition are Attenuated by Ketanserin in Healthy Human Volunteers. Neuropsychopharmacology. 37(3):630–40 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 19.Vollenweider FX, Vollenweider-Scherpenhuyzen MFI, Bäbler A, Vogel H, Hell D. 1998. Psilocybin induces schizophrenia-like psychosis in humans via a serotonin-2 agonist action. NeuroReport. 9(17):3897. [DOI] [PubMed] [Google Scholar]
  • 20.Fiorella D, Rabin RA, Winter JC. 1995. The role of the 5-HT2A and 5-HT2C receptors in the stimulus effects of hallucinogenic drugs I: Antagonist correlation analysis. Psychopharmacology. 121(3):347–56 [DOI] [PubMed] [Google Scholar]
  • 21.Madsen MK, Fisher PM, Burmester D, Dyssegaard A, Stenbæk DS, et al. 2019. Psychedelic effects of psilocybin correlate with serotonin 2A receptor occupancy and plasma psilocin levels. Neuropsychopharmacol. 44(7):1328–34 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 22.González-Maeso J, Weisstaub NV, Zhou M, Chan P, Ivic L, et al. 2007. Hallucinogens Recruit Specific Cortical 5-HT2A Receptor-Mediated Signaling Pathways to Affect Behavior. Neuron. 53(3):439–52 [DOI] [PubMed] [Google Scholar]
  • 23.Canal CE, Olaghere da Silva UB, Gresch PJ, Watt EE, Sanders-Bush E, Airey DC. 2010. The serotonin 2C receptor potently modulates the head-twitch response in mice induced by a phenethylamine hallucinogen. Psychopharmacology (Berl). 209(2):163–74 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 24.Cornea-Hébert V, Riad M, Wu C, Singh SK, Descarries L. 1999. Cellular and subcellular distribution of the serotonin 5-HT2A receptor in the central nervous system of adult rat. Journal of Comparative Neurology. 409(2):187–209 [DOI] [PubMed] [Google Scholar]
  • 25.Weber ET, Andrade R. 2010. Htr2a Gene and 5-HT2A Receptor Expression in the Cerebral Cortex Studied Using Genetically Modified Mice. Front Neurosci. 4:36. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 26.Pazos A, Probst A, Palacios JM. 1987. Serotonin receptors in the human brain—IV. Autoradiographic mapping of serotonin-2 receptors. Neuroscience. 21(1):123–39 [DOI] [PubMed] [Google Scholar]
  • 27.Beliveau V, Ganz M, Feng L, Ozenne B, Højgaard L, et al. 2017. A High-Resolution In Vivo Atlas of the Human Brain’s Serotonin System. J Neurosci. 37(1):120–28 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 28.Porter RHP, Benwell KR, Lamb H, Malcolm CS, Allen NH, et al. 1999. Functional characterization of agonists at recombinant human 5-HT2A, 5-HT2B and 5-HT2C receptors in CHO-K1 cells. British Journal of Pharmacology. 128(1):13–20 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 29.Egan CT, Herrick-Davis K, Miller K, Glennon RA, Teitler M. 1998. Agonist activity of LSD and lisuride at cloned 5HT2A and 5HT2C receptors. Psychopharmacology. 136(4):409–14 [DOI] [PubMed] [Google Scholar]
  • 30.Cussac D, Boutet-Robinet E, Ailhaud M-C, Newman-Tancredi A, Martel J-C, et al. 2008. Agonist-directed trafficking of signalling at serotonin 5-HT2A, 5-HT2B and 5-HT2C-VSV receptors mediated Gq/11 activation and calcium mobilisation in CHO cells. European Journal of Pharmacology. 594(1):32–38 [DOI] [PubMed] [Google Scholar]
  • 31.Masson J, Emerit MB, Hamon M, Darmon M. 2012. Serotonergic signaling: multiple effectors and pleiotropic effects. Wiley Interdisciplinary Reviews: Membrane Transport and Signaling. 1(6):685–713 [Google Scholar]
  • 32.Vargas MV, Dunlap LE, Dong C, Carter SJ, Tombari RJ, et al. 2023. Psychedelics promote neuroplasticity through the activation of intracellular 5-HT2A receptors. Science. 379(6633):700–706 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 33.Schmitz G, Chiu Y-T, König G, Kostenis E, Roth B, Herman M. 2022. Psychedelic compounds directly excite 5-HT2ALayer 5 Pyramidal Neurons in the Prefrontal Cortex through a 5-HT2AGq -mediated activation mechanism. BioRxiv. DOI: 10.1101/2022.11.15.516655 [DOI] [Google Scholar]
  • 34.Cameron LP, Tombari RJ, Lu J, Pell AJ, Hurley ZQ, et al. 2021. A non-hallucinogenic psychedelic analogue with therapeutic potential. Nature. 589(7842):474–79 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 35.Cao D, Yu J, Wang H, Luo Z, Liu X, et al. 2022. Structure-based discovery of nonhallucinogenic psychedelic analogs. Science. 375(6579):403–11 [DOI] [PubMed] [Google Scholar]
  • 36.Lewis V, Bonniwell EM, Lanham JK, Ghaffari A, Sheshbaradaran H, et al. 2023. A non-hallucinogenic LSD analog with therapeutic potential for mood disorders. Cell Reports. 42(3):112203. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 37.Faillace LA, Vourlekis A, Szara S. 1967. Clinical evaluation of some hallucinogenic tryptamine derivatives. The Journal of Nervous and Mental Disease. 145(4):306. [DOI] [PubMed] [Google Scholar]
  • 38.Dunlap LE, Azinfar A, Ly C, Cameron LP, Viswanathan J, et al. 2020. Identification of Psychoplastogenic N,N-Dimethylaminoisotryptamine (isoDMT) Analogues through Structure–Activity Relationship Studies. J. Med. Chem 63(3):1142–55 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 39.Cunningham MJ, Bock HA, Serrano IC, Bechand B, Vidyadhara DJ, et al. 2023. Pharmacological Mechanism of the Non-hallucinogenic 5-HT2A Agonist Ariadne and Analogs. ACS Chem. Neurosci 14(1):119–35 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 40.Kaplan AL, Confair DN, Kim K, Barros-Álvarez X, Rodriguiz RM, et al. 2022. Bespoke library docking for 5-HT2A receptor agonists with antidepressant activity. Nature. 610(7932):582–91 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 41.Rabin RA, Regina M, Doat M, Winter JC. 2002. 5-HT2A receptor-stimulated phosphoinositide hydrolysis in the stimulus effects of hallucinogens. Pharmacology Biochemistry and Behavior. 72(1):29–37 [DOI] [PubMed] [Google Scholar]
  • 42.Dong C, Ly C, Dunlap LE, Vargas MV, Sun J, et al. 2021. Psychedelic-inspired drug discovery using an engineered biosensor. Cell. 184(10):2779–2792.e18 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 43.Rodriguiz RM, Nadkarni V, Means CR, Pogorelov VM, Chiu Y-T, et al. 2021. LSD-stimulated behaviors in mice require β-arrestin 2 but not β-arrestin 1. Sci Rep. 11(1):17690. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 44.Schmid CL, Raehal KM, Bohn LM. 2008. Agonist-directed signaling of the serotonin 2A receptor depends on β-arrestin-2 interactions in vivo. Proc Natl Acad Sci U S A. 105(3):1079–84 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 45.Garcia EE, Smith RL, Sanders-Bush E. 2007. Role of Gq protein in behavioral effects of the hallucinogenic drug 1-(2,5-dimethoxy-4-iodophenyl)-2-aminopropane. Neuropharmacology. 52(8):1671–77 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 46.González-Maeso J, Ang RL, Yuen T, Chan P, Weisstaub NV, et al. 2008. Identification of a serotonin/glutamate receptor complex implicated in psychosis. Nature. 452(7183):93–97 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 47.Łukasiewicz S, Polit A, Kędracka-Krok S, Wędzony K, Maćkowiak M, Dziedzicka-Wasylewska M. 2010. Hetero-dimerization of serotonin 5-HT2A and dopamine D2 receptors. Biochimica et Biophysica Acta (BBA) - Molecular Cell Research. 1803(12):1347–58 [DOI] [PubMed] [Google Scholar]
  • 48.Viñals X, Moreno E, Lanfumey L, Cordomí A, Pastor A, et al. 2015. Cognitive Impairment Induced by Delta9-tetrahydrocannabinol Occurs through Heteromers between Cannabinoid CB1 and Serotonin 5-HT2A Receptors. PLoS Biol. 13(7):e1002194. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 49.Moutkine I, Quentin E, Guiard BP, Maroteaux L, Doly S. 2017. Heterodimers of serotonin receptor subtypes 2 are driven by 5-HT2C protomers. J Biol Chem. 292(15):6352–68 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 50.Borroto-Escuela DO, Romero-Fernandez W, Narvaez M, Oflijan J, Agnati LF, Fuxe K. 2014. Hallucinogenic 5-HT2AR agonists LSD and DOI enhance dopamine D2R protomer recognition and signaling of D2-5-HT2A heteroreceptor complexes. Biochemical and Biophysical Research Communications. 443(1):278–84 [DOI] [PubMed] [Google Scholar]
  • 51.Schindler EAD, Dave KD, Smolock EM, Aloyo VJ, Harvey JA. 2012. Serotonergic and dopaminergic distinctions in the behavioral pharmacology of (±)-1-(2,5-dimethoxy-4-iodophenyl)-2-aminopropane (DOI) and lysergic acid diethylamide (LSD). Pharmacol Biochem Behav. 101(1):69–76 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 52.Moreno JL, Holloway T, Albizu L, Sealfon SC, González-Maeso J. 2011. Metabotropic glutamate mGlu2 receptor is necessary for the pharmacological and behavioral effects induced by hallucinogenic 5-HT2A receptor agonists. Neurosci Lett. 493(3):76–79 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 53.Barnes NM, Sharp T. 1999. A review of central 5-HT receptors and their function. Neuropharmacology. 38(8):1083–1152 [DOI] [PubMed] [Google Scholar]
  • 54.Doly S, Valjent E, Setola V, Callebert J, Hervé D, et al. 2008. Serotonin 5-HT2B Receptors Are Required for 3,4-Methylenedioxymethamphetamine-Induced Hyperlocomotion and 5-HT Release In Vivo and In Vitro. J. Neurosci 28(11):2933–40 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 55.Burns CM, Chu H, Rueter SM, Hutchinson LK, Canton H, et al. 1997. Regulation of serotonin-2C receptor G-protein coupling by RNA editing. Nature. 387(6630):303–8 [DOI] [PubMed] [Google Scholar]
  • 56.Gumpper RH, Fay JF, Roth BL. 2022. Molecular insights into the regulation of constitutive activity by RNA editing of 5HT2C serotonin receptors. Cell Reports. 40(7):111211. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 57.De Gregorio D, Posa L, Ochoa-Sanchez R, McLaughlin R, Maione S, et al. 2016. The hallucinogen d-lysergic diethylamide (LSD) decreases dopamine firing activity through 5-HT1A, D2 and TAAR1 receptors. Pharmacological Research. 113:81–91 [DOI] [PubMed] [Google Scholar]
  • 58.Tepper SJ, Rapoport AM, Sheftell FD. 2002. Mechanisms of Action of the 5-HT1B/1D Receptor Agonists. Archives of Neurology. 59(7):1084–88 [DOI] [PubMed] [Google Scholar]
  • 59.Bunzow JR, Sonders MS, Arttamangkul S, Harrison LM, Zhang G, et al. 2001. Amphetamine, 3,4-Methylenedioxymethamphetamine, Lysergic Acid Diethylamide, and Metabolites of the Catecholamine Neurotransmitters Are Agonists of a Rat Trace Amine Receptor. Mol Pharmacol. 60(6):1181–88 [DOI] [PubMed] [Google Scholar]
  • 60.Fontanilla D, Johannessen M, Hajipour AR, Cozzi NV, Jackson MB, Ruoho AE. 2009. The Hallucinogen N,N-Dimethyltryptamine (DMT) Is an Endogenous Sigma-1 Receptor Regulator. Science. 323(5916):934–37 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 61.Moliner R, Girych M, Brunello CA, Kovaleva V, Biojone C, et al. 2023. Psychedelics promote plasticity by directly binding to BDNF receptor TrkB. Nat Neurosci. 26(6):1032–41 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 62.Nichols CD, Sanders-Bush E. 2002. A Single Dose of Lysergic Acid Diethylamide Influences Gene Expression Patterns within the Mammalian Brain. Neuropsychopharmacol. 26(5):634–42 [DOI] [PubMed] [Google Scholar]
  • 63.González-Maeso J, Yuen T, Ebersole BJ, Wurmbach E, Lira A, et al. 2003. Transcriptome Fingerprints Distinguish Hallucinogenic and Nonhallucinogenic 5-Hydroxytryptamine 2A Receptor Agonist Effects in Mouse Somatosensory Cortex. J Neurosci. 23(26):8836–43 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 64.Leslie RA, Moorman JM, Coulson A, Grahame-Smith DG. 1993. Serotonin2/1 C receptor activation causes a localized expression of the immediate-early gene c-fos in rat brain: evidence for involvement of dorsal raphe nucleus projection fibres. Neuroscience. 53(2):457–63 [DOI] [PubMed] [Google Scholar]
  • 65.Gresch PJ, Strickland LV, Sanders-Bush E. 2002. Lysergic acid diethylamide-induced Fos expression in rat brain: role of serotonin-2A receptors. Neuroscience. 114(3):707–13 [DOI] [PubMed] [Google Scholar]
  • 66.Nichols CD, Sanders-Bush E. 2004. Molecular genetic responses to lysergic acid diethylamide include transcriptional activation of MAP kinase phosphatase-1, C/EBP-β and ILAD-1, a novel gene with homology to arrestins. Journal of Neurochemistry. 90(3):576–84 [DOI] [PubMed] [Google Scholar]
  • 67.Pei Q, Tordera R, Sprakes M, Sharp T. 2004. Glutamate receptor activation is involved in 5-HT2 agonist-induced Arc gene expression in the rat cortex. Neuropharmacology. 46(3):331–39 [DOI] [PubMed] [Google Scholar]
  • 68.Pei Q, Lewis L, Sprakes ME, Jones EJ, Grahame-Smith DG, Zetterström TSC. 2000. Serotonergic regulation of mRNA expression of Arc, an immediate early gene selectively localized at neuronal dendrites. Neuropharmacology. 39(3):463–70 [DOI] [PubMed] [Google Scholar]
  • 69.Davoudian PA, Shao L-X, Kwan AC. 2022. Shared and distinct brain regions targeted for immediate early gene expression by ketamine and psilocybin. ACS Chem. Neurosci 14(3):468–480 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 70.Vaidya VA, Marek GJ, Aghajanian GK, Duman RS. 1997. 5-HT2A Receptor-Mediated Regulation of Brain-Derived Neurotrophic Factor mRNA in the Hippocampus and the Neocortex. J Neurosci. 17(8):2785–95 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 71.Benekareddy M, Nair AR, Dias BG, Suri D, Autry AE, et al. 2013. Induction of the plasticity-associated immediate early gene Arc by stress and hallucinogens: role of brain-derived neurotrophic factor. International Journal of Neuropsychopharmacology. 16(2):405–15 [DOI] [PubMed] [Google Scholar]
  • 72.Gewirtz JC, Chen AC, Terwilliger R, Duman RC, Marek GJ. 2002. Modulation of DOI-induced increases in cortical BDNF expression by group II mGlu receptors. Pharmacology Biochemistry and Behavior. 73(2):317–26 [DOI] [PubMed] [Google Scholar]
  • 73.Zhai Y, George CA, Zhai J, Nisenbaum ES, Johnson MP, Nisenbaum LK. 2003. Group II Metabotropic Glutamate Receptor Modulation of DOI-induced c-fos mRNA and Excitatory Responses in the Cerebral Cortex. Neuropsychopharmacol. 28(1):45–52 [DOI] [PubMed] [Google Scholar]
  • 74.Scruggs JL, Patel S, Bubser M, Deutch AY. 2000. DOI-Induced Activation of the Cortex: Dependence on 5-HT2A Heteroceptors on Thalamocortical Glutamatergic Neurons. J Neurosci. 20(23):8846–52 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 75.Desouza LA, Benekareddy M, Fanibunda SE, Mohammad F, Janakiraman B, et al. 2021. The Hallucinogenic Serotonin2A Receptor Agonist, 2,5-Dimethoxy-4-Iodoamphetamine, Promotes cAMP Response Element Binding Protein-Dependent Gene Expression of Specific Plasticity-Associated Genes in the Rodent Neocortex. Front Mol Neurosci. 14:790213. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 76.Raval NR, Johansen A, Donovan LL, Ros NF, Ozenne B, et al. 2021. A Single Dose of Psilocybin Increases Synaptic Density and Decreases 5-HT2A Receptor Density in the Pig Brain. Int J Mol Sci. 22(2):835. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 77.Jefsen OH, Elfving B, Wegener G, Müller HK. 2021. Transcriptional regulation in the rat prefrontal cortex and hippocampus after a single administration of psilocybin. J Psychopharmacol. 35(4):483–93 [DOI] [PubMed] [Google Scholar]
  • 78.Wojtas A, Bysiek A, Wawrzczak-Bargiela A, Szych Z, Majcher-Maślanka I, et al. 2022. Effect of Psilocybin and Ketamine on Brain Neurotransmitters, Glutamate Receptors, DNA and Rat Behavior. Int J Mol Sci. 23(12):6713. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 79.Dakic V, Minardi Nascimento J, Costa Sartore R, Maciel R de M, de Araujo DB, et al. 2017. Short term changes in the proteome of human cerebral organoids induced by 5-MeO-DMT. Sci Rep. 7(1):12863. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 80.Ornelas IM, Cini FA, Wießner I, Marcos E, Araújo DB, et al. 2022. Nootropic effects of LSD: Behavioral, molecular and computational evidence. Experimental Neurology. 356:114148. [DOI] [PubMed] [Google Scholar]
  • 81.Revenga M de la F, Zhu B, Guevara CA, Naler LB, Saunders JM, et al. 2021. Prolonged epigenomic and synaptic plasticity alterations following single exposure to a psychedelic in mice. Cell Rep. 37(3):109836. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 82.Inserra A, Campanale A, Cheishvili D, Dymov S, Wong A, et al. 2022. Modulation of DNA methylation and protein expression in the prefrontal cortex by repeated administration of D-lysergic acid diethylamide (LSD): Impact on neurotropic, neurotrophic, and neuroplasticity signaling. Progress in Neuro-Psychopharmacology and Biological Psychiatry. 119:110594. [DOI] [PubMed] [Google Scholar]
  • 83.Olson DE. 2018. Psychoplastogens: A Promising Class of Plasticity-Promoting Neurotherapeutics. J Exp Neurosci. 12:1179069518800508. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 84.Ly C, Greb AC, Cameron LP, Wong JM, Barragan EV, et al. 2018. Psychedelics Promote Structural and Functional Neural Plasticity. Cell reports. 23(11):3170. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 85.Jones KA, Srivastava DP, Allen JA, Strachan RT, Roth BL, Penzes P. 2009. Rapid modulation of spine morphology by the 5-HT2A serotonin receptor through kalirin-7 signaling. Proc Natl Acad Sci U S A. 106(46):19575–80 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 86.Mi Z, Si T, Kapadia K, Li Q, Muma NA. 2017. Receptor-stimulated transamidation induces activation of Rac1 and Cdc42 and the regulation of dendritic spines. Neuropharmacology. 117:93–105 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 87.Yoshida H, Kanamaru C, Ohtani A, Li F, Senzaki K, Shiga T. 2011. Subtype specific roles of serotonin receptors in the spine formation of cortical neurons in vitro. Neuroscience Research. 71(3):311–14 [DOI] [PubMed] [Google Scholar]
  • 88.Ohtani A, Kozono N, Senzaki K, Shiga T. 2014. Serotonin 2A receptor regulates microtubule assembly and induces dynamics of dendritic growth cones in rat cortical neurons in vitro. Neuroscience Research. 81–82:11–20 [DOI] [PubMed] [Google Scholar]
  • 89.Cameron LP, Patel SD, Vargas MV, Barragan EV, Saeger HN, et al. 2023. 5-HT2ARs Mediate Therapeutic Behavioral Effects of Psychedelic Tryptamines. ACS Chem. Neurosci 14(3):351–358 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 90.Ly C, Greb AC, Vargas MV, Duim WC, Grodzki ACG, et al. 2021. Transient Stimulation with Psychoplastogens Is Sufficient to Initiate Neuronal Growth. ACS Pharmacol. Transl. Sci 4(2):452–60 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 91.De Gregorio D, Inserra A, Enns JP, Markopoulos A, Pileggi M, et al. 2022. Repeated lysergic acid diethylamide (LSD) reverses stress-induced anxiety-like behavior, cortical synaptogenesis deficits and serotonergic neurotransmission decline. Neuropsychopharmacol. 47(6):1188–98 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 92.Cameron LP, Benson CJ, DeFelice BC, Fiehn O, Olson DE. 2019. Chronic, Intermittent Microdoses of the Psychedelic N,N-Dimethyltryptamine (DMT) Produce Positive Effects on Mood and Anxiety in Rodents. ACS Chem. Neurosci 10(7):3261–70 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 93.Shao L-X, Liao C, Gregg I, Davoudian PA, Savalia NK, et al. 2021. Psilocybin induces rapid and persistent growth of dendritic spines in frontal cortex in vivo. Neuron. 109(16):2535–2544.e4 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 94.Jefferson SJ, Gregg I, Dibbs M, Liao C, Wu H, et al. 2023. 5-MeO-DMT modifies innate behaviors and promotes structural neural plasticity in mice. Neuropsychopharmacology. Advance online publication. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 95.Olson DE. 2022. Biochemical Mechanisms Underlying Psychedelic-Induced Neuroplasticity. Biochemistry. 61(3):127–36 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 96.De Gregorio D, Popic J, Enns JP, Inserra A, Skalecka A, et al. 2021. Lysergic acid diethylamide (LSD) promotes social behavior through mTORC1 in the excitatory neurotransmission. Proceedings of the National Academy of Sciences. 118(5):e2020705118. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 97.Qu Y, Chang L, Ma L, Wan X, Hashimoto K. 2023. Rapid antidepressant-like effect of non-hallucinogenic psychedelic analog lisuride, but not hallucinogenic psychedelic DOI, in lipopolysaccharide-treated mice. Pharmacology Biochemistry and Behavior. 222:173500. [DOI] [PubMed] [Google Scholar]
  • 98.Lu J, Tjia M, Mullen B, Cao B, Lukasiewicz K, et al. 2021. An analog of psychedelics restores functional neural circuits disrupted by unpredictable stress. Mol Psychiatry. 26(11):6237–52 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 99.Vargas MV, Meyer R, Avanes AA, Rus M, Olson DE. 2021. Psychedelics and Other Psychoplastogens for Treating Mental Illness. Frontiers in Psychiatry. 12:727117. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 100.Nichols CD, Garcia EE, Sanders-Bush E. 2003. Dynamic changes in prefrontal cortex gene expression following lysergic acid diethylamide administration. Molecular Brain Research. 111(1):182–88 [DOI] [PubMed] [Google Scholar]
  • 101.Fanibunda SE, Deb S, Maniyadath B, Tiwari P, Ghai U, et al. 2019. Serotonin regulates mitochondrial biogenesis and function in rodent cortical neurons via the 5-HT2A receptor and SIRT1–PGC-1α axis. Proceedings of the National Academy of Sciences. 116(22):11028–37 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 102.Fujimoto M, Hayashi T, Urfer R, Mita S, Su T-P. 2012. Sigma-1 receptor chaperones regulate the secretion of brain-derived neurotrophic factor. Synapse. 66(7): 10.1002/syn.21549 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 103.Kimura Y, Fujita Y, Shibata K, Mori M, Yamashita T. 2013. Sigma-1 Receptor Enhances Neurite Elongation of Cerebellar Granule Neurons via TrkB Signaling. PLOS ONE. 8(10):e75760. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 104.Ka M, Kook Y-H, Liao K, Buch S, Kim W-Y. 2016. Transactivation of TrkB by Sigma-1 receptor mediates cocaine-induced changes in dendritic spine density and morphology in hippocampal and cortical neurons. Cell Death Dis. 7(10):e2414–e2414 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 105.Barnabé-Heider F, Miller FD. 2003. Endogenously Produced Neurotrophins Regulate Survival and Differentiation of Cortical Progenitors via Distinct Signaling Pathways. J Neurosci. 23(12):5149–60 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 106.Kowiański P, Lietzau G, Czuba E, Waśkow M, Steliga A, Moryś J. 2018. BDNF: A Key Factor with Multipotent Impact on Brain Signaling and Synaptic Plasticity. Cell Mol Neurobiol. 38(3):579–93 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 107.Szabo A, Kovacs A, Riba J, Djurovic S, Rajnavolgyi E, Frecska E. 2016. The Endogenous Hallucinogen and Trace Amine N,N-Dimethyltryptamine (DMT) Displays Potent Protective Effects against Hypoxia via Sigma-1 Receptor Activation in Human Primary iPSC-Derived Cortical Neurons and Microglia-Like Immune Cells. Front Neurosci. 10:423. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 108.Nardai S, László M, Szabó A, Alpár A, Hanics J, et al. 2020. N,N-dimethyltryptamine reduces infarct size and improves functional recovery following transient focal brain ischemia in rats. Experimental Neurology. 327:113245. [DOI] [PubMed] [Google Scholar]
  • 109.Lima da Cruz RV, Moulin TC, Petiz LL, Leão RN. 2018. A Single Dose of 5-MeO-DMT Stimulates Cell Proliferation, Neuronal Survivability, Morphological and Functional Changes in Adult Mice Ventral Dentate Gyrus. Front Mol Neurosci. 11:312. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 110.Jha S, Rajendran R, Fernandes KA, Vaidya VA. 2008. 5-HT2A/2C receptor blockade regulates progenitor cell proliferation in the adult rat hippocampus. Neuroscience Letters. 441(2):210–14 [DOI] [PubMed] [Google Scholar]
  • 111.Catlow BJ, Song S, Paredes DA, Kirstein CL, Sanchez-Ramos J. 2013. Effects of psilocybin on hippocampal neurogenesis and extinction of trace fear conditioning. Exp Brain Res. 228(4):481–91 [DOI] [PubMed] [Google Scholar]
  • 112.Morales-Garcia JA, Calleja-Conde J, Lopez-Moreno JA, Alonso-Gil S, Sanz-SanCristobal M, et al. 2020. N,N-dimethyltryptamine compound found in the hallucinogenic tea ayahuasca, regulates adult neurogenesis in vitro and in vivo. Transl Psychiatry. 10:331. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 113.Aghajanian GK, Marek GJ. 1999. Serotonin, via 5-HT2A receptors, increases EPSCs in layer V pyramidal cells of prefrontal cortex by an asynchronous mode of glutamate release. Brain Research. 825(1):161–71 [DOI] [PubMed] [Google Scholar]
  • 114.Lambe EK, Aghajanian GK. 2006. Hallucinogen-Induced UP States in the Brain Slice of Rat Prefrontal Cortex: Role of Glutamate Spillover and NR2B-NMDA Receptors. Neuropsychopharmacol. 31(8):1682–89 [DOI] [PubMed] [Google Scholar]
  • 115.Aghajanian GK, Marek GJ. 1997. Serotonin induces excitatory postsynaptic potentials in apical dendrites of neocortical pyramidal cells. Neuropharmacology. 36(4–5):589–99 [DOI] [PubMed] [Google Scholar]
  • 116.Kłodzinska A, Bijak M, Tokarski K, Pilc A. 2002. Group II mGlu receptor agonists inhibit behavioural and electrophysiological effects of DOI in mice. Pharmacology Biochemistry and Behavior. 73(2):327–32 [DOI] [PubMed] [Google Scholar]
  • 117.Arvanov VL, Liang X, Magro P, Roberts R, Wang RY. 1999. A pre- and postsynaptic modulatory action of 5-HT and the 5-HT2A, 2C receptor agonist DOB on NMDA-evoked responses in the rat medial prefrontal cortex. European Journal of Neuroscience. 11(8):2917–34 [DOI] [PubMed] [Google Scholar]
  • 118.Riga MS, Soria G, Tudela R, Artigas F, Celada P. 2014. The natural hallucinogen 5-MeO-DMT, component of Ayahuasca, disrupts cortical function in rats: reversal by antipsychotic drugs. International Journal of Neuropsychopharmacology. 17(8):1269–82 [DOI] [PubMed] [Google Scholar]
  • 119.Willins DL, Deutch AY, Roth BL. 1997. Serotonin 5-HT2A receptors are expressed on pyramidal cells and interneurons in the rat cortex. Synapse. 27(1):79–82 [DOI] [PubMed] [Google Scholar]
  • 120.De Almeida J, Mengod G. 2007. Quantitative analysis of glutamatergic and GABAergic neurons expressing 5-HT2A receptors in human and monkey prefrontal cortex. Journal of Neurochemistry. 103(2):475–86 [DOI] [PubMed] [Google Scholar]
  • 121.Marek GJ, Aghajanian GK. 1996. LSD and the phenethylamine hallucinogen DOI are potent partial agonists at 5-HT2A receptors on interneurons in rat piriform cortex. J Pharmacol Exp Ther. 278(3):1373–82 [PubMed] [Google Scholar]
  • 122.Rasmussen K, Aghajanian GK. 1986. Effect of hallucinogens on spontaneous and sensory-evoked locus coeruleus unit activity in the rat: reversal by selective 5-HT2antagonists. Brain Research. 385(2):395–400 [DOI] [PubMed] [Google Scholar]
  • 123.Aghajanian GK, Foote WE, Sheard MH. 1968. Lysergic Acid Diethylamide: Sensitive Neuronal Units in the Midbrain Raphe. Science. 161(3842):706–8 [DOI] [PubMed] [Google Scholar]
  • 124.Trulson ME, Heym J, Jacobs BL. 1981. Dissociations between the effects of hallucinogenic drugs on behavior and raphe unit activity in freely moving cats. Brain Research. 215(1):275–93 [DOI] [PubMed] [Google Scholar]
  • 125.Aghajanian GK, Foote WE, Sheard MH. 1970. Action of Psychotogenic Drugs on Single Midbrain Raphe Neurons. J Pharmacol Exp Ther. 171(2):178–87 [PubMed] [Google Scholar]
  • 126.Hesselgrave N, Troppoli TA, Wulff AB, Cole AB, Thompson SM. 2021. Harnessing psilocybin: antidepressant-like behavioral and synaptic actions of psilocybin are independent of 5-HT2R activation in mice. Proc Natl Acad Sci U S A. 118(17):e2022489118. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 127.Muschamp JW, Regina MJ, Hull EM, Winter JC, Rabin RA. 2004. Lysergic acid diethylamide and [−]-2,5-dimethoxy-4-methylamphetamine increase extracellular glutamate in rat prefrontal cortex. Brain Research. 1023(1):134–40 [DOI] [PubMed] [Google Scholar]
  • 128.Scruggs JL, Schmidt D, Deutch AY. 2003. The hallucinogen 1-[2,5-dimethoxy-4-iodophenyl]-2-aminopropane (DOI) increases cortical extracellular glutamate levels in rats. Neuroscience Letters. 346(3):137–40 [DOI] [PubMed] [Google Scholar]
  • 129.Herian M, Wojtas A, Kamińska K, Świt P, Wach A, Gołembiowska K. 2019. Hallucinogen-Like Action of the Novel Designer Drug 25I-NBOMe and Its Effect on Cortical Neurotransmitters in Rats. Neurotox Res. 36(1):91–100 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 130.Gouzoulis-Mayfrank E, Schreckenberger M, Sabri O, Arning C, Thelen B, et al. 1999. Neurometabolic Effects of Psilocybin, 3,4-Methylenedioxyethylamphetamine (MDE) and d-Methamphetamine in Healthy Volunteers A Double-Blind, Placebo-Controlled PET Study with [18F]FDG. Neuropsychopharmacol. 20(6):565–81 [DOI] [PubMed] [Google Scholar]
  • 131.Vollenweider FX, Leenders KL, Scharfetter C, Maguire P, Stadelmann O, Angst J. 1997. Positron Emission Tomography and Fluorodeoxyglucose Studies of Metabolic Hyperfrontality and Psychopathology in the Psilocybin Model of Psychosis. Neuropsychopharmacol. 16(5):357–72 [DOI] [PubMed] [Google Scholar]
  • 132.Abi-Saab WM, Bubser M, Roth RH, Deutch AY. 1999. 5-HT2 Receptor Regulation of Extracellular GABA Levels in the Prefrontal Cortex. Neuropsychopharmacol. 20(1):92–96 [DOI] [PubMed] [Google Scholar]
  • 133.de Castro-Neto EF, da Cunha RH, da Silveira DX, Yonamine M, Gouveia TLF, et al. 2013. Changes in aminoacidergic and monoaminergic neurotransmission in the hippocampus and amygdala of rats after ayahuasca ingestion. World J Biol Chem. 4(4):141–47 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 134.Bortolozzi A, Díaz-Mataix L, Scorza MC, Celada P, Artigas F. 2005. The activation of 5-HT2A receptors in prefrontal cortex enhances dopaminergic activity. Journal of Neurochemistry. 95(6):1597–1607 [DOI] [PubMed] [Google Scholar]
  • 135.Bowers BJ, Henry MB, Thielen RJ, McBride WJ. 2000. Serotonin 5-HT2 Receptor Stimulation of Dopamine Release in the Posterior but Not Anterior Nucleus Accumbens of the Rat. Journal of Neurochemistry. 75(4):1625–33 [DOI] [PubMed] [Google Scholar]
  • 136.Sakashita Y, Abe K, Katagiri N, Kambe T, Saitoh T, et al. 2015. Effect of Psilocin on Extracellular Dopamine and Serotonin Levels in the Mesoaccumbens and Mesocortical Pathway in Awake Rats. Biological and Pharmaceutical Bulletin. 38(1):134–38 [DOI] [PubMed] [Google Scholar]
  • 137.Erdtmann-Vourliotis M, Mayer P, Riechert U, Höllt V. 1999. Acute injection of drugs with low addictive potential (Δ9-tetrahydrocannabinol, 3,4-methylenedioxymethamphetamine, lysergic acid diamide) causes a much higher c-fos expression in limbic brain areas than highly addicting drugs (cocaine and morphine). Molecular Brain Research. 71(2):313–24 [DOI] [PubMed] [Google Scholar]
  • 138.Crick FC, Koch C. 2005. What is the function of the claustrum? Philos Trans R Soc Lond B Biol Sci. 360(1458):1271–79 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 139.Atlan G, Terem A, Peretz-Rivlin N, Groysman M, Citri A. 2017. Mapping synaptic cortico-claustral connectivity in the mouse. Journal of Comparative Neurology. 525(6):1381–1402 [DOI] [PubMed] [Google Scholar]
  • 140.White MG, Cody PA, Bubser M, Wang H-D, Deutch AY, Mathur BN. 2017. Cortical Hierarchy Governs Rat Claustrocortical Circuit Organization. J Comp Neurol. 525(6):1347–62 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 141.Torgerson CM, Irimia A, Goh SYM, Van Horn JD. 2015. The DTI connectivity of the human claustrum. Human Brain Mapping. 36(3):827–38 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 142.Celada P, Puig MV, Díaz-Mataix L, Artigas F. 2008. The Hallucinogen DOI Reduces Low-Frequency Oscillations in Rat Prefrontal Cortex: Reversal by Antipsychotic Drugs. Biological Psychiatry. 64(5):392–400 [DOI] [PubMed] [Google Scholar]
  • 143.Riga MS, Lladó-Pelfort L, Artigas F, Celada P. 2018. The serotonin hallucinogen 5-MeO-DMT alters cortico-thalamic activity in freely moving mice: Regionally-selective involvement of 5-HT1A and 5-HT2A receptors. Neuropharmacology. 142:219–30 [DOI] [PubMed] [Google Scholar]
  • 144.Wood J, Kim Y, Moghaddam B. 2012. Disruption of Prefrontal Cortex Large Scale Neuronal Activity by Different Classes of Psychotomimetic Drugs. J. Neurosci 32(9):3022–31 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 145.Golden CT, Chadderton P. 2022. Psilocybin reduces low frequency oscillatory power and neuronal phase-locking in the anterior cingulate cortex of awake rodents. Sci Rep. 12(1):12702. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 146.Michaiel AM, Parker PRL, Niell CM. 2019. A Hallucinogenic Serotonin-2A Receptor Agonist Reduces Visual Response Gain and Alters Temporal Dynamics in Mouse V1. Cell Reports. 26(13):3475–3483.e4 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 147.Yu Z-P, Li Q, Wu Z-X, Tang Z-H, Zhang X-Q, et al. 2023. The high frequency oscillation in orbitofrontal cortex is susceptible to phenethylamine psychedelic 25C–NBOMe in male rats. Neuropharmacology. 227:109452. [DOI] [PubMed] [Google Scholar]
  • 148.Vejmola Č, Tylš F, Piorecká V, Koudelka V, Kadeřábek L, et al. 2021. Psilocin, LSD, mescaline, and DOB all induce broadband desynchronization of EEG and disconnection in rats with robust translational validity. Transl Psychiatry. 11(1):506. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 149.Watakabe A, Komatsu Y, Sadakane O, Shimegi S, Takahata T, et al. 2009. Enriched Expression of Serotonin 1B and 2A Receptor Genes in Macaque Visual Cortex and their Bidirectional Modulatory Effects on Neuronal Responses. Cerebral Cortex. 19(8):1915–28 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 150.Rose D, Horn G. 1977. Effects of LSD on the responses of single units in cat visual cortex. Exp Brain Res. 27(1):71–80 [DOI] [PubMed] [Google Scholar]
  • 151.Cichon J, Wasilczuk AZ, Looger LL, Contreras D, Kelz MB, Proekt A. 2023. Ketamine triggers a switch in excitatory neuronal activity across neocortex. Nat Neurosci. 26(1):39–52 [DOI] [PMC free article] [PubMed] [Google Scholar]

RESOURCES