Abstract
The resurgence of interest in the therapeutic potential of psychedelics for treating psychiatric disorders has rekindled efforts to elucidate their mechanism of action. In this Perspective, we focus on the ability of psychedelics to promote neural plasticity, postulated to be central to their therapeutic activity. We begin with a brief overview of the history and behavioral effects of the classical psychedelics. We then summarize our current understanding of the cellular and subcellular mechanisms underlying these drugs’ behavioral effects, their effects on neural plasticity, and the roles of stress and inflammation in the acute and long-term effects of psychedelics. The signaling pathways activated by psychedelics couple to numerous potential mechanisms for producing long-term structural changes in the brain, a complexity that has barely begun to be disentangled. This complexity is mirrored by that of the neural mechanisms underlying psychiatric disorders and the transformations of consciousness, mood, and behavior that psychedelics promote in health and disease. Thus, beyond changes in the brain, psychedelics catalyze changes in our understanding of the neural basis of psychiatric disorders, as well as consciousness and human behavior.
INTRODUCTION
Human use of psychedelic drugs traces a remarkable historical arc, extending from the prehistoric realm of myth to the laboratories and clinical treatment rooms of the modern day, where these compounds’ mysteries are finally yielding to concerted inquiry. And yet our purpose and that of our ancestors are one and the same: to harness the potential of these compounds for insight and healing.
Evidence for human consumption of psychedelics in traditional medicine and religious ceremonies stretches back into prehistory (Sayin, 2014; Guerra-Doce, 2015; Froese et al., 2016). These naturally occurring psychedelics include ayahuasca, a brew of several plants indigenous to South America containing N,N-dimethyltryptamine (DMT); 5-MeO-DMT, found in a variety of plant species as well as in the toxic secretions of the Sonoran Desert toad; and psilocybin, found in a myriad of fungus species, mostly of the genus Psilocybe. By contrast, lysergic acid diethylamide (LSD), an alkaloid derivative of the fungus ergot, was first synthesized in 1938 and marketed as an adjunct to psychotherapy that could enhance introspection and self-awareness. Indeed, until psychedelics were listed by the U.S. Drug Enforcement Agency as Schedule I drugs in 1970, thousands of studies were published on the therapeutic potential of psychedelics for psychiatric disorders and on their research potential for understanding psychosis (Nichols and Walter, 2020).
Over the past two decades, as human subject research on psychedelics has begun to revive, several small clinical trials have shown the remarkable potential of psychedelics to treat psychiatric disorders (Goldberg et al., 2020; Reiff et al., 2020). A parallel research effort has explored the neural mechanisms of psychedelics for both their profound acute effects on perception and cognition and post–acute effects underlying long-term changes in mental health. In this Perspective, we lay out the case for psychedelics as catalysts of change, linking the experience induced by psychedelics and their effects on neural plasticity. We focus on the current understanding of the cellular and subcellular mechanisms underlying these drugs’ actions. But first, to provide context, we discuss the profound effects these drugs have on the human mind and body.
BEHAVIORAL AND PHYSIOLOGICAL EFFECTS OF PSYCHEDELICS IN HUMAN SUBJECTS
The acute behavioral effects of psychedelics (i.e., the “psychedelic experience”) constitute a profound alteration in consciousness with both pleasant and challenging components (Carbonaro et al., 2016; Roseman et al., 2017). These challenging aspects are mirrored by an acute physiological stress response shortly after drug administration, as indicated by increases in plasma corticosteroid levels, heart rate, and blood pressure (Hasler et al., 2004; Strajhar et al., 2016). Although there are concerns of negative long-term consequences with drugs that produce such powerful effects, there is no evidence for increased incidence of suicidality or psychiatric illnesses in the months following psychedelic experiences; in fact, studies show the opposite (Hendricks et al., 2015; Johansen and Krebs, 2015). Similarly, in spite of widespread recreational use and characterization of psychedelics as “drugs of abuse,” psychedelics present low addictive potential (Garcia-Romeu et al., 2016). Several studies have also demonstrated the safety of psychedelics when administered in controlled settings (Brown et al., 2017).
Multiple clinical trials over the past 20 years have demonstrated the potential of these agents for treating psychiatric disorders (Reiff et al., 2020), including depression and anxiety (Muttoni et al., 2019; Goldberg et al., 2020), substance use (Johnson et al., 2014; Bogenschutz et al., 2015), and obsessive-compulsive disorders (Moreno et al., 2006), with the degree of therapeutic benefit correlating with the intensity of the patient’s psychedelic experience (Roseman et al., 2017). Remarkably, one or two administered doses of psilocybin result in rapid and prolonged changes in mood and outlook, with symptomatic relief lasting at least 3–12 months (Carhart-Harris et al., 2016a; Griffiths et al., 2016; Ross et al., 2016; Johnson et al., 2017). In healthy subjects, psilocybin promotes long-lasting increases in well-being and positive perspective on life experiences (Griffiths et al., 2011; Nicholas et al., 2018). Thus, psychedelics facilitate long-lasting behavioral changes, presumably mediated by long-lasting structural changes in the brain. To understand how that might happen requires a finer-grained exploration of their actions in the brain.
PHARMACOLOGY AND 5-HT 2A RECEPTOR SIGNALING
Psychedelics have a chemical structure similar to serotonin (5-HT), and are divided into two broad categories that differ in their receptor subtype specificity. The indolealkylamines (e.g., LSD, psilocybin, DMT, 5-MeO-DMT) include tryptamine- and lysergic acid derivatives and β-carbolines and exhibit comparatively less specificity for the 5-HT type 2A receptor (5-HT2AR); in particular, they show significant agonism at 5-HT1ARs as well as adrenergic and dopaminergic receptors (Halberstadt and Geyer, 2011; Rickli et al., 2016). The phenylalkylamines (e.g., mescaline, DOM, DOI) bind more specifically to 5-HT2ARs as well as 5-HT2CRs. DOI in particular is commonly used in animal studies, but there are no published studies on its effects in human subjects.
Despite their expansive binding profile, the effects of psychedelics in humans strongly correlate with 5-HT2AR occupancy in the central nervous system (CNS) and are almost entirely blocked by 5-HT2A antagonists (Vollenweider et al., 1998; Valle et al., 2016; Madsen et al., 2019; Preller et al., 2020), thus motivating focus on the 5-HT2AR. Indeed, psychedelics are typically defined as psychoactive agonists of the serotonin 5-HT2A receptor (5-HT2AR). It is important to note, however, that 5-HT2AR antagonists are not perfectly selective either, and there is some evidence suggesting the involvement of other receptors in the neurophysiological effects of psychedelics (Halberstadt and Geyer, 2011; Pokorny et al., 2016). Thus, the involvement of other receptors in the behavioral and therapeutic effects in humans should not be entirely excluded.
Expression of 5-HT2AR mRNA and protein in the brain is widespread. It is most prevalent in excitatory neurons in the neocortex but is also expressed in inhibitory interneurons (Jakab and Goldman-Rakic, 1998; Weber and Andrade, 2010). Importantly, depression and other mood disorders are associated with increased 5-HT2AR density in the brain, especially in the prefrontal cortex (PFC) (Meyer et al., 2003; Bhagwagar et al., 2006), and antidepressant treatments are associated with decreased density (Yatham et al., 1999; Meyer et al., 2001), as is the administration of psychedelics (Buckholtz et al., 1988).
The 5-HT2A receptor is a Class A, rhodopsin-like, G-protein–coupled receptor (Lopez-Gimenez and Gonzalez-Maeso, 2018) for which 5-HT, the endogenous ligand, is a full agonist with a Kd of just over 1 nM (Sleight et al., 1996). Canonically, 5-HT2ARs couple with the G-protein Gq (Figure 1), catalyzing the production of phospholipase C and inositol triphosphate to mobilize intracellular calcium, activate calcineurin, and inhibit type 1.2 voltage-gated calcium channels (Hoyer et al., 1994; Day et al., 2002). In addition, 5-HT2AR signaling through G-proteins from the Gi family and subsequent inhibition of cAMP formation have also been observed (Garnovskaya et al., 1995). Either of these pathways can contribute to neural plasticity (Tedford and Zamponi, 2006; Betke et al., 2012).
FIGURE 1:
Molecular, cellular, and systems support for psychedelic-induced long-term changes. Psychedelic compounds (LSD, psilocin, mescaline) and serotonin bind with high affinity to serotonin 2A receptors. In mammalian systems, direct (solid arrows) and indirect (dashed arrows) consequences of G-protein and β-arrestin signaling downstream of serotonin 2A receptor activation intersect with glutamate release to yield enhanced neural plasticity. The relative engagement of intracellular signaling pathways (arrow weight) is distinct for psychedelics (red arrows) vs. serotonin (blue arrows). Neural plasticity is further supported by neuropeptide synthesis and release, structural changes to neuronal architecture, and altered expression, localization, and phosphorylation of ionotropic glutamate receptors. The consequences of this neural plasticity are modified by psychedelic-induced, large-scale changes in brain activity and attendant perceptual and cognitive changes in the processing of information. When psychedelic drugs are given in the context of a psychotherapeutic support model, these changes appear to ultimately support long-term changes in behavior and promotion of mental well-being.
BIASED AGONISM MODEL FOR PSYCHEDELIC ACTIONS
Psychedelic agonists of 5-HT2AR are associated with profound changes in perception and cognition, whereas other ligands such as 5-HT are not. The basis for this difference is commonly explained using the ternary complex model of receptor activity, which suggests that drug molecules act to shift the equilibrium between a receptor’s different conformational states. As each of these receptor states has a different affinity for various downstream binding partners, this can result in a drug exhibiting “bias,” such as preference for activating G-protein–dependent versus β-arrestin–dependent signaling (Kenakin, 2012). Long-standing evidence indicates that 5-HT2AR signaling efficacy is ligand-dependent and prone to biased agonism across these pathways (Roth et al., 1997; Berg et al., 1998; Fitzgerald et al., 1999; Egan et al., 2000; Cussac et al., 2008; Lopez-Gimenez and Gonzalez-Maeso, 2018). Molecular modeling, site-directed mutagenesis, and x-ray crystallography have identified the distinct psychedelic and nonpsychedelic ligand binding sites that ultimately lead to these differential functional signaling consequences (Wang et al., 1993; Shapiro et al., 2000; Kanagarajadurai et al., 2009).
In the case of Gq-dependent signaling, both psychedelic (DOI) and nonpsychedelic (lisuride) agonists of 5-HT2AR activate several shared downstream pathways, including pPLC, pPKC, pERK, and pCREB; however, the magnitude of these Gq-mediated responses is greater for psychedelic ligands (Figure 1) (Banerjee and Vaidya, 2020). Non-Gq pathways likely contribute to psychedelic effects as well. For example, in mice lacking Gq expression, the response to DOI is blunted but not eliminated (Garcia et al., 2007). Furthermore, psychedelic-dependent phosphoproteomic and transcriptomic signatures are pertussis toxin–sensitive (Gonzalez-Maeso et al., 2007; Karaki et al., 2014), suggesting the involvement of Gi/o signaling. Notably, Gi/o-dependent outcomes may depend on participation of additional signaling partners, as 5-HT2ARs do not appear to participate directly in functional coupling with Gi/o (Kim et al., 2020).
Beyond G-protein–coupled pathways, agonism at 5-HT2ARs can also engage in β-arrestin signaling (Pottie et al., 2020) via PI3K, SRC, and AKT (Figure 1), although the relevance of such β-arrestin–dependent signaling for promoting psychedelic effects is still unclear. For example, while LSD’s ability to promote a β-arrestin–biased conformation at 5-HT2ARs supports its psychedelic effects (Wacker et al., 2017; Kim et al., 2020), other psychedelics are functionally insensitive to β-arrestin knockout in mice (Schmid et al., 2008; Schmid and Bohn, 2010). Close attention to differences in off-target binding profile (Halberstadt and Geyer, 2011; Rickli et al., 2016) and time-dependent evolution in signaling (Wacker et al., 2017) is needed for future studies to yield a more comprehensive picture of β-arrestin–induced effects across the full spectrum of psychedelic ligands.
INTERACTION BETWEEN 5-HT 2AR AND GLUTAMATE RECEPTOR SIGNALING
Glutamate signaling deficits are a prominent feature of schizophrenia (Iwata et al., 2015) and major depressive disorder (Wise et al., 2018) and can be ameliorated by antidepressant administration (Gonzalez-Burgos and Lewis, 2012; Sanacora et al., 2012). Interestingly, psychedelics cause an increase in extracellular glutamate in the PFC (Scruggs et al., 2003; Muschamp et al., 2004), resulting in the release of neurotrophic factors that promote neural plasticity (de Almeida et al., 2019; Holze et al., 2021; Hutten et al., 2021), as well as the activation of ionotropic glutamate receptors, which also contribute to neural plasticity (Lisman et al., 2012).
Glutamate release also activates metabotropic glutamate receptors (mGluR), which regulate neural plasticity (Mukherjee and Manahan-Vaughan, 2013) and modulate the effects of psychedelic agonism at 5-HT2ARs. The agonists for two subtypes, mGluR2 and mGluR3, suppress the response to psychedelics, reducing their effects on neural activity and cellular signaling and the behavioral manifestations of psychedelics, as does (paradoxically) knocking out mGluR2 (Zhai et al., 2003; Gonzalez-Maeso et al., 2008; Moreno et al., 2011; Benvenga et al., 2018).
This interplay between mGluR2 and 5-HT2AR signaling may arise from physical (transmembrane) interactions between the receptors (Gonzalez-Maeso et al., 2008), which are expressed within close molecular proximity (Marek et al., 2000; Hanks and González-Maeso, 2013). Further, several studies indicate that 5-HT2AR and mGluR2 form heterodimeric complexes, integrating glutamatergic and serotonergic signaling and modulating subsequent G-protein coupling and downstream effects (Zhai et al., 2003; Gonzalez-Maeso et al., 2008; Moreno et al., 2016). For example, when cells coexpressing mGluR2 and 5-HT2AR receptors are treated with mGluR2/3 agonists, signaling is propagated through Gq/11, rather than the Gi/o pathway usually activated by mGluR2. This effect is reduced in 5-HT2AR knockout mice (Moreno et al., 2016), suggesting that 5-HT2ARs and mGluR2 and mGluR3 can form heterocomplexes that have a distinct signaling profile. Importantly, signaling through this complex may facilitate the rapid antidepressant effects observed with psychedelics, as mGluR2 and mGluR3 have been implicated in the regulation of neural plasticity in the PFC, amygdala, and hippocampus, brain regions implicated in mood disorders, along with fear- and stress-associated learning (Walker et al., 2015).
CELLULAR AND NETWORK-LEVEL ELECTROPHYSIOLOGICAL EFFECTS OF PSYCHEDELICS
The signaling pathways activated by psychedelic agonism at the 5-HT2AR provide a basis for structural changes in the brain following the psychedelic experience (Figure 1). How these long-term changes relate to the acute effects of psychedelics is a critical but unresolved question. Psychedelics acutely modulate neural activity and connectivity, and these changes correlate with the phenomenology of the psychedelic experience (de Araujo et al., 2012; Carhart-Harris et al., 2016b; Valle et al., 2016). Effects on neural activity are broadly consistent with an increase in excitability (Muthukumaraswamy et al., 2013; Kometer et al., 2015; Valle et al., 2016; Timmermann et al., 2019) and a switch from externally driven to internally driven neural activity in sensory areas of the brain (Bravermanova et al., 2018; Timmermann et al., 2018; Michaiel et al., 2019). Psychedelics increase brain signal complexity (an indirect measure of signal diversity and information content), and these changes also correlate with specific aspects of the psychedelic experience (Schartner et al., 2017). These changes in complexity likely reflect increased information flow along pathways that are not typically engaged in the absence of the drug. Similarly, synesthesia, in which sensory stimuli in one modality trigger sensations in another, is a common element of the psychedelic experience (Studerus et al., 2010; Schmid et al., 2015), thought to rely on existing but dormant connections between areas of the brain that typically process distinct sensations, for example, vision or audition (Maurer et al., 2020).
Experiments confirm that psychedelics alter connectivity in the brain (Tagliazucchi et al., 2016; Preller et al., 2018, 2019), and this form of neural plasticity may underlie their therapeutic activity. For example, excessive amygdala reactivity and decreased connectivity between the amygdala and other brain regions, especially the PFC, are linked to psychiatric disorders (Quirk and Gehlert, 2003; Negron-Oyarzo et al., 2016). Psychedelics alter connectivity between the amygdala and key brain regions during emotional stimulus processing, effects that may contribute to altered behavioral and neural responses to these stimuli characteristic of mood disorders (Rocha et al., 2019; Barrett et al., 2020a). Psychedelics acutely reduce connectivity within the default mode network (Carhart-Harris et al., 2012; Palhano-Fontes et al., 2015), a set of brain regions whose activity and connectivity correlate with self-referential mental activity in healthy subjects (Davey et al., 2016) and rumination and depression symptoms in patients (Sheline et al., 2009; Berman et al., 2011). More broadly, psychedelics administered to healthy volunteers alter connectivity in multiple higher-order brain regions (Roseman et al., 2014; Tagliazucchi et al., 2016; Sampedro et al., 2017; Preller et al., 2018, 2020; Barrett et al., 2020b). Thus, psychedelics induce acute changes in connectivity in brain regions that are implicated in multiple psychiatric disorders, but the mechanisms of these changes and whether these changes persist postacutely remain areas of active inquiry (Barrett et al., 2020a).
NEURAL AND BEHAVIORAL PLASTICITY
The psychedelic-induced changes in neural activity and connectivity in human subjects described above are manifestations of acute effects on neural plasticity. These acute effects have also been studied in animal models, where DOI acts on both pre- and postsynaptic 5-HT2ARs to modulate glutamate signaling (Barre et al., 2016; Berthoux et al., 2019). Rodent models have also revealed long-term structural changes induced by psychedelics. Many of these changes manifest in structural changes at synapses, such as the growth of neurites, the cellular processes that connect neurons, and changes in dendritic spines, the specialized postsynaptic structures found most commonly at excitatory synapses on excitatory neurons (Holtmaat and Svoboda, 2009). Studies of cultured cortical neurons indicate that DOI acts on 5-HT2ARs to increase dendritic spine diameter (Jones et al., 2009). Furthermore, psychedelics enhance de novo neurite and dendritic spine formation through the TrkB, mTOR, and 5-HT2A pathway (Ly et al., 2018), all implicated in promoting neuroplastic changes in the PFC (Meunier et al., 2017). Finally, psychedelics induce neurogenesis in the rodent hippocampus (Catlow et al., 2013; Morales-Garcia et al., 2017; Lima da Cruz et al., 2018).
These long-term effects of psychedelics are likely mediated at least in part by changes in gene expression (Martin and Nichols, 2018). A single administered dose of psilocybin induces changes in neural plasticity–related gene expression in the hippocampus and PFC (Jefsen et al., 2020), and 5-MeO-DMT induces large-scale changes in protein expression in cultured human brain tissue (Dakic et al., 2017). Psychedelics interact particularly with signaling by the neural plasticity–associated protein brain-derived neurotrophic factor (BDNF). DOI acts on 5-HT2ARs to modulate the level of BDNF mRNA in the neocortex and hippocampus, likely via an activity-dependent mechanism and CREB signaling (Vaidya et al., 1997; Musazzi et al., 2014; Jaggar and Vaidya, 2018). DOI increases expression of mRNA for the neuroplasticity-associated protein Arc (Korb and Finkbeiner, 2011) via a pathway that depends on glutamate and BDNF signaling (Pei et al., 2000, 2004; Benekareddy et al., 2013). Psychedelics trigger increases in plasma levels of BDNF in human subjects (de Almeida et al., 2019; Holze et al., 2021; Hutten et al., 2021). Thus, psychedelics are able to mobilize multiple signaling pathways to promote neuroplastic changes in the brain. Given the importance in clinical trials of post–acute psychotherapy sessions, and the extended time course of some of the signaling cascades mentioned here, it seems likely that the pro-neuroplastic effects of psychedelics extend beyond the dosing session, that is, that psychedelics open a window of neural plasticity that lasts for days or weeks. However, this idea has not yet been tested experimentally.
Consistent with these effects on neural plasticity at the cellular level, there is considerable evidence that psychedelics modify learning and memory in vivo (Zhang and Stackman, 2015; Healy, 2021). This is likely a consequence of the role in cognitive function of 5-HT2ARs, whose activation enhances various types of learning and memory (Williams et al., 2002; Harvey, 2003). For example, psychedelics enhance both the acquisition and extinction of fear conditioning in rodents (Catlow et al., 2013; Zhang et al., 2013). These studies are important in two regards: First, they suggest that psychedelics can promote long-term changes in behavior in the absence of preconceived notions about their potential therapeutic benefit, an emerging concern in clinical trials (Noorani, 2020). Second, they demonstrate that psychedelics have the potential to support learning of new behaviors, while also facilitating extinction of older learned behaviors. This suggests a window for enhanced but nonspecific neural plasticity that can be exploited for therapeutic benefit in partnership with mental health professionals.
STRESS AND INFLAMMATION
The intersection between psychiatric disorders, stress, and psychedelics is an emerging area of interest (Murnane, 2019; Brouwer and Carhart-Harris, 2020). Chronic stress is a major precipitating factor in the etiology of many psychiatric disorders that psychedelics have shown clinical efficacy in treating (McEwen, 2004). Animal models have shown that chronic stress induces behavioral and neural changes that can be reversed by antidepressants (Campos et al., 2013; Moda-Sava et al., 2019; Planchez et al., 2019). Thus, it is both surprising and intriguing that acute stress may play a role in promoting the neuroplastic effects of psychedelics in the context of treating these same disorders. Like psychedelics, acute stress is pro-neuroplastic (Huang et al., 2005; Cadle and Zoladz, 2015), and signaling at the 5-HT2AR plays a role in this neural plasticity (Murnane, 2019). Along with this convergence on neuroplastic mechanisms to modify behavior, psychedelics trigger an acute biochemical stress response consisting of catecholamine and glucocorticoid release (Hemrick-Luecke and Evans, 2002; Hasler et al., 2004; Galvão et al., 2018), and it has been postulated that this stress response is critical for the transformative nature of the psychedelic experience (Brouwer and Carhart-Harris, 2020).
The mechanism underlying this stress response is unclear, but there are two obvious (and not necessarily mutually exclusive) possibilities. First, psychedelics may act directly at 5-HT2ARs in the hypothalamus to induce expression and/or release of corticotrophin-releasing hormone, elevating plasma concentrations of stress-associated glucocorticoids (Van de Kar et al., 2001; Jorgensen et al., 2002). This possibility is consistent with the established regulatory role of 5-HT in the hypothalamic–pituitary–adrenal axis, the primary system involved in stress regulation (Contesse et al., 2000). Alternatively, the psychedelic-induced altered state of consciousness itself may trigger the acute stress response, as such states frequently include components that are challenging (e.g., fear- or anxiety-provoking). This raises the possibility that the pro-neuroplastic effects of psychedelics depend in part on the acute stress response that they induce, but whether the stressful components of the psychedelic experience are hindrances to or foundational for therapeutic benefit is in dispute (Carbonaro et al., 2016; Roseman et al., 2017; Wolff et al., 2020).
The therapeutic and pro-neuroplastic effects of psychedelics may also be linked to their anti-inflammatory action (Inserra et al., 2021). Inflammation and immune system alterations are commonly associated with psychiatric disorders (Beurel et al., 2020; Thompson and Szabo, 2020), and 5-HT plays a key role immune system function (Herr et al., 2017). Recent work shows that psychedelics have potent inhibitory effects on cytokines, which are immune signaling molecules that directly modulate synaptic function and neural plasticity (Wang et al., 2012; Vezzani and Viviani, 2015). 5-MeO-DMT acutely reduces levels of proinflammatory cytokines in humans (Uthaug et al., 2020), and DOI has been shown to inhibit peripheral cytokine cascades through 5-HT2AR activation (Yu et al., 2008; Nau et al., 2013). These data suggest that psychedelics may be useful in treating inflammatory diseases generally, but there may be more direct effects on inflammation in the brain as well. Microglia, the resident immune cells in the brain, express several 5-HT receptor subtypes (Krabbe et al., 2012; Glebov et al., 2015) and respond directly to DOI treatment (Krabbe et al., 2012). This raises the possibility that systemically administered psychedelics could directly modulate microglial function, which becomes aberrant in mood disorders (Haroon et al., 2017) as well as in neurodegenerative disorders (Perry and Holmes, 2014). The therapeutic potential for the anti-inflammatory effects of psychedelics is just beginning to be explored, including a phase 1 clinical trial in healthy older volunteers with the goal of using LSD to treat Alzheimer’s disease (Family et al., 2020).
GLOSSARY
Amygdala is a region of the brain involved in regulation and expression of emotions, as well as memory and decision making. It is tightly coupled to the hippocampus and prefrontal cortex.
Connectivity refers to the connection strength between neurons, groups of neurons, or regions of the brain.
Dendritic spines are protrusions from the dendrites of some types of neurons (usually excitatory cells) that are the loci of excitatory synaptic terminals. Changes in spine size are indicative of changes in synaptic strength.
Glutamate signaling refers to communication between neurons via the neurotransmitter glutamate. Glutamate receptors can be ionotropic, that is, ligand-gated ion channels, or metabotropic, that is, G-protein–coupled receptors. Glutamate signaling is commonly modified by neural plasticity.
G-protein–coupled receptors are proteins that span the plasma membrane of cells (e.g., neurons) that induce intracellular signaling changes in response to extracellular chemical signals.
Hippocampus is a region of the brain located near the amygdala that is involved in memory formation and retrieval. It is tightly coupled to the amygdala and prefrontal cortex.
Neocortex is a mammal-specific part of the brain that is organized into regions with specific functions including sensation, speech and language production, planning, and reason.
Neural plasticity (adj. neuroplastic) refers to changes in connectivity between neurons, including the amount of neurotransmitter released or the magnitude of response to the neurotransmitter. Proneuroplastic effects refer to increases in neural plasticity.
Neurogenesis is the process of creating new neurons in the brain. In humans, neurogenesis is generally limited following the completion of embryonic brain development
Prefrontal cortex (PFC) is a region in the neocortex that is highly developed in primates and especially humans. It contributes to attention, executive control, regulation of mood and emotion, and monitoring the current state of the body.
Psychedelic experience refers to the acute effects of psychedelics while the drug is present in the CNS. This experience can include distorted perception of time, altered sensory perception, heightened emotional response to music, hallucinations, altered sense of self, ego dissolution, and a sense of connection to a larger truth or absolute reality not typically glimpsed in everyday life (the so-called “mystical experience”). Challenging aspects of the psychedelic experience include anxiety, fear, and confusion.
CONCLUSIONS
As psychedelic science emerges from the shadows and attracts new resources, further research will deepen our understanding and advance the responsible therapeutic use of these compounds. Psychedelics induce acute effects on gene expression, neurotransmitter and neuroendocrine release, neural activity, connectivity, and perception and cognition, ultimately leading to profound long-term effects on mood and behavior (Figure 1). Elucidating the mechanisms that connect the acute effects of psychedelics to these long-term changes is critical but is complicated by our limited mechanistic understanding of the psychiatric disorders that these drugs are effective at treating. Thus, understanding how these drugs work in clinical settings may also provide needed insight into the neurobiological basis for psychiatric illnesses. Within the context of psychedelics as change agents, an important next step is to identify long-term changes in brain structure that relate to therapeutic activity, which will require support from both clinical and preclinical research programs.
Studies should focus on neural plasticity, both its locus and acute and long-term mechanisms. Critical unresolved questions include the following. Do psychedelics open a window of neural plasticity? If so, is this effect nonspecific or focused in regions of the brain that are particularly relevant to behavioral phenotypes associated with human psychiatric disorders? How long does the window last? Which signaling pathways are essential for facilitating neural plasticity? Are there alternative strategies for promoting neural plasticity that may translate into more effective therapeutic interventions in patients? Despite (or perhaps because of) all the data accumulated so far, the field is left with many more questions than it started with. We predict that over the next decade, our understanding of the therapeutic potential of psychedelics will be considerably expanded.
Abbreviations used:
- AKT
protein kinase B
- BDNF
brain-derived neurotrophic factor
- cAMP
cyclic adenosine monophosphate
- CREB
cyclic adenosine monophosphate response element-binding protein
- DMT
N, N-dimethyltryptamine
- DOI
2,5-dimethoxy-4-iodoamphetamine
- DOM
2,5-dimethoxy-4-methylamphetamine
- 5-HT
serotonin
- 5-HT 1AR
serotonin type 1A receptor
- 5-HT 2AR
serotonin type 2A receptor
- 5-HT 2CR
serotonin type 2C receptor
- 5-MeO-DMT
5-Methoxy- N, N-dimethyltryptamine
- K d
dissociation constant
- LSD
lysergic acid diethylamide
- mGluR
metabotropic glutamate receptor
- mRNA
messenger ribonucleic acid
- mTOR
mechanistic target of rapamycin kinase
- pCREB
phosphorylated cyclic adenosine monophosphate response element-binding protein
- pERK
phosphorylated extracellular signal-regulated kinase
- PFC
prefrontal cortex
- PI3K
phosphatidylinositol 3-kinase
- pPKC
phosphorylated protein kinase C
- pPLC
phosphorylated phospholipase C
- SRC
sarcoma proto-oncogene, non-receptor tyrosine kinase
- TrkB
tropomyosin receptor kinase B.
Footnotes
REFERENCES
- Banerjee AA, Vaidya VA (2020). Differential signaling signatures evoked by DOI versus lisuride stimulation of the 5-HT(2A) receptor. Biochem Biophys Res Commun 531, 609–614. [DOI] [PubMed] [Google Scholar]
- Barre A, Berthoux C, De Bundel D, Valjent E, Bockaert J, Marin P, Becamel C (2016). Presynaptic serotonin 2A receptors modulate thalamocortical plasticity and associative learning. Proc Natl Acad Sci USA 113, E1382–E1391. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Barrett FS, Doss MK, Sepeda ND, Pekar JJ, Griffiths RR (2020a). Emotions and brain function are altered up to one month after a single high dose of psilocybin. Sci Rep 10, 2214. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Barrett FS, Krimmel SR, Griffiths RR, Seminowicz DA, Mathur BN (2020b). Psilocybin acutely alters the functional connectivity of the claustrum with brain networks that support perception, memory, and attention. Neuroimage 218, 116980. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Benekareddy M, Nair AR, Dias BG, Suri D, Autry AE, Monteggia LM, Vaidya VA (2013). Induction of the plasticity-associated immediate early gene Arc by stress and hallucinogens: role of brain-derived neurotrophic factor. Int J Neuropsychopharmacol 16, 405–415. [DOI] [PubMed] [Google Scholar]
- Benvenga MJ, Chaney SF, Baez M, Britton TC, Hornback WJ, Monn JA, Marek GJ (2018). Metabotropic glutamate(2) receptors play a key role in modulating head twitches induced by a serotonergic hallucinogen in mice. Front Pharmacol 9, 208. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Berg KA, Maayani S, Goldfarb J, Scaramellini C, Leff P, Clarke WP (1998). Effector pathway-dependent relative efficacy at serotonin type 2A and 2C receptors: evidence for agonist-directed trafficking of receptor stimulus. Mol Pharmacol 54, 94–104. [PubMed] [Google Scholar]
- Berman MG, Peltier S, Nee DE, Kross E, Deldin PJ, Jonides J (2011). Depression, rumination and the default network. Soc Cogn Affect Neurosci 6, 548–555. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Berthoux C, Barre A, Bockaert J, Marin P, Becamel C (2019). Sustained activation of postsynaptic 5-HT2A receptors gates plasticity at prefrontal cortex synapses. Cereb Cortex 29, 1659–1669. [DOI] [PubMed] [Google Scholar]
- Betke KM, Wells CA, Hamm HE (2012). GPCR mediated regulation of synaptic transmission. Prog Neurobiol 96, 304–321. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Beurel E, Toups M, Nemeroff CB (2020). The bidirectional relationship of depression and inflammation: double trouble. Neuron 107, 234–256. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Bhagwagar Z, Hinz R, Taylor M, Fancy S, Cowen P, Grasby P (2006). Increased 5-HT(2A) receptor binding in euthymic, medication-free patients recovered from depression: a positron emission study with [(11)C]MDL 100,907. Am J Psychiatry 163, 1580–1587. [DOI] [PubMed] [Google Scholar]
- Bogenschutz MP, Forcehimes AA, Pommy JA, Wilcox CE, Barbosa PC, Strassman RJ (2015). Psilocybin-assisted treatment for alcohol dependence: a proof-of-concept study. J Psychopharmacol 29, 289–299. [DOI] [PubMed] [Google Scholar]
- Bravermanova A, Viktorinova M, Tyls F, Novak T, Androvicova R, Korcak J, Horacek J, Balikova M, Griskova-Bulanova I, Danielova D, et al. (2018). Psilocybin disrupts sensory and higher order cognitive processing but not pre-attentive cognitive processing-study on P300 and mismatch negativity in healthy volunteers. Psychopharmacology (Berl) 235, 491–503. [DOI] [PubMed] [Google Scholar]
- Brouwer A, Carhart-Harris RL (2020). Pivotal mental states. J Psychopharmacol, 269881120959637. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Brown RT, Nicholas CR, Cozzi NV, Gassman MC, Cooper KM, Muller D, Thomas CD, Hetzel SJ, Henriquez KM, Ribaudo AS, Hutson PR (2017). Pharmacokinetics of escalating doses of oral psilocybin in healthy adults. Clin Pharmacokinet 56, 1543–1554. [DOI] [PubMed] [Google Scholar]
- Buckholtz NS, Zhou DF, Freedman DX (1988). Serotonin2 agonist administration down-regulates rat brain serotonin2 receptors. Life Sci 42, 2439–2445. [DOI] [PubMed] [Google Scholar]
- Cadle CE, Zoladz PR (2015). Stress time-dependently influences the acquisition and retrieval of unrelated information by producing a memory of its own. Front Psychol 6, 910. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Campos AC, Fogaca MV, Aguiar DC, Guimaraes FS (2013). Animal models of anxiety disorders and stress. Braz J Psychiatry 35(Suppl 2), S101–S111. [DOI] [PubMed] [Google Scholar]
- Carbonaro TM, Bradstreet MP, Barrett FS, MacLean KA, Jesse R, Johnson MW, Griffiths RR (2016). Survey study of challenging experiences after ingesting psilocybin mushrooms: acute and enduring positive and negative consequences. J Psychopharmacol 30, 1268–1278. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Carhart-Harris RL, Bolstridge M, Rucker J, Day CM, Erritzoe D, Kaelen M, Bloomfield M, Rickard JA, Forbes B, Feilding A, et al. (2016a). Psilocybin with psychological support for treatment-resistant depression: an open-label feasibility study. Lancet Psychiatry 3, 619–627. [DOI] [PubMed] [Google Scholar]
- Carhart-Harris RL, Erritzoe D, Williams T, Stone JM, Reed LJ, Colasanti A, Tyacke RJ, Leech R, Malizia AL, Murphy K, et al. (2012). Neural correlates of the psychedelic state as determined by fMRI studies with psilocybin. Proc Natl Acad Sci USA 109, 2138–2143. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Carhart-Harris RL, Muthukumaraswamy S, Roseman L, Kaelen M, Droog W, Murphy K, Tagliazucchi E, Schenberg EE, Nest T, Orban C, et al. (2016b). Neural correlates of the LSD experience revealed by multimodal neuroimaging. Proc Natl Acad Sci USA 113, 4853–4858. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 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, 481–491. [DOI] [PubMed] [Google Scholar]
- Contesse V, Lefebvre H, Lenglet S, Kuhn JM, Delarue C, Vaudry H (2000). Role of 5-HT in the regulation of the brain-pituitary-adrenal axis: effects of 5-HT on adrenocortical cells. Can J Physiol Pharmacol 78, 967–983. [PubMed] [Google Scholar]
- Cussac D, Boutet-Robinet E, Ailhaud MC, Newman-Tancredi A, Martel JC, Danty N, Rauly-Lestienne I (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. Eur J Pharmacol 594, 32–38. [DOI] [PubMed] [Google Scholar]
- Dakic V, Minardi Nascimento J, Costa Sartore R, Maciel RM, de Araujo DB, Ribeiro S, Martins-de-Souza D, Rehen SK (2017). Short term changes in the proteome of human cerebral organoids induced by 5-MeO-DMT. Sci Rep 7, 12863. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Davey CG, Pujol J, Harrison BJ (2016). Mapping the self in the brain’s default mode network. Neuroimage 132, 390–397. [DOI] [PubMed] [Google Scholar]
- Day M, Olson PA, Platzer J, Striessnig J, Surmeier DJ (2002). Stimulation of 5-HT(2) receptors in prefrontal pyramidal neurons inhibits Ca(v)1.2 L type Ca(2+) currents via a PLCbeta/IP3/calcineurin signaling cascade. J Neurophysiol 87, 2490–2504. [DOI] [PubMed] [Google Scholar]
- de Almeida RN, Galvão ACM, da Silva FS, Silva E, Palhano-Fontes F, Maia-de-Oliveira JP, de Araújo LB, Lobão-Soares B, Galvão-Coelho NL (2019). Modulation of serum brain-derived neurotrophic factor by a single dose of ayahuasca: observation from a randomized controlled trial. Front Psychol 10, 1234. [DOI] [PMC free article] [PubMed] [Google Scholar]
- de Araujo DB, Ribeiro S, Cecchi GA, Carvalho FM, Sanchez TA, Pinto JP, de Martinis BS, Crippa JA, Hallak JE, Santos AC (2012). Seeing with the eyes shut: neural basis of enhanced imagery following ayahuasca ingestion. Hum Brain Mapp 33, 2550–2560. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Egan C, Grinde E, Dupre A, Roth BL, Hake M, Teitler M, Herrick-Davis K (2000). Agonist high and low affinity state ratios predict drug intrinsic activity and a revised ternary complex mechanism at serotonin 5-HT(2A) and 5-HT(2C) receptors. Synapse 35, 144–150. [DOI] [PubMed] [Google Scholar]
- Family N, Maillet EL, Williams LTJ, Krediet E, Carhart-Harris RL, Williams TM, Nichols CD, Goble DJ, Raz S (2020). Safety, tolerability, pharmacokinetics, and pharmacodynamics of low dose lysergic acid diethylamide (LSD) in healthy older volunteers. Psychopharmacology (Berl) 237, 841–853. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Fitzgerald LW, Conklin DS, Krause CM, Marshall AP, Patterson JP, Tran DP, Iyer G, Kostich WA, Largent BL, Hartig PR (1999). High-affinity agonist binding correlates with efficacy (intrinsic activity) at the human serotonin 5-HT2A and 5-HT2C receptors: evidence favoring the ternary complex and two-state models of agonist action. J Neurochem 72, 2127–2134. [DOI] [PubMed] [Google Scholar]
- Froese T, Guzmán G, Guzmán-Dávalos L (2016). On the origin of the genus psilocybe and its potential ritual use in ancient Africa and Europe. Econ Bot 70, 103–114. [Google Scholar]
- Galvão ACM, de Almeida RN, Silva E, Freire FAM, Palhano-Fontes F, Onias H, Arcoverde E, Maia-de-Oliveira JP, de Araújo DB, Lobão-Soares B, Galvão-Coelho NL (2018). Cortisol modulation by ayahuasca in patients with treatment resistant depression and healthy controls. Front Psychiatry 9, 185. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Garcia EE, Smith RL, Sanders-Bush E (2007). Role of G(q) protein in behavioral effects of the hallucinogenic drug 1-(2,5-dimethoxy-4-iodophenyl)-2-aminopropane. Neuropharmacology 52, 1671–1677. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Garcia-Romeu A, Kersgaard B, Addy PH (2016). Clinical applications of hallucinogens: a review. Exp Clin Psychopharmacol 24, 229–268. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Garnovskaya MN, Nebigil CG, Arthur JM, Spurney RF, Raymond JR (1995). 5-Hydroxytryptamine2A receptors expressed in rat renal mesangial cells inhibit cyclic AMP accumulation. Mol Pharmacol 48, 230–237. [PubMed] [Google Scholar]
- Glebov K, Löchner M, Jabs R, Lau T, Merkel O, Schloss P, Steinhäuser C, Walter J (2015). Serotonin stimulates secretion of exosomes from microglia cells. Glia 63, 626–634. [DOI] [PubMed] [Google Scholar]
- Goldberg SB, Pace BT, Nicholas CR, Raison CL, Hutson PR (2020). The experimental effects of psilocybin on symptoms of anxiety and depression: a meta-analysis. Psychiatry Res 284, 112749. [DOI] [PubMed] [Google Scholar]
- Gonzalez-Burgos G, Lewis DA (2012). NMDA receptor hypofunction, parvalbumin-positive neurons, and cortical gamma oscillations in schizophrenia. Schizophr Bull 38, 950–957. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Gonzalez-Maeso J, Ang RL, Yuen T, Chan P, Weisstaub NV, Lopez-Gimenez JF, Zhou M, Okawa Y, Callado LF, Milligan G, et al. (2008). Identification of a serotonin/glutamate receptor complex implicated in psychosis. Nature 452, 93–97. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Gonzalez-Maeso J, Weisstaub NV, Zhou M, Chan P, Ivic L, Ang R, Lira A, Bradley-Moore M, Ge Y, Zhou Q, et al. (2007). Hallucinogens recruit specific cortical 5-HT(2A) receptor-mediated signaling pathways to affect behavior. Neuron 53, 439–452. [DOI] [PubMed] [Google Scholar]
- Griffiths RR, Johnson MW, Carducci MA, Umbricht A, Richards WA, Richards BD, Cosimano MP, Klinedinst MA (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, 1181–1197. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Griffiths RR, Johnson MW, Richards WA, Richards BD, McCann U, Jesse R (2011). Psilocybin occasioned mystical-type experiences: immediate and persisting dose-related effects. Psychopharmacology (Berl) 218, 649–665. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Guerra-Doce E (2015). Psychoactive substances in prehistoric times: examining the archaeological evidence. Time Mind 8, 91–112. [Google Scholar]
- Halberstadt AL, Geyer MA (2011). Multiple receptors contribute to the behavioral effects of indoleamine hallucinogens. Neuropharmacology 61, 364–381. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Hanks JB, González-Maeso J (2013). Animal models of serotonergic psychedelics. ACS Chem Neurosci 4, 33–42. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Haroon E, Miller AH, Sanacora G (2017). Inflammation, glutamate, and glia: a trio of trouble in mood disorders. Neuropsychopharmacology 42, 193–215. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Harvey JA (2003). Role of the serotonin 5-HT(2A) receptor in learning. Learn Mem 10, 355–362. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Hasler F, Grimberg U, Benz MA, Huber T, Vollenweider FX (2004). Acute psychological and physiological effects of psilocybin in healthy humans: a double-blind, placebo-controlled dose-effect study. Psychopharmacology (Berl) 172, 145–156. [DOI] [PubMed] [Google Scholar]
- Healy CJ (2021). The acute effects of classic psychedelics on memory in humans. Psychopharmacology (Berl) 238, 639–653. [DOI] [PubMed] [Google Scholar]
- Hemrick-Luecke SK, Evans DC (2002). Comparison of the potency of MDL 100,907 and SB 242084 in blocking the serotonin (5-HT)(2) receptor agonist-induced increases in rat serum corticosterone concentrations: evidence for 5-HT(2A) receptor mediation of the HPA axis. Neuropharmacology 42, 162–169. [DOI] [PubMed] [Google Scholar]
- Hendricks PS, Thorne CB, Clark CB, Coombs DW, Johnson MW (2015). Classic psychedelic use is associated with reduced psychological distress and suicidality in the United States adult population. J Psychopharmacol 29, 280–288. [DOI] [PubMed] [Google Scholar]
- Herr N, Bode C, Duerschmied D (2017). The effects of serotonin in immune cells. Front Cardiovasc Med 4, 48. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Holtmaat A, Svoboda K (2009). Experience-dependent structural synaptic plasticity in the mammalian brain. Nat Rev Neurosci 10, 647–658. [DOI] [PubMed] [Google Scholar]
- Holze F, Vizeli P, Ley L, Muller F, Dolder P, Stocker M, Duthaler U, Varghese N, Eckert A, Borgwardt S, Liechti ME (2021). Acute dose-dependent effects of lysergic acid diethylamide in a double-blind placebo-controlled study in healthy subjects. Neuropsychopharmacology 46, 537–544. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Hoyer D, Clarke DE, Fozard JR, Hartig PR, Martin GR, Mylecharane EJ, Saxena PR, Humphrey PP (1994). International Union of Pharmacology classification of receptors for 5-hydroxytryptamine (serotonin). Pharmacol Rev 46, 157–203. [PubMed] [Google Scholar]
- Huang CC, Yang CH, Hsu KS (2005). Do stress and long-term potentiation share the same molecular mechanisms? Mol Neurobiol 32, 223–235. [DOI] [PubMed] [Google Scholar]
- Hutten NRPW, Mason NL, Dolder PC, Theunissen EL, Holze F, Liechti ME, Varghese N, Eckert A, Feilding A, Ramaekers JG, Kuypers KPC (2021). Low doses of LSD acutely increase BDNF blood plasma levels in healthy volunteers. ACS Pharmacol Transl Sci 4, 461–466. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Inserra A, De Gregorio D, Gobbi G (2021). Psychedelics in psychiatry: neuroplastic, immunomodulatory, and neurotransmitter mechanisms. Pharmacol Rev 73, 202–277. [DOI] [PubMed] [Google Scholar]
- Iwata Y, Nakajima S, Suzuki T, Keefe RS, Plitman E, Chung JK, Caravaggio F, Mimura M, Graff-Guerrero A, Uchida H (2015). Effects of glutamate positive modulators on cognitive deficits in schizophrenia: a systematic review and meta-analysis of double-blind randomized controlled trials. Mol Psychiatry 20, 1151–1160. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Jaggar M, Vaidya V (2018). 5-HT2A receptors and BDNF regulation: implications for psychopathology. In: 5-HT2A Receptors in the Central Nervous System. The Receptors, vol. 32, ed Guiard B., Di Giovanni G., Cham, Switzerland: Humana Press, 395–438. [Google Scholar]
- Jakab RL, Goldman-Rakic PS (1998). 5-Hydroxytryptamine2A serotonin receptors in the primate cerebral cortex: possible site of action of hallucinogenic and antipsychotic drugs in pyramidal cell apical dendrites. Proc Natl Acad Sci USA 95, 735–740. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Jefsen OH, Elfving B, Wegener G, Müller HK (2020). Transcriptional regulation in the rat prefrontal cortex and hippocampus after a single administration of psilocybin. J Psychopharmacol, 0269881120959614. [DOI] [PubMed] [Google Scholar]
- Johansen P, Krebs TS (2015). Psychedelics not linked to mental health problems or suicidal behavior: a population study. J Psychopharmacol 29, 270–279. [DOI] [PubMed] [Google Scholar]
- Johnson MW, Garcia-Romeu A, Cosimano MP, Griffiths RR (2014). Pilot study of the 5-HT2AR agonist psilocybin in the treatment of tobacco addiction. J Psychopharmacol 28, 983–992. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Johnson MW, Garcia-Romeu A, Griffiths RR (2017). Long-term follow-up of psilocybin-facilitated smoking cessation. Am J Drug Alcohol Abuse 43, 55–60. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 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 USA 106, 19575–19580. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Jorgensen H, Knigge U, Kjaer A, Moller M, Warberg J (2002). Serotonergic stimulation of corticotropin-releasing hormone and pro-opiomelanocortin gene expression. J Neuroendocrinol 14, 788–795. [DOI] [PubMed] [Google Scholar]
- Kanagarajadurai K, Malini M, Bhattacharya A, Panicker MM, Sowdhamini R (2009). Molecular modeling and docking studies of human 5-hydroxytryptamine 2A (5-HT2A) receptor for the identification of hotspots for ligand binding. Mol Biosyst 5, 1877–1888. [DOI] [PubMed] [Google Scholar]
- Karaki S, Becamel C, Murat S, Mannoury la Cour C, Millan MJ, Prézeau L, Bockaert J, Marin P, Vandermoere F (2014). Quantitative phosphoproteomics unravels biased phosphorylation of serotonin 2A receptor at Ser280 by hallucinogenic versus nonhallucinogenic agonists. Mol Cell Proteomics 13, 1273–1285. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Kenakin TP (2012). Biased signalling and allosteric machines: new vistas and challenges for drug discovery. Br J Pharmacol 165, 1659–1669. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Kim K, Che T, Panova O, DiBerto JF, Lyu J, Krumm BE, Wacker D, Robertson MJ, Seven AB, Nichols DE, et al. (2020). Structure of a hallucinogen-activated Gq-coupled 5-HT(2A) serotonin receptor. Cell 182, 1574–1588.e1519. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Kometer M, Pokorny T, Seifritz E, Volleinweider FX (2015). Psilocybin-induced spiritual experiences and insightfulness are associated with synchronization of neuronal oscillations. Psychopharmacology (Berl) 232, 3663–3676. [DOI] [PubMed] [Google Scholar]
- Korb E, Finkbeiner S (2011). Arc in synaptic plasticity: from gene to behavior. Trends Neurosci 34, 591–598. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Krabbe G, Matyash V, Pannasch U, Mamer L, Boddeke HW, Kettenmann H (2012). Activation of serotonin receptors promotes microglial injury-induced motility but attenuates phagocytic activity. Brain Behav Immun 26, 419–428. [DOI] [PubMed] [Google Scholar]
- Lima da Cruz RV, Moulin TC, Petiz LL, Leao 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]
- Lisman J, Yasuda R, Raghavachari S (2012). Mechanisms of CaMKII action in long-term potentiation. Nat Rev Neurosci 13, 169–182. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Lopez-Gimenez JF, Gonzalez-Maeso J (2018). Hallucinogens and serotonin 5-HT2A receptor-mediated signaling pathways. Curr Top Behav Neurosci 36, 45–73. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Ly C, Greb AC, Cameron LP, Wong JM, Barragan EV, Wilson PC, Burbach KF, Soltanzadeh Zarandi S, Sood A, Paddy MR, et al. (2018). Psychedelics promote structural and functional neural plasticity. Cell Rep 23, 3170–3182. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Madsen MK, Fisher PM, Burmester D, Dyssegaard A, Stenbaek DS, Kristiansen S, Johansen SS, Lehel S, Linnet K, Svarer C, et al. (2019). Psychedelic effects of psilocybin correlate with serotonin 2A receptor occupancy and plasma psilocin levels. Neuropsychopharmacology 44, 1328–1334. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Marek GJ, Wright RA, Schoepp DD, Monn JA, Aghajanian GK (2000). Physiological antagonism between 5-hydroxytryptamine(2A) and group II metabotropic glutamate receptors in prefrontal cortex. J Pharmacol Exp Ther 292, 76–87. [PubMed] [Google Scholar]
- Martin DA, Nichols CD (2018). The effects of hallucinogens on gene expression. Curr Top Behav Neurosci 36, 137–158. [DOI] [PubMed] [Google Scholar]
- Maurer D, Ghloum JK, Gibson LC, Watson MR, Chen LM, Akins K, Enns JT, Hensch TK, Werker JF (2020). Reduced perceptual narrowing in synesthesia. Proc Natl Acad Sci USA 117, 10089–10096. [DOI] [PMC free article] [PubMed] [Google Scholar]
- McEwen BS (2004). Protection and damage from acute and chronic stress: allostasis and allostatic overload and relevance to the pathophysiology of psychiatric disorders. Ann NY Acad Sci 1032, 1–7. [DOI] [PubMed] [Google Scholar]
- Meunier CN, Chameau P, Fossier PM (2017). Modulation of synaptic plasticity in the cortex needs to understand all the players. Front Synaptic Neurosci 9, 2. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Meyer JH, Kapur S, Eisfeld B, Brown GM, Houle S, DaSilva J, Wilson AA, Rafi-Tari S, Mayberg HS, Kennedy SH (2001). The effect of paroxetine on 5-HT(2A) receptors in depression: an [(18)F]setoperone PET imaging study. Am J Psychiatry 158, 78–85. [DOI] [PubMed] [Google Scholar]
- Meyer JH, McMain S, Kennedy SH, Korman L, Brown GM, DaSilva JN, Wilson AA, Blak T, Eynan-Harvey R, Goulding VS, et al. (2003). Dysfunctional attitudes and 5-HT2 receptors during depression and self-harm. Am J Psychiatry 160, 90–99. [DOI] [PubMed] [Google Scholar]
- 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 Rep 26, 3475–3483.e3474. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Moda-Sava RN, Murdock MH, Parekh PK, Fetcho RN, Huang BS, Huynh TN, Witztum J, Shaver DC, Rosenthal DL, Alway EJ, et al. (2019). Sustained rescue of prefrontal circuit dysfunction by antidepressant-induced spine formation. Science 364. eaat8078.. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Morales-Garcia JA, de la Fuente Revenga M, Alonso-Gil S, Rodriguez-Franco MI, Feilding A, Perez-Castillo A, Riba J (2017). The alkaloids of Banisteriopsis caapi, the plant source of the Amazonian hallucinogen ayahuasca, stimulate adult neurogenesis in vitro. Sci Rep 7, 5309. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Moreno FA, Wiegand CB, Taitano EK, Delgado PL (2006). Safety, tolerability, and efficacy of psilocybin in 9 patients with obsessive-compulsive disorder. J Clin Psychiatry 67, 1735–1740. [DOI] [PubMed] [Google Scholar]
- 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, 76–79. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Moreno JL, Miranda-Azpiazu P, García-Bea A, Younkin J, Cui M, Kozlenkov A, Ben-Ezra A, Voloudakis G, Fakira AK, Baki L, et al. (2016). Allosteric signaling through an mGlu2 and 5-HT2A heteromeric receptor complex and its potential contribution to schizophrenia. Sci Signal 9, ra5. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Mukherjee S, Manahan-Vaughan D (2013). Role of metabotropic glutamate receptors in persistent forms of hippocampal plasticity and learning. Neuropharmacology 66, 65–81. [DOI] [PubMed] [Google Scholar]
- Murnane KS (2019). Serotonin 2A receptors are a stress response system: implications for post-traumatic stress disorder. Behav Pharmacol 30, 151–162. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Musazzi L, Rimland JM, Ieraci A, Racagni G, Domenici E, Popoli M (2014). Pharmacological characterization of BDNF promoters I, II and IV reveals that serotonin and norepinephrine input is sufficient for transcription activation. Int J Neuropsychopharmacol 17, 779–791. [DOI] [PubMed] [Google Scholar]
- 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 Res 1023, 134–140. [DOI] [PubMed] [Google Scholar]
- Muthukumaraswamy SD, Carhart-Harris RL, Moran RJ, Brookes MJ, Williams TM, Errtizoe D, Sessa B, Papadopoulos A, Bolstridge M, Singh KD, et al. (2013). Broadband cortical desynchronization underlies the human psychedelic state. J Neurosci 33, 15171–15183. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Muttoni S, Ardissino M, John C (2019). Classical psychedelics for the treatment of depression and anxiety: a systematic review. J Affect Disord 258, 11–24. [DOI] [PubMed] [Google Scholar]
- Nau F Jr, Yu B, Martin D, Nichols CD (2013). Serotonin 5-HT2A receptor activation blocks TNF-α mediated inflammation in vivo. PLoS One 8, e75426. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Negron-Oyarzo I, Aboitiz F, Fuentealba P (2016). Impaired functional connectivity in the prefrontal cortex: a mechanism for chronic stress-induced neuropsychiatric disorders. Neural Plast 2016, 7539065. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Nicholas CR, Henriquez KM, Gassman MC, Cooper KM, Muller D, Hetzel S, Brown RT, Cozzi NV, Thomas C, Hutson PR (2018). High dose psilocybin is associated with positive subjective effects in healthy volunteers. J Psychopharmacol 32, 770–778. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Nichols DE, Walter H (2020). The history of psychedelics in psychiatry. Pharmacopsychiatry, doi: 10.1055/a-1310-3990. [DOI] [PubMed] [Google Scholar]
- Noorani T (2020). The Pollan effect: psychedelic research between world and word. Fieldsights, https://culanth.org/fieldsights/the-pollan-effect-psychedelic-research-between-world-and-word. [Google Scholar]
- Palhano-Fontes F, Andrade KC, Tofoli LF, Santos AC, Crippa JA, Hallak JE, Ribeiro S, de Araujo DB (2015). The psychedelic state induced by ayahuasca modulates the activity and connectivity of the default mode network. PLoS One 10, e0118143. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Pei Q, Lewis L, Sprakes ME, Jones EJ, Grahame-Smith DG, Zetterström TS (2000). Serotonergic regulation of mRNA expression of Arc, an immediate early gene selectively localized at neuronal dendrites. Neuropharmacology 39, 463–470. [DOI] [PubMed] [Google Scholar]
- 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, 331–339. [DOI] [PubMed] [Google Scholar]
- Perry VH, Holmes C (2014). Microglial priming in neurodegenerative disease. Nat Rev Neurol 10, 217–224. [DOI] [PubMed] [Google Scholar]
- Planchez B, Surget A, Belzung C (2019). Animal models of major depression: drawbacks and challenges. J Neural Transm (Vienna) 126, 1383–1408. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Pokorny T, Preller KH, Kraehenmann R, Vollenweider FX (2016). Modulatory effect of the 5-HT1A agonist buspirone and the mixed non-hallucinogenic 5-HT1A/2A agonist ergotamine on psilocybin-induced psychedelic experience. Eur Neuropsychopharmacol 26, 756–766. [DOI] [PubMed] [Google Scholar]
- Pottie E, Dedecker P, Stove CP (2020). Identification of psychedelic new psychoactive substances (NPS) showing biased agonism at the 5-HT(2A)R through simultaneous use of β-arrestin 2 and miniGα(q) bioassays. Biochem Pharmacol 182, 114251. [DOI] [PubMed] [Google Scholar]
- Preller KH, Burt JB, Ji JL, Schleifer CH, Adkinson BD, Stämpfli P, Seifritz E, Repovs G, Krystal JH, Murray JD, et al. (2018). Changes in global and thalamic brain connectivity in LSD-induced altered states of consciousness are attributable to the 5-HT2A receptor. eLife 7, e35082. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Preller KH, Duerler P, Burt JB, Ji JL, Adkinson B, Stampfli P, Seifritz E, Repovs G, Krystal JH, Murray JD, et al. (2020). Psilocybin induces time-dependent changes in global functional connectivity. Biol Psychiatry 88, 197–207. [DOI] [PubMed] [Google Scholar]
- Preller KH, Razi A, Zeidman P, Stampfli P, Friston KJ, Vollenweider FX (2019). Effective connectivity changes in LSD-induced altered states of consciousness in humans. Proc Natl Acad Sci USA 116, 2743–2748. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Quirk GJ, Gehlert DR (2003). Inhibition of the amygdala: key to pathological states? Ann NY Acad Sci 985, 263–272. [DOI] [PubMed] [Google Scholar]
- Reiff CM, Richman EE, Nemeroff CB, Carpenter LL, Widge AS, Rodriguez CI, Kalin NH, McDonald WM, the Work Group on Biomarkers and Novel Treatments (2020). Psychedelics and psychedelic-assisted psychotherapy. Am J Psychiatry 177, 391–410. [DOI] [PubMed] [Google Scholar]
- Rickli A, Moning OD, Hoener MC, Liechti ME (2016). Receptor interaction profiles of novel psychoactive tryptamines compared with classic hallucinogens. Eur Neuropsychopharmacol 26, 1327–1337. [DOI] [PubMed] [Google Scholar]
- Rocha JM, Osorio FL, Crippa JAS, Bouso JC, Rossi GN, Hallak JEC, Dos Santos RG (2019). Serotonergic hallucinogens and recognition of facial emotion expressions: a systematic review of the literature. Ther Adv Psychopharmacol 9, 2045125319845774. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Roseman L, Leech R, Feilding A, Nutt DJ, Carhart-Harris RL (2014). The effects of psilocybin and MDMA on between-network resting state functional connectivity in healthy volunteers. Front Hum Neurosci 8, 204. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Roseman L, Nutt DJ, Carhart-Harris RL (2017). Quality of acute psychedelic experience predicts therapeutic efficacy of psilocybin for treatment-resistant depression. Front Pharmacol 8, 974. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Ross S, Bossis A, Guss J, Agin-Liebes G, Malone T, Cohen B, Mennenga SE, Belser A, Kalliontzi K, Babb J, et al. (2016). Rapid and sustained symptom reduction following psilocybin treatment for anxiety and depression in patients with life-threatening cancer: a randomized controlled trial. J Psychopharmacol 30, 1165–1180. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Roth BL, Choudhary MS, Khan N, Uluer AZ (1997). High-affinity agonist binding is not sufficient for agonist efficacy at 5-hydroxytryptamine2A receptors: evidence in favor of a modified ternary complex model. J Pharmacol Exp Ther 280, 576–583. [PubMed] [Google Scholar]
- Sampedro F, de la Fuente Revenga M, Valle M, Roberto N, Dominguez-Clave E, Elices M, Luna LE, Crippa JAS, Hallak JEC, de Araujo DB, et al. (2017). Assessing the psychedelic “after-glow” in ayahuasca users: post-acute neurometabolic and functional connectivity changes are associated with enhanced mindfulness capacities. Int J Neuropsychopharmacol 20, 698–711. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Sanacora G, Treccani G, Popoli M (2012). Towards a glutamate hypothesis of depression: an emerging frontier of neuropsychopharmacology for mood disorders. Neuropharmacology 62, 63–77. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Sayin U (2014). The consumption of psychoactive plants in ancient global and Anatolian cultures during religious rituals: the roots of the eruption of mythological figures and common symbols in religions and myths. NeuroQuantology 12, 2. [Google Scholar]
- Schartner MM, Pigorini A, Gibbs SA, Arnulfo G, Sarasso S, Barnett L, Nobili L, Massimini M, Seth AK, Barrett AB (2017). Global and local complexity of intracranial EEG decreases during NREM sleep. Neurosci Conscious 2017, niw022. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Schmid CL, Bohn LM (2010). Serotonin, but not N-methyltryptamines, activates the serotonin 2A receptor via a ß-arrestin2/Src/Akt signaling complex in vivo. J Neurosci 30, 13513–13524. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Schmid CL, Raehal KM, Bohn LM (2008). Agonist-directed signaling of the serotonin 2A receptor depends on beta-arrestin-2 interactions in vivo. Proc Natl Acad Sci USA 105, 1079–1084. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Schmid Y, Enzler F, Gasser P, Grouzmann E, Preller KH, Vollenweider FX, Brenneisen R, Muller F, Borgwardt S, Liechti ME (2015). Acute effects of lysergic acid diethylamide in healthy subjects. Biol Psychiatry 78, 544–553. [DOI] [PubMed] [Google Scholar]
- 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. Neurosci Lett 346, 137–140. [DOI] [PubMed] [Google Scholar]
- Shapiro DA, Kristiansen K, Kroeze WK, Roth BL (2000). Differential modes of agonist binding to 5-hydroxytryptamine(2A) serotonin receptors revealed by mutation and molecular modeling of conserved residues in transmembrane region 5. Mol Pharmacol 58, 877–886. [DOI] [PubMed] [Google Scholar]
- Sheline YI, Barch DM, Price JL, Rundle MM, Vaishnavi SN, Snyder AZ, Mintun MA, Wang S, Coalson RS, Raichle ME (2009). The default mode network and self-referential processes in depression. Proc Natl Acad Sci USA 106, 1942–1947. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Sleight AJ, Stam NJ, Mutel V, Vanderheyden PM (1996). Radiolabelling of the human 5-HT2A receptor with an agonist, a partial agonist and an antagonist: effects on apparent agonist affinities. Biochem Pharmacol 51, 71–76. [DOI] [PubMed] [Google Scholar]
- Strajhar P, Schmid Y, Liakoni E, Dolder PC, Rentsch KM, Kratschmar DV, Odermatt A, Liechti ME (2016). Acute effects of lysergic acid diethylamide on circulating steroid levels in healthy subjects. J Neuroendocrinol 28, 12374. [DOI] [PubMed] [Google Scholar]
- Studerus E, Gamma A, Vollenweider FX (2010). Psychometric evaluation of the altered states of consciousness rating scale (OAV). PLoS One 5, e12412. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Tagliazucchi E, Roseman L, Kaelen M, Orban C, Muthukumaraswamy SD, Murphy K, Laufs H, Leech R, McGonigle J, Crossley N, et al. (2016). Increased global functional connectivity correlates with LSD-induced ego dissolution. Curr Biol 26, 1043–1050. [DOI] [PubMed] [Google Scholar]
- Tedford HW, Zamponi GW (2006). Direct G protein modulation of Cav2 calcium channels. Pharmacol Rev 58, 837–862. [DOI] [PubMed] [Google Scholar]
- Thompson C, Szabo A (2020). Psychedelics as a novel approach to treating autoimmune conditions. Immunol Lett 228, 45–54. [DOI] [PubMed] [Google Scholar]
- Timmermann C, Roseman L, Schartner M, Milliere R, Williams LTJ, Erritzoe D, Muthukumaraswamy S, Ashton M, Bendrioua A, Kaur O, et al. (2019). Neural correlates of the DMT experience assessed with multivariate EEG. Sci Rep 9, 16324. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Timmermann C, Spriggs MJ, Kaelen M, Leech R, Nutt DJ, Moran RJ, Carhart-Harris RL, Muthukumaraswamy SD (2018). LSD modulates effective connectivity and neural adaptation mechanisms in an auditory oddball paradigm. Neuropharmacology 142, 251–262. [DOI] [PubMed] [Google Scholar]
- Uthaug MV, Lancelotta R, Szabo A, Davis AK, Riba J, Ramaekers JG (2020). Prospective examination of synthetic 5-methoxy-N,N-dimethyltryptamine inhalation: effects on salivary IL-6, cortisol levels, affect, and non-judgment. Psychopharmacology (Berl) 237, 773–785. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 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, 2785–2795. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Valle M, Maqueda AE, Rabella M, Rodríguez-Pujadas A, Antonijoan RM, Romero S, Alonso JF, Mañanas M, Barker S, Friedlander P, et al. (2016). Inhibition of alpha oscillations through serotonin-2A receptor activation underlies the visual effects of ayahuasca in humans. Eur Neuropsychopharmacol 26, 1161–1175. [DOI] [PubMed] [Google Scholar]
- Van de Kar LD, Javed A, Zhang Y, Serres F, Raap DK, Gray TS (2001). 5-HT2A receptors stimulate ACTH, corticosterone, oxytocin, renin, and prolactin release and activate hypothalamic CRF and oxytocin-expressing cells. J Neurosci 21, 3572–3579. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Vezzani A, Viviani B (2015). Neuromodulatory properties of inflammatory cytokines and their impact on neuronal excitability. Neuropharmacology 96, 70–82. [DOI] [PubMed] [Google Scholar]
- Vollenweider FX, Vollenweider-Scherpenhuyzen MF, Babler A, Vogel H, Hell D (1998). Psilocybin induces schizophrenia-like psychosis in humans via a serotonin-2 agonist action. Neuroreport 9, 3897–3902. [DOI] [PubMed] [Google Scholar]
- Wacker D, Wang S, McCorvy JD, Betz RM, Venkatakrishnan AJ, Levit A, Lansu K, Schools ZL, Che T, Nichols DE, et al. (2017). Crystal structure of an LSD-bound human serotonin receptor. Cell 168, 377–389.e312. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Walker AG, Wenthur CJ, Xiang Z, Rook JM, Emmitte KA, Niswender CM, Lindsley CW, Conn PJ (2015). Metabotropic glutamate receptor 3 activation is required for long-term depression in medial prefrontal cortex and fear extinction. Proc Natl Acad Sci USA 112, 1196–1201. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Wang CD, Gallaher TK, Shih JC (1993). Site-directed mutagenesis of the serotonin 5-hydroxytrypamine2 receptor: identification of amino acids necessary for ligand binding and receptor activation. Mol Pharmacol 43, 931–940. [PubMed] [Google Scholar]
- Wang DS, Zurek AA, Lecker I, Yu J, Abramian AM, Avramescu S, Davies PA, Moss SJ, Lu WY, Orser BA (2012). Memory deficits induced by inflammation are regulated by α5-subunit-containing GABAA receptors. Cell Rep 2, 488–496. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Weber ET, Andrade R (2010). Htr2a gene and 5-HT(2A) receptor expression in the cerebral cortex studied using genetically modified mice. Front Neurosci 4, 36.. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Williams GV, Rao SG, Goldman-Rakic PS (2002). The physiological role of 5-HT2A receptors in working memory. J Neurosci 22, 2843–2854. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Wise T, Taylor MJ, Herane-Vives A, Gammazza AM, Cappello F, Lythgoe DJ, Williams SC, Young AH, Cleare AJ, Arnone D (2018). Glutamatergic hypofunction in medication-free major depression: secondary effects of affective diagnosis and relationship to peripheral glutaminase. J Affect Disord 234, 214–219. [DOI] [PubMed] [Google Scholar]
- Wolff M, Evens R, Mertens LJ, Koslowski M, Betzler F, Grunder G, Jungaberle H (2020). Learning to let go: a cognitive-behavioral model of how psychedelic therapy promotes acceptance. Front Psychiatry 11, 5. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Yatham LN, Liddle PF, Dennie J, Shiah IS, Adam MJ, Lane CJ, Lam RW, Ruth TJ (1999). Decrease in brain serotonin 2 receptor binding in patients with major depression following desipramine treatment: a positron emission tomography study with fluorine-18-labeled setoperone. Arch Gen Psychiatry 56, 705–711. [DOI] [PubMed] [Google Scholar]
- Yu B, Becnel J, Zerfaoui M, Rohatgi R, Boulares AH, Nichols CD (2008). Serotonin 5-hydroxytryptamine(2A) receptor activation suppresses tumor necrosis factor-alpha-induced inflammation with extraordinary potency. J Pharmacol Exp Ther 327, 316–323. [DOI] [PubMed] [Google Scholar]
- 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. Neuropsychopharmacology 28, 45–52. [DOI] [PubMed] [Google Scholar]
- Zhang G, Asgeirsdottir HN, Cohen SJ, Munchow AH, Barrera MP, Stackman RW Jr (2013). Stimulation of serotonin 2A receptors facilitates consolidation and extinction of fear memory in C57BL/6J mice. Neuropharmacology 64, 403–413. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Zhang G, Stackman RW Jr (2015). The role of serotonin 5-HT2A receptors in memory and cognition. Front Pharmacol 6, 225. [DOI] [PMC free article] [PubMed] [Google Scholar]

