Buscar en
International Journal of Clinical and Health Psychology
Toda la web
Inicio International Journal of Clinical and Health Psychology Seeking the Psilocybiome: Psychedelics meet the microbiota-gut-brain axis
Journal Information
Vol. 23. Issue 2.
(April - June 2023)
Share
Share
Download PDF
More article options
Visits
2599
Vol. 23. Issue 2.
(April - June 2023)
Original article
Open Access
Seeking the Psilocybiome: Psychedelics meet the microbiota-gut-brain axis
Visits
2599
John R. Kellya,b,
Corresponding author
kellyjr@tcd.ie

Corresponding author at: Trinity Centre for Health Sciences, Trinity College Dublin and Tallaght University Hospital, Dublin 24, Ireland.
, Gerard Clarkec,d, Andrew Harkine, Sinead C. Corrd,f, Stephen Galvina, Vishnu Pradeepa,b, John F. Cryanc,d, Veronica O'Keanea,b,e, Timothy G. Dinanc,d
a Department of Psychiatry, Trinity College, Dublin, Ireland
b Tallaght University Hospital, Dublin, Ireland
c Department of Psychiatry and Neurobehavioral Science, University College Cork, Ireland
d APC Microbiome Ireland, University College Cork, Cork, Ireland
e Trinity College Institute of Neuroscience, Ireland
f Department of Microbiology, Trinity College Dublin, Ireland
This item has received

Under a Creative Commons license
Article information
Abstract
Full Text
Bibliography
Download PDF
Statistics
Figures (1)
Special issue
This article is part of special issue:
Contributions from the Neurosciences of Consciousness to Clinical Psychology

Edited by: Óscar F. Gonçalves

More info
Abstract

Moving towards a systems psychiatry paradigm embraces the inherent complex interactions across all levels from micro to macro and necessitates an integrated approach to treatment. Cortical 5-HT2A receptors are key primary targets for the effects of serotonergic psychedelics. However, the therapeutic mechanisms underlying psychedelic therapy are complex and traverse molecular, cellular, and network levels, under the influence of biofeedback signals from the periphery and the environment. At the interface between the individual and the environment, the gut microbiome, via the gut-brain axis, plays an important role in the unconscious parallel processing systems regulating host neurophysiology. While psychedelic and microbial signalling systems operate over different timescales, the microbiota-gut-brain (MGB) axis, as a convergence hub between multiple biofeedback systems may play a role in the preparatory phase, the acute administration phase, and the integration phase of psychedelic therapy. In keeping with an interconnected systems-based approach, this review will discuss the gut microbiome and mycobiome and pathways of the MGB axis, and then explore the potential interaction between psychedelic therapy and the MGB axis and how this might influence mechanism of action and treatment response. Finally, we will discuss the possible implications for a precision medicine-based psychedelic therapy paradigm.

Keywords:
psychedelics
hallucinogens
psilocybin
Lysergic acid diethylamide (LSD)
Dimethyltryptamine (DMT)
microbiome
microbiota-gut-brain axis
Full Text
Introduction

“If the doors of perception were cleansed everything would appear to man (sic) as it is, Infinite. For man has closed himself up, till he sees all things thro’ narrow chinks of his cavern”

- William Blake's The Marriage of Heaven and Hell (1790)

Varying degrees of dysfunction across different temporal and spatial scales can result in disorders of mental health. Re-calibration of dysregulated systems across the various levels in order to re-establish homeostasis, depends on a combination of conditions unique to the individual as well as the presence of universal conditions conducive to homeostasis (McDaid et al., 2019; WHO, 2021). An integrated approach across all levels from micro to macro may lead to a more complete understanding of mental health disorders and potentially to improved therapeutic outcomes.

The last two decades have seen major advances in microbiome (Cryan & Dinan, 2012; Cryan et al., 2019) and psychedelic science research (Nichols, 2016; Nutt et al., 2020). Serotonergic psychedelics administered in the context of psychological support can deliver therapeutic benefits for disorders with overly-restricted and/or maladaptive patterns of emotion, behaviour, and cognition (Carhart-Harris & Friston, 2019). Accruing clinical evidence suggests that psychedelic therapy can improve outcomes in depression (Carhart-Harris et al., 2021; Davis et al., 2021; Gukasyan et al., 2022), treatment-resistant depression (TRD) (Carhart-Harris et al., 2018, 2016; Goodwin, et al., 2020) and addiction disorders (Bogenschutz et al., 2022; Johnson et al., 2014).

The therapeutic mechanisms underlying psychedelic therapy are complex and traverse molecular, cellular, and network levels. Central activation of the 5-HT2A receptors, particularly in cortical layers are thought to initiate the cascade of altered information processing necessary for therapeutic effects. However, biofeedback signals from peripheral networks and the environment to central networks are key in both the modulation of acute effects and the anticipated trajectory towards sustained therapeutic psychedelic responses. An expanding knowledge of host-microbe interactions may further open the “doors of perception” to reveal patterns of interconnectivity to potentially enhance therapeutic response rates.

It has long been established that the microbial ecosystem is an integral part of host health, particularly in programming immune, metabolic and endocrine systems (de Vos et al., 2022). Preclinical evidence has shown that gut microbes via the gut-brain axis play a role in the regulation of brain development and function (Cryan et al., 2019).

The signalling pathways of the microbiota-gut-brain (MGB) axis operate throughout life, but are especially important during early development, in shaping aspects of the stress response (Sudo et al., 2004), cognition (Desbonnet et al., 2015), and social (Buffington et al., 2016; Desbonnet et al., 2014; Wu et al., 2021) and emotional domains (Cowan et al., 2016).

In humans, perturbations in the gut microbiota are associated with a diverse range of psychiatric disorders. Deciphering the direction of causality is a major challenge (Cryan & Mazmanian, 2022), but there may be general gut microbiota patterns, including depletion of certain anti-inflammatory butyrate-producing bacteria and enrichment of pro-inflammatory bacteria that signify suboptimal homeostasis across psychiatric disorders (Borkent et al., 2022; Kelly et al., 2021; McGuinness et al., 2022; Nguyen et al., 2018; Nikolova et al., 2021; Valles-Colomer et al., 2019).

Psychedelic therapy is comprised of three phases; preparation, administration/dosing, and integration. While the MGB axis signalling system operates over different timescales to the centrally acting serotonergic psychedelics, there are several ways by which the integration of the MGB axis, as a convergence hub between multiple biofeedback systems, could play an indirect role in the modulation of acute and sustained symptomatic responses in psychedelic therapy. Baseline MGB axis activity, together with other peripheral mediators and psychological measures, could serve as composite psycho-biomarkers that could be mapped onto the continuum of responses to psychedelic therapy in order to stratify those who may preferentially respond to psychedelic therapy (Kelly et al., 2021). Given the plasticity of MGB axis signalling, which can be rapidly altered following dietary and other approaches (Zhu et al., 2020), it is feasible that the MGB axis, during the preparatory phase or in the pre-preparatory phase, could be shifted into a state which might better respond to psychedelic therapy. In the acute administration phase, the MGB axis may play a subtle role in the inter-individual variability of psychedelic drug metabolism (Clarke et al., 2019). In the integration phase, re-enforcing peripheral mechanisms via synergistic MGB-axis signalling could contribute to the instigation and maintenance of behavioural changes that are more optimal to homeostasis over longer time frames.

A systems-based approach that encompasses the MGB axis and its relationship to the continuum of acute and sustained responses to psychedelic therapy provides an intriguing and potentially clinically useful avenue of further exploration. It has previously been proposed that sustained therapeutic effects of “regular and low doses of psychedelics” particularly those related to sleep, diet and lifestyle factors, could be mediated via the MGB axis (Kuypers, 2019), and that modulation of the gut microbiome by psychedelics (Inserra 2022) may influence immune functions (Thompson & Szabo, 2020). This review of the psilocybiome as a broad concept of reciprocal host-microbiota-psychedelic interactions (Fig. 1) will first discuss the microbiome and pathways of the MGB axis, before moving on to an exploration of the interaction between psychedelic therapy and the MGB axis and how this might influence acute and sustained changes in stress-, social- and reward-processing systems. Finally, we will discuss the implications of the MGB axis for a precision medicine-based psychedelic therapy paradigm.

Fig. 1.

The possible reciprocal host-microbiota-psychedelic interactions of the Psilocybiome. The MGB axis, as a convergence hub between multiple biofeedback systems, could play a role in the modulation of acute and sustained psychedelic responses in psychedelic therapy. In the preparatory phase, baseline MGB axis activity, together with other peripheral mediators and psychological measures, could serve as composite psycho-biomarkers that could be mapped onto the continuum of responses to psychedelic therapy in order to stratify those who may preferentially respond to psychedelic therapy. During the pre-preparatory or preparatory phase the MGB axis could be shifted into a state which might better respond to psychedelic therapy. In the acute administration phase, the MGB axis may play a subtle role in the inter-individual variability of psychedelic drug metabolism. In the longer-term, re-enforcing peripheral mechanisms via synergistic MGB-axis signalling could contribute to the instigation and maintenance of behavioural changes that are more optimal to homeostasis.

(0.55MB).
Evolutionary perspective

Microbes have inhabited the planet for approximately 3.5–3.7 billion of the earth's 4.5 billion years (Grosberg & Strathmann, 2007; Nutman et al., 2016). The first animals emerged 800 million years ago, with homo sapiens relative latecomers at 350,000 years old (Hublin et al., 2017). Every stage of the co-evolutionary timeline of increasing complexity occurred in a microbial world, which has led to an interconnected physiology between microbes and hosts. There are as many microbial cells in the human body as human cells, numbering 40 trillion (Sender & Fuchs, 2016). This complex composite of microbial and human cells and genes, known as the “holobiont”, expands the organismic boundaries and confines of the self to encompass bidirectional information exchange processes with our microbial ecosystem (Cryan et al., 2019; Theis et al., 2016).

This co-evolutionary lineage of increasing layers of interconnected material complexity and communication pathways between micro and macro levels – each influencing each other, to eventually culminate in processing systems that are able to reflect on the entire (known) evolutionary journey and alter its direction – is all the more important in this era of ecosystem disequilibrium (Almond & Petersen, 2020; Ceballos et al., 2017; Cobb, 2020; Johnson et al., 2020; Lent, 2017; Sheldrake, 2020; Watts et al., 2021). Analogous to the proposed synergistic relationship between contact with the natural environment and psychedelics (Gandy et al., 2020; Kettner et al., 2019; Lyons & Carhart-Harris, 2018), exploration of the molecular constellations and associated information pathways of interconnectivity between the microbial ecosystem and psychedelic therapy is at the forefront of systems neuroscience.

Fungi and psilocybin

At 1 billion years old, fungi are evolutionary younger microorganisms than bacteria (Bonneville et al., 2020; Loron et al., 2019). They form enmeshed relationships with bacteria (Deveau et al., 2018) and plants (Kiers et al., 2011) and are especially important for decomposition and regeneration, including the degradation of various environmental pollutants (Akhtar & Mannan, 2020; Dusengemungu et al., 2021; Sheldrake, 2020). There are 120,000 known species of fungi, but escalating estimates based on high-throughput sequencing techniques suggest as many as 12 million, revised from previous estimates of 5.1 million (Hawksworth, 2001; Wu et al., 2019). Mushrooms containing psilocybin, numbering approximately 200, mostly from the genus Psilocybe, are estimated to be 10–20 million years old and may have evolved separately more than once (Kosentka et al., 2013). It is not known why some mushrooms produce psilocybin. An intriguing, though tenuous, evolutionary theory based on a comparative genomic analysis of hallucinogenic fungi with non-hallucinogenic relatives posited that psilocybin may have provided a fitness advantage in certain environments by altering the behaviour of predatory insects, to decrease the chances of the fungi getting eaten (Awan et al., 2018; Reynolds et al., 2018).

Other fungi also alter host behaviour. Massospora is a fungus that infects insects. When it infects cicadas, it produces psilocybin and the amphetamine cathinone, which are associated with the alteration of some of its hosts behavioural patterns (Boyce et al., 2019; Cooley et al., 2018). Similarly, the fungus Ophiocordyceps unilateralis manipulates behaviour in certain species of ants (de Bekker et al., 2014; Mangold et al., 2019). Even more remarkable is that the fungal behavioural control in some ant species may occur in the periphery, via extensive networks in muscles (Fredericksen et al., 2017).

Psychoactive fungi can also alter human behaviour. Accidental consumption of the alkaloid ergotamine – a precursor to lysergic acid diethylamide (LSD), which is produced by the fungus Claviceps purpurea that grows on grains/rye – can alter human behaviour and perception (Correia et al., 2003; Eadie, 2003). More subtle unintentional ingestion of toxic fungal secondary metabolites or mycotoxins in food contaminants occurs today (Eskola et al., 2020; Lindell et al., 2022). While the contribution of psychedelic mushrooms to the evolutionary shaping of human faculties is speculative (Rodríguez Arce & Winkelman, 2021), intentional consumption in ritual/ceremonial settings by humans is thought to have occurred for several millennia (Miller et al., 2019; Robinson et al., 2020; Rodríguez Arce & Winkelman, 2021).

Gut microbiome and mycobiome

Most of the human body's microbes reside in the gastrointestinal tract (GIT). The vast majority are bacteria (Zhernakova et al., 2016). Viruses and bacteriophages, protozoa, archaea, and indeed fungi are also present but in much smaller proportions (Lankelma et al., 2015). Fungal populations or the mycobiome within the microbiota comprise 0.001–0.1% of the total gut microbiome and are dominated by yeasts (Hallen-Adams & Suhr, 2017; Nash et al., 2017). The mycobiome is less stable than the microbiome, with less, if any colonization continuity (Auchtung et al., 2018; Hallen-Adams & Suhr, 2017; Lavrinienko et al., 2021; Suhr & Hallen-Adams, 2015). Similar to the gut microbiome, the gut mycobiome is shaped by geography, urbanization, ethnicity, and diet (Sun et al., 2021). Both the microbiome and the less studied mycobiome interact in complex ways to collectively shape host homeostasis, especially that of immune (Leonardi et al., 2018; van Tilburg Bernardes et al., 2020) and metabolic systems (Mims et al., 2021; Sanna et al., 2019).

Defining the optimal gut microbiome is challenging given the extensive individual differences between the microbiomes of healthy people (Falony et al., 2016; Gacesa et al., 2022; The Human Microbiome Project et al., 2012). However, it is generally thought that diverse gut microbiome composition and stability are beneficial for human health (Bäckhed et al., 2012; Lozupone et al., 2012). There are some indicators that certain gut microbial species have been lost and alpha-diversity (distribution of species abundances within a sample) reduced as humans entered the industrialized age, but the precise implications for human health are not fully known (De Filippo et al., 2010; Moeller et al., 2014; Olm et al., 2022; Smits et al., 2017).

Communication channels: microbiota-gut-brain (MGB) axis

Over the last two decades, notwithstanding the aforementioned challenges in translation, there have been advances in the mechanistic understanding of how the gut microbiota communicate with the brain (Forssten et al., 2022; Nagpal & Cryan, 2021). Germ-free (GF) rodents (born and raised without microbes), antibiotics, probiotics, gastrointestinal (GI) infection studies, and Fecal Microbiota Transplantation (FMT) rodent studies have progressed the understanding of the mechanisms by which the gut microbiota can influence brain development and function via the gut-brain axis. This MGB signalling system communicates with the brain via stimulation of immune pathways (Erny et al., 2015), vagus nerve activation (Fülling et al., 2019), production of circulating microbial metabolites and/or microbial induction of host molecules (short-chain fatty acids (SCFA's), (O'Riordan et al., 2022), amino acid derivatives (catecholamines, and other indole derivatives), and secondary bile acids, as well as stimulation of enteroendocrine cells and enteric nervous system signalling (Ye et al., 2021), tryptophan metabolism (O'Mahony et al., 2015) and the HPA axis (Sudo et al., 2004). Some of these pathways may serve as overlapping, indirect points of convergence in the trajectory of psychedelic therapy.

Tryptamines and the MGB axis

Psilocybin and psilocin are tryptamines or tryptophan indole-based monoamine alkaloids (indolalkylamines) (Lenz et al., 2021). Tryptamines are derived from the decarboxylation of tryptophan, which is an essential amino acid acquired from the diet, and the precursor for serotonin (5-hydroxytryptamine) and melatonin (Fricke et al., 2017; Sherwood et al., 2020). After ingestion, psilocybin is rapidly dephosphorylated by alkaline phosphatases in the intestine and liver to the biologically active psilocin, which is then absorbed from the stomach and small intestine into circulation and can cross the blood brain barrier (BBB) (Dinis-Oliveira, 2017). Psilocin can be further metabolized by a demethylation and oxidative deamination catalysed by liver Monoamine oxidases (MAO) or aldehyde dehydrogenase (Dinis-Oliveira, 2017). Further metabolism of psilocin by glucuronidation of the hydroxyl group to psilocin O-glucuronide occurs by UDP glucuronosyltransferases (UGT)1A10 in the small intestine and likely UGT1A9 in the circulation (Dinis-Oliveira, 2017; Manevski et al., 2010). (For a detailed review of the metabolism of psilocybin and psilocin see Dinis-Oliveira 2017). Certain psychotropics are subject to glucuronidation, a process which may be influenced by the gut microbiota (Clarke, et al., 2019; Cussotto et al., 2021). But it is not yet known whether microbes play a role in the glucuronidation of psilocin.

The structural similarity of psilocybin and psilocin to other indoles suggests the possibility that these are interkingdom signalling molecules (Lee et al., 2015). The largest reserve of 5-HT is located in enterochromaffin cells in the GIT (Mawe & Hoffman, 2013). 5-HT is an important signalling molecule in the gut-brain-axis under the direct or indirect influence of the gut microbiota (Clarke et al., 2013; O'Mahony et al., 2015; Yano et al., 2015). Gut bacteria-produced tryptamine can activate the epithelial G-protein-coupled receptor (GPCR) 5-HT4 to increase GI transit (Agus et al., 2018; Bhattarai et al., 2018). Tryptamine can induce the release of 5-HT by enterochromaffin cells, and enteric neurons are able to take up tryptamine, which displaces 5-HT in intracellular synaptic vesicles, resulting in 5-HT release (Gheorghe et al., 2019; Israelyan et al., 2019). Furthermore, gut microbes, such as Clostridium sporogenes and Ruminococcus gnavus can decarboxylate tryptophan to tryptamine which can activate host receptors (Bhattarai et al., 2018; Cryan et al., 2018; Williams et al., 2014). Bifidobacterium longum subsp. longum 35,624 has been shown to increase plasma tryptophan levels in rats (Desbonnet et al., 2008) and increase Tryptophan Hydroxylase expression and secretion of 5-hydroxytryptophan in vitro (Tian et al., 2019). Certain gut microbes can also produce tyramine, a trace amine derived from the amino acid tyrosine, that acts as a neurotransmitter and catecholamine-releasing agent and which can activate dopaminergic GPCRs (Colosimo et al., 2019).

Monoamine oxidases (MAO) A and B are the primary enzymes involved in tryptamine metabolism to produce indole-3-acetaldehyde (MetaCyc, 2022). A recent study indicated that some Psilocybe species (P. cubensis, P. Mexicana, P. semilanceata, P. cyanescens) produce harmane, harmine, and other l-tryptophan-derived β-carbolines (Blei et al., 2020), which act as reversible MAO-A inhibitors. There are indications that host-microbiota-metabolome interactions may have implications for inter-individual variability in the metabolism of tryptamines. For example, a study in mice showed that Morganella morganii, a gram-negative gut commensal, converted the essential amino acid L-Pheinto phenethylamine into the potent psychoactive trace amine phenethylamine (Chen et al., 2019). When combined with the non-selective and irreversible MAO inhibitor (MAO-I) phenelzine, M. morganii triggered phenethylamine toxicity (Chen et al., 2019). Taken together, it is conceivable that certain gut microbiota configurations and associated signalling pathways may play a subtle role in the inter-individual variability of psychedelic metabolism. However, the landscape for host-microbiota-psychedelic interactions in drug metabolism has yet to be fully explored.

Kynurenine pathway

The kynurenine pathway (KP) is an alternative metabolic route for tryptophan. A large majority of tryptophan is metabolized along the KP giving rise to molecules collectively referred to as “kynurenines” (Gheorghe et al., 2019). Enzymes of the KP are immune- and stress-responsive (O'Mahony et al., 2015) and are partially regulated by the gut microbiota (O'Mahony et al., 2015). As discussed above, the availability of tryptophan is also altered by gut microbes generating either indole and its derivatives, tryptamine or 5-HT, which can affect GI function (Gheorghe et al., 2019). Dysregulated tryptophan metabolism can occur under conditions of stress or inflammation in both the gut and the brain to alter gut-brain axis signalling. During stress or immune activation, tryptophan can be preferentially converted to kynurenine rather than 5-HT in the gut, with implications for GI motility and function. Tryptophan is metabolized along the KP by microglia and astrocytes in the brain leading to the formation of either kynurenic acid (KYNA) or quinolinic acid (QUIN) (Gheorghe et al., 2019). The KYNA and QUIN balance has implications for psychiatric disorders (Gheorghe et al., 2019; Savitz, 2020). For a detailed review on microbial regulation of tryptophan and KP metabolism, see (Gheorghe et al., 2019).

Previous studies suggest that KP metabolites may play a role in SSRI (Bhattacharyya et al., 2019; Erabi et al., 2020) and ketamine treatment response (Verdonk et al., 2019; Zhou et al., 2018). In a single-arm, open-label study of 84 medicated patients with unipolar and bipolar depression, intravenous (IV) ketamine increased serum KYNA levels and the KYNA/kynurenine ratio at 24 h following the first infusion in responders compared to non-responders, and this elevation lasted up to 14 days, associated with improvements in depressive symptoms (Zhou et al., 2018). In mice, a single dose of intraperitoneal (IP) ketamine restored lipopolysaccharide (LPS) induced depressive-like alterations and decreased cytokine production, microglial activation in the prefrontal cortex (PFC) and QUIN production (Verdonk et al., 2019). In the human part of the same study, which included 15 medicated inpatients with TRD (and 15 age and sex matched healthy controls), plasma KYNA/QUIN predicted (IV) ketamine response, and a reduction in QUIN was associated with a reduction in depressive symptoms (Verdonk et al., 2019). These studies did not explore the gut microbiota, but preclinical studies indicate that ketamine can alter the gut microbiota (Table 1). It is not yet known whether the serotonergic psychedelics influence host-microbe control over tryptophan and KP metabolism, and whether this would have any clinically relevant impact on response rates in psychedelic therapy.

Table 1.

Gut microbiota and psychotropics.

Ketamine
Design  Currently proposed primary mechanism of action  Effect on the gut microbiome 
male rats, ketamine 2.5 mg/kg 7 days, IP  N-methyl-d-aspartate receptor (NMDAR) antagonist  increase in Lactobacillus, Turicibacter and Sarcina and decrease in opportunistic pathogens Mucispirillum and Ruminococcus vs saline. alpha diversity: no significant difference (Getachew et al., 2018
in vitro (ketamine and propofol mix)    antimicrobial activity against E.coli, P.aeruginosa and C.albicans. S.aureus not inhibited (Begec et al., 2013
in vitro    dose dependent antimicrobial activity against: S.aureus, S.pyogenes, S.epidermidis, E.faecalis, P.aeruginosa and E.coliS.aureus and S.pyogenes most sensitive (Gocmen et al., 2008
in vitro    antimicrobial activity against Stachybotrys chartarum, Staphylococcus epidermidis and Borrelia burgdorferi (Torres et al., 2018
MDMA     
male rats, MDMA 20 mg/kg, single dose, subcutaneous  inhibits serotonin (SERT), norepinephrine (NET), and dopamine transporters (DAT)  increase in cecal Proteus mirabilis vs placebo, antibiotics reduced MDMA-induced hyperthermia (Ridge et al., 2019
SSRI's     
male mice, escitalopram 10 mg/kg, oral gavage, daily stress (CUMS)  inhibits SERT  responder group: increase in Prevotellaceae UCG-003, and decrease in Ruminococcaceae and Lactobacillaceae vs non-responder group. escitalopram increased alpha-diversity (Duan et al., 2021
male mice, amitriptyline (25 mg/kg/d) or fluoxetine (12 mg/kg/d) for 6 weeks, oral gavage, daily stress (CUMS)    increase in Bacteroidetes and decrease in Firmicutes, increase in Porphyromonadaceaeincrease in Bacteroidaceae associated with Ami, not Fluincrease in Parabacteroides, Butyricimonas, and Alistipes in Ami and Flualpha diversity increased in Ami and Flu (Zhang et al., 2021
male rats, 28 days fluoxetine 10 mg/kg/day, escitalopram 5 mg/kg/day, in drinking water    fluoxetine inhibited growth of cecal Succinivibrio and PrevotellaSSRIs: increase ileal but not colonic permeabilityin vitro: escitalopram antimicrobial activity against E.coli, but not L.rhamnosus.Fluoxetine antimicrobial activity against L.rhamnosus and E.coliIn vitro: escitalopram had antimicrobial effect on E. coli, but no effect on L. rhamnosusFluoxetine: strong dose-dependent antimicrobial activity against L. rhamnosus and E. coli (Cussotto et al., 2019
male mice, 29 days fluoxetine 20 mg/kg/day, oral gavage    decrease in L.johnsonii and Bacteroidales S24–7alpha diversity: no significant difference (Lyte et al., 2019
male mice, 21 days fluoxetine and escitalopram 10 mg/kg/day, IP injections    decrease in Ruminococcus (Lukić et al., 2019), alpha diversity: decrease 
male rats, 21 days of 2 mg/kg/day, oral gavage, daily stress (Fluoxetine)    decrease in Prevotellaceae Ga6A1 and increase in Ruminoclostridium 6 and Ruminococcaceae NK4A214alpha diversity: no significant difference (Zhu et al., 2019
male and female mice, 7 days fluoxetine 10 mg/kg/day, oral gavage    decrease in Turicibacter (Fung et al., 2019
male mice, 21 days fluoxetine 12 mg/kg/day, oral gavage, daily stress (CUMS)    Fluoxetine altered Erysipelotrichia; Alphaproteobacteria, Betaproteobacteria, Deltaproteobacteria, and Epsilonproteobacteria (Sun et al., 2019
female rat model of maternal vulnerability, heterozygous SERT knockout (SERT+/−), 10 mg/kg fluoxetine, oral gavage    increase in Prevotella and Ruminococcus during pregnancy and lactation in fluoxetine, and decreased fecal amino acids.amino acid concentrations correlated negatively with Prevotella and Bacteroides (Ramsteijn et al., 2020
Sertraline, in vitro    Potent antimicrobial against E. coli (Bohnert et al., 2011)Inhibits the growth of S. aureus, E. coli and P. aeruginosa (Ayaz et al., 2015)Potent antifungal activity against Cryptococcus neoformans, Coccidioides immitis and Candida spp. (Lass-Flörl et al., 2003; Paul et al., 2016; Rossato et al., 2016; Treviño-Rangel Rde et al., 2016; Zhai et al., 2012)Inhibits Leishmania donovani (Palit & Ali, 2008
Phencyclidine     
male rats, 7 days phencyclidine 5 mg/kg IP  N-methyl-d-aspartate receptor (NMDAR) antagonist  impaired novel object recognition, ampicillin abolished the subPCP-induced memory deficitincrease in Lachnospiraceae and Clostridiaceae, including the Roseburia and Odoribacter genus in subPCP (Pyndt Jorgensen et al., 2015
DOI     
male mice, offspring of maternal immune activation mouse model (influenza virus), DOI 0.5 mg/kg IP  5-HT2A, 5-HT2B, 5-HT2C agonist  adult offspring exhibited altered gut microbiota profiles, up-regulated frontal cortex 5-HT2A receptors,increased DOI induced head twitch behaviour and cognitive deficits in the novel object recognition test5-HT2A receptor density positively correlated with Ruminococcaceae and Candidatus (saccharibacteria), trend negative correlation with Lactobacillaceaestreptomycin in the prepubertal stage prevented cognitive impairment, but not 5-HT2AR changes (Saunders et al., 2020

MDMA; 3,4-methylenedioxymethamphetamine, DOI; 2,5-dimethoxy-4-iodophenyl-2-aminopropane, IP; intraperitoneally, SSRI; Selective serotonin reuptake inhibitor, CUMS; chronic unpredictable mild stress, Ami; amitriptyline, Flu; Fluoxetine, PCP; phencyclidine.

Immune pathways

The gut microbiota plays a key role in regulating immune system homeostasis by maintaining pro-inflammatory and anti-inflammatory signalling in the GIT (Ansaldo et al., 2021; Schirmer et al., 2016). The immune system is regulated by a plethora of ligands and signalling molecules produced by microorganisms in the gut, such as SCFA's, including butyrate, propionate, and acetate. Other examples include polyamines, linoleic acid metabolites, tryptophan metabolites, trimethylamine-N-oxide, vitamins, and secondary bile acids (Hertli & Zimmermann, 2022). Lack of balanced gut microbiota plays a role in various autoimmune diseases and given the role of T cells in classical neuroinflammatory diseases, there is a growing awareness of the likely relevance of the gut-brain axis in immune dysregulation that affects CNS function via microglial activation, pro-inflammatory cytokine expression, molecular mimicry, anti-neuronal antibodies, self-reactive T cells and disturbance of intestinal and BBB in psychiatric disorders (Ivanov et al., 2022; Medina-Rodriguez et al., 2020; Pape et al., 2019; Pearson-Leary et al., 2020).

Serotonergic psychedelics have immuno-modulatory properties (Szabo, 2015; Szabo et al., 2014, 2016; Thompson & Szabo, 2020). In vitro studies have demonstrated that psilocin and dimethyltryptamine (DMT) reduced levels of TLR4, p65 and CD80 proteins, and upregulated TREM2 in mouse primary microglia cells (Kozłowska et al., 2021), whereas extracts from psilocybin-containing mushrooms inhibited LPS-induced production of TNF-α and IL-1β in human U937 macrophage cells (Nkadimeng et al., 2021). (For a review of immune related proteins see Beumer et al. 2012). In the small intestine, the selective 5-HT2A/2C receptor agonist 2,5-dimethoxy-4-iodoamphetamine (R)-DOI inhibited a variety of TNF-α induced mRNA expression of pro-inflammatory markers mediated via the 5-HT2A receptor (Nau et al., 2013).

Similar anti-inflammatory effects, via 5-HT2A receptor activation, were observed in rodent aortic smooth muscle (Yu et al., 2008) and in models of asthma (Flanagan et al., 2019) and cardiovascular disease (Flanagan et al., 2019). The gut microbiota may further moderate the immuno-modulatory properties of serotonergic psychedelics. A maternal immune activation mouse model demonstrated a link between the gut microbiota and responses to DOI (Saunders et al., 2020). Adult male offspring exhibited altered gut microbiota profiles, up-regulated 5-HT2A receptors in the frontal cortex, increased DOI induced head-twitch behaviour (rapid side-to-side rotational head movement that occurs in rodents after administration of 5-HT2A agonists), and cognitive deficits in the novel object recognition test (Saunders et al., 2020). 5-HT2A receptor density correlated with gut Ruminococcaceae (Saunders et al., 2020). Oral antibiotics in the prepubertal stage prevented the cognitive impairment, but not the change in 5-HT2A receptor density or head-twitch behaviour in response to challenge with DOI (Saunders et al., 2020).

Interestingly, the prebiotic (galacto-oligosaccharides) prevented an LPS-mediated increase in 5-HT2A receptor density in the frontal cortex in mice (Savignac et al., 2016), whereas the antibiotic ampicillin abolished an impairment in novel object recognition induced by sub-chronic doses of the non-competitive N-methyl-D-aspartate (NMDA) receptor antagonist phencyclidine in rats (Pyndt Jorgensen et al., 2015) (Table 1). In humans, an exploratory study of the short-acting psychedelic 5-MeO-DMT in healthy volunteers (n = 11) found a significant decrease in salivary IL-6, but no changes in CRP and IL-1β, and increases in salivary cortisol levels (Uthaug et al., 2020) (Table 2). Ayahuasca reduced CRP levels in both TRD patients, and a comparison group of healthy adults compared to placebo, though there were no significant changes in IL-6 levels (Galvão-Coelho et al., 2020; Palhano-Fontes et al., 2019). Further exploration of the immuno-modulatory effects of serotonergic psychedelics and how they may be influenced by the gut microbiota could advance mechanistic insights.

Table 2.

Serotonergic psychedelics and acute changes in peripheral markers in humans.

Psychedelic  Group  Outcomes 
Ayahuasca(single dose)  TRD (n = 29), HC (n = 45)antidepressant freeparallel arm, randomized double-blinded placebo-controlled  decrease in blood CRP in TRD and HCs vs placebo, no significant changes in IL-6correlation between larger reductions of CRP and lower depressive symptoms 48 h after aya in TRDincrease in salivary cortisol levels in both TRD and HCs, 48hours after aya: no differences in CAR or plasma cortisol between the groups (Palhano-Fontes et al., 2019; Galvão et al., 2018; Galvão-Coelho et al., 2020)increase in BDNF at day 2 vs placebo in TRD and HC, but no significant differences between TRD and HC (de Almeida et al., 2019
Ayahuasca(single dose)  HC (n = 22)randomized, placebo-controlled  no change in blood BDNF (Rocha et al., 2021
Ayahuasca(single dose)  Social anxiety disorder (n = 17)Randomized, placebo-controlled  no change in blood BDNF (Dos Santos et al., 2021
Psilocybin (45, 115, 215, 315 µg/kg)  HC (n = 8)double-blind placebo-controlled  acute increase in plasma ACTH, cortisol, prolactin and TSH (Hasler, et al., 2004
5-MeO-DMT (inhaled)  HC (n = 11)non-controlled  significant increase in salivary cortisol and decrease in IL-6 (30 min before & after 90 min after 5-MeO-DMT)no changes in CRP and IL-1β (Uthaug, et al., 2020
N,N DMT (IV)  HC (n = 12)experienced psychedelic usersrandomized placebo-controlled  acute dose dependent increase in blood cortisol, corticotropin, prolactin, growth hormone and ß-endorphinall endocrine markers returned to baseline 5 h post N,N DMTno change in serum melatonin (Strassman & Qualls, 1994; Strassman et al., 1996
LSD(200 μg)  HC (n = 16)randomized, double-blind, placebo-controlled cross-over  increase in plasma cortisol, cortisone, corticosterone, 11-dehydrocorticosterone, and androgen dehydroepiandrosterone vs placebono changes in other androgens, progestogens or mineralocorticoids vs placebo (Strajhar et al., 2016
LSD(200 μg)  HC (n = 16)double-blind, randomized, placebo-controlled, crossover  increase in plasma cortisol, prolactin, oxytocin, and epinephrine (Schmid, et al., 2015
LSD(25, 50, 100, 200 µg)  HC (n = 16)double-blind, randomized, placebo-controlled, crossover  increase in blood BDNF at 200 µg vs placebo (Holze, et al., 2021
LSD(5, 10, 20 μg)  HC (n = 23, n = 5 all-time points)within-subject placebo-controlled  increase in blood BDNF at 4 h (5 μg) and 6 h (5, 20 μg) vs placebo (Hutten et al., 2021
LSD(100 µg)  HC (n = 17)randomized, placebo-controlled crossover  increase in sympathetic activity (electrocardiographic recordings) (Schmid, et al., 2015

TRD, HC, CRP; reduced C-reactive protein, IL; interleukin, TSH; thyroid stimulating hormone, h; hours, CAR; cortisol awakening response, BDNF; brain-derived neurotrophic factor, ACTH; adrenocorticotropic hormone, 5-MeO-DMT; 5-methoxy-N,N-dimethyltryptamine, N,N-dimethyltryptamine, min; minutes, IV; intravenous, vs; versus.

MGB axis and the stress response

It has long been established that chronic exposure to stress, which exceeds the individual's coping capacity (allostatic load) can lead to adverse mental health outcomes (McEwen, 2017). The MGB axis is one of the systems that plays a role in stress regulation. Stress alters the gut microbiota, and alteration of the microbiota mediates stress responsivity (Foster et al., 2017), particularly the HPA axis (Sudo et al., 2004), the main neuroendocrine regulator of stress responses. Early life stressors are especially potent sensitizers of the stress response system, including the MGB axis (Clarke et al., 2013).

Administration of serotonergic psychedelics acutely stimulates the HPA axis in animals and humans via central and peripheral actions (Galvão-Coelho et al., 2020; Hasler et al., 2004; Schindler et al., 2018; Uthaug et al., 2020). 5-HT2A, 5-HT2C and 5-HT1A receptors are expressed in the hypothalamus, and serotonergic psychedelics can stimulate corticotropin releasing hormone release and dose dependent increases in serum adrenocorticotropic hormone and corticosterone/cortisol (Schindler et al., 2018). (For a review of the neuroendocrine effects of serotonergic psychedelics see (Schindler et al., 2018). A study in male mice suggested that the acute anxiogenic effects induced by psilocybin were associated with the post-acute anxiolytic effects (Jones et al., 2020). This study showed that chronic corticosterone administration suppressed the psilocybin induced acute corticosterone and behavioural changes (Jones et al., 2020). The authors postulated that psilocybin may act as an initial stressor that provides resilience to subsequent stress (Jones et al., 2020).

Stress-induced alterations in amygdala 5-HT2A receptor-signalling may be one point of convergence between the serotonergic psychedelics and the MGB axis. Preclinical data suggests that the gut microbiota influences amygdala structure and function (Hoban et al., 2017; Luczynski et al., 2016; Pinto-Sanchez et al., 2017; Stilling et al., 2018). Psychedelics acutely alter amygdala reactivity, with sustained effects of at least one month in some people (Barrett et al., 2020). This may be associated with a shift in emotional reactivity from a negative to a positive bias (Grimm et al., 2018; Kometer et al., 2012; Kraehenmann et al., 2015). Given the aforementioned perturbations of the gut microbiota in stress-related psychiatric disorders (Borkent et al., 2022; McGuinness et al., 2022; Nguyen et al., 2018; Nikolova et al., 2021; Valles-Colomer et al., 2019), exploration of the interaction of the MGB axis and peripheral and central components of the stress response system could potentially increase overlapping resilience strategies to decreased rates of relapse in stress-related psychiatric disorders after psychedelic therapy.

MGB axis and social processing

Social processes involve a complex interplay of social reward circuits, molecular signalling pathways, and environmental cues. Social network interconnectivity includes socially distributed symbiotic microbes, required for the development and maintenance of social processes (Buffington et al., 2016; Desbonnet et al., 2014; Sherwin et al., 2019; Stilling et al., 2018). In a human study (n = 655), people with larger social networks had more diverse gut microbiota composition (Johnson, 2020). Conversely, anxiety and stress were associated with reduced gut microbiota profiles (Johnson, 2020). A number of pathways of the MGB axis can influence social behaviour. The HPA axis is intertwined with the expression of social behaviours. A recent study in male mice showed that reducing HPA axis-mediated production of corticosterone via the gut microbiota altered social behaviour (Wu et al., 2021). The same study suggested that Enterococcus faecalis restored social deficits and reduced corticosterone levels following social stress (Wu et al., 2021).

Psychedelic therapy, heavily dependent on context, can alter social processing and potentially foster increased levels of perceived connectivity (Kettner et al., 2021; Watts et al., 2017). Higher levels of perceived connectivity, whether social or otherwise, can strengthen resilience against stressors. Social isolation (and perceived sense of disconnection) can precipitate stress. An 8-week social isolation model in juvenile marmosets, resulted in decreased fecal cortisol levels in both ayahuasca and saline treated groups, though in the male animals, ayahuasca reduced scratching (a stress related stereotypic behaviour) and increased feeding behaviour (da Silva et al., 2019).

In addition to the HPA stress response, other overlapping signalling pathways, such as oxytocin, which are under the partial influence of the gut microbiota, influence social processes (Erdman, 2021; Sgritta et al., 2019). Serotonergic psychedelics can increase oxytocin (Schmid et al., 2015), but similar to acute activation of the HPA axis, the implications for psychedelic therapy have yet to be established. Although not a classical psychedelic, 3,4-ethylenedioxymethamphetamine (MDMA) can modulate oxytocin-mediated synaptic plasticity in the nucleus accumbens in mice, which is necessary for social reward learning (Nardou et al., 2019). The gut microbiota was not measured in this study, but another preclinical study showed that MDMA increased Proteus mirabilis in the caecum, while treatment with antibiotics reduced MDMA-induced hyperthermia, suggesting a role of the MGB axis in thermoregulation (Ridge et al., 2019). These studies prompt further investigation into the MGB axis and its interaction with neuroendocrine responses in contributing to the potential alterations in social processing over the trajectory of psychedelic therapy.

MGB axis and reward

Preliminary clinical studies show that psychedelic therapy may have a therapeutic role in overcoming alcohol use disorder (Bogenschutz et al., 2022; Krebs & Johansen, 2012) and nicotine addiction (Garcia-Romeu et al., 2014; Johnson et al., 2014; Noorani et al., 2018). Several trials are underway to explore psychedelic therapy for other addiction disorders (e.g. cocaine, methamphetamine, opioids). There has been increasing interest in the role of the MGB axis in the various aspects of reward processing (García-Cabrerizo et al., 2021; Han et al., 2018). Microbiome depletion antibiotic rodent models indicate that the microbiome may play a role in sensitising rodents to cocaine reward (Kiraly et al., 2016), and variability in gut microbiota profiles may be associated with altered cocaine consumption in rats (Suess et al., 2021). Similarly, microbiome depletion rodent models suggest the gut microbiome alters brain circuitry in opioid (oxycodone) intoxication and withdrawal (Simpson et al., 2020).

Alcohol also alters the gut microbiota (Peterson et al., 2017), which may contribute to alcohol addiction cycles via bidirectional communication pathways involving immuno-kynurenine (Leclercq et al., 2021), intestinal permeability (Leclercq et al., 2014), metabolic pathways (Leclercq et al., 2020), and alterations in dopaminergic signalling (Carbia et al., 2021; González-Arancibia et al., 2019; Jadhav et al., 2018). Recently, a translational study using antibiotic and FMT rodent models showed that metabolic disturbance and weight gain associated with smoking cessation were partially dependent on the gut microbiome (Fluhr et al., 2021).

While the central activity of psychedelic compounds are primary, these studies propose that the MGB axis might play an overlapping role, albeit over different time scales, with psychedelic therapy in gating reward valence and therefore in the modulation of complex reward-related behavioural patterns or habits (Dong et al., 2022; García-Cabrerizo et al., 2021; Meckel & Kiraly, 2019; Salavrakos et al., 2021). This synergistic combination of MGB axis and psychedelic therapy could help to restructure reward-processing pathways to overcome various addiction cycles, particularly in maintaining abstinence in the post-psychedelic therapy phase.

The gut microbiota, dietary and physical activity patterns

There is increasing recognition that diet, via multiple overlapping biological pathways may be a modifiable risk factor for mental health disorders. Notwithstanding subtle personalised gut microbiota signatures and associated metabolic function (Liu et al., 2020), there are broad dietary inputs that are optimal for system homeostasis, such as plant-based diets with a high content of grains/fibres, fermented foods and fish (Deehan et al., 2020; Dinan et al., 2019; Reynolds et al., 2019). Conversely, it is established that processed or fried foods, refined grains and sugary products are broadly detrimental to mental health (Khambadkone et al., 2020; Oddy et al., 2018).

Diet is the primary contributing factor to the gut microbiome and can rapidly alter its composition (David et al., 2014). Certain dietary patterns are associated with gut microbial signatures (De Filippis et al., 2016). Lower gut microbiota diversity is associated with low-fiber western diets (Sonnenburg & Sonnenburg, 2019; Vangay et al., 2018), whereas high-fermented-food diets may increase microbiota diversity and decrease inflammatory markers (Wastyk et al., 2021) and alter tryptophan metabolites (Zhu et al., 2020). A high-fat diet rodent model of depression showed that plasma tryptophan indole-metabolites and bile acids inversely correlated with gut microbiota signatures (Abildgaard et al., 2021).

Dietary or feeding patterns are governed by a balance between homeostatic and reward seeking systems (Murray et al., 2014; Rossi & Stuber, 2018). The meso-corticolimbic circuit and serotonergic and dopaminergic signalling are essential for homeostatic and reward-associated regulation of feeding behaviour and systemic energy metabolism. Preclinical studies indicate that MGB axis signalling contributes not only to homeostatic and energy metabolism regulation, but also the regulation of feeding behaviour (Alcock et al., 2014; Gabanyi et al., 2022; García-Cabrerizo et al., 2021; Leitão-Gonçalves et al., 2017; Trevelline & Kohl, 2022).

A re-appraisal of maladaptive or aberrant or unhealthy eating habits/dietary patterns, across the spectrum from overly restrictive to uncontrolled, triggered by psychedelic therapy (Foldi et al., 2020; Spriggs et al., 2021a, 2021b) could harness the reciprocal homeostatic and reward-processing cycles of the diet-MGB axis (Berding et al., 2021; Boscaini et al., 2022; Marx et al., 2021; Urrutia-Piñones et al., 2018). Similarly, given the overlapping relationship between diet, exercise and the MGB axis, psychedelic therapy-induced reappraisal of maladaptive physical activity patterns could be primed by optimal diet-MGB axis signalling before psychedelic therapy and then maintained after psychedelic therapy (Barton et al., 2018; Clarke et al., 2014; Clauss et al., 2021; O'Sullivan et al., 2015).

At this point, however, there is limited data on dietary and physical activity patterns pre- and post-psychedelic therapy (Teixeira et al., 2022). While self-reported survey data must be considered in the context of inherent self-selection and recall biases, there are some tentative indicators that diet, and exercise patterns may improve due to psychedelic experiences. 63% of participants endorsed “improved diet”, and 55% reported “increased exercise” in a survey primarily focussed on people who self-reported to have reduced (or stopped) alcohol consumption and misuse after a psychedelic experience (Garcia-Romeu et al., 2019). Similarly, 59% endorsed “improved diet”, and 58% endorsed “increased exercise” as a result of their psychedelic experience in a survey of people who self-reported to have stopped or reduced cannabis, opioid, or stimulant misuse after a psychedelic experience (Garcia-Romeu et al., 2020). It is also interesting to note that 96% of respondents who met criteria for substance use disorder before the psychedelic experience, decreased to 27% after the experience (Garcia-Romeu, et al., 2020).

A similarly cautious approach is warranted for the interpretation of a small open label study in TRD in which nearly half of the participants reported improvements in diet and exercise, and reductions in alcohol consumption (Carhart-Harris et al., 2016; Watts et al., 2017). While mild and transient GI disturbance can occur in approximately one fifth of people (Goodwin, et al., 2020), it is interesting to note one participant's account of the experience (Watts et al., 2017). This participant, who had pre-existing “stomach problems/food sensitivities”, re-appraised his relationship with food after psilocybin therapy, despite having no “lessons about diet” (Watts et al., 2017). While this participant reported changing his mindset and then his diet, during psilocybin dosing he initially experienced stomach pains and interpreted this as the psilocybin “going where it needed to go” (Teixeira et al., 2022; Watts et al., 2017).

Of course, each psychedelic therapy experience is complex, unique, and dependent on the individual's psycho-biologically encoded life narrative and priors. Yet, it raises interesting implications for conscious and unconscious information processing pathways between central and peripheral systems. The degree to which intent or conscious priors drive the self-reported improvements in diet and exercise in the aforementioned studies is not clear (Garcia-Romeu et al., 2020, 2019). But, as articulated by Teixeira and colleagues, this may open opportunities for behavioural therapy to further improve lifestyle factors (Teixeira et al., 2022). Moreover, psychedelic therapy induced expansion of the confines of the self to encompass an appreciation of the complex interconnectivity between micro and macro levels, inclusive of the microbiome, could potentially enhance the re-establishment and maintenance of diet-MGB and exercise-MBG axis patterns that are more homeostatically favourable to the individual.

The gut microbiota and adverse effects of psychedelics

When discussing side-effects, it is important to draw distinctions between clinical trials in which participants are thoroughly screened and supported and the potential side-effects from recreational use (Schlag et al., 2022). For example, most clinical studies exclude people with a family history of psychosis and major cardiac problems. Even with recreational use, reports of psychosis (Dos Santos et al., 2017), mania (Hendin & Penn, 2021), disturbed/unpredictable behaviour (Giancola et al., 2021; Honyiglo et al., 2019), serotonin syndrome (Schifano et al., 2021) and Hallucinogen Persisting Perception Disorder (HPPD) (Litjens et al., 2014) are seemingly rare. Data from the 2017 Global Drug Survey of 9233 past year magic mushroom users, 0.2% of respondents reported having sought emergency medical treatment, mainly for anxiety/panic and paranoia/suspiciousness (Kopra et al., 2022). This was associated with inadequate set and setting and consumption with other substances (Kopra et al., 2022).

The emergence of clinical trial data is beginning to provide a clearer scientific picture of the side-effects of psilocybin therapy, though large studies with extended periods of follow-up are still lacking. Although limited by a relatively small sample of 89 healthy volunteers, there were no serious adverse effects and no treatment-emergent adverse events (TEAEs) that led to study withdrawal in a randomised, double-blind, placebo-controlled study of a single oral dose of psilocybin 10 mg or 25 mg or placebo (Rucker et al., 2022). Preliminary data from the largest double-blind placebo-controlled dosing finding psilocybin therapy trial (n = 233) reported the most common side effects in the 25 mg group on dosing day were headache (24.1%), nausea (21.5%), dizziness (6.3%), and fatigue (6.3%), with 3.8% experiencing anxiety (Goodwin, et al., 2020). In the subgroup of TRD who continued SSRI's (n = 19), psilocybin was well-tolerated, with 11 (58%) participants reporting TEAEs, of which a majority (81%) were of mild severity (COMPASS, 2021). Given the role of serotonin in the stimulation of gut motility, it raises interesting questions of whether the diet-MGB axis may have subtle implications for the modulation of transient GI side-effects or autonomic nervous system effects in psychedelic therapy (Holze et al., 2021; Mörkl et al., 2022; Olbrich et al., 2021).

Psilocybiome: towards a personalised psychedelic treatment

The operation of clinical psychiatry requires a reductionistic approach to demarcate subjective experiences and observable behaviour according to severity of impairment and distress, and the application of the best available evidence-based treatment strategies. However, this “discipline of the unexplained” (Kelly, 2021) will only fully be explicated by understanding the complex constellation of fluctuating transitions of information processing across all levels from micro to macro (Fried, 2022; Fried & Robinaugh, 2020; Topol, 2014). In keeping with this interconnected systems-based approach, the microbiome has been proposed as an additional transdiagnostic unit of analysis in the Research Domain Criteria (RDoC) framework (Kelly et al., 2018). This evolving neuroscientific framework aims to integrate developmental processes and environmental inputs over the trajectory of the life course to determine the mechanisms underlying normal-range functioning, and then how disruptions correspond to dimensional psychopathology (Insel et al., 2010). It is hoped that the identification of targetable biosignatures that either cut across traditional disorder categories or that are unique to specific clinical phenomenona will lead to personalised treatment strategies and improve outcomes for people with mental health disorders (Krystal et al., 2020; Medeiros et al., 2020).

There is growing interest in incorporating the gut microbiome into personalised medicine schedules (Gupta et al., 2020). Gut microbiota signatures play a contributing role in the prediction of plasma glucose levels (Schüssler-Fiorenza Rose et al., 2019; Zeevi et al., 2015), glycaemic responses to exercise, (Liu et al., 2020) and lipid levels (Fu et al., 2015). Accruing data is starting to reveal the contributing influence of the gut microbiome on the variability of blood metabolites, also impacted by diet/lifestyle and genetic factors (Bar et al., 2020; Diener et al., 2022; Wilmanski et al., 2019). Microbial interventions also have an individualized impact on mucosal community structure and gut transcriptome (Johnson et al., 2019; Zmora et al., 2018). A recent example of a personalised microbiome approach, from a randomised, single-blinded, placebo-controlled trial of the prebiotic inulin in 106 people with obesity showed minimal changes in mood or cognition over placebo in the total group (Leyrolle et al., 2021). However, when stratified into responders and non-responders, according to differences between the positive and negative score in the Positive and Negative Affect Schedule, responders (who had worse baseline mood scores) had increased baseline levels of Coprococcus, IL-8 and altered glucose homeostasis (Leyrolle et al., 2021). After treatment, increases in Bifidobacterium and Haemophilus were larger in the responder group, thus highlighting the intricacies and potential applications of personalised microbiome strategies (Leyrolle et al., 2021).

Future clinical studies could explore whether configurations of the microbiome architecture preferentially respond to psychedelic therapy. Previous studies have suggested that responses to standard antidepressant approaches are influenced by peripheral mediators such as immune (Drevets et al., 2022; Yang et al., 2019), kynurenine pathway metabolites (Bhattacharyya et al., 2019; Erabi et al., 2020) and metabolomic profiles (Caspani et al., 2021). As discussed throughout this review, the multi-system capabilities of the MGB axis interact with these pathways, and may play a role in unravelling the complex multi-factorial reasons for the continuum of responses to conventional antidepressant (Donoso et al., 2022) and psychedelic therapy, particularly in people with dysregulated neuroimmune, neuro-endocrine and metabolic profiles. Indeed, emerging preclinical data indicates that the gut microbiome is also associated with divergent responses to antidepressants (Duan et al., 2021; Zhang et al., 2021) (Table 1).

The individual variability in psychotropic drug response and disposition is also partially influenced by the gut microbiota (Clarke et al., 2019; Cussotto et al., 2021; Maini Rekdal et al., 2019; Vich Vila et al., 2020; Zimmermann et al., 2019), though the physiological implications for serotonergic psychedelics have yet to be meaningfully explored or defined. Preclinical studies of single and interval dosing regimens of serotonergic psychedelics across multiple dose ranges using GF, antibiotic, psychobiotic, FMT and dietary manipulation rodent models, together with measures of intestinal and BBB permeability and brain neurochemistry, could assess the mechanistic relevance of the MGB axis to the effects of psychedelics (Kelly et al., 2015; Knox et al., 2022).

Clinical psychedelic research is still at a relatively early stage, and it is essential to continuously improve the methodological rigour of clinical trials, with particular attention to challenges related to treatment masking/blinding, self-selection bias, and patient expectations (Aday et al., 2022; Hayes et al., 2022). Preliminary results from the largest and most methodologically rigorous trial to date in TRD, which compared single doses of 25 mg, 10 mg and 1 mg showed a 37% response rate (MADRS) at 3 weeks in the 25 mg group compared to 18% in the 10 mg group and 17% in the 1 mg group (Goodwin, et al., 2020). Interestingly, in a separate open label pilot study of TRD participants who received psilocybin (25 mg) along with their SSRI medications (of at least 6 weeks) showed a similar response rate of 42% at 3 weeks (COMPASS, 2021). It remains to be seen whether the incorporation of the diet-MGB and exercise-MGB axis into psychedelic therapy will improve these response rates.

Summary, conclusions, and perspectives

The complex therapeutic mechanisms underlying psychedelic therapy traverse molecular, cellular, and network levels, under the influence of biofeedback signals from peripheral networks and the environment. Deciphering the clinical relevance of acute and transient changes in peripheral markers in healthy people and in those with dysregulated profiles associated with mental health disorders and their relationship with central markers in psychedelic therapy could lead to clinical benefits. As we have outlined, the gut microbial matrix influences overlapping neuroimmune, neuro-endocrine and transmitter signalling pathways of indirect applicability to the preparatory phase, the acute administration phase, and the integration phase of psychedelic therapy. This may occur even in the context of 1–3 dosing sessions.

Deeper mechanistic insights into the role of the MGB axis in modulating the effects of serotonergic psychedelics could be advanced by various microbiome manipulation approaches in rodent models. Exploration of reciprocal host-microbiota-psychedelic interactions, including diet-MGB and exercise-MGB axis interactions, perhaps amplified by components of behavioural therapy and digital health platforms, could, over the course of psychedelic therapy and into the longer-term, open avenues to counteract the negative consequences of stress-related feedforward cycles of maladaptive behavioural patterns to enhance therapeutic response rates.

Taken together, the MGB axis holds promise as an adjunctive modifiable translational target across the trajectory of psychedelic therapy, which in conjunction with other strategies could be leveraged to influence the continuum of therapeutic responses and improve outcomes in psychedelic therapy. Longitudinal clinical studies exploring the MGB axis over the trajectory of psychedelic therapy and into the long term, using different psychedelic doses compared to placebo, with central markers of brain activity, and then investigating various MGB axis modulation strategies (diet, probiotics, prebiotics, and antibiotics) particularly in the preparatory and pre-dosing phase, could help define whether the gut microbiome is a possible mediator and thus clinically relevant to the effects of psychedelics.

From a mindful microbes perspective, it is interesting to note that it has taken 350 years from the first human observation of microbes (Lane, 2015) to the current burgeoning implementation of the microbial system into the complex algorithms attempting to predict human emotion, cognition, and behaviour. Elucidating the precise constellation of molecular configurations and associated information signalling pathways of the psilocybiome, as an example of systems interconnectivity, will require an integrated multiomics approach. The future integration of the gut microbiome in studies implementing a dimensional approach would push the frontiers of an interconnected precise-personalised-systems based psychedelic therapy paradigm and further open the “doors of perception”.

Limitations

This is a narrative review exploring the potential role of the MGB axis in psychedelic therapy.

Funding

JK was the sub-PI on the COMPASS trials (COMP 001, 003, 004) in Ireland. JFC & TGD have research funding from Dupont Nutrition Biosciences APS, Cremo SA, Mead Johnson Nutrition, Nutricia Danone. JFC, TGD & GC have spoken at meetings sponsored by food and pharmaceutical companies. VO'K was supported through HRB Grant Code: 201,651.12553 and the Meath Foundation, Tallaght University Hospital. VO'K was the Principal Investigator (PI) on the COMPASS trials (COMP001, 003, 004) in Ireland.

CRediT authorship contribution statement

John R. Kelly: Writing – original draft. Gerard Clarke: Writing – review & editing. Andrew Harkin: Writing – review & editing. Sinead C. Corr: Writing – review & editing. Stephen Galvin: Writing – review & editing. Vishnu Pradeep: Writing – review & editing. John F. Cryan: Writing – review & editing. Veronica O'Keane: Writing – review & editing. Timothy G. Dinan: Writing – review & editing.

Acknowledgments

We would like to acknowledge Paul Quinlan who designed Fig. 1 and Christopher Connolly for his review of the manuscript.

References
[Abildgaard et al., 2021]
A. Abildgaard, T. Kern, O. Pedersen, T. Hansen, S. Lund, G. Wegener.
A diet-induced gut microbiota component and related plasma metabolites are associated with depressive-like behaviour in rats.
European Neuropsychopharmacology, 43 (2021), pp. 10-21
[Aday et al., 2022]
J.S. Aday, B.D. Heifets, S.D. Pratscher, E. Bradley, R. Rosen, J.D. Woolley.
Great Expectations: Recommendations for improving the methodological rigor of psychedelic clinical trials.
Psychopharmacology, 239 (2022), pp. 1989-2010
[Agus et al., 2018]
A. Agus, J. Planchais, H. Sokol.
Gut microbiota regulation of tryptophan metabolism in health and disease.
Cell Host Microbe, 23 (2018), pp. 716-724
[Akhtar and Mannan, 2020]
N. Akhtar, M.A. Mannan.
Mycoremediation: Expunging environmental pollutants.
Biotechnology Reports (Amst), 26 (2020), pp. e00452
[Alcock et al., 2014]
J. Alcock, C.C. Maley, C.A. Aktipis.
Is eating behavior manipulated by the gastrointestinal microbiota? Evolutionary pressures and potential mechanisms.
BioEssays: News and Reviews in Molecular, Cellular and Developmental Biology, 36 (2014), pp. 940-949
[Almond and Petersen, 2020]
Almond, R.E.A.M.G., & Petersen, T. (2020). Living planet report 2020 - bending the curve of biodiversity loss, from https://www.wwf.org.uk/sites/default/files/2020-09/LPR20_Full_report.pdf
[Ansaldo et al., 2021]
E. Ansaldo, T.K. Farley, Y. Belkaid.
Control of immunity by the microbiota.
Annual Review of Immunology, 39 (2021), pp. 449-479
[Auchtung et al., 2018]
T.A. Auchtung, T.Y. Fofanova, C.J. Stewart, A.K. Nash, M.C. Wong, J.R. Gesell, et al.
Investigating colonization of the healthy adult gastrointestinal tract by fungi.
mSphere., 3 (2018),
[Awan et al., 2018]
Awan, A.R., Winter, J.M., Turner, D., Shaw, W.M., Suz, L.M., Bradshaw, A.J., et al. (2018). Convergent evolution of psilocybin biosynthesis by psychedelic mushrooms. bioRxiv., 374199.
[Ayaz et al., 2015]
M. Ayaz, F. Subhan, J. Ahmed, A.U. Khan, F. Ullah, I. Ullah, et al.
Sertraline enhances the activity of antimicrobial agents against pathogens of clinical relevance.
Journal of Biological Research (Thessalon), 22 (2015), pp. 4
[Bäckhed et al., 2012]
F. Bäckhed, C.M. Fraser, Y. Ringel, M.E. Sanders, R.B. Sartor, P.M. Sherman, et al.
Defining a healthy human gut microbiome: Current concepts, future directions, and clinical applications.
Cell Host Microbe, 12 (2012), pp. 611-622
[Bar et al., 2020]
N. Bar, T. Korem, O. Weissbrod, D. Zeevi, D. Rothschild, S. Leviatan, et al.
A reference map of potential determinants for the human serum metabolome.
Nature, 588 (2020), pp. 135-140
[Barrett et al., 2020]
F.S. Barrett, M.K. Doss, N.D. Sepeda, J.J. Pekar, R.R. Griffiths.
Emotions and brain function are altered up to one month after a single high dose of psilocybin.
Scientific Reports, 10 (2020), pp. 2214
[Barton et al., 2018]
W. Barton, N.C. Penney, O. Cronin, I. Garcia-Perez, M.G. Molloy, E. Holmes, et al.
The microbiome of professional athletes differs from that of more sedentary subjects in composition and particularly at the functional metabolic level.
[Begec et al., 2013]
Z. Begec, A. Yucel, Y. Yakupogullari, M.A. Erdogan, Y. Duman, M. Durmus, et al.
Efeitos antimicrobianos de cetamina em combinação com propofol: Um estudo in vitro.
Brazilian Journal of Anesthesiology, 63 (2013), pp. 461-465
[Berding et al., 2021]
K. Berding, K. Vlckova, W. Marx, H. Schellekens, C. Stanton, G. Clarke, et al.
Diet and the Microbiota-Gut-Brain Axis: Sowing the seeds of good mental health.
Advances in Nutrition, 12 (2021), pp. 1239-1285
[Beumer et al., 2012]
W. Beumer, S.M. Gibney, R.C. Drexhage, L. Pont-Lezica, J. Doorduin, H.C. Klein, et al.
The immune theory of psychiatric diseases: A key role for activated microglia and circulating monocytes.
Journal of Leukocyte Biology, 92 (2012), pp. 959-975
[Bhattacharyya et al., 2019]
S. Bhattacharyya, A.T. Ahmed, M. Arnold, D. Liu, C. Luo, H. Zhu, et al.
Metabolomic signature of exposure and response to citalopram/escitalopram in depressed outpatients.
Translational Psychiatry, 9 (2019), pp. 173
[Bhattarai et al., 2018]
Y. Bhattarai, B.B. Williams, E.J. Battaglioli, W.R. Whitaker, L. Till, M. Grover, et al.
Gut microbiota-produced tryptamine activates an epithelial G-protein-coupled receptor to increase colonic secretion.
Cell Host Microbe, 23 (2018), pp. 775-785
[Blei et al., 2020]
F. Blei, S. Dörner, J. Fricke, F. Baldeweg, F. Trottmann, A. Komor, et al.
Simultaneous Production of psilocybin and a cocktail of β-carboline monoamine oxidase inhibitors in "Magic" mushrooms.
Chemistry (Weinheim An Der Bergstrasse, Germany), 26 (2020), pp. 729-734
[Bogenschutz et al., 2022]
M.P. Bogenschutz, S. Ross, S. Bhatt, T. Baron, A.A. Forcehimes, E. Laska, et al.
Percentage of heavy drinking days following psilocybin-assisted psychotherapy vs placebo in the treatment of adult patients with alcohol use disorder: A randomized clinical trial.
JAMA Psychiatry, 79 (2022), pp. 953-962
[Bohnert et al., 2011]
J.A. Bohnert, M. Szymaniak-Vits, S. Schuster, W.V. Kern.
Efflux inhibition by selective serotonin reuptake inhibitors in Escherichia coli.
Journal of Antimicrobial Chemotherapy, 66 (2011), pp. 2057-2060
[Bonneville et al., 2020]
S. Bonneville, F. Delpomdor, A. Préat, C. Chevalier, T. Araki, M. Kazemian, et al.
Molecular identification of fungi microfossils in a Neoproterozoic shale rock.
Science Advances, 6 (2020), pp. eaax7599
[Borkent et al., 2022]
J. Borkent, M. Ioannou, J.D. Laman, B.C.M. Haarman, I.E.C. Sommer.
Role of the gut microbiome in three major psychiatric disorders.
Psychological Medicine, 52 (2022), pp. 1222-1242
[Boscaini et al., 2022]
S. Boscaini, S.-J. Leigh, A. Lavelle, R. García-Cabrerizo, T. Lipuma, G. Clarke, et al.
Microbiota and body weight control: Weight watchers within?.
Molecular Metabolism, 57 (2022),
[Boyce et al., 2019]
G.R. Boyce, E. Gluck-Thaler, J.C. Slot, J.E. Stajich, W.J. Davis, T.Y. James, et al.
Psychoactive plant- and mushroom-associated alkaloids from two behavior modifying cicada pathogens.
Fungal Ecology, 41 (2019), pp. 147-164
[Buffington et al., 2016]
S.A. Buffington, G.V. Di Prisco, T.A. Auchtung, N.J. Ajami, J.F. Petrosino, M. Costa-Mattioli.
Microbial reconstitution reverses maternal diet-induced social and synaptic deficits in offspring.
Cell, 165 (2016), pp. 1762-1775
[Carbia et al., 2021]
C. Carbia, S. Lannoy, P. Maurage, E. López-Caneda, K.J. O'Riordan, T.G. Dinan, et al.
A biological framework for emotional dysregulation in alcohol misuse: from gut to brain.
Molecular Psychiatry, 26 (2021), pp. 1098-1118
[Carhart-Harris et al., 2018]
R.L. Carhart-Harris, M. Bolstridge, C.M.J. Day, J. Rucker, R. Watts, D.E. Erritzoe, et al.
Psilocybin with psychological support for treatment-resistant depression: Six-month follow-up.
Psychopharmacology, 235 (2018), pp. 399-408
[Carhart-Harris et al., 2016]
R.L. Carhart-Harris, M. Bolstridge, J. Rucker, C.M. Day, D. Erritzoe, M. Kaelen, et al.
Psilocybin with psychological support for treatment-resistant depression: An open-label feasibility study.
Lancet Psychiatry, 3 (2016), pp. 619-627
[Carhart-Harris and Friston, 2019]
R.L. Carhart-Harris, K.J. Friston.
REBUS and the anarchic brain: Toward a Unified model of the brain action of psychedelics.
Pharmacological Reviews, 71 (2019), pp. 316-344
[Carhart-Harris et al., 2021]
R. Carhart-Harris, B. Giribaldi, R. Watts, M. Baker-Jones, A. Murphy-Beiner, R. Murphy, et al.
Trial of psilocybin versus escitalopram for depression.
New England Journal of Medicine, 384 (2021), pp. 1402-1411
[Caspani et al., 2021]
G. Caspani, G. Turecki, R.W. Lam, R.V. Milev, B.N. Frey, G.M. MacQueen, et al.
Metabolomic signatures associated with depression and predictors of antidepressant response in humans: A CAN-BIND-1 report.
Communications Biology, 4 (2021), pp. 903
[Ceballos et al., 2017]
G. Ceballos, P.R. Ehrlich, R. Dirzo.
Biological annihilation via the ongoing sixth mass extinction signaled by vertebrate population losses and declines.
Proceedings of the National Academy of Sciences, 114 (2017), pp. E6089-E6096
[Chen et al., 2019]
H. Chen, P.-K. Nwe, Y. Yang, C.E. Rosen, A.A. Bielecka, M. Kuchroo, et al.
A forward chemical genetic screen reveals gut microbiota metabolites that modulate host physiology.
Cell, 177 (2019), pp. 1217-1231
[Clarke et al., 2013]
G. Clarke, S. Grenham, P. Scully, P. Fitzgerald, R.D. Moloney, F. Shanahan, et al.
The microbiome-gut-brain axis during early life regulates the hippocampal serotonergic system in a sex-dependent manner.
Molecular Psychiatry, 18 (2013), pp. 666-673
[Clarke et al., 2019]
G. Clarke, K.V. Sandhu, B.T. Griffin, T.G. Dinan, J.F. Cryan, N.P. Hyland.
Gut reactions: Breaking down xenobiotic-microbiome interactions.
Pharmacological Reviews, 71 (2019), pp. 198-224
[Clarke et al., 2014]
S.F. Clarke, E.F. Murphy, O. O'Sullivan, A.J. Lucey, M. Humphreys, A. Hogan, et al.
Exercise and associated dietary extremes impact on gut microbial diversity.
[Clauss et al., 2021]
M. Clauss, P. Gérard, A. Mosca, M. Leclerc.
Interplay between exercise and gut microbiome in the context of human health and performance.
Frontiers in Nutrition, 8 (2021),
[Cobb, 2020]
M. Cobb.
The idea of the brain: The past and future of neuroscience.
Basic Books, (2020),
[Colosimo et al., 2019]
D.A. Colosimo, J.A. Kohn, P.M. Luo, F.J. Piscotta, S.M. Han, A.J. Pickard, et al.
Mapping interactions of microbial metabolites with human G-protein-coupled receptors.
Cell Host & Microbe, 26 (2019), pp. 273-282
[COMPASS 2021]
COMPASS. (2021). COMP360 psilocybin therapy in treatment-resistant depression: phase IIb results. Psilocybin therapy as adjunct to SSRI antidepressants in Treatment Resistant Depression, from https://compasspathways.com/category/articles/compass-news/.
[Cooley et al., 2018]
J.R. Cooley, D.C. Marshall, K.B.R. Hill.
A specialized fungal parasite (Massospora cicadina) hijacks the sexual signals of periodical cicadas (Hemiptera: Cicadidae: Magicicada).
Scientific Reports, 8 (2018), pp. 1432
[Correia et al., 2003]
T. Correia, N. Grammel, I. Ortel, U. Keller, P. Tudzynski.
Molecular cloning and analysis of the ergopeptine assembly system in the ergot fungus Claviceps purpurea.
Chemistry & Biology, 10 (2003), pp. 1281-1292
[Cowan et al., 2016]
C.S. Cowan, B.L. Callaghan, R. Richardson.
The effects of a probiotic formulation (Lactobacillus rhamnosus and L. helveticus) on developmental trajectories of emotional learning in stressed infant rats.
Translational Psychiatry, 6 (2016), pp. e823
[Cryan et al., 2018]
J.F. Cryan, G. Clarke, T.G. Dinan, H. Schellekens.
A microbial drugstore for motility.
Cell Host & Microbe, 23 (2018), pp. 691-692
[Cryan and Dinan, 2012]
J.F. Cryan, T.G. Dinan.
Mind-altering microorganisms: The impact of the gut microbiota on brain and behaviour.
Nature Reviews Neuroscience, 13 (2012), pp. 701-712
[Cryan and Mazmanian, 2022]
J.F. Cryan, S.K. Mazmanian.
Microbiota-brain axis: Context and causality.
Science, 376 (2022), pp. 938-939
[Cryan et al., 2019]
J.F. Cryan, K.J. O'Riordan, C.S.M. Cowan, K.V. Sandhu, T.F.S. Bastiaanssen, M. Boehme, et al.
The microbiota-gut-brain axis.
Physiological Reviews, 99 (2019), pp. 1877-2013
[Cussotto et al., 2021]
S. Cussotto, G. Clarke, T.G. Dinan, J.F. Cryan.
Psychotropic drugs and the microbiome.
Mod Trends Psychiatry, 32 (2021), pp. 113-133
[Cussotto et al., 2019]
S. Cussotto, C.R. Strain, F. Fouhy, R.G. Strain, V.L. Peterson, G. Clarke, et al.
Differential effects of psychotropic drugs on microbiome composition and gastrointestinal function.
Psychopharmacology, 236 (2019), pp. 1671-1685
[Cussotto et al., 2021]
S. Cussotto, J. Walsh, A.V. Golubeva, A.V. Zhdanov, C.R. Strain, F. Fouhy, et al.
The gut microbiome influences the bioavailability of olanzapine in rats.
EBioMedicine, 66 (2021),
[da Silva et al., 2019]
F.S. da Silva, E.A.S. Silva, G.M. Sousa Jr., J.P. Maia-de-Oliveira, V.P. Soares-Rachetti, D.B. de Araujo, et al.
Acute effects of ayahuasca in a juvenile non-human primate model of depression.
Brazilian Journal of Psychiatry, 41 (2019), pp. 280-288
[David et al., 2014]
L.A. David, C.F. Maurice, R.N. Carmody, D.B. Gootenberg, J.E. Button, B.E. Wolfe, et al.
Diet rapidly and reproducibly alters the human gut microbiome.
Nature, 505 (2014), pp. 559-563
[Davis et al., 2021]
A.K. Davis, F.S. Barrett, D.G. May, M.P. Cosimano, N.D. Sepeda, M.W. Johnson, et al.
Effects of psilocybin-assisted therapy on major depressive disorder: a randomized clinical trial.
JAMA Psychiatry, 78 (2021), pp. 481-489
[de Almeida et al., 2019]
R.N. de Almeida, A.C.M. Galvão, F.S. da Silva, E. Silva, F. Palhano-Fontes, J.P. Maia-de-Oliveira, et al.
Modulation of serum brain-derived neurotrophic factor by a single dose of ayahuasca: Observation from a randomized controlled trial.
Frontiers in Psychology, 10 (2019), pp. 1234
[de Bekker et al., 2014]
C. de Bekker, L.E. Quevillon, P.B. Smith, K.R. Fleming, D. Ghosh, A.D. Patterson, et al.
Species-specific ant brain manipulation by a specialized fungal parasite.
BMC Evolution. Biology, 14 (2014), pp. 166
[De Filippis et al., 2016]
F. De Filippis, N. Pellegrini, L. Vannini, I.B. Jeffery, A. La Storia, L. Laghi, et al.
High-level adherence to a Mediterranean diet beneficially impacts the gut microbiota and associated metabolome.
[De Filippo et al., 2010]
C. De Filippo, D. Cavalieri, M. Di Paola, M. Ramazzotti, J.B. Poullet, S. Massart, et al.
Impact of diet in shaping gut microbiota revealed by a comparative study in children from Europe and rural Africa.
Proceedings of the National Academy of Sciences, 107 (2010), pp. 14691-14696
[de Vos et al., 2022]
W.M. de Vos, H. Tilg, M. Van Hul, P.D. Cani.
Gut microbiome and health: mechanistic insights.
Gut, (2022),
[Deehan et al., 2020]
E.C. Deehan, C. Yang, M.E. Perez-Muñoz, N.K. Nguyen, C.C. Cheng, L. Triador, et al.
Precision microbiome modulation with discrete dietary fiber structures directs short-chain fatty acid production.
Cell Host & Microbe, 27 (2020), pp. 389-404
[Desbonnet et al., 2014]
L. Desbonnet, G. Clarke, F. Shanahan, T.G. Dinan, J.F. Cryan.
Microbiota is essential for social development in the mouse.
Molecular Psychiatry, 19 (2014), pp. 146-148
[Desbonnet et al., 2015]
L. Desbonnet, G. Clarke, A. Traplin, O. O'Sullivan, F. Crispie, R.D. Moloney, et al.
Gut microbiota depletion from early adolescence in mice: Implications for brain and behaviour.
Brain, Behavior, and Immunity, 48 (2015), pp. 165-173
[Desbonnet et al., 2008]
L. Desbonnet, L. Garrett, G. Clarke, J. Bienenstock, T.G. Dinan.
The probiotic bifidobacteria infantis: an assessment of potential antidepressant properties in the rat.
Journal of Psychiatric Research, 43 (2008), pp. 164-174
[Deveau et al., 2018]
A. Deveau, G. Bonito, J. Uehling, M. Paoletti, M. Becker, S. Bindschedler, et al.
Bacterial–fungal interactions: Ecology, mechanisms and challenges.
FEMS Microbiology Reviews, 42 (2018), pp. 335-352
[Diener et al., 2022]
Diener, C., Dai, C.L., Wilmanski, T., Baloni, P., Smith, B., Rappaport, N., et al. (2022). Genome-microbiome interplay provides insight into the determinants of the human blood metabolome. bioRxiv, 2022.2002.2004.479172.
[Dinan et al., 2019]
T.G. Dinan, C. Stanton, C. Long-Smith, P. Kennedy, J.F. Cryan, C.S.M. Cowan, et al.
Feeding melancholic microbes: MyNewGut recommendations on diet and mood.
Clinical Nutrition, 38 (2019), pp. 1995-2001
[Dinis-Oliveira, 2017]
R.J. Dinis-Oliveira.
Metabolism of psilocybin and psilocin: clinical and forensic toxicological relevance.
Drug Metabolism Reviews, 49 (2017), pp. 84-91
[Dong et al., 2022]
T.S. Dong, M. Guan, E.A. Mayer, J. Stains, C. Liu, P. Vora, et al.
Obesity is associated with a distinct brain-gut microbiome signature that connects Prevotella and Bacteroides to the brain's reward center.
Gut Microbes, 14 (2022),
[Donoso et al., 2022]
F. Donoso, J.F. Cryan, L. Olavarría-Ramírez, Y.M. Nolan, G. Clarke.
Inflammation, lifestyle factors, and the microbiome-gut-brain axis: Relevance to depression and antidepressant action.
Clinical Pharmacology & Therapeutics, (2022),
[Dos Santos et al., 2017]
R.G. Dos Santos, J.C. Bouso, J.E.C Hallak.
Ayahuasca, dimethyltryptamine, and psychosis: A systematic review of human studies.
Ther Adv Psychopharmacol, 7 (2017), pp. 141-157
[Dos Santos et al., 2021]
R.G. Dos Santos, F.L. Osório, J.M. Rocha, G.N. Rossi, J.C. Bouso, L.S. Rodrigues, et al.
Ayahuasca Improves self-perception of speech performance in subjects with social anxiety disorder: A pilot, proof-of-concept, randomized, placebo-controlled trial.
Journal of Clinical Psychopharmacology, 41 (2021), pp. 540-550
[Drevets et al., 2022]
W.C. Drevets, G.M. Wittenberg, E.T. Bullmore, H.K. Manji.
Immune targets for therapeutic development in depression: towards precision medicine.
Nature Reviews Drug Discovery, 21 (2022), pp. 224-244
[Duan et al., 2021]
J. Duan, Y. Huang, X. Tan, T. Chai, J. Wu, H. Zhang, et al.
Characterization of gut microbiome in mice model of depression with divergent response to escitalopram treatment.
Translational Psychiatry, 11 (2021), pp. 303
[Dusengemungu et al., 2021]
L. Dusengemungu, G. Kasali, C. Gwanama, B. Mubemba.
Overview of fungal bioleaching of metals.
Environmental Advances, 5 (2021),
[Eadie, 2003]
M.J. Eadie.
Convulsive ergotism: Epidemics of the serotonin syndrome?.
The Lancet Neurology, 2 (2003), pp. 429-434
[Erabi et al., 2020]
H. Erabi, G. Okada, C. Shibasaki, D. Setoyama, D. Kang, M. Takamura, et al.
Kynurenic acid is a potential overlapped biomarker between diagnosis and treatment response for depression from metabolome analysis.
Scientific Reports, 10 (2020), pp. 16822
[Erdman, 2021]
S.E. Erdman.
Oxytocin and the microbiome.
Current Opinion in Endocrine and Metabolic Research, 19 (2021), pp. 8-14
[Erny et al., 2015]
D. Erny, A.L. Hrabe de Angelis, D. Jaitin, P. Wieghofer, O. Staszewski, E. David, et al.
Host microbiota constantly control maturation and function of microglia in the CNS.
Nature Neuroscience, 18 (2015), pp. 965-977
[Eskola et al., 2020]
M. Eskola, G. Kos, C.T. Elliott, J. Hajšlová, S. Mayar, R. Krska.
Worldwide contamination of food-crops with mycotoxins: Validity of the widely cited ‘FAO estimate’ of 25%.
Critical Reviews in Food Science and Nutrition, 60 (2020), pp. 2773-2789
[Falony et al., 2016]
G. Falony, M. Joossens, S. Vieira-Silva, J. Wang, Y. Darzi, K. Faust, et al.
Population-level analysis of gut microbiome variation.
Science, 352 (2016), pp. 560-564
[Flanagan et al., 2019]
T.W. Flanagan, M.N. Sebastian, D.M. Battaglia, T.P. Foster, S.A. Cormier, C.D. Nichols.
5-HT2 receptor activation alleviates airway inflammation and structural remodeling in a chronic mouse asthma model.
Life Sciences, 236 (2019),
[Flanagan et al., 2019]
T.W. Flanagan, M.N. Sebastian, D.M. Battaglia, T.P. Foster, E.L. Maillet, C.D. Nichols.
Activation of 5-HT(2) receptors reduces inflammation in vascular tissue and cholesterol levels in high-fat diet-fed apolipoprotein E knockout mice.
Science Reports, 9 (2019), pp. 13444
[Fluhr et al., 2021]
L. Fluhr, U. Mor, A.A. Kolodziejczyk, M. Dori-Bachash, A. Leshem, S. Itav, et al.
Gut microbiota modulates weight gain in mice after discontinued smoke exposure.
Nature, 600 (2021), pp. 713-719
[Foldi et al., 2020]
C.J. Foldi, P. Liknaitzky, M. Williams, B.J. Oldfield.
Rethinking therapeutic strategies for anorexia nervosa: Insights from psychedelic medicine and animal models. [Mini Review].
Frontiers in Neuroscience, 14 (2020),
[Forssten et al., 2022]
S.D. Forssten, A.C. Ouwehand, S.M. Griffin, E. Patterson.
One giant leap from mouse to man: The microbiota–gut–brain axis in mood disorders and translational challenges moving towards human clinical trials.
Nutrients, 14 (2022), pp. 568
[Foster et al., 2017]
J.A. Foster, L. Rinaman, J.F. Cryan.
Stress & the gut-brain axis: Regulation by the microbiome.
Neurobiology of Stress, 7 (2017), pp. 124-136
[Fredericksen et al., 2017]
M.A. Fredericksen, Y. Zhang, M.L. Hazen, R.G. Loreto, C.A. Mangold, D.Z. Chen, et al.
Three-dimensional visualization and a deep-learning model reveal complex fungal parasite networks in behaviorally manipulated ants.
Proceedings of the National Academy of Sciences, 114 (2017), pp. 12590-12595
[Fricke et al., 2017]
J. Fricke, F. Blei, D. Hoffmeister.
Enzymatic Synthesis of Psilocybin.
Angewandte Chemie International Edition, 56 (2017), pp. 12352-12355
[Fried and Robinaugh, 2020]
E.I. Fried, D.J. Robinaugh.
Systems all the way down: Embracing complexity in mental health research.
BMC Medicine, 18 (2020), pp. 205
[Fried, E. I. (2022)]
Fried, E. I. (2022). Studying mental health problems as systems, not syndromes. Current Directions in Psychological Science, 09637214221114089.
[Fu et al., 2015]
J. Fu, J. Bonder Marc, C. Cenit María, F. Tigchelaar Ettje, A. Maatman, A.M. Dekens Jackie, et al.
The gut microbiome contributes to a substantial proportion of the variation in blood lipids.
Circulation Research, 117 (2015), pp. 817-824
[Fülling et al., 2019]
C. Fülling, T.G. Dinan, J.F. Cryan.
Gut Microbe to brain signaling: What happens in vagus….
Neuron, 101 (2019), pp. 998-1002
[Fung et al., 2019]
T.C. Fung, H.E. Vuong, C.D.G. Luna, G.N. Pronovost, A.A. Aleksandrova, N.G. Riley, et al.
Intestinal serotonin and fluoxetine exposure modulate bacterial colonization in the gut.
Nature Microbiology, 4 (2019), pp. 2064-2073
[Gabanyi et al., 2022]
I. Gabanyi, G. Lepousez, R. Wheeler, A. Vieites-Prado, A. Nissant, S. Wagner, et al.
Bacterial sensing via neuronal Nod2 regulates appetite and body temperature.
Science, 376 (2022), pp. eabj3986
[Gacesa et al., 2022]
R. Gacesa, A. Kurilshikov, A. Vich Vila, T. Sinha, M.A.Y. Klaassen, L.A. Bolte, et al.
Environmental factors shaping the gut microbiome in a Dutch population.
Nature, 604 (2022), pp. 732-739
[Galvão et al., 2018]
A.C.M. Galvão, R.N. de Almeida, E. Silva, F.A.M. Freire, F. Palhano-Fontes, H. Onias, et al.
Cortisol modulation by ayahuasca in patients with treatment resistant depression and healthy controls.
Frontiers in Psychiatry, 9 (2018), pp. 185
[Galvão-Coelho et al., 2020]
N.L. Galvão-Coelho, A.C. de Menezes Galvão, R.N. de Almeida, F. Palhano-Fontes, I. Campos Braga, B. Lobão Soares, et al.
Changes in inflammatory biomarkers are related to the antidepressant effects of Ayahuasca.
Journal of Psychopharmacology (Oxford, England), 34 (2020), pp. 1125-1133
[Gandy et al., 2020]
S. Gandy, M. Forstmann, R.L. Carhart-Harris, C. Timmermann, D. Luke, R. Watts.
The potential synergistic effects between psychedelic administration and nature contact for the improvement of mental health.
Health Psychology Open, 7 (2020),
[García-Cabrerizo et al., 2021]
R. García-Cabrerizo, C. Carbia, O.R. KJ, H. Schellekens, J.F. Cryan.
Microbiota-gut-brain axis as a regulator of reward processes.
Journal of Neurochemistry, 157 (2021), pp. 1495-1524
[Garcia-Romeu et al., 2020]
A. Garcia-Romeu, A.K. Davis, E. Erowid, F. Erowid, R.R. Griffiths, M.W. Johnson.
Persisting reductions in cannabis, opioid, and stimulant misuse after naturalistic psychedelic use: An online survey. [Original research].
Frontiers in Psychiatry, 10 (2020), pp. 955
[Garcia-Romeu et al., 2019]
A. Garcia-Romeu, A.K. Davis, F. Erowid, E. Erowid, R.R. Griffiths, M.W. Johnson.
Cessation and reduction in alcohol consumption and misuse after psychedelic use.
Journal of Psychopharmacology, 33 (2019), pp. 1088-1101
[Garcia-Romeu et al., 2014]
A. Garcia-Romeu, R.R. Griffiths, M.W. Johnson.
Psilocybin-occasioned mystical experiences in the treatment of tobacco addiction.
Current Drug Abuse Reviews, 7 (2014), pp. 157-164
[Getachew et al., 2018]
B. Getachew, J.I. Aubee, R.S. Schottenfeld, A.B. Csoka, K.M. Thompson, Y. Tizabi.
Ketamine interactions with gut-microbiota in rats: Relevance to its antidepressant and anti-inflammatory properties.
BMC Microbiology, 18 (2018), pp. 222
[Gheorghe et al., 2019]
C.E. Gheorghe, J.A. Martin, F.V. Manriquez, T.G. Dinan, J.F. Cryan, G. Clarke.
Focus on the essentials: Tryptophan metabolism and the microbiome-gut-brain axis.
Current Opinion in Pharmacology, 48 (2019), pp. 137-145
[Giancola et al., 2021]
N.B. Giancola, C.J. Korson, J.P. Caplan, C.A. McKnight.
A “Trip” to the intensive care unit: An intravenous injection of psilocybin.
Journal of the Academy of Consultation-Liaison Psychiatry, 62 (2021), pp. 370-371
[Gocmen et al., 2008]
S. Gocmen, U. Buyukkocak, O. Caglayan.
In vitro investigation of the antibacterial effect of ketamine.
Upsala Journal of Medical Sciences, 113 (2008), pp. 39-46
[Goodwin et al., 2019]
G.M. Goodwin, S.T. Aaronson, O. Alvarez, P.C. Arden, A. Baker, J.C. Bennett, et al.
Single-dose psilocybin for a treatment-resistant episode of major depression.
New England Journal of Medicine, 387 (2022), pp. 1637-1648
[González-Arancibia et al., 2019]
C. González-Arancibia, J. Urrutia-Piñones, J. Illanes-González, J. Martinez-Pinto, R. Sotomayor-Zárate, M. Julio-Pieper, et al.
Do your gut microbes affect your brain dopamine?.
Psychopharmacology, 236 (2019), pp. 1611-1622
[Grimm et al., 2018]
O. Grimm, R. Kraehenmann, K.H. Preller, E. Seifritz, F.X. Vollenweider.
Psilocybin modulates functional connectivity of the amygdala during emotional face discrimination.
European Neuropsychopharmacology, 28 (2018), pp. 691-700
[Grosberg and Strathmann, 2007]
R.K. Grosberg, R.R. Strathmann.
The evolution of multicellularity: A minor major transition?.
Annual Review of Ecology, Evolution, and Systematics, 38 (2007), pp. 621-654
[Gukasyan et al., 2022]
N. Gukasyan, A.K. Davis, F.S. Barrett, M.P. Cosimano, N.D. Sepeda, M.W. Johnson, et al.
Efficacy and safety of psilocybin-assisted treatment for major depressive disorder: Prospective 12-month follow-up.
Journal of Psychopharmacology, 36 (2022), pp. 151-158
[Gupta et al., 2020]
V.K. Gupta, M. Kim, U. Bakshi, K.Y. Cunningham, J.M. Davis, K.N. Lazaridis, et al.
A predictive index for health status using species-level gut microbiome profiling.
Nature Communications, 11 (2020), pp. 4635
[Hallen-Adams and Suhr, 2017]
H.E. Hallen-Adams, M.J. Suhr.
Fungi in the healthy human gastrointestinal tract.
Virulence, 8 (2017), pp. 352-358
[Han et al., 2018]
W. Han, L.A. Tellez, M.H. Perkins, I.O. Perez, T. Qu, J. Ferreira, et al.
A neural circuit for gut-induced reward.
[Hasler et al., 2004]
F. Hasler, U. Grimberg, M.A. Benz, T. Huber, F.X. Vollenweider.
Acute psychological and physiological effects of psilocybin in healthy humans: A double-blind, placebo-controlled dose-effect study.
Psychopharmacology, 172 (2004), pp. 145-156
[Hawksworth, 2001]
D.L. Hawksworth.
The magnitude of fungal diversity: the 1.5 million species estimate revisited* *paper presented at the Asian Mycological Congress 2000 (AMC 2000), incorporating the 2nd asia-pacific mycological congress on biodiversity and biotechnology, and held at the University of Hong Kong on 9-13 July 2000.
Mycological Research, 105 (2001), pp. 1422-1432
[Hayes et al., 2022]
C. Hayes, M. Wahba, S. Watson.
Will psilocybin lose its magic in the clinical setting?.
Therapeutic Advances in Psychopharmacology, 12 (2022),
[Hendin and Penn, 2021]
H.M. Hendin, A.D. Penn.
An episode of mania following self-reported ingestion of psilocybin mushrooms in a woman previously not diagnosed with bipolar disorder: A case report.
Bipolar Disorders, 23 (2021), pp. 733-735
[Hertli and Zimmermann, 2022]
S. Hertli, P. Zimmermann.
Molecular interactions between the intestinal microbiota and the host.
Molecular Microbiology, 117 (2022), pp. 1297-1307
[Hoban et al., 2017]
A.E. Hoban, R.M. Stilling, M.M. G, R.D. Moloney, F. Shanahan, T.G. Dinan, et al.
Microbial regulation of microRNA expression in the amygdala and prefrontal cortex.
[Holze et al., 2021]
F. Holze, P. Vizeli, L. Ley, F. Müller, P. Dolder, M. Stocker, et al.
Acute dose-dependent effects of lysergic acid diethylamide in a double-blind placebo-controlled study in healthy subjects.
Neuropsychopharmacology, 46 (2021), pp. 537-544
[Honyiglo et al., 2019]
E. Honyiglo, A. Franchi, N. Cartiser, C. Bottinelli, A.S. Advenier, F. Bévalot, et al.
Unpredictable behavior under the influence of "Magic mushrooms": A case report and review of the literature.
Journal of Forensic Science, 64 (2019), pp. 1266-1270
[Hublin et al., 2017]
J.J. Hublin, A. Ben-Ncer, S.E. Bailey, S.E. Freidline, S. Neubauer, M.M. Skinner, et al.
New fossils from Jebel Irhoud, Morocco and the pan-African origin of Homo sapiens.
Nature, 546 (2017), pp. 289-292
[Hutten et al., 2021]
N. Hutten, N.L. Mason, P.C. Dolder, E.L. Theunissen, F. Holze, M.E. Liechti, et al.
Low doses of LSD acutely increase BDNF blood plasma levels in healthy volunteers.
ACS Pharmacology & Translational Science, 4 (2021), pp. 461-466
[Huttenhower et al., 2012]
C. Huttenhower, D. Gevers, R. Knight, S. Abubucker, J.H. Badger, The Human Microbiome Project, C, et al.
Structure, function and diversity of the healthy human microbiome. [Article].
Nature, 486 (2012), pp. 207
[Insel et al., 2010]
T. Insel, B. Cuthbert, M. Garvey, R. Heinssen, D.S. Pine, K. Quinn, et al.
Research domain criteria (RDoC): Toward a new classification framework for research on mental disorders.
American Journal of Psychiatry, 167 (2010), pp. 748-751
[Israelyan et al., 2019]
N. Israelyan, A. Del Colle, Z. Li, Y. Park, A. Xing, J.P.R. Jacobsen, et al.
Effects of serotonin and slow-release 5-hydroxytryptophan on gastrointestinal motility in a mouse model of depression.
Gastroenterology, 157 (2019), pp. 507-521
[Inserra 2022]
Inserra, A., Giorgini, G., Lacroix, S., Bertazzo, A., Choo, J., Markopolous, A., et al. (2022). Effects of Repeated Lysergic Acid Diethylamide (LSD) on the Mouse Brain Endocannabinoidome and Gut Microbiome. Br J Pharmacol.
[Ivanov et al., 2022]
II Ivanov, T. Tuganbaev, A.N. Skelly, K. Honda.
T cell responses to the microbiota.
Annual Review of Immunology, 40 (2022), pp. 559-587
[Jadhav et al., 2018]
K.S. Jadhav, V.L. Peterson, O. Halfon, G. Ahern, F. Fouhy, C. Stanton, et al.
Gut microbiome correlates with altered striatal dopamine receptor expression in a model of compulsive alcohol seeking.
Neuropharmacology, 141 (2018), pp. 249-259
[Johnson et al., 2019]
A.J. Johnson, P. Vangay, G.A. Al-Ghalith, B.M. Hillmann, T.L. Ward, R.R. Shields-Cutler, et al.
Daily sampling reveals personalized diet-microbiome associations in humans.
Cell Host Microbe, 25 (2019), pp. 789-802
[Johnson et al., 2020]
C.K. Johnson, P.L. Hitchens, P.S. Pandit, J. Rushmore, T.S. Evans, C.C.W. Young, et al.
Global shifts in mammalian population trends reveal key predictors of virus spillover risk.
Proceedings of the Royal Society B: Biological Sciences, 287 (2020),
[Johnson, 2020]
K.V.A. Johnson.
Gut microbiome composition and diversity are related to human personality traits.
Human Microbiome Journal, 15 (2020),
[Johnson et al., 2014]
M.W. Johnson, A. Garcia-Romeu, M.P. Cosimano, R.R. Griffiths.
Pilot study of the 5-HT2AR agonist psilocybin in the treatment of tobacco addiction.
Journal of Psychopharmacology, 28 (2014), pp. 983-992
[Jones et al., 2020]
Jones, N.T., Zahid, Z., Grady, S.M., Sultan, Z.W., Zheng, Z., Banks, M.I., et al. (2020). Delayed anxiolytic-like effects of psilocybin in male mice are supported by acute glucocorticoid release. bioRxiv, 2020.2008.2012.248229.
[Kelly, 2021]
B.D. Kelly.
Psychiatry is essential for now but might eventually disappear (although this is unlikely to happen any time soon).
Australasian Psychiatry, (2021),
[Kelly et al., 2018]
J.R. Kelly, G. Clarke, J.F. Cryan, T.G. Dinan.
Dimensional thinking in psychiatry in the era of the Research Domain Criteria (RDoC).
Irish Journal of Psychological Medicine, 35 (2018), pp. 89-94
[Kelly et al., 2021]
J.R. Kelly, C.M. Gillan, J. Prenderville, C. Kelly, A. Harkin, G. Clarke, et al.
Psychedelic therapy's transdiagnostic effects: A research domain criteria (RDoC) perspective.
Frontiers in Psychiatry, 12 (2021),
[Kelly et al., 2015]
J.R. Kelly, P.J. Kennedy, J.F. Cryan, T.G. Dinan, G. Clarke, N.P. Hyland.
Breaking down the barriers: the gut microbiome, intestinal permeability and stress-related psychiatric disorders.
Frontiers in Cellular Neuroscience, 9 (2015), pp. 392
[Kelly et al., 2021]
J.R. Kelly, C. Minuto, J.F. Cryan, G. Clarke, T.G. Dinan.
The role of the gut microbiome in the development of schizophrenia.
Schizophrenia Research, 234 (2021), pp. 4-23
[Kettner et al., 2019]
H. Kettner, S. Gandy, E.C.H.M. Haijen, R.L. Carhart-Harris.
From egoism to ecoism: Psychedelics increase nature relatedness in a state-mediated and context-dependent manner.
International Journal of Environmental Research and Public Health, 16 (2019), pp. 5147
[Kettner et al., 2021]
H. Kettner, F.E. Rosas, C. Timmermann, L. Kärtner, R.L. Charhart-Harris, L. Roseman.
Psychedelic communitas: intersubjective experience during psychedelic group sessions predicts enduring changes in psychological wellbeing and social connectedness. [Original research].
Frontiers in Pharmacology, 12 (2021),
[Khambadkone et al., 2020]
S.G. Khambadkone, Z.A. Cordner, F. Dickerson, E.G. Severance, E. Prandovszky, M. Pletnikov, et al.
Nitrated meat products are associated with mania in humans and altered behavior and brain gene expression in rats.
Molecular Psychiatry, 25 (2020), pp. 560-571
[Kiers et al., 2011]
E.T. Kiers, M. Duhamel, Y. Beesetty, J.A. Mensah, O. Franken, E. Verbruggen, et al.
Reciprocal rewards stabilize cooperation in the mycorrhizal symbiosis.
Science, 333 (2011), pp. 880-882
[Kiraly et al., 2016]
D.D. Kiraly, D.M. Walker, E.S. Calipari, B. Labonte, O. Issler, C.J. Pena, et al.
Alterations of the host microbiome affect behavioral responses to cocaine.
Scientific Reports, 6 (2016), pp. 35455
[Knox et al., 2022]
E.G. Knox, M.R. Aburto, G. Clarke, J.F. Cryan, C.M O'Driscoll.
The blood-brain barrier in aging and neurodegeneration.
Molecular Psychiatry, 27 (2022), pp. 2659-2673
[Kometer et al., 2012]
M. Kometer, A. Schmidt, R. Bachmann, E. Studerus, E. Seifritz, F.X. Vollenweider.
Psilocybin biases facial recognition, goal-directed behavior, and mood state toward positive relative to negative emotions through different serotonergic subreceptors.
Biological Psychiatry, 72 (2012), pp. 898-906
[Kopra et al., 2022]
E.I. Kopra, J.A. Ferris, A.R. Winstock, A.H. Young, J.J. Rucker.
Adverse experiences resulting in emergency medical treatment seeking following the use of magic mushrooms.
Journal of Psychopharmacology, 36 (2022), pp. 965-973
[Kosentka et al., 2013]
P. Kosentka, S.L. Sprague, M. Ryberg, J. Gartz, A.L. May, S.R. Campagna, et al.
Evolution of the toxins muscarine and psilocybin in a family of mushroom-forming fungi.
[Kozłowska et al., 2021]
Kozłowska, U., Klimczak, A., Wiatr, K., & Figiel, M. (2021). The DMT and psilocin treatment changes CD11b+ activated microglia immunological phenotype. bioRxiv, 2021.2003.2007.434103.
[Kraehenmann et al., 2015]
R. Kraehenmann, K.H. Preller, M. Scheidegger, T. Pokorny, O.G. Bosch, E. Seifritz, et al.
Psilocybin-induced decrease in amygdala reactivity correlates with enhanced positive mood in healthy volunteers.
Biological Psychiatry, 78 (2015), pp. 572-581
[Krebs and Johansen, 2012]
T.S. Krebs, P.O. Johansen.
Lysergic acid diethylamide (LSD) for alcoholism: Meta-analysis of randomized controlled trials.
Journal of Psychopharmacology, 26 (2012), pp. 994-1002
[Krystal et al., 2020]
A.D. Krystal, D.A. Pizzagalli, M. Smoski, S.J. Mathew, J. Nurnberger, S.H. Lisanby, et al.
A randomized proof-of-mechanism trial applying the ‘fast-fail’ approach to evaluating κ-opioid antagonism as a treatment for anhedonia.
Nature Medicine, 26 (2020), pp. 760-768
[Kuypers, 2019]
K.P.C. Kuypers.
Psychedelic medicine: The biology underlying the persisting psychedelic effects.
Medical Hypotheses, 125 (2019), pp. 21-24
[Lane, 2015]
N. Lane.
The unseen world: Reflections on Leeuwenhoek (1677) 'concerning little animals'.
Philosophical Transactions of the Royal Society of London. Series B: Biological Sciences, 370 (2015),
[Lankelma et al., 2015]
J.M. Lankelma, M. Nieuwdorp, W.M. de Vos, W.J. Wiersinga.
The gut microbiota in internal medicine: Implications for health and disease.
Netherlands Journal of Medicine, 73 (2015), pp. 61-68
[Lass-Flörl et al., 2003]
C. Lass-Flörl, M. Ledochowski, D. Fuchs, C. Speth, L. Kacani, M.P. Dierich, et al.
Interaction of sertraline with Candida species selectively attenuates fungal virulence in vitro.
Fems Immunology and Medical Microbiology, 35 (2003), pp. 11-15
[Lavrinienko et al., 2021]
A. Lavrinienko, T. Scholier, S.T. Bates, A.N. Miller, P.C. Watts.
Defining gut mycobiota for wild animals: A need for caution in assigning authentic resident fungal taxa.
Animal Microbiome, 3 (2021), pp. 75
[Leclercq et al., 2020]
S. Leclercq, T. Le Roy, S. Furgiuele, V. Coste, L.B. Bindels, Q. Leyrolle, et al.
Gut microbiota-induced changes in β-hydroxybutyrate metabolism are linked to altered sociability and depression in alcohol use disorder.
Cell Reports, 33 (2020),
[Leclercq et al., 2014]
S. Leclercq, S. Matamoros, P.D. Cani, A.M. Neyrinck, F. Jamar, P. Stärkel, et al.
Intestinal permeability, gut-bacterial dysbiosis, and behavioral markers of alcohol-dependence severity.
Proceedings of the National Academy of Sciences, 111 (2014), pp. E4485-E4493
[Leclercq et al., 2021]
S. Leclercq, M. Schwarz, N.M. Delzenne, P. Stärkel, P. de Timary.
Alterations of kynurenine pathway in alcohol use disorder and abstinence: A link with gut microbiota, peripheral inflammation and psychological symptoms.
Translational Psychiatry, 11 (2021), pp. 503
[Lee et al., 2015]
J.H. Lee, T.K. Wood, J. Lee.
Roles of Indole as an Interspecies and interkingdom signaling molecule.
Trends in Microbiology, 23 (2015), pp. 707-718
[Leitão-Gonçalves et al., 2017]
R. Leitão-Gonçalves, Z. Carvalho-Santos, A.P. Francisco, G.T. Fioreze, M. Anjos, C. Baltazar, et al.
Commensal bacteria and essential amino acids control food choice behavior and reproduction.
PLOS Biology, 15 (2017),
[Lent, 2017]
Lent, J.R. (2017). The patterning instinct : A cultural history of humanity's search for meaning.
[Lenz et al., 2021]
C. Lenz, A. Sherwood, R. Kargbo, D. Hoffmeister.
Taking different roads: l-tryptophan as the origin of psilocybe natural products.
ChemPlusChem, 86 (2021), pp. 28-35
[Leonardi et al., 2018]
I. Leonardi, X. Li, A. Semon, D. Li, I. Doron, G. Putzel, et al.
CX3CR1(+) mononuclear phagocytes control immunity to intestinal fungi.
Science, 359 (2018), pp. 232-236
[Leyrolle et al., 2021]
Q. Leyrolle, R. Cserjesi, D.G.H.M. M, G. Zamariola, S. Hiel, M.A. Gianfrancesco, et al.
Prebiotic effect on mood in obese patients is determined by the initial gut microbiota composition: A randomized, controlled trial.
Brain, Behavior, and Immunity, 94 (2021), pp. 289-298
[Lindell et al., 2022]
A.E. Lindell, M. Zimmermann-Kogadeeva, K.R. Patil.
Multimodal interactions of drugs, natural compounds and pollutants with the gut microbiota.
Nature Reviews Microbiology, 20 (2022), pp. 431-443
[Litjens et al., 2014]
R.P. Litjens, T.M. Brunt, G.J. Alderliefste, R.H. Westerink.
Hallucinogen persisting perception disorder and the serotonergic system: A comprehensive review including new MDMA-related clinical cases.
European Neuropsychopharmacology, 24 (2014), pp. 1309-1323
[Liu et al., 2020]
Y. Liu, Y. Wang, Y. Ni, C.K.Y. Cheung, K.S.L. Lam, Y. Wang, et al.
Gut microbiome fermentation determines the efficacy of exercise for diabetes prevention.
Cell Metabolism, 31 (2020), pp. 77-91
[Loron et al., 2019]
C.C. Loron, C. François, R.H. Rainbird, E.C. Turner, S. Borensztajn, E.J. Javaux.
Early fungi from the Proterozoic era in Arctic Canada.
Nature, 570 (2019), pp. 232-235
[Lozupone et al., 2012]
C.A. Lozupone, J.I. Stombaugh, J.I. Gordon, J.K. Jansson, R. Knight.
Diversity, stability and resilience of the human gut microbiota.
Nature, 489 (2012), pp. 220-230
[Luczynski et al., 2016]
P. Luczynski, S.O. Whelan, C. O'Sullivan, G. Clarke, F. Shanahan, T.G. Dinan, et al.
Adult microbiota-deficient mice have distinct dendritic morphological changes: Differential effects in the amygdala and hippocampus.
European Journal of Neuroscience, 44 (2016), pp. 2654-2666
[Lukić et al., 2019]
I. Lukić, D. Getselter, O. Ziv, O. Oron, E. Reuveni, O. Koren, et al.
Antidepressants affect gut microbiota and ruminococcus flavefaciens is able to abolish their effects on depressive-like behavior.
Translational Psychiatry, 9 (2019), pp. 133
[Lyons and Carhart-Harris, 2018]
T. Lyons, R.L. Carhart-Harris.
Increased nature relatedness and decreased authoritarian political views after psilocybin for treatment-resistant depression.
Journal of Psychopharmacology, 32 (2018), pp. 811-819
[Lyte et al., 2019]
M. Lyte, K.M. Daniels, S. Schmitz-Esser.
Fluoxetine-induced alteration of murine gut microbial community structure: Evidence for a microbial endocrinology-based mechanism of action responsible for fluoxetine-induced side effects.
PeerJ, 7 (2019), pp. e6199
[Maini Rekdal et al., 2019]
V. Maini Rekdal, E.N. Bess, J.E. Bisanz, P.J. Turnbaugh, E.P. Balskus.
Discovery and inhibition of an interspecies gut bacterial pathway for Levodopa metabolism.
Science, 364 (2019),
[Manevski et al., 2010]
N. Manevski, M. Kurkela, C. Höglund, T. Mauriala, M.H. Court, J. Yli-Kauhaluoma, et al.
Glucuronidation of psilocin and 4-hydroxyindole by the human UDP-glucuronosyltransferases.
Drug Metabolism and Disposition: The Biological Fate of Chemicals, 38 (2010), pp. 386-395
[Mangold et al., 2019]
C.A. Mangold, M.J. Ishler, R.G. Loreto, M.L. Hazen, D.P. Hughes.
Zombie ant death grip due to hypercontracted mandibular muscles.
The Journal of Experimental Biology, 222 (2019),
[Marx et al., 2021]
W. Marx, M. Lane, M. Hockey, H. Aslam, M. Berk, K. Walder, et al.
Diet and depression: Exploring the biological mechanisms of action.
Molecular Psychiatry, 26 (2021), pp. 134-150
[Mawe and Hoffman, 2013]
G.M. Mawe, J.M. Hoffman.
Serotonin signalling in the gut–functions, dysfunctions and therapeutic targets.
Nature Reviews. Gastroenterology & Hepatology, 10 (2013), pp. 473-486
[McDaid et al., 2019]
D. McDaid, A.L. Park, K. Wahlbeck.
The economic case for the prevention of mental illness.
Annual Review of Public Health, 40 (2019), pp. 373-389
[McEwen, 2017]
B.S. McEwen.
Allostasis and the epigenetics of brain and body health over the life course: The brain on stress.
JAMA Psychiatry, 74 (2017), pp. 551-552
[McGuinness et al., 2022]
A.J. McGuinness, J.A. Davis, S.L. Dawson, A. Loughman, F. Collier, M. O'Hely, et al.
A systematic review of gut microbiota composition in observational studies of major depressive disorder, bipolar disorder and schizophrenia.
Molecular Psychiatry, 27 (2022), pp. 1920-1935
[Meckel and Kiraly, 2019]
K.R. Meckel, D.D. Kiraly.
A potential role for the gut microbiome in substance use disorders.
Psychopharmacology, 236 (2019), pp. 1513-1530
[Medeiros et al., 2020]
G.C. Medeiros, A.J. Rush, M. Jha, T. Carmody, J.L. Furman, A.H. Czysz, et al.
Positive and negative valence systems in major depression have distinct clinical features, response to antidepressants, and relationships with immunomarkers.
Depression and Anxiety, 37 (2020), pp. 771-783
[Medina-Rodriguez et al., 2020]
E.M. Medina-Rodriguez, D. Madorma, G. O'Connor, B.L. Mason, D. Han, S.K. Deo, et al.
Identification of a signaling mechanism by which the microbiome regulates Th17 cell-mediated depressive-like behaviors in mice.
American Journal of Psychiatry, 177 (2020), pp. 974-990
[MetaCyc 2022]
MetaCyc. (2022). MetaCyc pathway: L-tryptophan degradation VI (via tryptamine). from https://biocyc.org/META/new-image?object=PWY-3181MetaCyc
[Miller et al., 2019]
M.J. Miller, J. Albarracin-Jordan, C. Moore, J.M. Capriles.
Chemical evidence for the use of multiple psychotropic plants in a 1000-year-old ritual bundle from South America.
Proceedings of the National Academy of Sciences, 116 (2019), pp. 11207-11212
[Mims et al., 2021]
T.S. Mims, Q.A. Abdallah, J.D. Stewart, S.P. Watts, C.T. White, T.V. Rousselle, et al.
The gut mycobiome of healthy mice is shaped by the environment and correlates with metabolic outcomes in response to diet.
Communications Biology, 4 (2021), pp. 281
[Moeller et al., 2014]
A.H. Moeller, Y. Li, E. Mpoudi Ngole, S. Ahuka-Mundeke, E.V. Lonsdorf, A.E. Pusey, et al.
Rapid changes in the gut microbiome during human evolution.
Proceedings of the National Academy of Sciences of the United States of America, 111 (2014), pp. 16431-16435
[Mörkl et al., 2022]
S. Mörkl, A. Oberascher, J.M. Tatschl, S. Lackner, T.F.S. Bastiaanssen, M.I. Butler, et al.
Cardiac vagal activity is associated with gut-microbiome patterns in women-an exploratory pilot study.
Dialogues in Clinical Neuroscience, 24 (2022), pp. 1-9
[Murray et al., 2014]
S. Murray, A. Tulloch, M.S. Gold, N.M. Avena.
Hormonal and neural mechanisms of food reward, eating behaviour and obesity.
Nature Reviews Endocrinology, 10 (2014), pp. 540-552
[Nagpal and Cryan, 2021]
J. Nagpal, J.F. Cryan.
Microbiota-brain interactions: Moving toward mechanisms in model organisms.
Neuron, 109 (2021), pp. 3930-3953
[Nardou et al., 2019]
R. Nardou, E.M. Lewis, R. Rothhaas, R. Xu, A. Yang, E. Boyden, et al.
Oxytocin-dependent reopening of a social reward learning critical period with MDMA.
Nature, 569 (2019), pp. 116-120
[Nash et al., 2017]
A.K. Nash, T.A. Auchtung, M.C. Wong, D.P. Smith, J.R. Gesell, M.C. Ross, et al.
The gut mycobiome of the human microbiome project healthy cohort.
[Nau et al., 2013]
F. Nau Jr., B. Yu, D. Martin, C.D Nichols.
Serotonin 5-HT2A receptor activation blocks TNF-α mediated inflammation in vivo.
[Nguyen et al., 2018]
T.T. Nguyen, T. Kosciolek, L.T. Eyler, R. Knight, D.V. Jeste.
Overview and systematic review of studies of microbiome in schizophrenia and bipolar disorder.
Journal of Psychiatric Research, 99 (2018), pp. 50-61
[Nichols, 2016]
D.E. Nichols.
Psychedelics.
Pharmacological Reviews, 68 (2016), pp. 264-355
[Nikolova et al., 2021]
V.L. Nikolova, M.R.B. Smith, L.J. Hall, A.J. Cleare, J.M. Stone, A.H. Young.
Perturbations in gut microbiota composition in psychiatric disorders: A review and meta-analysis.
JAMA Psychiatry, 78 (2021), pp. 1343-1354
[Nkadimeng et al., 2021]
S.M. Nkadimeng, C.M.L. Steinmann, J.N. Eloff.
Anti-inflammatory effects of four psilocybin-containing magic mushroom water extracts in vitro on 15-lipoxygenase activity and on lipopolysaccharide-induced cyclooxygenase-2 and inflammatory cytokines in human U937 macrophage cells.
Journal of Inflammation Research, 14 (2021), pp. 3729-3738
[Noorani et al., 2018]
T. Noorani, A. Garcia-Romeu, T.C. Swift, R.R. Griffiths, M.W. Johnson.
Psychedelic therapy for smoking cessation: Qualitative analysis of participant accounts.
Journal of Psychopharmacology, 32 (2018), pp. 756-769
[Nutman et al., 2016]
A.P. Nutman, V.C. Bennett, C.R.L. Friend, M.J. Van Kranendonk, A.R. Chivas.
Rapid emergence of life shown by discovery of 3700-million-year-old microbial structures.
Nature, 537 (2016), pp. 535-538
[Nutt et al., 2020]
D. Nutt, D. Erritzoe, R. Carhart-Harris.
Psychedelic psychiatry's brave new world.
[Oddy et al., 2018]
W.H. Oddy, K.L. Allen, G.S.A. Trapp, G.L. Ambrosini, L.J. Black, R.C. Huang, et al.
Dietary patterns, body mass index and inflammation: Pathways to depression and mental health problems in adolescents.
Brain, Behavior, and Immunity, 69 (2018), pp. 428-439
[Olbrich et al., 2021]
S. Olbrich, K.H. Preller, F.X. Vollenweider.
LSD and ketanserin and their impact on the human autonomic nervous system.
Psychophysiology, 58 (2021), pp. e13822
[Olm et al., 2022]
M.R. Olm, D. Dahan, M.M. Carter, B.D. Merrill, F.B. Yu, S. Jain, et al.
Robust variation in infant gut microbiome assembly across a spectrum of lifestyles.
Science, 376 (2022), pp. 1220-1223
[O'Mahony et al., 2015]
S.M. O'Mahony, G. Clarke, Y.E. Borre, T.G. Dinan, J.F. Cryan.
Serotonin, tryptophan metabolism and the brain-gut-microbiome axis.
Behavioural Brain Research, 277 (2015), pp. 32-48
[O'Riordan et al., 2022]
K.J. O'Riordan, M.K. Collins, G.M. Moloney, E.G. Knox, M.R. Aburto, C. Fülling, et al.
Short chain fatty acids: Microbial metabolites for gut-brain axis signalling.
Molecular and Cellular Endocrinology, 546 (2022),
[O'Sullivan et al., 2015]
O. O'Sullivan, O. Cronin, S.F. Clarke, E.F. Murphy, M.G. Molloy, F. Shanahan, et al.
Exercise and the microbiota.
Gut Microbes, 6 (2015), pp. 131-136
[Palhano-Fontes et al., 2019]
F. Palhano-Fontes, D. Barreto, H. Onias, K.C. Andrade, M.M. Novaes, J.A. Pessoa, et al.
Rapid antidepressant effects of the psychedelic ayahuasca in treatment-resistant depression: A randomized placebo-controlled trial.
Psychological Medicine, 49 (2019), pp. 655-663
[Palit and Ali, 2008]
P. Palit, N. Ali.
Oral therapy with sertraline, a selective serotonin reuptake inhibitor, shows activity against leishmania donovani.
Journal of Antimicrobial Chemotherapy, 61 (2008), pp. 1120-1124
[Pape et al., 2019]
K. Pape, R. Tamouza, M. Leboyer, F. Zipp.
Immunoneuropsychiatry - novel perspectives on brain disorders.
Nature Reviews Neurology, 15 (2019), pp. 317-328
[Paul et al., 2016]
S. Paul, R.B. Mortimer, M. Mitchell.
Sertraline demonstrates fungicidal activity in vitro for coccidioides immitis.
[Pearson-Leary et al., 2020]
J. Pearson-Leary, C. Zhao, K. Bittinger, D. Eacret, S. Luz, A.S. Vigderman, et al.
The gut microbiome regulates the increases in depressive-type behaviors and in inflammatory processes in the ventral hippocampus of stress vulnerable rats.
Molecular Psychiatry, 25 (2020), pp. 1068-1079
[Peterson et al., 2017]
V.L. Peterson, N.J. Jury, R. Cabrera-Rubio, L.A. Draper, F. Crispie, P.D. Cotter, et al.
Drunk bugs: Chronic vapour alcohol exposure induces marked changes in the gut microbiome in mice.
Behavioural Brain Research, 323 (2017), pp. 172-176
[Pinto-Sanchez et al., 2017]
M.I. Pinto-Sanchez, G.B. Hall, K. Ghajar, A. Nardelli, C. Bolino, J.T. Lau, et al.
Probiotic bifidobacterium longum NCC3001 reduces depression scores and alters brain activity: A pilot study in patients with irritable bowel syndrome.
Gastroenterology, 153 (2017), pp. 448-459
[Pyndt Jorgensen et al., 2015]
B. Pyndt Jorgensen, L. Krych, T.B. Pedersen, N. Plath, J.P. Redrobe, A.K. Hansen, et al.
Investigating the long-term effect of subchronic phencyclidine-treatment on novel object recognition and the association between the gut microbiota and behavior in the animal model of schizophrenia.
Physiology & Behavior, 141 (2015), pp. 32-39
[Ramsteijn et al., 2020]
A.S. Ramsteijn, E. Jašarević, D.J. Houwing, T.L. Bale, J.D.A. Olivier.
Antidepressant treatment with fluoxetine during pregnancy and lactation modulates the gut microbiome and metabolome in a rat model relevant to depression.
Gut Microbes, 11 (2020), pp. 735-753
[Reynolds et al., 2019]
A. Reynolds, J. Mann, J. Cummings, N. Winter, E. Mete, L. Te Morenga.
Carbohydrate quality and human health: a series of systematic reviews and meta-analyses.
The Lancet, 393 (2019), pp. 434-445
[Reynolds et al., 2018]
H.T. Reynolds, V. Vijayakumar, E. Gluck-Thaler, H.B. Korotkin, P.B. Matheny, J.C. Slot.
Horizontal gene cluster transfer increased hallucinogenic mushroom diversity.
Evolution Letters, 2 (2018), pp. 88-101
[Ridge et al., 2019]
E.A. Ridge, S. Pachhain, S.R. Choudhury, S.R. Bodnar, R.A. Larsen, V. Phuntumart, et al.
The influence of the host microbiome on 3,4-methylenedioxymethamphetamine (MDMA)-induced hyperthermia and vice versa.
Scientific Reports, 9 (2019), pp. 4313
[Robinson et al., 2020]
D.W. Robinson, K. Brown, M. McMenemy, L. Dennany, M.J. Baker, P. Allan, et al.
Datura quids at Pinwheel Cave, California, provide unambiguous confirmation of the ingestion of hallucinogens at a rock art site.
PNAS, 117 (2020), pp. 31026-31037
[Rocha et al., 2021]
J.M. Rocha, G.N. Rossi, F. de Lima Osório, J.C. Bouso, G. de Oliveira Silveira, M. Yonamine, et al.
Effects of ayahuasca on the recognition of facial expressions of emotions in naive healthy volunteers: A pilot, proof-of-concept, randomized controlled trial.
Journal of Clinical Psychopharmacology, 41 (2021), pp. 267-274
[Rodríguez Arce and Winkelman, 2021]
J.M. Rodríguez Arce, M.J. Winkelman.
Psychedelics, sociality, and human evolution.
Frontiers in Psychology, 12 (2021),
[Rossato et al., 2016]
L. Rossato, É, S. Loreto, R.A. Zanette, F. Chassot, J.M. Santurio, S.H Alves.
In vitro synergistic effects of chlorpromazine and sertraline in combination with amphotericin B against Cryptococcus neoformans var. grubii.
Folia Microbiologica, 61 (2016), pp. 399-403
[Rossi and Stuber, 2018]
M.A. Rossi, G.D. Stuber.
Overlapping brain circuits for homeostatic and hedonic feeding.
Cell Metabolism, 27 (2018), pp. 42-56
[Rucker et al., 2022]
J.J. Rucker, L. Marwood, R.L.J. Ajantaival, C. Bird, H. Eriksson, J. Harrison, et al.
The effects of psilocybin on cognitive and emotional functions in healthy participants: Results from a phase 1, randomised, placebo-controlled trial involving simultaneous psilocybin administration and preparation.
Journal of Psychopharmacology, 36 (2022), pp. 114-125
[Salavrakos et al., 2021]
M. Salavrakos, S. Leclercq, P. De Timary, G. Dom.
Microbiome and substances of abuse.
Progress in Neuro-Psychopharmacology and Biological Psychiatry, 105 (2021),
[Sanna et al., 2019]
S. Sanna, N.R. van Zuydam, A. Mahajan, A. Kurilshikov, A. Vich Vila, U. Võsa, et al.
Causal relationships among the gut microbiome, short-chain fatty acids and metabolic diseases.
Nature Genetics, 51 (2019), pp. 600-605
[Saunders et al., 2020]
J.M. Saunders, J.L. Moreno, D. Ibi, M. Sikaroodi, D.J. Kang, R. Muñoz-Moreno, et al.
Gut microbiota manipulation during the prepubertal period shapes behavioral abnormalities in a mouse neurodevelopmental disorder model.
Scientific Reports, 10 (2020), pp. 4697
[Savignac et al., 2016]
H.M. Savignac, Y. Couch, M. Stratford, D.M. Bannerman, G. Tzortzis, D.C. Anthony, et al.
Prebiotic administration normalizes lipopolysaccharide (LPS)-induced anxiety and cortical 5-HT2A receptor and IL1-β levels in male mice.
Brain, Behavior, and Immunity, 52 (2016), pp. 120-131
[Savitz, 2020]
J. Savitz.
The kynurenine pathway: A finger in every pie.
Molecular Psychiatry, 25 (2020), pp. 131-147
[Schifano et al., 2021]
F. Schifano, S. Chiappini, A. Miuli, J.M. Corkery, N. Scherbaum, F. Napoletano, et al.
New psychoactive substances (NPS) and serotonin syndrome onset: A systematic review.
Experimental Neurology, 339 (2021),
[Schindler et al., 2018]
E.A.D. Schindler, R.M. Wallace, J.A. Sloshower, D.C D'Souza.
Neuroendocrine associations underlying the persistent therapeutic effects of classic serotonergic psychedelics.
Frontiers in Pharmacology, 9 (2018), pp. 177
[Schirmer et al., 2016]
M. Schirmer, S.P. Smeekens, H. Vlamakis, M. Jaeger, M. Oosting, E.A. Franzosa, et al.
Linking the Human gut microbiome to inflammatory cytokine production capacity.
Cell, 167 (2016), pp. 1125-1136
[Schlag et al., 2022]
A.K. Schlag, J. Aday, I. Salam, J.C. Neill, D.J. Nutt.
Adverse effects of psychedelics: From anecdotes and misinformation to systematic science.
Journal of Psychopharmacology (Oxford, England), 36 (2022), pp. 258-272
[Schmid et al., 2015]
Y. Schmid, F. Enzler, P. Gasser, E. Grouzmann, K.H. Preller, F.X. Vollenweider, et al.
Acute effects of lysergic acid diethylamide in healthy subjects.
Biological Psychiatry, 78 (2015), pp. 544-553
[Schüssler-Fiorenza Rose et al., 2019]
S.M. Schüssler-Fiorenza Rose, K. Contrepois, K.J. Moneghetti, W. Zhou, T. Mishra, S. Mataraso, et al.
A longitudinal big data approach for precision health.
Nature Medicine, 25 (2019), pp. 792-804
[Sender and Fuchs, 2016]
Sender, R., & Fuchs, S. (2016). Revised estimates for the number of human and bacteria cells in the body. 14(8), e1002533.
[Sgritta et al., 2019]
M. Sgritta, S.W. Dooling, S.A. Buffington, E.N. Momin, M.B. Francis, R.A. Britton, et al.
Mechanisms underlying microbial-mediated changes in social behavior in mouse models of autism spectrum disorder.
[Sheldrake, 2020]
M. Sheldrake.
Entangled life.
Bodley Head, (2020),
[Sherwin et al., 2019]
E. Sherwin, S.R. Bordenstein, J.L. Quinn, T.G. Dinan, J.F. Cryan.
Microbiota and the social brain.
Science, 366 (2019),
[Sherwood et al., 2020]
A.M. Sherwood, A.L. Halberstadt, A.K. Klein, J.D. McCorvy, K.W. Kaylo, R.B. Kargbo, et al.
Synthesis and biological evaluation of tryptamines found in hallucinogenic mushrooms: Norbaeocystin, baeocystin, norpsilocin, and aeruginascin.
Journal of Natural Products, 83 (2020), pp. 461-467
[Simpson et al., 2020]
S. Simpson, A. Kimbrough, B. Boomhower, R. McLellan, M. Hughes, K. Shankar, et al.
Depletion of the microbiome alters the recruitment of neuronal ensembles of oxycodone intoxication and withdrawal.
Eneuro, 7 (2020),
[Smits et al., 2017]
S.A. Smits, J. Leach, E.D. Sonnenburg, C.G. Gonzalez, J.S. Lichtman, G. Reid, et al.
Seasonal cycling in the gut microbiome of the Hadza hunter-gatherers of Tanzania.
Science (New York, N.Y.), 357 (2017), pp. 802-806
[Sonnenburg and Sonnenburg, 2019]
E.D. Sonnenburg, J.L. Sonnenburg.
The ancestral and industrialized gut microbiota and implications for human health.
Nature Reviews Microbiology, 17 (2019), pp. 383-390
[Spriggs et al., 2021a]
M.J. Spriggs, H.M. Douglass, R.J. Park, T. Read, J.L. Danby, F.J.C. de Magalhães, et al.
Study protocol for “psilocybin as a treatment for anorexia nervosa: a pilot study”. [Study Protocol].
Front Psychiatry, 12 (2021),
[Spriggs et al., 2021b]
M.J. Spriggs, H. Kettner, R.L. Carhart-Harris.
Positive effects of psychedelics on depression and wellbeing scores in individuals reporting an eating disorder.
Eating and Weight Disorders, 26 (2021), pp. 1265-1270
[Stilling et al., 2018]
R.M. Stilling, G.M. Moloney, F.J. Ryan, A.E. Hoban, T.F. Bastiaanssen, F. Shanahan, et al.
Social interaction-induced activation of RNA splicing in the amygdala of microbiome-deficient mice.
Elife, 7 (2018),
[Strajhar et al., 2016]
P. Strajhar, Y. Schmid, E. Liakoni, P.C. Dolder, K.M. Rentsch, D.V. Kratschmar, et al.
Acute effects of lysergic acid diethylamide on circulating steroid levels in healthy subjects.
Journal of Neuroendocrinology, 28 (2016), pp. 12374
[Strassman and Qualls, 1994]
R.J. Strassman, C.R. Qualls.
Dose-response study of N,N-dimethyltryptamine in humans. I. Neuroendocrine, autonomic, and cardiovascular effects.
Archives of General Psychiatry, 51 (1994), pp. 85-97
[Strassman et al., 1996]
R.J. Strassman, C.R. Qualls, L.M. Berg.
Differential tolerance to biological and subjective effects of four closely spaced doses of N,N-dimethyltryptamine in humans.
Biological Psychiatry, 39 (1996), pp. 784-795
[Sudo et al., 2004]
N. Sudo, Y. Chida, Y. Aiba, J. Sonoda, N. Oyama, X.N. Yu, et al.
Postnatal microbial colonization programs the hypothalamic-pituitary-adrenal system for stress response in mice.
Journal of Physiology, 558 (2004), pp. 263-275
[Suess et al., 2021]
Suess, G.J., Kasiah, J., Simpson, S., Brennan, M., Conlisk, D., Maturin, L., et al. (2021). The gut microbiome is associated with cocaine behavior and predicts addiction vulnerability in adult male rats. bioRxiv., 2021.2007.2020.453110.
[Suhr and Hallen-Adams, 2015]
M.J. Suhr, H.E. Hallen-Adams.
The human gut mycobiome: Pitfalls and potentials–a mycologist's perspective.
Mycologia, 107 (2015), pp. 1057-1073
[Sun et al., 2019]
L. Sun, H. Zhang, Y. Cao, C. Wang, C. Zhao, H. Wang, et al.
Fluoxetine ameliorates dysbiosis in a depression model induced by chronic unpredicted mild stress in mice.
International Journal of Medical Sciences, 16 (2019), pp. 1260-1270
[Sun et al., 2021]
Y. Sun, T. Zuo, C.P. Cheung, W. Gu, Y. Wan, F. Zhang, et al.
Population-level configurations of gut mycobiome across 6 ethnicities in urban and rural China.
Gastroenterology, 160 (2021), pp. 272-286
[Szabo, 2015]
A. Szabo.
Psychedelics and immunomodulation: novel approaches and therapeutic opportunities.
Frontiers in Immunology, 6 (2015), pp. 358
[Szabo et al., 2014]
A. Szabo, A. Kovacs, E. Frecska, E. Rajnavolgyi.
Psychedelic N,N-dimethyltryptamine and 5-methoxy-N,N-dimethyltryptamine modulate innate and adaptive inflammatory responses through the sigma-1 receptor of human monocyte-derived dendritic cells.
Plos One, 9 (2014),
[Szabo et al., 2016]
A. Szabo, A. Kovacs, J. Riba, S. Djurovic, E. Rajnavolgyi, E. Frecska.
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.
Frontiers in Neuroscience, 10 (2016), pp. 423
[Teixeira et al., 2022]
P.J. Teixeira, M.W. Johnson, C. Timmermann, R. Watts, D. Erritzoe, H. Douglass, et al.
Psychedelics and health behaviour change.
Journal of Psychopharmacology, 36 (2022), pp. 12-19
[Theis et al., 2016]
K.R. Theis, N.M. Dheilly, J.L. Klassen, R.M. Brucker, J.F. Baines, T.C.G. Bosch, et al.
Getting the hologenome concept right: An eco-evolutionary framework for hosts and their microbiomes.
mSystems, 1 (2016), pp. e00028
[Thompson and Szabo, 2020]
C. Thompson, A. Szabo.
Psychedelics as a novel approach to treating autoimmune conditions.
Immunology Letters, 228 (2020), pp. 45-54
[Tian et al., 2019]
P. Tian, G. Wang, J. Zhao, H. Zhang, W. Chen.
Bifidobacterium with the role of 5-hydroxytryptophan synthesis regulation alleviates the symptom of depression and related microbiota dysbiosis.
The Journal of Nutritional Biochemistry, 66 (2019), pp. 43-51
[Topol, 2014]
E.J. Topol.
Individualized medicine from prewomb to tomb.
[Torres et al., 2018]
G. Torres, C.L. Hoehmann, J.A. Cuoco, K. Hitscherich, C. Pavia, M. Hadjiargyrou, et al.
Ketamine intervention limits pathogen expansion in vitro.
Pathogens and Disease, 76 (2018),
[Trevelline and Kohl, 2022]
B.K. Trevelline, K.D. Kohl.
The gut microbiome influences host diet selection behavior.
Proceedings of the National Academy of Sciences, 119 (2022),
[Treviño-Rangel Rde et al., 2016]
J. Treviño-Rangel Rde, H. Villanueva-Lozano, P. Hernández-Rodríguez, M.F. Martínez-Reséndez, J. García-Juárez, H. Rodríguez-Rocha, et al.
Activity of sertraline against Cryptococcus neoformans: In vitro and in vivo assays.
Medical Mycology, 54 (2016), pp. 280-286
[Urrutia-Piñones et al., 2018]
J. Urrutia-Piñones, J. Illanes-González, A. López-Aguilera, M. Julio-Pieper, J.A. Bravo.
Do obese bacteria make us “Want them”? Intestinal microbiota, mesocorticolimbic circuit and non-homeostatic feeding.
Current Behavioral Neuroscience Reports, 5 (2018), pp. 211-217
[Uthaug et al., 2020]
M.V. Uthaug, R. Lancelotta, A. Szabo, A.K. Davis, J. Riba, J.G. Ramaekers.
Prospective examination of synthetic 5-methoxy-N,N-dimethyltryptamine inhalation: effects on salivary IL-6, cortisol levels, affect, and non-judgment.
Psychopharmacology, 237 (2020), pp. 773-785
[Valles-Colomer et al., 2019]
M. Valles-Colomer, G. Falony, Y. Darzi, E.F. Tigchelaar, J. Wang, R.Y. Tito, et al.
The neuroactive potential of the human gut microbiota in quality of life and depression.
Nature Microbiology, 4 (2019), pp. 623-632
[van Tilburg Bernardes et al., 2020]
E. van Tilburg Bernardes, V.K. Pettersen, M.W. Gutierrez, I. Laforest-Lapointe, N.G. Jendzjowsky, J.B. Cavin, et al.
Intestinal fungi are causally implicated in microbiome assembly and immune development in mice.
Nature Communications, 11 (2020), pp. 2577
[Vangay et al., 2018]
P. Vangay, A.J. Johnson, T.L. Ward, G.A. Al-Ghalith, R.R. Shields-Cutler, B.M. Hillmann, et al.
US immigration westernizes the human gut microbiome.
[Verdonk et al., 2019]
F. Verdonk, A.C. Petit, P. Abdel-Ahad, F. Vinckier, G. Jouvion, P. de Maricourt, et al.
Microglial production of quinolinic acid as a target and a biomarker of the antidepressant effect of ketamine.
Brain, Behavior, and Immunity, 81 (2019), pp. 361-373
[Vich Vila et al., 2020]
A. Vich Vila, V. Collij, S. Sanna, T. Sinha, F. Imhann, A.R. Bourgonje, et al.
Impact of commonly used drugs on the composition and metabolic function of the gut microbiota.
Nature Communications, 11 (2020), pp. 362
[Wastyk et al., 2021]
H.C. Wastyk, G.K. Fragiadakis, D. Perelman, D. Dahan, B.D. Merrill, F.B. Yu, et al.
Gut-microbiota-targeted diets modulate human immune status.
Cell, 184 (2021), pp. 4137-4153
[Watts et al., 2021]
N. Watts, M. Amann, N. Arnell, S. Ayeb-Karlsson, J. Beagley, K. Belesova, et al.
The 2020 report of the lancet countdown on health and climate change: Responding to converging crises.
The Lancet, 397 (2021), pp. 129-170
[Watts et al., 2017]
R. Watts, C. Day, J. Krzanowski, D. Nutt, R. Carhart-Harris.
Patients’ accounts of increased “connectedness” and “acceptance” after psilocybin for treatment-resistant depression.
Journal of Humanistic Psychology, 57 (2017), pp. 520-564
[WHO 2021]
WHO. (2021). Comprehensive mental health action plan 2013-2030.
[Williams et al., 2014]
B.B. Williams, A.H. Van Benschoten, P. Cimermancic, M.S. Donia, M. Zimmermann, M. Taketani, et al.
Discovery and characterization of gut microbiota decarboxylases that can produce the neurotransmitter tryptamine.
Cell Host Microbe, 16 (2014), pp. 495-503
[Wilmanski et al., 2019]
T. Wilmanski, N. Rappaport, J.C. Earls, A.T. Magis, O. Manor, J. Lovejoy, et al.
Blood metabolome predicts gut microbiome α-diversity in humans.
Nature Biotechnology, 37 (2019), pp. 1217-1228
[Wu et al., 2019]
B. Wu, M. Hussain, W. Zhang, M. Stadler, X. Liu, M. Xiang.
Current insights into fungal species diversity and perspective on naming the environmental DNA sequences of fungi.
Mycology, 10 (2019), pp. 127-140
[Wu et al., 2021]
W.L. Wu, M.D. Adame, C.W. Liou, J.T. Barlow, T.T. Lai, G. Sharon, et al.
Microbiota regulate social behaviour via stress response neurons in the brain.
Nature, 595 (2021), pp. 409-414
[Yang et al., 2019]
C. Yang, K.J. Wardenaar, F.J. Bosker, J. Li, R.A. Schoevers.
Inflammatory markers and treatment outcome in treatment resistant depression: A systematic review.
Journal of Affective Disorders, 257 (2019), pp. 640-649
[Yano et al., 2015]
J.M. Yano, K. Yu, G.P. Donaldson, G.G. Shastri, P. Ann, L. Ma, et al.
Indigenous bacteria from the gut microbiota regulate host serotonin biosynthesis.
[Ye et al., 2021]
L. Ye, M. Bae, C.D. Cassilly, S.V. Jabba, D.W. Thorpe, A.M. Martin, et al.
Enteroendocrine cells sense bacterial tryptophan catabolites to activate enteric and vagal neuronal pathways.
Cell Host Microbe, 29 (2021), pp. 179-196
[Yu et al., 2008]
B. Yu, J. Becnel, M. Zerfaoui, R. Rohatgi, A.H. Boulares, C.D. Nichols.
Serotonin 5-hydroxytryptamine(2A) receptor activation suppresses tumor necrosis factor-alpha-induced inflammation with extraordinary potency.
Journal of Pharmacology and Experimental Therapeutics, 327 (2008), pp. 316-323
[Zeevi et al., 2015]
D. Zeevi, T. Korem, N. Zmora, D. Israeli, D. Rothschild, A. Weinberger, et al.
Personalized nutrition by prediction of glycemic responses.
Cell, 163 (2015), pp. 1079-1094
[Zhai et al., 2012]
B. Zhai, C. Wu, L. Wang, M.S. Sachs, X. Lin.
The antidepressant sertraline provides a promising therapeutic option for neurotropic cryptococcal infections.
Antimicrobial Agents and Chemotherapy, 56 (2012), pp. 3758-3766
[Zhang et al., 2021]
W. Zhang, W. Qu, H. Wang, H. Yan.
Antidepressants fluoxetine and amitriptyline induce alterations in intestinal microbiota and gut microbiome function in rats exposed to chronic unpredictable mild stress.
Translational Psychiatry, 11 (2021), pp. 131
[Zhernakova et al., 2016]
A. Zhernakova, A. Kurilshikov, M.J. Bonder, E.F. Tigchelaar, M. Schirmer, T. Vatanen, et al.
Population-based metagenomics analysis reveals markers for gut microbiome composition and diversity.
Science, 352 (2016), pp. 565-569
[Zhou et al., 2018]
Y. Zhou, W. Zheng, W. Liu, C. Wang, Y. Zhan, H. Li, et al.
Antidepressant effect of repeated ketamine administration on kynurenine pathway metabolites in patients with unipolar and bipolar depression.
Brain, Behavior, and Immunity, 74 (2018), pp. 205-212
[Zhu et al., 2020]
C. Zhu, L. Sawrey-Kubicek, E. Beals, C.H. Rhodes, H.E. Houts, R. Sacchi, et al.
Human gut microbiome composition and tryptophan metabolites were changed differently by fast food and Mediterranean diet in 4 days: a pilot study.
Nutrition Research, 77 (2020), pp. 62-72
[Zhu et al., 2019]
H.Z. Zhu, Y.D. Liang, Q.Y. Ma, W.Z. Hao, X.J. Li, M.S. Wu, et al.
Xiaoyaosan improves depressive-like behavior in rats with chronic immobilization stress through modulation of the gut microbiota.
Biomedicine & Pharmacotherapy, 112 (2019),
[Zimmermann et al., 2019]
M. Zimmermann, M. Zimmermann-Kogadeeva, R. Wegmann, A.L. Goodman.
Mapping human microbiome drug metabolism by gut bacteria and their genes.
Nature, 570 (2019), pp. 462-467
[Zmora et al., 2018]
N. Zmora, G. Zilberman-Schapira, J. Suez, U. Mor, M. Dori-Bachash, S. Bashiardes, et al.
Personalized gut mucosal colonization resistance to empiric probiotics is associated with unique host and microbiome features.
Cell, 174 (2018), pp. 1388-1405
Copyright © 2022. The Authors
Article options
Tools