Hostname: page-component-f554764f5-44mx8 Total loading time: 0 Render date: 2025-04-18T10:02:39.766Z Has data issue: false hasContentIssue false

Is there a place for psychedelics in sports practice?

Published online by Cambridge University Press:  21 March 2025

Marina A.M. Portes
Affiliation:
Department of Pharmacology, Federal University of Santa Catarina, Florianopolis, Santa Catarina, Brazil
Isabel Werle
Affiliation:
Department of Pharmacology, Federal University of Santa Catarina, Florianopolis, Santa Catarina, Brazil
Leandro J. Bertoglio*
Affiliation:
Department of Pharmacology, Federal University of Santa Catarina, Florianopolis, Santa Catarina, Brazil
*
Corresponding author: Leandro Jose Bertoglio; Email: [email protected]
Rights & Permissions [Opens in a new window]

Abstract

Growing evidence suggests that psychedelic-assisted therapies can alleviate depression, anxiety, posttraumatic stress, and substance use disorder, offering relatively safe profiles, enhanced efficacy, and lasting effects after a few applications. Athletes often experience high levels of stress and pressure, making them susceptible to these psychiatric conditions. However, the effects of psychedelic substances on athletic performance remain largely unknown. Before potential acceptance, evaluating their impact on physical and physiological measures beyond mental health outcomes is crucial. Here, we aim to explore this topic and highlight research directions to advance our understanding. Preclinical studies suggest that psilocybin/psilocin, lysergic acid diethylamide (LSD), N,N-dimethyltryptamine (DMT), and ayahuasca possess anti-inflammatory and anti-nociceptive properties. Studies investigating the effects of classical psychedelics or 3,4-methylenedioxymethamphetamine (MDMA) on factors such as muscle strength, motor coordination, locomotion, endurance, fluid and electrolyte balance, hormonal regulation, and metabolism are still scarce. While adhering to regulatory frameworks, further research in animal models, athletes, and non-athletes is needed to address these gaps, compare psychedelics with commonly used psychoactive drugs, and explore the potential prophylactic and regenerative benefits of specific interventions.

Type
Perspective
Creative Commons
Creative Common License - CCCreative Common License - BY
This is an Open Access article, distributed under the terms of the Creative Commons Attribution licence (https://creativecommons.org/licenses/by/4.0/), which permits unrestricted re-use, distribution and reproduction, provided the original article is properly cited.
Copyright
© The Author(s), 2025. Published by Cambridge University Press on behalf of Scandinavian College of Neuropsychopharmacology

>Highlights

  • Athletes frequently experience intense stress and pressure, increasing their vulnerability to mental health challenges such as depression, anxiety, and sports-related trauma.

  • While psychedelics hold the potential for alleviating these issues, their impact on physical and physiological performance in athletes remains largely unexplored.

  • This perspective explores the effects of psilocybin, LSD, DMT, and MDMA on mental and physical health, identifying key knowledge gaps and proposing future research directions using rodent models relevant to athletic populations.

Summations

  • Psychedelic-assisted therapies are increasingly known for their potential to mitigate symptoms of various psychiatric conditions.

  • Psychedelics may offer intriguing possibilities for enhancing resilience, aiding recovery, and treating sports-related trauma.

  • As scientific understanding evolves, specific psychedelic substances could emerge as complementary tools in sports medicine.

Perspectives

  • Research on the effects of psychedelics on physical performance and physiological parameters is still limited in rodents and humans.

  • Permitting specific psychedelics in sports competitions will require a strong scientific foundation and a revision of anti-doping regulations.

  • Establishing proper guidelines, dosages, and usage contexts will be crucial to ensure their responsible application.

Introduction

Psychedelics are currently defined as psychoactive substances that alter sensory perception, thought patterns, mood, and emotional experiences, affecting numerous cognitive processes (Nichols, Reference Nichols2016). They induce profound changes in consciousness, including visual and auditory hallucinations, an altered perception of time, and a heightened sense of interconnectedness – effects often attributed to serotonin (5-HT) transmission in the brain (Osmond, Reference Osmond1957; Wittmann et al., Reference Wittmann, Carter, Hasler, Cahn, Grimberg, Spring, Hell, Flohr and Vollenweider2007; Nichols, Reference Nichols2016; Yanakieva et al., Reference Yanakieva, Polychroni, Family, Williams, Luke and Terhune2019; Vollenweider & Preller, Reference Vollenweider and Preller2020).

Psychedelic compounds can be classified according to their chemical structure or mechanism of action (Mitchell & Anderson, Reference Mitchell and Anderson2024). Serotonergic psychedelics fall into two main structural categories, characterised by modifications in the tryptamine or the phenethylamine group (Mendes et al., Reference Mendes, Costa, Wiltenburg, Morales-Lima, Fernandes and Filev2022). The first category includes psilocybin (psilocin is the active metabolite) found in certain mushrooms, N,N-dimethyltryptamine (DMT) present in ayahuascaFootnote 1 , and 5-methoxy-N,N-DMT (5-MeO-DMT) derived from certain toad species. The second comprises mescaline, the primary psychoactive component of peyote cacti, and synthetic compounds such as (±)-2,5-dimethoxy-4-iodoamphetamine hydrochloride (DOI). Lysergic acid diethylamide (LSD) is an ergoline-derived compound.

Classical psychedelics (psilocybin, DMT, 5-MeO-DMT, mescaline, and LSD) act as partial or full agonists at 5-HT receptors, primarily 5-HT2A, 5-HT1A, and 5-HT2C (Werle & Bertoglio, Reference Werle and Bertoglio2024). In contrast, compounds like DOI are relatively more selective agonists at 5-HT2A receptors (Werle & Bertoglio, Reference Werle and Bertoglio2024). Some substances associated with psychedelics act through distinct mechanisms. For example, 3,4-methylenedioxymethamphetamine (MDMA) produces psychoactive effects primarily by releasing monoamines (5-HT, noradrenaline, and dopamine) and inhibiting their reuptake; ketamine is a glutamate N-methyl-D-aspartate (NMDA) receptor antagonist; and ibogaine (noribogaine is the active metabolite) interacts with multiple molecular targets, including 5-HT2A receptors, NMDA receptors, and monoamine transporters (Johnson et al., Reference Johnson, Hendricks, Barrett and Griffiths2019; Mendes et al., Reference Mendes, Costa, Wiltenburg, Morales-Lima, Fernandes and Filev2022).

Activating 5-HT2A receptors, primarily those expressed in the apical dendrites of human layer V cortical pyramidal neurones, is essential for the perceptual effects of psychedelic experiences (Madsen et al., Reference Madsen, Fisher, Burmester, Dyssegaard, Stenbæk, Kristiansen, Johansen, Lehel, Linnet, Svarer, Erritzoe, Ozenne and Knudsen2019). The canonical 5-HT2A receptor signalling pathway involves the activation of Gαq/11-proteins and subsequent activation of the enzyme phospholipase C, leading to hydrolysis of phosphatidylinositol-4,5-bisphosphate and the release of inositol triphosphate and diacylglycerol. 5-HT2A receptors also interact with arrestins, recruiting intracellular signalling pathways dependent on these proteins (Kim et al., Reference Kim, Che, Panova, DiBerto, Lyu, Krumm, Wacker, Robertson, Seven, Nichols, Shoichet, Skiniotis and Roth2020; McClure-Begley & Roth, Reference McClure-Begley and Roth2022; Wallach et al., Reference Wallach, Cao, Calkins, Heim, Lanham, Bonniwell, Hennessey, Bock, Anderson, Sherwood, Morris, de Klein, Klein, Cuccurazzu, Gamrat, Fannana, Zauhar, Halberstadt and McCorvy2023). The psychedelic potential of some phenethylamine analogues is associated with the efficacy of 5-HT2A-Gq but not 5-HT2A-β-arrestin-2 recruitment (Wallach et al., Reference Wallach, Cao, Calkins, Heim, Lanham, Bonniwell, Hennessey, Bock, Anderson, Sherwood, Morris, de Klein, Klein, Cuccurazzu, Gamrat, Fannana, Zauhar, Halberstadt and McCorvy2023).

Increasing evidence suggests that 5-HT2A receptor agonism does not fully explain the pharmacological effects of psychedelics (Inserra et al., Reference Inserra, De Gregorio and Gobbi2021; Mendes et al., Reference Mendes, Costa, Wiltenburg, Morales-Lima, Fernandes and Filev2022; Werle et al., Reference Werle, Nascimento, Dos Santos, Soares, Dos Santos, Hallak and Bertoglio2024). Their action also involves the brain activation of other serotonergic and dopaminergic receptor subtypes (Werle & Bertoglio, Reference Werle and Bertoglio2024), tropomyosin receptor kinase B (TrkB) (Moliner et al., Reference Moliner, Girych, Brunello, Kovaleva, Biojone, Enkavi, Antenucci, Kot, Goncharuk, Kaurinkoski, Kuutti, Fred, Elsilä, Sakson, Cannarozzo, Diniz, Seiffert, Rubiolo, Haapaniemi, Meshi, Nagaeva, Öhman, Róg, Kankuri, Vilar, Varjosalo, Korpi, Permi, Mineev, Saarma, Vattulainen, Casarotto and Castrén2023; Shafiee et al., Reference Shafiee, Arabzadeh Bahri, Rafiei, Esmaeilpur Abianeh, Razmara, Jafarabady and Amini2024), ionotropic glutamate receptor interactions (Heresco-Levy & Lerer, Reference Heresco-Levy and Lerer2024), neurotransmitters release (White et al., Reference White, Obradovic, Imel and Wheaton1996; Mason et al., Reference Mason, Kuypers, Müller, Reckweg, Tse, Toennes, Hutten, Jansen, Stiers, Feilding and Ramaekers2020), increased expression of the brain-derived neurotrophic factor (BDNF; He et al., Reference He, McGough, Ravindranathan, Jeanblanc, Logrip, Phamluong, Janak and Ron2005; de Almeida et al., Reference de Almeida, Galvão, da Silva, Silva, Palhano-Fontes, Maia-de-Oliveira, de Araújo, Lobão-Soares and Galvão-Coelho2019; Marton et al., Reference Marton, González, Rodríguez-Bottero, Miquel, Martínez-Palma, Pazos, Prieto, Rodríguez, Sames, Seoane, Scorza, Cassina and Carrera2019; Hutten et al., Reference Hutten, Mason, Dolder, Theunissen, Holze, Liechti, Varghese, Eckert, Feilding, Ramaekers and Kuypers2020b; Shafiee et al., Reference Shafiee, Arabzadeh Bahri, Rafiei, Esmaeilpur Abianeh, Razmara, Jafarabady and Amini2024), and epigenetic changes (Inserra et al., Reference Inserra, Campanale, Rezai, Romualdi and Rubino2024). How psychedelics influence the abovementioned targets/mechanisms is complex, with each substance exhibiting particular features (Ray, Reference Ray2010; Cameron et al., Reference Cameron, Benetatos, Lewis, Bonniwell, Jaster, Moliner, Castrén, McCorvy, Palner and Aguilar-Valles2023).

The ability to induce adaptive structural and functional changes in the brain is a common feature of psychedelics shown in both preclinical and clinical studies (Ly et al., Reference Ly, Greb, Cameron, Wong, Barragan, Wilson, Burbach, Soltanzadeh Zarandi, Sood, Paddy, Duim, Dennis, McAllister, Ori-McKenney, Gray and Olson2018; Lukasiewicz et al., Reference Lukasiewicz, Baker, Zuo and Lu2021; de Vos et al., Reference de Vos, Mason and Kuypers2021; Liao et al., Reference Liao, Dua, Wojtasiewicz, Liston and Kwan2025). These substances induce neuroplasticity in response to intrinsic or extrinsic stimuli, modifying the strength and efficacy of synaptic transmission (Calder & Hasler, Reference Calder and Hasler2023). The cascade of cellular and molecular events implicated includes transmembrane and cytosolic receptor activation (Preller et al., Reference Preller, Burt, Ji, Schleifer, Adkinson, Stämpfli, Seifritz, Repovs, Krystal, Murray, Vollenweider and Anticevic2018; Moliner et al., Reference Moliner, Girych, Brunello, Kovaleva, Biojone, Enkavi, Antenucci, Kot, Goncharuk, Kaurinkoski, Kuutti, Fred, Elsilä, Sakson, Cannarozzo, Diniz, Seiffert, Rubiolo, Haapaniemi, Meshi, Nagaeva, Öhman, Róg, Kankuri, Vilar, Varjosalo, Korpi, Permi, Mineev, Saarma, Vattulainen, Casarotto and Castrén2023; Vargas et al., Reference Vargas, Dunlap, Dong, Carter, Tombari, Jami, Cameron, Patel, Hennessey, Saeger, McCorvy, Gray, Tian and Olson2023), recruitment of secondary messengers and proteins (Olson, Reference Olson2022), changes in the number or complexity of dendritic spines (Ly et al., Reference Ly, Greb, Cameron, Wong, Barragan, Wilson, Burbach, Soltanzadeh Zarandi, Sood, Paddy, Duim, Dennis, McAllister, Ori-McKenney, Gray and Olson2018; Shao et al., Reference Shao, Liao, Gregg, Davoudian, Savalia, Delagarza and Kwan2021), generation of new neurones (Lima da Cruz et al., Reference Lima da Cruz, Moulin, Petiz and Leão2018; Morales-Garcia et al., Reference Morales-Garcia, Calleja-Conde, Lopez-Moreno, Alonso-Gil, Sanz-SanCristobal, Riba and Perez-Castillo2020), among others. Moreover, psychedelics can induce varying effects on functional connectivity across brain networks, such as decreased connectivity within the default mode network associated with self-referential thoughts and the sense of ego (Carhart-Harris et al., Reference Carhart-Harris, Erritzoe, Williams, Stone, Reed, Colasanti, Tyacke, Leech, Malizia, Murphy, Hobden, Evans, Feilding, Wise and Nutt2012; Palhano-Fontes et al., Reference Palhano-Fontes, Andrade, Tofoli, Santos, Crippa, Hallak, Ribeiro and de Araujo2015; Carhart-Harris, Muthukumaraswamy, et al., Reference Carhart-Harris, Muthukumaraswamy, Roseman, Kaelen, Droog, Murphy, Tagliazucchi, Schenberg, Nest, Orban, Leech, Williams, Williams, Bolstridge, Sessa, McGonigle, Sereno, Nichols, Hellyer, Hobden, Evans, Singh, Wise, Curran, Feilding and Nutt2016; Preller et al., Reference Preller, Duerler, Burt, Ji, Adkinson, Stämpfli, Seifritz, Repovš, Krystal, Murray, Anticevic and Vollenweider2020; Daws et al., Reference Daws, Timmermann, Giribaldi, Sexton, Wall, Erritzoe, Roseman, Nutt and Carhart-Harris2022; Siegel et al., Reference Siegel, Subramanian, Perry, Kay, Gordon, Laumann, Reneau, Metcalf, Chacko, Gratton, Horan, Krimmel, Shimony, Schweiger, Wong, Bender, Scheidter, Whiting, Padawer-Curry, Shinohara, Chen, Moser, Yacoub, Nelson, Vizioli, Fair, Lenze, Carhart-Harris, Raison, Raichle, Snyder, Nicol and Dosenbach2024). These changes may shift rigid thought patterns into more integrated and flexible thinking, potentially leading individuals to new insights and perspectives on life experiences. The altered states of consciousness induced by psychedelics may also affect emotional processing and facilitate coping with difficult emotions or traumatic experiences, leading to improved mental health outcomes and even therapeutic benefits (Kraehenmann et al., Reference Kraehenmann, Preller, Scheidegger, Pokorny, Bosch, Seifritz and Vollenweider2015; Barrett et al., Reference Barrett, Doss, Sepeda, Pekar and Griffiths2020; Mertens et al., Reference Mertens, Wall, Roseman, Demetriou, Nutt and Carhart-Harris2020; Arruda Sanchez et al., Reference Arruda Sanchez, Ramos, Araujo, Schenberg, Yonamine, Lobo, de Araujo and Luna2024; Stoliker et al., Reference Stoliker, Novelli, Vollenweider, Egan, Preller and Razi2024; Melani et al., Reference Melani, Bonaso, Biso, Zucchini, Conversano and Scarselli2025).

Psychedelics were categorised as Schedule I substances under the Controlled Substances Act by the United States Drug Enforcement Administration in the 1970s, a decision mirrored by regulatory agencies in other countries. This classification significantly restricted academic and clinical research. However, over the past ten years, scientific and medical interest has been resurgent in exploring the pharmacological effects of these substances. As described in the following two sections, studies indicate that psychedelics have a relatively good safety profile, produce rapid benefits, and exert enduring effects after just a few doses (Riba et al., Reference Riba, Valle, Urbano, Yritia, Morte and Barbanoj2003; Palhano-Fontes et al., Reference Palhano-Fontes, Barreto, Onias, Andrade, Novaes, Pessoa, Mota-Rolim, Osório, Sanches, Dos Santos, Tófoli, de Oliveira Silveira, Yonamine, Riba, Santos, Silva-Junior, Alchieri, Galvão-Coelho, Lobão-Soares, Hallak, Arcoverde, Maia-de-Oliveira and Araújo2019; Mitchell et al., Reference Mitchell, Bogenschutz, Lilienstein, Harrison, Kleiman, Parker-Guilbert, Ot’alora, Garas, Paleos, Gorman, Nicholas, Mithoefer, Carlin, Poulter, Mithoefer, Quevedo, Wells, Klaire, van der Kolk, Tzarfaty, Amiaz, Worthy, Shannon, Woolley, Marta, Gelfand, Hapke, Amar, Wallach, Brown, Hamilton, Wang, Coker, Matthews, de Boer, Yazar-Klosinski, Emerson and Doblin2021; Gukasyan et al., Reference Gukasyan, Davis, Barrett, Cosimano, Sepeda, Johnson and Griffiths2022; Rhee et al., Reference Rhee, Davoudian, Sanacora and Wilkinson2023; Dos Santos & Hallak, Reference Dos Santos and Hallak2024; Hinkle et al., Reference Hinkle, Graziosi, Nayak and Yaden2024). As a result, their therapeutic potential has been explored, presenting a promising approach for treating various psychiatric disorders, as detailed in Section 3 and Table 1. The subsequent sections will evaluate the potential applications of psychedelics for maintaining and improving mental wellness in athletes, their effects on physical and physiological parameters pertinent to athletic performance, and the relevant legal and regulatory frameworks.

Table 1. Effects of single or repeated administration of psilocybin, LSD, ayahuasca, DMT, or MDMA on the mental health of individuals diagnosed with selected psychiatric disorders

Legend: ↔ = relatively no changes; ↓ = reduction; ♂ = men; ♀ = women; AUD = alcohol use disorder; D-B = double-blind; i.v. = intravenous route; MDD = major depressive disorder; MDMA = 3,4-methylenedioxymethamphetamine; n.d. = not described; DMT = N,N-dimethyltryptamine; P-C = placebo-controlled; PTSD = posttraumatic stress disorder; R = randomised; SAD = social anxiety disorder; SD = standard deviation; SEM = standard error of mean; SUD = substance use disorder; TR-MDD = treatment-resistant major depressive disorder.

On the safety of psychedelics

The acute toxicity of psychedelics is considered low. Reports of fatal overdoses associated with their use are rare (Haden & Woods, Reference Haden and Woods2020; Darke et al., Reference Darke, Duflou, Peacock, Farrell, Hall and Lappin2024; Thomas, Reference Thomas2024), with deaths primarily linked to relatively high doses (i.e. ≥ 20 times the typical dose) or the combination of psychedelics with other drugs or ethanol (Schlag et al., Reference Schlag, Aday, Salam, Neill and Nutt2022; Lake & Lucas, Reference Lake and Lucas2024; Kopra et al., Reference Kopra, Penttinen, Rucker and Copeland2025). Clinical studies conducted in supervised settings have also demonstrated low addictive potential (Johnson et al., Reference Johnson, Richards and Griffiths2008; Johansen & Krebs, Reference Johansen and Krebs2015; Johnson et al., Reference Johnson, Griffiths, Hendricks and Henningfield2018; Schlag et al., Reference Schlag, Aday, Salam, Neill and Nutt2022; Hinkle et al., Reference Hinkle, Graziosi, Nayak and Yaden2024). Compared to ethanol, opioids, cocaine, crack, amphetamines, and some psychostimulants, they have a low risk of addiction and intoxication (Johnson et al., Reference Johnson, Griffiths, Hendricks and Henningfield2018). Noteworthy, clinical evidence suggests that psychedelics can alleviate psychological and physiological symptoms associated with dependence on other psychoactive substances (Vamvakopoulou & Nutt, Reference Vamvakopoulou and Nutt2024; Yao et al., Reference Yao, Guo, Lu, Liu, Huang, Diao, Li, Zhang, Kosten, Shi, Bao, Lu and Han2024).

Challenging emotional experiences (e.g. anxiety and panic attacks), sensory and spatial distortions, headache, nausea and vomiting, and elevations in heart rate and blood pressure are changes induced by psychedelics as transient effects observed after administering usual doses but infrequently manifest in protocols using microdoses (Nichols, Reference Nichols2016; Polito & Stevenson, Reference Polito and Stevenson2019; Schlag et al., Reference Schlag, Aday, Salam, Neill and Nutt2022; Wsół, Reference Wsół2023; Murphy et al., Reference Murphy, Muthukumaraswamy and de Wit2024; Neumann et al., Reference Neumann, Dhein, Kirchhefer, Hofmann and Gergs2024; Yerubandi et al., Reference Yerubandi, Thomas, Bhuiya, Harrington, Villa Zapata and Caballero2024) or when used in controlled settings with appropriate inclusion criteria (Rhee et al., Reference Rhee, Davoudian, Sanacora and Wilkinson2023; Hinkle et al., Reference Hinkle, Graziosi, Nayak and Yaden2024; Klaiber et al., Reference Klaiber, Humbert-Droz, Ley, Schmid and Liechti2024; Romeo et al., Reference Romeo, Kervadec, Fauvel, Strika-Bruneau, Amirouche, Verroust, Piolino and Benyamina2024; Simon et al., Reference Simon, Olsen, Hoyte, Black, Reynolds, Dart and Monte2024; Sabé et al., Reference Sabé, Sulstarova, Glangetas, De Pieri, Mallet, Curtis, Richard-Lepouriel, Penzenstadler, Seragnoli, Thorens, Zullino, Preller, Böge, Leucht, Correll, Solmi, Kaiser and Kirschner2025). These relatively limited adverse reactions are associated with stimulating various 5-HT receptors (Johnson et al., Reference Johnson, Richards and Griffiths2008; Family et al., Reference Family, Hendricks, Williams, Luke, Krediet, Maillet and Raz2022; Holze et al., Reference Holze, Caluori, Vizeli and Liechti2022). For example, the potential cardiovascular risk associated with serotonergic psychedelics is attributed to their interaction with 5-HT1B, 5-HT2B, and 5-HT4 receptors (Wsół, Reference Wsół2023). However, no associations have been established between the lifetime use of classical psychedelics and the development of cardiometabolic diseases (Simonsson et al., Reference Simonsson, Osika, Carhart-Harris and Hendricks2021).

The relationship between psychedelic use and the risk of seizures is not fully understood, as clinical studies typically exclude individuals with a history of seizures or convulsions. While psychedelics may theoretically increase the risk in predisposed individuals due to cortical 5-HT2A receptor hyperstimulation, most studies suggest that these substances have a low epileptogenic potential when used in controlled settings. The risk may be elevated when psychedelics are combined with factors common in athletic environments, such as sleep deprivation, stimulant use, or high-stress conditions, particularly in susceptible individuals. However, further investigation is needed to understand better the underlying mechanisms and associated risk factors (Freidel et al., Reference Freidel, Kreuder, Rabinovitch, Chen, Huang and Lewis2024; Lewis et al., Reference Lewis, Jaeger, Girn, Omene, Brendle and Argento2024; Soto-Angona et al., Reference Soto-Angona, Fortea, Fortea, Martínez-Ramírez, Santamarina, López, Knudsen and Ona2024). Based on this, caution is advised for athletes with a history of seizures and those using medications (e.g. bupropion) or supplements (e.g. high-dose caffeine) that may lower the seizure threshold.

The use of psychedelics, particularly MDMA, has been associated with an increased risk of hyponatremia in humans, primarily due to increased antidiuretic hormone (also known as vasopressin) release from the posterior pituitary and excessive fluid intake, leading to water retention and sodium dilution (Atila et al., Reference Atila, Straumann, Vizeli, Beck, Monnerat, Holze, Liechti and Christ-Crain2024). This mechanism is attributed to MDMA’s elevation of hypothalamic 5-HT and dopamine levels, stimulating vasopressin release and promoting water retention via vasopressin-2 receptors in the kidneys. Excessive water intake, driven by hyperthermia, dry mouth, and stimulant effects in physically demanding or hot environments, may exacerbate sodium dilution. Although this effect is self-limiting and observed mainly acutely, this condition may be particularly concerning for endurance athletes. Temporary hyponatremia outside of competition may contribute to longer-term consequences, potentially predisposing them to a higher risk of injuries or reduced performance in subsequent training or competitions.

Evidence from both rodent and human studies has demonstrated an association between MDMA use and an increased risk of hyperthermia and rhabdomyolysis. MDMA-treated rodents exhibited significant increases in body temperature, sustained muscle contraction, and muscle damage resembling rhabdomyolysis. These effects were related to increases in neurotransmitters, primarily 5-HT and dopamine, and activation of the sympathetic nervous system (Sprague et al., Reference Sprague, Brutcher, Mills, Caden and Rusyniak2004; Duarte et al., Reference Duarte, Leão, Magalhães, Ascensão, Bastos, Amado, Vilarinho, Quelhas, Appell and Carvalho2005; Rusyniak et al., Reference Rusyniak, Tandy, Hekmatyar, Mills, Smith, Bansal, MacLellan, Harper and Sprague2005; Sprague et al., Reference Sprague, Moze, Caden, Rusyniak, Holmes, Goldstein and Mills2005; de Bragança et al., Reference de Bragança, Moreau, de Brito, Shimizu, Canale, de Jesus, Silva, Gois, Seguro and Magaldi2017). In humans, clinical and observational studies have reported similar effects, especially in intoxication or recreational settings involving prolonged physical activity, crowded environments, and inadequate thermoregulation (Screaton et al., Reference Screaton, Singer, Cairns, Thrasher, Sarner and Cohen1992; Lehmann et al., Reference Lehmann, Thom and Croft1995; Halachanova et al., Reference Halachanova, Sansone and McDonald2001; Sue et al., Reference Sue, Lee and Huang2002; Vanden Eede et al., Reference Vanden Eede, Montenij, Touw and Norris2012; Doyle et al., Reference Doyle, Meyer, Breen and Hunt2020). This could be relevant for endurance athletes if their MDMA use and physical exercise are not adequately spaced apart, as MDMA-induced hyperthermia and rhabdomyolysis can be exacerbated by the physiological demands of prolonged exertion, increasing the risk of severe complications and impairing athletic performance. Although the cited articles did not assess athletes under the acute effects of psychedelic substances, their findings indirectly underscore the importance of understanding the risks associated with MDMA use in physically demanding contexts, as well as the need for proper monitoring of signs and symptoms.

Evidence indicates that 5-HT is a key neuromodulator of locomotor activity (Bacqué-Cazenave et al., Reference Bacqué-Cazenave, Bharatiya, Barrière, Delbecque, Bouguiyoud, Di Giovanni, Cattaert and De Deurwaerdère2020; Flaive et al., Reference Flaive, Fougère, van der Zouwen and Ryczko2020). As reviewed by Werle and Bertoglio (Reference Werle and Bertoglio2024), published studies have demonstrated the biphasic effects of psychedelic substances on locomotion. In the open-field test, rats and mice exhibit either hyperlocomotion or hypolocomotion, depending on the dose. These effects are mediated by mechanisms involving the activation of 5-HT1A, 5-HT2C, and 5-HT2A receptors (in the case of MDMA, they also involve the release of 5-HT and dopamine). Each substance has its particularities, although hypolocomotor effects (suggestive of sedation) generally predominate at moderate to high doses (Werle & Bertoglio, Reference Werle and Bertoglio2024). While it is unlikely and strongly discouraged for individuals to participate in sports while under the acute influence of psychedelics, it is worth noting that rodent studies suggest psilocybin, LSD, DMT, ayahuasca, and MDMA can influence locomotor activity.

Psychedelics and mental health

Psychedelics can provide significant benefits across multiple domains of mental health and well-being in healthy individuals (Lebedev et al., Reference Lebedev, Kaelen, Lövdén, Nilsson, Feilding, Nutt and Carhart-Harris2016; Schmid & Liechti, Reference Schmid and Liechti2018; Hutten et al., Reference Hutten, Mason, Dolder, Theunissen, Holze, Liechti, Feilding, Ramaekers and Kuypers2020a; Perkins et al., Reference Perkins, Pagni, Sarris, Barbosa and Chenhall2022). Of particular relevance to athletes are several potential effects, including reduced pain (Ramaekers et al., Reference Ramaekers, Hutten, Mason, Dolder, Theunissen, Holze, Liechti, Feilding and Kuypers2021; Askey et al., Reference Askey, Lasrado, Maiarú and Stephens2024; Strand et al., Reference Strand, Whitney, Johnson, Dunn, Attanti, Maloney, Misra, Gomez, Viswanath, Emami and Leathem2025) and improvements in sleep (Allen et al., Reference Allen, Jeremiah, Murphy, Sumner, Forsyth, Hoeh, Menkes, Evans, Muthukumaraswamy, Sundram and Roop2024). Additionally, psychedelics may enhance stress management by reducing anxiety levels and promoting greater emotional resilience (Griffiths et al., Reference Griffiths, Johnson, Richards, Richards, McCann and Jesse2011; Arruda Sanchez et al., Reference Arruda Sanchez, Ramos, Araujo, Schenberg, Yonamine, Lobo, de Araujo and Luna2024).

The growing interest and acceptance of psychedelic substances have driven clinical trials, advancing our understanding of their potential benefits (Nichols, Reference Nichols2016; Reiff et al., Reference Reiff, Richman, Nemeroff, Carpenter, Widge, Rodriguez, Kalin and McDonald2020; Nutt & Carhart-Harris, Reference Nutt and Carhart-Harris2021; McClure-Begley & Roth, Reference McClure-Begley and Roth2022). Their contribution to alleviating symptoms of depression, anxiety, posttraumatic stress disorder (PTSD), eating disorders, and substance use disorders has been documented (Reiff et al., Reference Reiff, Richman, Nemeroff, Carpenter, Widge, Rodriguez, Kalin and McDonald2020; Barber & Aaronson, Reference Barber and Aaronson2022; Brewerton et al., Reference Brewerton, Wang, Lafrance, Pamplin, Mithoefer, Yazar-Klosinki, Emerson and Doblin2022; Cavarra et al., Reference Cavarra, Falzone, Ramaekers, Kuypers and Mento2022; Cuerva et al., Reference Cuerva, Spirou, Cuerva, Delaquis and Raman2024; Dos Santos & Hallak, Reference Dos Santos and Hallak2024; Doss et al., Reference Doss, DeMarco, Dunsmoor, Cisler, Fonzo and Nemeroff2024; Zaretsky et al., Reference Zaretsky, Jagodnik, Barsic, Antonio, Bonanno, MacLeod, Pierce, Carney, Morrison, Saylor, Danias, Lepow and Yehuda2024). Table 1 presents the details and primary findings of human studies examining the effects of psilocybin, LSD, DMT, ayahuasca, and MDMA on the mental health of individuals diagnosed with the aforementioned psychiatric conditions. Noteworthy, the association of psychedelics with psychotherapeutic support (i.e. psychedelic-assisted psychotherapy) has been shown to improve the integration of psychedelic experiences (Luoma et al., Reference Luoma, Chwyl, Bathje, Davis and Lancelotta2020).

Some of the studies reviewed (Table 1) also report that these substances are associated with significant and long-lasting symptom reduction, with therapeutic effects persisting for weeks or months following only a few administrations, even in patients resistant to typical pharmacological treatment. Psychedelics have also presented a favourable safety profile, as indicated by the relatively low incidence of severe adverse reactions when administered under controlled clinical conditions. Such features may be particularly relevant for health care in athletes, who often endure high levels of physical and mental stress and are vulnerable to various psychiatric disorders (Edwards, Reference Edwards2024). Hypothetically, psychedelic therapy could serve as a valuable tool for enhancing well-being in this population with minimal risk of impairing performance.

However, it is essential to address the methodological limitations of the studies published to date, as well as the gaps that still need to be clarified to enable a responsible application of psychedelic therapies in clinical practice. Some reviewed studies included small sample sizes and lacked double-blind methodologies or inactive placebos, which limits the generalisability of the observed results and increases the chance of confirmation bias. Furthermore, the majority of participants were White or Caucasian, which may limit the extrapolation of findings to other ethnic groups with distinct cultural or genetic characteristics, thus impacting the representativeness of these results when psychedelics are applied on a larger scale. Another issue is the variability in study protocols (e.g. dosage, number of administrations, and intervals between treatments). Greater methodological rigour and standardisation are needed to understand better the actual clinical impact of psychedelic therapy on both the general population and athletes. Future research should also incorporate more objective evaluation methods, ideally including physiological or neurobiological measurements that can be correlated with the health status (or psychiatric disorder under investigation).

Mental health issues in athletes

Studies indicate that the prevalence of psychiatric disorders in high-performance athletes (both amateur and professional) may be similar or even higher than in the general population, which likely arises from intense physical and emotional stressors often experienced (Gouttebarge et al., Reference Gouttebarge, Castaldelli-Maia, Gorczynski, Hainline, Hitchcock, Kerkhoffs, Rice and Reardon2019; Reardon et al., Reference Reardon, Hainline, Aron, Baron, Baum, Bindra, Budgett, Campriani, Castaldelli-Maia, Currie, Derevensky, Glick, Gorczynski, Gouttebarge, Grandner, Han, McDuff, Mountjoy, Polat, Purcell, Putukian, Rice, Sills, Stull, Swartz, Zhu and Engebretsen2019; Glick et al., Reference Glick, Stillman and McDuff2020; Marí-Sanchis et al., Reference Marí-Sanchis, Burgos-Balmaseda and Hidalgo-Borrajo2022; McDonald et al., Reference McDonald, Losty and MacCarthy2023; Smith et al., Reference Smith, Buadze, Colangelo and Liebrenz2023; Thuany et al., Reference Thuany, Viljoen, Gomes, Knechtle and Scheer2023; Beable, Reference Beable2024). Among them are the high demand for physical and sports performance, overtraining, interpersonal conflicts in competitions, the imbalance between personal life and training, injuries, and early retirement (Chang et al., Reference Chang, Putukian, Aerni, Diamond, Hong, Ingram, Reardon and Wolanin2020). Furthermore, due to self-pressure to demonstrate mental resilience, athletes may not report their health concerns, accept professional assistance, or adhere to treatment. Additionally, athletes may often avoid pharmacological treatment due to concerns about doping, potential adverse reactions, and the effects of medication on athletic performance (Reardon, Reference Reardon2016; Bomfim, Reference Bomfim2020). As a result, a cycle of untreated suffering can develop, compromising both mental health and physical aspects. Early identification of these factors and appropriate clinical intervention are essential to ensure performance and longevity in sports practice, as well as the psychological well-being of athletes (Glick et al., Reference Glick, Stillman, Reardon and Ritvo2012; Chang et al., Reference Chang, Putukian, Aerni, Diamond, Hong, Ingram, Reardon and Wolanin2020). Consequently, there is growing interest in sports research to assess the mental health of athletes such as long-distance runners, cyclists, swimmers, triathletes, and others (Berger et al., Reference Berger, Best, Best, Lane, Millet, Barwood, Marcora, Wilson and Bearden2024).

Drugs currently available for the management of psychiatric disorders in athletes present significant limitations (Morris, Reference Morris2015; Reardon & Creado, Reference Reardon and Creado2016; Tso & Pelliccia, Reference Tso and Pelliccia2022). Antidepressants and anxiolytics currently approved for clinical use are administered daily and can cause side effects that negatively affect athletic performance, such as drowsiness, changes in appetite, and weight gain (Reardon, Reference Reardon2016; Reardon & Creado, Reference Reardon and Creado2016; Edwards, Reference Edwards2024). In addition, individual variability in response to these medications can hinder treatment effectiveness. For example, while approximately 15% of participants in clinical trials experience a significant antidepressant effect beyond that of a placebo (Stone et al., Reference Stone, Yaseen, Miller, Richardville, Kalaria and Kirsch2022), around 30% of individuals diagnosed with major depressive disorder are resistant to conventional treatment, further increasing the social and economic burden of this condition (McIntyre et al., Reference McIntyre, Alsuwaidan, Baune, Berk, Demyttenaere, Goldberg, Gorwood, Ho, Kasper, Kennedy, Ly-Uson, Mansur, McAllister-Williams, Murrough, Nemeroff, Nierenberg, Rosenblat, Sanacora, Schatzberg, Shelton, Stahl, Trivedi, Vieta, Vinberg, Williams, Young and Maj2023). In this scenario, psychedelic therapy could emerge as either a complementary or an alternative for the treatment of psychiatric disorders in athletes.

Psychedelics to maintain and improve mental health in athletes

Several clinical studies have demonstrated the efficacy of psychedelic-assisted psychotherapy (Table 1; Nichols, Reference Nichols2016; Reiff et al., Reference Reiff, Richman, Nemeroff, Carpenter, Widge, Rodriguez, Kalin and McDonald2020; Nutt & Carhart-Harris, Reference Nutt and Carhart-Harris2021; Cavarra et al., Reference Cavarra, Falzone, Ramaekers, Kuypers and Mento2022; Knudsen, Reference Knudsen2023). Following approval by the Therapeutic Goods Administration in 2023, Australia became the first country to authorise and regulate the medicinal use of psilocybin and MDMA for the treatment of depression and PTSD, respectively (Nutt et al., Reference Nutt, Crome and Young2024). Similarly, Oregon and Colorado became the first American states to legalise psilocybin, issuing official licences to specialised mental healthcare service centres for use (Korthuis et al., Reference Korthuis, Hoffman, Wilson-Poe, Luoma, Bazinet, Pertl, Morgan, Cook, Bielavitz, Myers, Wolf, McCarty and Stauffer2024).

To date, the potential of psychedelics to enhance mental health or treat psychiatric disorders in athletes remains unknown. However, considering the evidence from the general population (Table 1), several aspects of psychedelic therapy may be beneficial for these individuals (Carhart-Harris & Goodwin, Reference Carhart-Harris and Goodwin2017; Barber & Aaronson, Reference Barber and Aaronson2022; Holze et al., Reference Holze, Singh, Liechti and D’Souza2024). In healthy athletes, the administration of psychedelics may offer benefits in promoting mental health and well-being, aiding in the management of psychological and emotional challenges. By enhancing resilience and emotional flexibility, psychedelic therapy could mitigate the effects of everyday stressors in high-performance sports, including intensive training routines, self-imposed demands for physical performance, and sustained competitiveness. Moreover, in athletes diagnosed with psychiatric disorders, psychedelic-assisted psychotherapy could offer some advantages over conventional treatments. Unlike daily medications, only a few sessions spaced over days to weeks are typically sufficient to promote long-term mental health benefits that are maintained over several months (Yao et al., Reference Yao, Guo, Lu, Liu, Huang, Diao, Li, Zhang, Kosten, Shi, Bao, Lu and Han2024). Furthermore, the half-life of these substances lasts only a few hours, not producing withdrawal symptoms. Although psychedelic therapy may result in adverse reactions, they are transient and manifest mainly in the following hours after administration. Thus, potential concerns associated with impaired sports performance can be reduced, even if athletes are in training or competition periods (Reardon & Creado, Reference Reardon and Creado2016; Edwards, Reference Edwards2024). Yousefi et al. (Reference Yousefi, Lietz, O’Higgins, Rippe, Hasler, van Elk and Enriquez-Geppert2025) have meta-analysed psilocybin’s acute effects on executive functions and attention. Psilocybin increased reaction times dose-dependently without significantly affecting accuracy, suggesting an impairment in executive function that may be relevant to specific sports. However, its impact on performance is potentially less concerning, as athletes are not expected to compete while under the influence of psychedelics.

Several psychedelic substances produce prosocial effects in rodent and human studies (Dumont et al., Reference Dumont, Sweep, van der Steen, Hermsen, Donders, Touw, van Gerven, Buitelaar and Verkes2009; Hysek et al., Reference Hysek, Schmid, Simmler, Domes, Heinrichs, Eisenegger, Preller, Quednow and Liechti2014; Kamilar-Britt & Bedi, Reference Kamilar-Britt and Bedi2015; Griffiths et al., Reference Griffiths, Johnson, Richards, Richards, Jesse, MacLean, Barrett, Cosimano and Klinedinst2018; De Gregorio et al., Reference De Gregorio, Popic, Enns, Inserra, Skalecka, Markopoulos, Posa, Lopez-Canul, He, Lafferty, Britt, Comai, Aguilar-Valles, Sonenberg and Gobbi2021; Bhatt & Weissman, Reference Bhatt and Weissman2024). While systematic research on psychedelics in sports is limited, their potential prospective effects may include improved social dynamics during training or competition, team cohesion, reduced anxiety, enhanced resilience among athletes, and sports-related mild traumatic brain injury (e.g. concussion) (VanderZwaag et al., Reference VanderZwaag, Garcia-Romeu and Garcia-Barrera2024). However, the use of psychedelics in sports raises potential issues. Serotonergic psychedelics and related compounds produce varying effects in tests of negative social interactions, often assessing aggression, in rodents through their actions on 5-HT2A and 5-HT1A receptors (Odland et al., Reference Odland, Kristensen and Andreasen2022). Future studies must establish optimal dosages, contexts, and protocols that maximise potential benefits while minimising risks.

Scientific evidence on the interactions between psychedelic substances and antidepressants, antipsychotics, anxiolytics, and mood stabilisers remains limited. However, it has been reported that psychedelics and certain psychiatric medications may share overlapping pharmacological targets, molecular pathways interactions, and hepatic metabolism via similar enzymes (Sarparast et al., Reference Sarparast, Thomas, Malcolm and Stauffer2022; Rhee et al., Reference Rhee, Davoudian, Sanacora and Wilkinson2023; Halman et al., Reference Halman, Kong, Sarris and Perkins2024). Consequently, drug interactions between psychedelic substances and medications already used by athletes should be considered, as they may potentiate or attenuate the actions of both substances. Therefore, adequate clinical monitoring will be essential to mitigate the risks of adverse reactions, toxicity, or inadequate management of psychiatric symptoms.

Effects of psychedelics on physical and physiological parameters

Administration of the psychedelic substance DOI has been shown to reduce circulating levels of total cholesterol and low-density lipoprotein (LDL) in a high-fat diet-fed apolipoprotein E knockout mice model without affecting food intake or body weight. DOI administration was also associated with a reduction in the increased serum levels of the pro-inflammatory cytokine CXCL10 induced by high-fat diet-fed and reduced expression of pro-inflammatory marker genes in the aortic arch (Flanagan et al., Reference Flanagan, Sebastian, Battaglia, Foster, Maillet and Nichols2019a). On the other hand, preclinical studies have shown potentially conflicting results of the psilocybin administration on metabolic parameters and body weight regulation. Although the administration of a high dose of psilocybin was associated with a modest but significant reduction in body weight, decreased consumption of the high-calorie diet, and decreased central adiposity in a rodent model of obesity (Huang et al., Reference Huang, Pham, Panenka, Honer and Barr2022), neither a single nor repeated administration of psilocybin had significant metabolic effects. It did not lower body weight or food intake in diet-induced obese mice or genetic mouse models of obesity (Fadahunsi et al., Reference Fadahunsi, Lund, Breum, Mathiesen, Larsen, Knudsen, Klein and Clemmensen2022). Moreover, increased creatine kinase, aspartate aminotransferase, and chloride have been reported in male and female mice treated with psilocybin (Shakir et al., Reference Shakir, Pedicini, Bullock, Hoen, Macias, Freiman, Pletnikov, Tamashiro and Cordner2024). Preclinical studies have shown that MDMA treatment may increase serum levels of total and LDL cholesterol, corticosterone, aspartate transaminase, alanine transaminase, or glucose in rodents (Graham et al., Reference Graham, Herring, Schaefer, Vorhees and Williams2010; Shahraki & Irani, Reference Shahraki and Irani2014; Golchoobian et al., Reference Golchoobian, Nabavizadeh, Roghani, Foroumadi and Mohammadian2017), although hypoglycaemia has also been reported (Soto-Montenegro et al., Reference Soto-Montenegro, Vaquero, Arango, Ricaurte, García-Barreno and Desco2007; Golchoobian et al., Reference Golchoobian, Nabavizadeh, Roghani, Foroumadi and Mohammadian2017).

In addition to regulating body weight, lipid metabolism is also essential for cellular mechanisms related to inflammation and nociception/pain, and the anti-inflammatory and immunomodulatory properties of psychedelics have also been reported (Flanagan & Nichols, Reference Flanagan and Nichols2022). Lipid mediators, including arachidonic acid (AA), can be metabolised by cyclooxygenase (COX), lipoxygenase (LOX), and cytochrome P450 (CYP450) enzymes and converted to pro-inflammatory metabolites such as prostaglandins, thromboxane, leukotrienes, and hydroxyeicosatetraenoic acids. In rodents, the psychedelic bufotenine has been shown to induce an anti-nociceptive effect and promote the downregulation of inflammatory mediators from COX, LOX, CYP450, linoleic acid, docosahexaenoic acid, and other pro-inflammatory pathways (Wang et al., Reference Wang, Xu, Shen, Zhou, Lv, Ma, Li, Wu and Duan2021a; Shen et al., Reference Shen, Lv, Yang, Deng, Liu, Zhou, Zhu and Ma2022). Askey et al. (Reference Askey, Lasrado, Maiarú and Stephens2024) have reviewed the psilocybin potential as an anti-nociceptive agent, focusing on preclinical animal models and exploring serotonergic mechanisms and neuroplastic actions that improve functional connectivity in brain regions involved in chronic pain. They also discuss its broader effects on pain and associated emotional and inflammatory components. The review by Strand et al. (Reference Strand, Whitney, Johnson, Dunn, Attanti, Maloney, Misra, Gomez, Viswanath, Emami and Leathem2025) has examined psilocybin, LSD, and ketamine as potential treatments for chronic pain. It focuses on their pharmacology, effects on neuropathic pain, clinical implications, safety profiles, and patient responses.

Preclinical and clinical data also indicate that psychedelics increase the release of anti-inflammatory interleukins (e.g. IL-10) and reduce the expression and activity of other pro-inflammatory markers, including IL-6, IL-1β, tumour necrosis factor-alpha (TNF-α), and nuclear factor kappa B (NF-kB). Thus, administration of these substances may attenuate the activation of genes and downstream signalling pathways that contribute to inflammation (dos Santos, Reference dos Santos2014; Boxler et al., Reference Boxler, Streun, Liechti, Schmid, Kraemer and Steuer2018; Flanagan & Nichols, Reference Flanagan and Nichols2022; Mason et al., Reference Mason, Szabo, Kuypers, Mallaroni, de la Torre Fornell, Reckweg, Tse, Hutten, Feilding and Ramaekers2023; Low et al., Reference Low, Ng, Lim, Goh and Kumari2025). DOI can inhibit TNF-α-induced inflammation by mitigating the expression of genes encoding intracellular adhesion molecule-1, vascular cell adhesion molecule-1, and IL-6 through serotonin 5-HT2A receptor activation in both in vitro and in vivo (Yu et al., Reference Yu, Becnel, Zerfaoui, Rohatgi, Boulares and Nichols2008; Nau et al., Reference Nau, Yu, Martin and Nichols2013). Furthermore, DOI administration blocked the activation and translocation of NF-kB and decreased nitric oxide synthase activity (Yu et al., Reference Yu, Becnel, Zerfaoui, Rohatgi, Boulares and Nichols2008).

In vitro studies have demonstrated that psilocybin-containing mushroom extracts inhibited lipopolysaccharide (LPS)-induced increases in TNF-α and IL-1β, besides decreasing COX-2 concentrations in treated human U937 macrophage cells (Nkadimeng et al., Reference Nkadimeng, Steinmann and Eloff2021; Laabi et al., Reference Laabi, LeMmon, Vogel, Chacon and Jimenez2024). In healthy volunteers, a single dose of psilocybin reduced plasma levels of TNF-α immediately after administration, and IL-6 and C-reactive protein were reduced in the psilocybin group seven days later. The persisting reductions in pro-inflammatory markers correlated with clinical improvement of mood and sociability (Mason et al., Reference Mason, Szabo, Kuypers, Mallaroni, de la Torre Fornell, Reckweg, Tse, Hutten, Feilding and Ramaekers2023).

Possible opposite effects have been reported regarding MDMA. Acute administration of MDMA appears to promote an anti-inflammatory effect. It impairs the secretion of IL-1β and TNF-α induced by LPS administration in rodents (Connor et al., Reference Connor, Kelly, McGee and Leonard2000), besides suppressing innate IFN-γ production by increasing IL-10 levels (Boyle & Connor, Reference Boyle and Connor2007). On the other hand, in human plasma samples collected at different time points after a single oral administration of MDMA, an increase in cortisol and lipidic mediators of inflammation was observed, suggesting stimulation of inflammatory pathways (Boxler et al., Reference Boxler, Streun, Liechti, Schmid, Kraemer and Steuer2018).

Immunomodulatory effects of psychedelic substances and other chemical compounds derived from ayahuasca have also been reported (dos Santos, Reference dos Santos2014; Galvão-Coelho et al., Reference Galvão-Coelho, de Menezes Galvão, de Almeida, Palhano-Fontes, Campos Braga, Lobão Soares, Maia-de-Oliveira, Perkins, Sarris and de Araujo2020). Harmine has been proposed to exert anti-inflammatory and antioxidant effects through several mechanisms, such as AMPK/Nrf2 pathway activation, reduced caspase-3 expression by repressing the Bax/Bcl2 ratio, inhibition of the c-Jun N-terminal kinase (JNK), downregulation of LC3B II/I, p38 MAPK, TLR4, and NF-κB levels. Furthermore, it appears to increase the expression of p62, Bcl-2, Beclin1, ULK1, and p-mTOR (Hamsa & Kuttan, Reference Hamsa and Kuttan2010; Liu et al., Reference Liu, Wu, Gong, Wang, Zhu, Tong, Chen, Ling and Huang2017a; Niu et al., Reference Niu, Yao, Li, Zang, Li, Zhao, Liu and Zhi2019; Ma et al., Reference Ma, Li, Yao, Liu, Yu, Zang, Wang, Zhou, Wen, Luo, Li and Niu2024; Tabaa et al., Reference Tabaa, Tabaa, Rashad, Elballal and Elazazy2024). Harmine also attenuated bone destruction induced by an inflammatory response. It shifted the polarisation of macrophages from M1 to M2 phenotypes both in vitro and in vivo in a murine model (Wang et al., Reference Wang, Wang, Wang, Ge, Xu, Zheng, Jiang, Zhao, Xu, Wang, Zhu and Geng2021b). A three-day ayahuasca treatment prevented anxiety and oxidative stress induced by an inflammatory insult in rats. Additionally, it increased cortical levels of the anti-inflammatory cytokine IL-4 and BDNF (de Camargo et al., Reference de Camargo, Joaquim, Machado, de Souza Ramos, da Rosa, de Novais Junior, Mathias, Maximiano, Strickert, Nord, Gava, Scarpari, Martins, Lins, Chaves, da Silva, de Oliveira, da Silva, Fernandes, Tiscoski, Piacentini, Santos, Inserra, Bobinski, Rezin, Yonamine, Petronilho and de Bitencourt2024).

Although the precise molecular mechanisms related to the effects of psychedelics on immunity and inflammatory responses remain to be elucidated, the involvement of 5-HT2A receptor activation has been proposed. 5-HT2A receptor is widely distributed in tissues and cells, regulating innate and adaptive immune responses, such as the spleen, thymus, circulating lymphocytes, T cells, eosinophils, and mononuclear cells (Herr et al., Reference Herr, Bode and Duerschmied2017; Thompson & Szabo, Reference Thompson and Szabo2020). While the 5-HT2A receptor activation by 5-HT primarily contributes to inflammation, psychedelics appear to recruit anti-inflammatory intracellular signalling pathways through activation of the same receptor, possibly by stabilising it in a slightly different structural and functional conformation, that is, biased agonism (Raote et al., Reference Raote, Bhattacharya, Panicker and Chattopadhyay2007; Shan et al., Reference Shan, Khelashvili, Mondal, Mehler and Weinstein2012; Flanagan et al., Reference Flanagan, Foster, Galbato, Lum, Louie, Song, Halberstadt, Billac and Nichols2024). The anti-inflammatory effects of psychedelics resulting from the activation of the 5-HT2A receptor have also been associated with improved respiratory and neurological function, demonstrating benefits in animal models of asthma (Stankevicius et al., Reference Stankevicius, Ferraz-de-Paula, Ribeiro, Pinheiro, Ligeiro de Oliveira, Damazo, Lapachinske, Moreau, Tavares de Lima and Palermo-Neto2012; Nau et al., Reference Nau, Miller, Saravia, Ahlert, Yu, Happel, Cormier and Nichols2015; Flanagan et al., Reference Flanagan, Sebastian, Battaglia, Foster, Cormier and Nichols2019b; Flanagan et al., Reference Flanagan, Billac, Landry, Sebastian, Cormier and Nichols2020) and attenuating the functional consequences of neuroinflammation (Zhong et al., Reference Zhong, Tao and Yang2015; Liu et al., Reference Liu, Li, Tan, Wang, Fan and Huang2017b; Sun et al., Reference Sun, Yang, Zhang, Zhang, Lu, Hu, Liu, Zhou and Chen2019; Nardai et al., Reference Nardai, László, Szabó, Alpár, Hanics, Zahola, Merkely, Frecska and Nagy2020; Xin et al., Reference Xin, Ma, Chen, Zhou, Dong, Wang and Ji2021; Goulart da Silva et al., Reference Goulart da Silva, Daros, Santos, Yonamine and de Bitencourt2022; Zanikov et al., Reference Zanikov, Gerasymchuk, Ghasemi Gojani, Robinson, Asghari, Groves, Haselhorst, Nandakumar, Stahl, Cameron, Li, Rodriguez-Juarez, Snelling, Hudson, Fiselier, Kovalchuk and Kovalchuk2023; Zheng et al., Reference Zheng, Lin, Lu, Cao, Liu, Lin, Yang, Zhang, Tu, Pan, Hu and Zhang2023; Floris et al., Reference Floris, Dabrowski, Zanda and Daws2024).

A significant knowledge gap in psychedelic research, particularly regarding their potential use in athletes, is the lack of studies evaluating their effects on physical health and metabolic parameters. Based on the evidence outlined above and its potential translational implications, treating athletes with psychedelic substances may offer benefits. The improved mental well-being and emotional control associated with psychedelic therapy could contribute to performance by making them more focused and resilient. At the same time, these substances’ anti-inflammatory and analgesic effects could mitigate physical stress, reduce muscle fatigue, and facilitate recovery after prolonged or intense exercise. By reducing inflammation, psychedelics could also improve mental health and reduce symptoms in individuals with psychiatric disorders such as depression or anxiety, as convergent evidence points to an increase in inflammatory markers in these clinical conditions and the significant role of inflammation in their pathophysiology (Bauer & Teixeira, Reference Bauer and Teixeira2019; Beurel et al., Reference Beurel, Toups and Nemeroff2020; Zeng et al., Reference Zeng, Chourpiliadis, Hammar, Seitz, Valdimarsdóttir, Fang, Song and Wei2024). Since research on the use of psychedelic substances in sports contexts is incipient (Fig. 1), far more studies are needed before potentially establishing guidelines on their safe and effective use.

Figure 1. An overview of the current landscape of psychedelics and athletic performance.

Psychedelics in sports competitions: legal and regulatory considerations

Psychedelic substances have been classified as prohibited or controlled substances in most countries, posing challenges for establishing potential guidelines that ensure treatment efficacy and safety under appropriate regulatory oversight. In sports competitions, the World Anti-Doping Agency (WADA; https://www.wada-ama.org/en/prohibited-list) does not list psychedelics as ‘prohibited substances’, except for MDMA, which is classified as a stimulant amphetamine.

For a substance or method to be included in WADA’s Prohibited Substances List under the World Anti-Doping Code, it must meet at least two of the following three criteria: (1) it enhances or has the potential to enhance sports performance, (2) it poses an actual or potential risk to athlete health, and (3) it violates the spirit of sport as defined in the Code (https://www.wada-ama.org/en/resources/world-anti-doping-code-and-international-standards/world-anti-doping-code). To date, no clinical evidence has suggested that psychedelics act as ergogenic aids. WADA regularly updates its prohibited and restricted substances list based on evolving scientific evidence. For example, while cannabis/Δ9-tetrahydrocannabinol remains prohibited in competition due to its potential to impair performance, pose safety risks, and violate the ’spirit of sport’, cannabidiol (CBD) has been permitted, as it lacks these properties. As research on psychedelics progresses, the regulatory status of specific compounds in sports may be reevaluated, potentially leading to updates similar to the removal of CBD from the prohibited list.

Conclusions and suggestions for future research

Several clinical studies have highlighted the mental health benefits of psychedelics and their potential role as therapeutic adjuncts to improve the quality of life, but significant considerations remain. A critical knowledge gap in evaluating these substances’ effects on physical health in humans persists. Similarly, the impact of psychedelics on physiological responses relevant to athletic performance, such as muscular strength, motor coordination, locomotion, endurance, cardiorespiratory capacity, fluid and electrolyte balance, hormonal regulation, fatigue, and reflexes, remains largely unexplored scientifically. Moreover, it is worth noting the ethical and legal concerns associated with performance-enhancing substances and the importance of distinguishing between the use of psychedelics within and outside the acute performance/sports context.

Rodent research can provide a valuable foundation for understanding the potential effects of psychedelic therapy on physical performance in humans (Fig. 2). The rotarod test has been used to assess motor coordination and balance in rodents. The gait analysis test provides a detailed assessment of movement patterns and gait symmetry, which is crucial for identifying motor coordination changes (Carter et al., Reference Carter, Morton and Dunnett2001; Deacon, Reference Deacon2013). Muscular strength is typically evaluated through the grip strength test, which measures the animal’s grip force by stimulating traction of the forelimbs or hind limbs (Munier et al., Reference Munier, Pank, Severino, Wang, Zhang, Vergnes and Reue2022). It provides a direct measure of muscle strength, relevant for assessing whether psychedelics could influence aspects of muscular endurance in humans, an essential factor in the performance of athletes. The treadmill running test (Dougherty et al., Reference Dougherty, Springer and Gershengorn2016; Castro & Kuang, Reference Castro and Kuang2017) is a tool for exploring the effects of psychedelic substances on endurance and cardiorespiratory capacity. Rodents are encouraged to run on a treadmill, allowing for analysis of aerobic capacity, fatigue, and prolonged exercise tolerance. These data help understand the potential of psychedelics to enhance aerobic performance and to observe possible indirect cardiorespiratory impacts from their administration. Stress resilience is also essential for high-performance athletes, and the forced swim test is a tool for assessing stress-coping strategy in rodents (Slattery & Cryan, Reference Slattery and Cryan2012; Commons et al., Reference Commons, Cholanians, Babb and Ehlinger2017). In this test, the duration of immobility in a forced swim scenario reflects the animal’s ability to persist under adverse conditions. Several psychedelics have been shown to decrease immobility and increase active behaviours, including swimming and climbing (Cameron et al., Reference Cameron, Benson, Dunlap and Olson2018; Hibicke et al., Reference Hibicke, Landry, Kramer, Talman and Nichols2020; Odland et al., Reference Odland, Kristensen and Andreasen2022; Rakoczy et al., Reference Rakoczy, Runge, Sen, Sandoval, Wells, Nguyen, Roberts, Sciortino, Gibbons, Friedberg, Jones and McMurray2024). Metabolic parameters and overall physical condition can be monitored through assessments such as food and water intake (to evaluate impacts on basal metabolism and caloric needs) and body condition scoring, which provides a qualitative assessment of the animal’s overall physical state by monitoring body composition and body mass index.

Figure 2. Helpful behavioural and physiological responses in rodents for inferring the physical effects of psychedelic drugs in humans.

Overall, these methods could provide preclinical evidence to elucidate the influence of psychedelics on motor, metabolic, and cardiorespiratory functions, as well as their impact on stress resilience. This knowledge could inform the design of safer and more effective clinical protocols to explore the potential benefits of psychedelics as adjunctive therapies in enhancing the mental health and physical performance of athletes and non-athletes. Such studies could also illuminate the underlying mechanisms of action, identify potential effects on organs and tissues beyond the central nervous system, and investigate potential sex differences or genetic and metabolic influences (Rakoczy et al., Reference Rakoczy, Runge, Sen, Sandoval, Wells, Nguyen, Roberts, Sciortino, Gibbons, Friedberg, Jones and McMurray2024; Werle & Bertoglio, Reference Werle and Bertoglio2024).

Acknowledgements

We thank the Brazilian Federal Government for free access to scientific articles through the Periodicals Portal of the Coordination for the Improvement of Higher Education Personnel (CAPES). Figures were created with BioRender.com.

Author contributions

  • M.A.M.P.: conception, methodology, data collection, writing, and editing of the text;

  • I.W.: data collection, writing, and editing of the text;

  • L.J.B.: conception, administration, project supervision, writing, and editing.

Funding statement

This study was supported by grants from the Conselho Nacional de Desenvolvimento Científico e Tecnológico (CNPq; 304851/2022-1; 140296/2023-9) and Fundação de Amparo à Pesquisa e Inovação do Estado Santa Catarina (FAPESC; 20/2024), which did not influence the review, collection, and interpretation of data or the decision to submit the article for publication.

Competing interests

The authors declare no conflicts of interest.

Declaration of generative AI and AI-assisted technologies in the writing process

In preparing this work, we leveraged ChatGPT and Grammarly, artificial intelligence-powered language technologies, to enhance readability, language, and style. We subsequently reviewed and edited the content for clarity and accuracy, and we take full responsibility for the final publication.

Footnotes

1 It is a psychoactive brew from the Amazon, typically made from Banisteriopsis caapi and Psychotria viridis. The β-carbolines harmine, harmaline, and tetrahydroharmine present in B. caapi act as monoamine oxidase inhibitors, preventing the first-pass metabolism of DMT found in P. viridis and allowing it to become orally active. It is also worth noting that DMT is found in various other plants worldwide.

References

Aaronson, ST, van der Vaart, A, Miller, T, LaPratt, J, Swartz, K, Shoultz, A, Lauterbach, M, Sackeim, HA and Suppes, T (2024) Single-dose synthetic psilocybin with psychotherapy for treatment-resistant bipolar type II major depressive episodes: a nonrandomized open-label trial. JAMA Psychiatry 81(6), 555562. DOI: 10.1001/jamapsychiatry.2023.4685.Google Scholar
Allen, N, Jeremiah, A, Murphy, R, Sumner, R, Forsyth, A, Hoeh, N, Menkes, DB, Evans, W, Muthukumaraswamy, S, Sundram, F and Roop, P (2024) LSD increases sleep duration the night after microdosing. Translational Psychiatry 14(1), 191. DOI: 10.1038/s41398-024-02900-4.Google Scholar
Arruda Sanchez, T, Ramos, LR, Araujo, F, Schenberg, EE, Yonamine, M, Lobo, I, de Araujo, DB and Luna, LE (2024) 2024Emotion regulation effects of ayahuasca in experienced subjects during implicit aversive stimulation: an fMRI study. Journal of Ethnopharmacology 320, 117430. DOI: 10.1016/j.jep.2023.117430.Google Scholar
Askey, T, Lasrado, R, Maiarú, M and Stephens, GJ (2024) Psilocybin as a novel treatment for chronic pain. British Journal of Pharmacology. DOI: 10.1111/bph.17420.Google Scholar
Atila, C, Straumann, I, Vizeli, P, Beck, J, Monnerat, S, Holze, F, Liechti, ME and Christ-Crain, M (2024) Oxytocin and the role of fluid restriction in MDMA-induced hyponatremia: a secondary analysis of 4 randomized clinical trials. JAMA Network Open 7(11), e2445278. DOI: 10.1001/jamanetworkopen.2024.45278.Google Scholar
Bacqué-Cazenave, J, Bharatiya, R, Barrière, G, Delbecque, JP, Bouguiyoud, N, Di Giovanni, G, Cattaert, D and De Deurwaerdère, P (2020) Serotonin in animal cognition and behavior. International Journal of Molecular Sciences 21(5), 1649. DOI: 10.3390/ijms21051649.Google Scholar
Barber, GS and Aaronson, ST (2022) The emerging field of psychedelic psychotherapy. Current Psychiatry Reports 24(10), 583590. DOI: 10.1007/s11920-022-01363-y.Google Scholar
Barrett, FS, Doss, MK, Sepeda, ND, Pekar, JJ and Griffiths, RR (2020) Emotions and brain function are altered up to one month after a single high dose of psilocybin. Scientific Reports 10(1), 2214. DOI: 10.1038/s41598-020-59282-y.Google Scholar
Bauer, ME and Teixeira, AL (2019) Inflammation in psychiatric disorders: what comes first? Annals of the New York Academy of Sciences 1437(1), 5767. DOI: 10.1111/nyas.13712.Google Scholar
Beable, SE (2024) Depressive disorders in athletes. Clinical Journal of Sport Medicine 43(1), 5370. DOI: 10.1016/j.csm.2023.06.011.Google Scholar
Berger, NJA, Best, R, Best, AW, Lane, AM, Millet, GY, Barwood, M, Marcora, S, Wilson, P and Bearden, S (2024) Limits of ultra: towards an interdisciplinary understanding of ultra-endurance running performance. Sports Medicine 54(1), 7393. DOI: 10.1007/s40279-023-01936-8.Google Scholar
Beurel, E, Toups, M and Nemeroff, CB (2020) The bidirectional relationship of depression and inflammation: double trouble. Neuron 107(2), 234256. DOI: 10.1016/j.neuron.2020.06.002.Google Scholar
Bhatt, KV and Weissman, CR (2024) The effect of psilocybin on empathy and prosocial behavior: a proposed mechanism for enduring antidepressant effects. Mental Health Research 3(1), 7. DOI: 10.1038/s44184-023-00053-8.Google Scholar
Bogenschutz, MP, Forcehimes, AA, Pommy, JA, Wilcox, CE, Barbosa, PC and Strassman, RJ (2015) Psilocybin-assisted treatment for alcohol dependence: a proof-of-concept study. Journal of Psychopharmacology 29(3), 289299. DOI: 10.1177/0269881114565144.Google Scholar
Bogenschutz, MP, Ross, S, Bhatt, S, Baron, T, Forcehimes, AA, Laska, E, Mennenga, SE, O’Donnell, K, Owens, LT, Podrebarac, S, Rotrosen, J, Tonigan, JS and Worth, L (2022) 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(10), 953962. DOI: 10.1001/jamapsychiatry.2022.2096.Google Scholar
Bomfim, JHGG (2020) Pharmaceutical care in sports. Pharmacy (Basel) 8(4), 218. DOI: 10.3390/pharmacy8040218.Google Scholar
Boxler, MI, Streun, GL, Liechti, ME, Schmid, Y, Kraemer, T and Steuer, AE (2018) Human metabolome changes after a single dose of 3,4-methylenedioxymethamphetamine (MDMA) with special focus on steroid metabolism and inflammation processes. Journal of Proteome Research 17(8), 29002907. DOI: 10.1021/acs.jproteome.8b00438.Google Scholar
Boyle, NT and Connor, TJ (2007) MDMA (“Ecstasy”) suppresses the innate IFN-gamma response in vivo: a critical role for the anti-inflammatory cytokine IL-10. European Journal of Pharmacology 572(2-3), 228238. DOI: 10.1016/j.ejphar.2007.07.020.Google Scholar
Breeksema, JJ, Niemeijer, A, Krediet, E, Karsten, T, Kamphuis, J, Vermetten, E, van den Brink, W and Schoevers, R (2024) Patient perspectives and experiences with psilocybin treatment for treatment-resistant depression: a qualitative study. Scientific Reports 14(1), 2929. DOI: 10.1038/s41598-024-53188-9.Google Scholar
Brewerton, TD, Wang, JB, Lafrance, A, Pamplin, C, Mithoefer, M, Yazar-Klosinki, B, Emerson, A and Doblin, R (2022) MDMA-assisted therapy significantly reduces eating disorder symptoms in a randomized placebo-controlled trial of adults with severe PTSD. Journal of Psychiatric Research 149, 128135. DOI: 10.1016/j.jpsychires.2022.03.008.Google Scholar
Calder, AE and Hasler, G (2023) Towards an understanding of psychedelic-induced neuroplasticity. Neuropsychopharmacology 48(1), 104112. DOI: 10.1038/s41386-022-01389-z.Google Scholar
Cameron, LP, Benetatos, J, Lewis, V, Bonniwell, EM, Jaster, AM, Moliner, R, Castrén, E, McCorvy, JD, Palner, M and Aguilar-Valles, A (2023) Beyond the 5-HT2A receptor: classic and nonclassic targets in psychedelic drug action. Journal of Neuroscience 43(45), 74727482. DOI: 10.1523/JNEUROSCI.1384-23.2023.Google Scholar
Cameron, LP, Benson, CJ, Dunlap, LE and Olson, DE (2018) Effects of N, N-dimethyltryptamine on rat behaviors relevant to anxiety and depression. ACS Chemical Neuroscience 9(7), 15821590. DOI: 10.1021/acschemneuro.8b00134.Google Scholar
Carhart-Harris, R, Giribaldi, B, Watts, R, Baker-Jones, M, Murphy-Beiner, A, Murphy, R, Martell, J, Blemings, A, Erritzoe, D and Nutt, DJ (2021) Trial of psilocybin versus escitalopram for depression. New England Journal of Medicine 384(15), 14021411. DOI: 10.1056/NEJMoa2032994.Google Scholar
Carhart-Harris, RL, Bolstridge, M, Day, CMJ, Rucker, J, Watts, R, Erritzoe, DE, Kaelen, M, Giribaldi, B, Bloomfield, M, Pilling, S, Rickard, JA, Forbes, B, Feilding, A, Taylor, D, Curran, HV and Nutt, DJ (2018) Psilocybin with psychological support for treatment-resistant depression: six-month follow-up. Psychopharmacology 235(2), 399408. DOI: 10.1007/s00213-017-4771-x.Google Scholar
Carhart-Harris, RL, Bolstridge, M, Rucker, J, Day, CM, Erritzoe, D, Kaelen, M, Bloomfield, M, Rickard, JA, Forbes, B, Feilding, A, Taylor, D, Pilling, S, Curran, VH and Nutt, DJ (2016) Psilocybin with psychological support for treatment-resistant depression: an open-label feasibility study. Lancet Psychiatry 3(7), 619627. DOI: 10.1016/S2215-0366(16)30065-7.Google Scholar
Carhart-Harris, RL, Erritzoe, D, Williams, T, Stone, JM, Reed, LJ, Colasanti, A, Tyacke, RJ, Leech, R, Malizia, AL, Murphy, K, Hobden, P, Evans, J, Feilding, A, Wise, RG and Nutt, DJ (2012) Neural correlates of the psychedelic state as determined by fMRI studies with psilocybin. National Academy of Sciences U.S.A 109(6), 21382143. DOI: 10.1073/pnas.1119598109-10.1073/pnas.1119598109.Google Scholar
Carhart-Harris, RL and Goodwin, GM (2017) The therapeutic potential of psychedelic drugs: past, present, and future. Neuropsychopharmacology 42(11), 21052113. DOI: 10.1038/npp.2017.84.Google Scholar
Carhart-Harris, RL, Muthukumaraswamy, S, Roseman, L, Kaelen, M, Droog, W, Murphy, K, Tagliazucchi, E, Schenberg, EE, Nest, T, Orban, C, Leech, R, Williams, LT, Williams, TM, Bolstridge, M, Sessa, B, McGonigle, J, Sereno, MI, Nichols, D, Hellyer, PJ, Hobden, P, Evans, J, Singh, KD, Wise, RG, Curran, HV, Feilding, A and Nutt, DJ (2016) Neural correlates of the LSD experience revealed by multimodal neuroimaging. National Academy of Sciences U.S.A 113(17), 48534858. DOI: 10.1073/pnas.1518377113.Google Scholar
Carhart-Harris, RL, Roseman, L, Bolstridge, M, Demetriou, L, Pannekoek, JN, Wall, MB, Tanner, M, Kaelen, M, McGonigle, J, Murphy, K, Leech, R, Curran, HV and Nutt, DJ (2017) Psilocybin for treatment-resistant depression: fMRI-measured brain mechanisms. Scientific Reports 7(1), 13187. DOI: 10.1038/s41598-017-13282-7.Google Scholar
Carter, RJ, Morton, J and Dunnett, SB (2001) Motor coordination and balance in rodents. Current Protocols in Neuroscience 8(8), 12. DOI: 10.1002/0471142301.ns0812s15.Google Scholar
Castro, B and Kuang, S (2017) Evaluation of muscle performance in mice by treadmill exhaustion test and whole-limb grip strength assay. Bio Protocol 7(8), e2237. DOI: 10.21769/BioProtoc.2237.Google Scholar
Cavarra, M, Falzone, A, Ramaekers, JG, Kuypers, KPC and Mento, C (2022) Psychedelic-assisted psychotherapy-A systematic review of associated psychological interventions. Frontiers of Psychology 13, 887255. DOI: 10.3389/fpsyg.2022.887255.Google Scholar
Chang, C, Putukian, M, Aerni, G, Diamond, A, Hong, G, Ingram, Y, Reardon, CL and Wolanin, A (2020) Mental health issues and psychological factors in athletes: detection, management, effect on performance and prevention: American medical society for sports medicine position statement-executive summary. British Journal of Sports Medicine 54(4), 216220. DOI: 10.1136/bjsports-2019-101583.Google Scholar
Ching, TH, Williams, MT, Wang, JB, Jerome, L, Yazar-Klosinski, B, Emerson, A and Doblin, R (2022) MDMA-assisted therapy for posttraumatic stress disorder: a pooled analysis of ethnoracial differences in efficacy and safety from two phase 2 open-label lead-in trials and a phase 3 randomized, blinded placebo-controlled trial. Journal of Psychopharmacology 36(8), 974986. DOI: 10.1177/02698811221104052.Google Scholar
Commons, KG, Cholanians, AB, Babb, JA and Ehlinger, DG (2017) The rodent forced swim test measures stress-coping strategy, not depression-like behavior. ACS Chemical Neuroscience 8(5), 955960. DOI: 10.1021/acschemneuro.7b00042.Google Scholar
Connor, TJ, Kelly, JP, McGee, M and Leonard, BE (2000) Methylenedioxymethamphetamine (MDMA; ecstasy) suppresses IL-1beta and TNF-alpha secretion following an in vivo lipopolysaccharide challenge. Life Science 67(13), 16011612. DOI: 10.1016/s0024-3205(00)00743-8.Google Scholar
Cuerva, K, Spirou, D, Cuerva, A, Delaquis, C and Raman, J (2024) Perspectives and preliminary experiences of psychedelics for the treatment of eating disorders: a systematic scoping review. European Eating Disorders Review 32(5), 9801001. DOI: 10.1002/erv.3101.Google Scholar
D’Souza, DC, Syed, SA, Flynn, LT, Safi-Aghdam, H, Cozzi, NV and Ranganathan, M (2022) Exploratory study of the dose-related safety, tolerability, and efficacy of dimethyltryptamine (DMT) in healthy volunteers and major depressive disorder. Neuropsychopharmacology 47(10), 18541862. DOI: 10.1038/s41386-022-01344-y.Google Scholar
Danforth, AL, Grob, CS, Struble, C, Feduccia, AA, Walker, N, Jerome, L, Yazar-Klosinski, B and Emerson, A (2018) Reduction in social anxiety after MDMA-assisted psychotherapy with autistic adults: a randomized, double-blind, placebo-controlled pilot study. Psychopharmacology (Berl) 235(11), 31373148. DOI: 10.1007/s00213-018-5010-9.Google Scholar
Darke, S, Duflou, J, Peacock, A, Farrell, M, Hall, W and Lappin, J (2024) A retrospective study of the characteristics and toxicology of cases of lysergic acid diethylamide (LSD)- and psilocybin-related death in Australia. Addiction 119(9), 15641571. DOI: 10.1111/add.16518.Google Scholar
Davis, AK, Barrett, FS, May, DG, Cosimano, MP, Sepeda, ND, Johnson, MW, Finan, PH and Griffiths, RR (2021) Effects of psilocybin-assisted therapy on major depressive disorder: a randomized clinical trial. JAMA Psychiatry 78(5), 481489. DOI: 10.1001/jamapsychiatry.2020.3285.Google Scholar
Daws, RE, Timmermann, C, Giribaldi, B, Sexton, JD, Wall, MB, Erritzoe, D, Roseman, L, Nutt, D and Carhart-Harris, R (2022) Increased global integration in the brain after psilocybin therapy for depression. Nature Medicine 28(4), 844851. DOI: 10.1038/s41591-022-01744-z.Google Scholar
de Almeida, RN, Galvão, ACM, da Silva, FS, Silva, EADS, Palhano-Fontes, F, Maia-de-Oliveira, JP, de Araújo, LB, Lobão-Soares, B and Galvão-Coelho, NL (2019) Modulation of serum brain-derived neurotrophic factor by a single dose of ayahuasca: observation from a randomized controlled trial. Frontiers of Psychology 10, 1234. DOI: 10.3389/fpsyg.2019.01234.Google Scholar
de Bragança, AC, Moreau, RLM, de Brito, T, Shimizu, MHM, Canale, D, de Jesus, DA, Silva, AMG, Gois, PH, Seguro, AC and Magaldi, AJ (2017) Ecstasy induces reactive oxygen species, kidney water absorption and rhabdomyolysis in normal rats. Effect of N-acetylcysteine and allopurinol in oxidative stress and muscle fiber damage. PLoS One 12(7), e0179199. DOI: 10.1371/journal.pone.0179199.Google Scholar
de Camargo, RW, Joaquim, L, Machado, RS, de Souza Ramos, S, da Rosa, LR, de Novais Junior, LR, Mathias, K, Maximiano, L, Strickert, YR, Nord, R, Gava, ML, Scarpari, E, Martins, HM, Lins, EMF, Chaves, JS, da Silva, LE, de Oliveira, MP, da Silva, MR, Fernandes, BB, Tiscoski, ADB, Piacentini, N, Santos, FP, Inserra, A, Bobinski, F, Rezin, GT, Yonamine, M, Petronilho, F and de Bitencourt, RM (2024, Ayahuasca pretreatment prevents sepsis-induced anxiety-like behavior, neuroinflammation, and oxidative stress, and increases brain-derived neurotrophic factor. Molecular neurobiology, Ayahuasca Pretreatment Prevents Sepsis-Induced Anxiety-Like Behavior, Neuroinflammation, and Oxidative Stress, and Increases Brain-Derived Neurotrophic Factor. Molecular Neurobiology. DOI: 10.1007/s12035-024-04597-4.Google Scholar
De Gregorio, D, Popic, J, Enns, JP, Inserra, A, Skalecka, A, Markopoulos, A, Posa, L, Lopez-Canul, M, He, Q, Lafferty, CK, Britt, JP, Comai, S, Aguilar-Valles, A, Sonenberg, N and Gobbi, G (2021) Lysergic acid diethylamide (LSD) promotes social behavior through mTORC1 in the excitatory neurotransmission. Proceedings of the National Academy of Sciences of the United States of America 118(5), e20207051188. DOI: 10.1073/pnas.2020705118.Google Scholar
de Vos, CMH, Mason, NL and Kuypers, KPC (2021) Psychedelics and neuroplasticity: a systematic review unraveling the biological underpinnings of psychedelics. Frontiers of Psychology 12, 724606. DOI: 10.3389/fpsyt.2021.724606.Google Scholar
Deacon, RM (2013) Measuring motor coordination in mice. Journal of Visualized Experiments 75(75), e2609. DOI: 10.3791/2609.Google Scholar
dos Santos, RG (2014) Immunological effects of ayahuasca in humans. Journal of Psychoactive Drugs 46(5), 383388. DOI: 10.1080/02791072.2014.960113.Google Scholar
Dos Santos, RG and Hallak, JEC (2024) Ayahuasca: pharmacology, safety, and therapeutic effects. CNS Spectrums 20(1), 19. DOI: 10.1017/s109285292400213x.Google Scholar
Dos Santos, RG, Osório, FL, Rocha, JM, Rossi, GN, Bouso, JC, Rodrigues, LS, de Oliveira Silveira, G, Yonamine, M and Hallak, JEC (2021) 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(5), 540550. DOI: 10.1097/JCP.0000000000001428.Google Scholar
Doss, MK, DeMarco, A, Dunsmoor, JE, Cisler, JM, Fonzo, GA and Nemeroff, CB (2024) How psychedelics modulate multiple memory mechanisms in posttraumatic stress disorder. Drugs 84(11), 14191443. DOI: 10.1007/s40265-024-02106-4.Google Scholar
Dougherty, JP, Springer, DA and Gershengorn, MC (2016) The treadmill fatigue test: a simple, high-throughput assay of fatigue-like behavior for the mouse. Journal of Visualized Experiments 31(111), 54052. DOI: 10.3791/54052.Google Scholar
Doyle, AJ, Meyer, J, Breen, K and Hunt, BJ (2020) N-methyl-3,4-methylendioxymethamphetamine (MDMA)-related coagulopathy and rhabdomyolysis: a case series and literature review. Research and Practice in Thrombosis and Haemostasis 4(5), 829834. DOI: 10.1002/rth2.12360.Google Scholar
Duarte, JA, Leão, A, Magalhães, J, Ascensão, A, Bastos, ML, Amado, FL, Vilarinho, L, Quelhas, D, Appell, HJ and Carvalho, F (2005) Strenuous exercise aggravates MDMA-induced skeletal muscle damage in mice. Toxicology 206(3), 349358. DOI: 10.1016/j.tox.2004.07.012.Google Scholar
Dumont, GJ, Sweep, FC, van der Steen, R, Hermsen, R, Donders, AR, Touw, DJ, van Gerven, JM, Buitelaar, JK and Verkes, RJ (2009) Increased oxytocin concentrations and prosocial feelings in humans after ecstasy (3,4-methylenedioxymethamphetamine) administration. Society for Neuroscience 4(4), 359366. DOI: 10.1080/17470910802649470.Google Scholar
Edwards, CD (2024) Management of mental health challenges in athletes: screening, pharmacology, and behavioral approaches. Clinical Sports Medicine 43(1), 1331. DOI: 10.1016/j.csm.2023.06.006.Google Scholar
Fadahunsi, N, Lund, J, Breum, AW, Mathiesen, CV, Larsen, IB, Knudsen, GM, Klein, AB and Clemmensen, C (2022) Acute and long-term effects of psilocybin on energy balance and feeding behavior in mice. Translational Psychiatry 12(1), 330. DOI: 10.1038/s41398-022-02103-9.Google Scholar
Family, N, Hendricks, PS, Williams, LT, Luke, D, Krediet, E, Maillet, EL and Raz, S (2022) Safety, tolerability, pharmacokinetics, and subjective effects of 50, 75, and 100 μg LSD in healthy participants within a novel intervention paradigm: a proof-of-concept study. Journal of Psychopharmacology 36(3), 321336. DOI: 10.1177/02698811211069103.Google Scholar
Flaive, A, Fougère, M, van der Zouwen, CI and Ryczko, D (2020) Serotonergic modulation of locomotor activity from basal vertebrates to mammals. Frontiers Neural Circuits 14, 590299. DOI: 10.3389/fncir.2020.590299.Google Scholar
Flanagan, TW, Billac, GB, Landry, AN, Sebastian, MN, Cormier, SA and Nichols, CD (2020) Structure-activity relationship analysis of psychedelics in a rat model of asthma reveals the anti-inflammatory pharmacophore. ACS Pharmacology & Translational Science 4(2), 488502. DOI: 10.1021/acsptsci.0c00063.Google Scholar
Flanagan, TW, Foster, TP, Galbato, TE, Lum, PY, Louie, B, Song, G, Halberstadt, AL, Billac, GB and Nichols, CD (2024) Serotonin-2 receptor agonists produce anti-inflammatory effects through functionally selective mechanisms that involve the suppression of disease-induced Arginase 1 expression. ACS Pharmacology & Translational Science 7(2), 478492. DOI: 10.1021/acsptsci.3c00297.Google Scholar
Flanagan, TW and Nichols, CD (2022) Psychedelics and anti-inflammatory activity in animal models. Current Topics in Behavioral Neurosciences 56, 229245. DOI: 10.1007/7854_2022_367.Google Scholar
Flanagan, TW, Sebastian, MN, Battaglia, DM, Foster, TP, Maillet, EL and Nichols, CD (2019a) Activation of 5-HT2 receptors reduces inflammation in vascular tissue and cholesterol levels in high-fat diet-fed apolipoprotein E knockout mice. Scientific Reports 9(1), 13444. DOI: 10.1038/s41598-019-49987-0.Google Scholar
Flanagan, TW, Sebastian, MN, Battaglia, DM, Foster, TP, Cormier, SA and Nichols, CD (2019b) 5-HT2 receptor activation alleviates airway inflammation and structural remodeling in a chronic mouse asthma model. Life Science 236, 116790. DOI: 10.1016/j.lfs.2019.116790.Google Scholar
Floris, G, Dabrowski, KR, Zanda, MT and Daws, SE (2024) Psilocybin reduces heroin seeking behavior and modulates inflammatory gene expression in the nucleus accumbens and prefrontal cortex of male rats. Molecular Psychiatry https://doi.org/10.1038/s41380-024-02788-y.Google Scholar
Freidel, N, Kreuder, L, Rabinovitch, BS, Chen, FY, Huang, RST and Lewis, EC (2024) Psychedelics, epilepsy, and seizures: a review. Frontiers of Psychology 14, 1326815. DOI: 10.3389/fphar.2023.1326815.Google Scholar
Galvão, ACM, de Almeida, RN, Silva, EADS, Freire, FAM, Palhano-Fontes, F, Onias, H, Arcoverde, E, Maia-de-Oliveira, JP, de Araújo, DB, Lobão-Soares, B and Galvão-Coelho, NL (2018) Cortisol modulation by Ayahuasca in patients with treatment resistant depression and healthy controls. Frontiers of Psychology 9, 185. DOI: 10.3389/fpsyt.2018.00185.Google Scholar
Galvão-Coelho, NL, de Menezes Galvão, AC, de Almeida, RN, Palhano-Fontes, F, Campos Braga, I, Lobão Soares, B, Maia-de-Oliveira, JP, Perkins, D, Sarris, J and de Araujo, DB (2020) Changes in inflammatory biomarkers are related to the antidepressant effects of Ayahuasca. Journal of Psychopharmacology 34(10), 11251133. DOI: 10.1177/0269881120936486.Google Scholar
Gasser, P, Holstein, D, Michel, Y, Doblin, R, Yazar-Klosinski, B, Passie, T and Brenneisen, R (2014) Safety and efficacy of lysergic acid diethylamide-assisted psychotherapy for anxiety associated with life-threatening diseases. Journal of Nervous and Mental Disease 202(7), 513520. DOI: 10.1097/NMD.0000000000000113.Google Scholar
Gasser, P, Kirchner, K and Passie, T (2015) LSD-assisted psychotherapy for anxiety associated with a life-threatening disease: a qualitative study of acute and sustained subjective effects. Journal of Psychopharmacology 29(1), 5768. DOI: 10.1177/0269881114555249.Google Scholar
Glick, ID, Stillman, MA and McDuff, D (2020) Update on integrative treatment of psychiatric symptoms and disorders in athletes. Physician and Sportsmedicine 48(4), 385391. DOI: 10.1080/00913847.2020.1757370.Google Scholar
Glick, ID, Stillman, MA, Reardon, CL and Ritvo, EC (2012) Managing psychiatric issues in elite athletes. Journal of Clinical Psychiatry 73(5), 640644. DOI: 10.4088/jcp.11r07381.Google Scholar
Golchoobian, R, Nabavizadeh, F, Roghani, M, Foroumadi, A and Mohammadian, M (2017) Alleviates MDMA-induced disturbance of serum glucose and lipids levels in the rat. Acta Medica Iranica 55(12), 736743.Google Scholar
Goodwin, GM, Aaronson, ST, Alvarez, O, Arden, PC, Baker, A, Bennett, JC, Bird, C, Blom, RE, Brennan, C, Brusch, D, Burke, L, Campbell-Coker, K, Carhart-Harris, R, Cattell, J, Daniel, A, DeBattista, C, Dunlop, BW, Eisen, K, Feifel, D, Forbes, M, Haumann, HM, Hellerstein, DJ, Hoppe, AI, Husain, MI, Jelen, LA, Kamphuis, J, Kawasaki, J, Kelly, JR, Key, RE, Kishon, R, Knatz Peck, S, Knight, G, Koolen, MHB, Lean, M, Licht, RW, Maples-Keller, JL, Mars, J, Marwood, L, McElhiney, MC, Miller, TL, Mirow, A, Mistry, S, Mletzko-Crowe, T, Modlin, LN, Nielsen, RE, Nielson, EM, Offerhaus, SR, O’Keane, V, Páleníček, T, Printz, D, Rademaker, MC, van Reemst, A, Reinholdt, F, Repantis, D, Rucker, J, Rudow, S, Ruffell, S, Rush, AJ, Schoevers, RA, Seynaeve, M, Shao, S, Soares, JC, Somers, M, Stansfield, SC, Sterling, D, Strockis, A, Tsai, J, Visser, L, Wahba, M, Williams, S, Young, AH, Ywema, P, Zisook, S and Malievskaia, E (2022) Single-dose psilocybin for a treatment-resistant episode of major depression. New England Journal of Medicine 387(18), 16371648. DOI: 10.1056/NEJMoa2206443.Google Scholar
Goodwin, GM, Croal, M, Feifel, D, Kelly, JR, Marwood, L, Mistry, S, O’Keane, V, Peck, SK, Simmons, H, Sisa, C, Stansfield, SC, Tsai, J, Williams, S and Malievskaia, E (2023) Psilocybin for treatment resistant depression in patients taking a concomitant SSRI medication. Neuropsychopharmacology 48(10), 14921499. DOI: 10.1038/s41386-023-01648-7.Google Scholar
Gorman, I, Belser, AB, Jerome, L, Hennigan, C, Shechet, B, Hamilton, S, Yazar-Klosinski, B, Emerson, A and Feduccia, AA (2020) Posttraumatic growth after MDMA-assisted psychotherapy for posttraumatic stress disorder. Journal of Traumatic Stress 33(2), 161170. DOI: 10.1002/jts.22479.Google Scholar
Goulart da Silva, M, Daros, GC, Santos, FP, Yonamine, M and de Bitencourt, RM (2022) Antidepressant and anxiolytic-like effects of ayahuasca in rats subjected to LPS-induced neuroinflammation. Behavioural Brain Research 434, 114007. DOI: 10.1016/j.bbr.2022.114007.Google Scholar
Gouttebarge, V, Castaldelli-Maia, JM, Gorczynski, P, Hainline, B, Hitchcock, ME, Kerkhoffs, GM, Rice, SM and Reardon, CL (2019) Occurrence of mental health symptoms and disorders in current and former elite athletes: a systematic review and meta-analysis. British Journal of Sports Medicine 53(11), 700706. DOI: 10.1136/bjsports-2019-100671.Google Scholar
Graham, DL, Herring, NR, Schaefer, TL, Vorhees, CV and Williams, MT (2010) Glucose and corticosterone changes in developing and adult rats following exposure to (+/-)-3,4-methylendioxymethamphetamine or 5-methoxydiisopropyltryptamine. Neurotoxicology and Teratology 32(2), 152157. DOI: 10.1016/j.ntt.2009.08.012.Google Scholar
Griffiths, RR, Johnson, MW, Carducci, MA, Umbricht, A, Richards, WA, Richards, BD, Cosimano, MP and Klinedinst, MA (2016) Psilocybin produces substantial and sustained decreases in depression and anxiety in patients with life-threatening cancer: a randomized double-blind trial. Journal of Psychopharmacology 30(12), 11811197. DOI: 10.1177/0269881116675513.Google Scholar
Griffiths, RR, Johnson, MW, Richards, WA, Richards, BD, Jesse, R, MacLean, KA, Barrett, FS, Cosimano, MP and Klinedinst, MA (2018) Psilocybin-occasioned mystical-type experience in combination with meditation and other spiritual practices produces enduring positive changes in psychological functioning and in trait measures of prosocial attitudes and behaviors. Journal of Psychopharmacology 32(1), 4969. DOI: 10.1177/0269881117731279.Google Scholar
Griffiths, RR, Johnson, MW, Richards, WA, Richards, BD, McCann, U and Jesse, R (2011) Psilocybin occasioned mystical-type experiences: immediate and persisting dose-related effects. Psychopharmacology (Berl) 218(4), 649665. DOI: 10.1007/s00213-011-2358-5.Google Scholar
Grob, CS, Danforth, AL, Chopra, GS, Hagerty, M, McKay, CR, Halberstadt, AL and Greer, GR (2011) Pilot study of psilocybin treatment for anxiety in patients with advanced-stage cancer. Archives Of General Psychiatry 68(1), 7178. DOI: 10.1001/archgenpsychiatry.2010.116.Google Scholar
Gukasyan, N, Davis, AK, Barrett, FS, Cosimano, MP, Sepeda, ND, Johnson, MW and Griffiths, RR (2022) Efficacy and safety of psilocybin-assisted treatment for major depressive disorder: prospective 12-month follow-up. Journal of Psychopharmacology 36(2), 151158. DOI: 10.1177/02698811211073759.Google Scholar
Haden, M and Woods, B (2020) LSD overdoses: three case reports. Journal of Studies on Alcohol and Drugs 81(1), 115118. DOI: 10.15288/jsad.2020.81.115.Google Scholar
Halachanova, V, Sansone, RA and McDonald, S (2001) Delayed rhabdomyolysis after ecstasy use. Mayo Clinic Proceedings 76(1), 112113. DOI: 10.4065/76.1.112.Google Scholar
Halman, A, Kong, G, Sarris, J and Perkins, D (2024) Drug-drug interactions involving classic psychedelics: a systematic review. Journal of Psychopharmacology 38(1), 318. DOI: 10.1177/02698811231211219.Google Scholar
Hamsa, TP and Kuttan, G (2010) Harmine inhibits tumour specific neo-vessel formation by regulating VEGF, MMP, TIMP and pro-inflammatory mediators both in vivo and in vitro. European Journal of Pharmacology 649(1-3), 6473. DOI: 10.1016/j.ejphar.2010.09.010.Google Scholar
He, DY, McGough, NN, Ravindranathan, A, Jeanblanc, J, Logrip, ML, Phamluong, K, Janak, PH and Ron, D (2005) Glial cell line-derived neurotrophic factor mediates the desirable actions of the anti-addiction drug ibogaine against alcohol consumption. Journal of Neurosciences 25(3), 619628. DOI: 10.1523/JNEUROSCI.3959-04.2005.Google Scholar
Heresco-Levy, U and Lerer, B (2024) Synergistic psychedelic - NMDAR modulator treatment for neuropsychiatric disorders. Molecular Psychiatry 29(1), 146152. DOI: 10.1038/s41380-023-02312-8.Google Scholar
Herr, N, Bode, C and Duerschmied, D (2017) The effects of serotonin in immune cells. Frontiers in Cardiovascular Medicine 4, 48. DOI: 10.3389/fcvm.2017.00048.Google Scholar
Hibicke, M, Landry, AN, Kramer, HM, Talman, ZK and Nichols, CD (2020) Psychedelics, but not ketamine, produce persistent antidepressant-like effects in a rodent experimental system for the study of depression. ACS Chemical Neuroscience 11(6), 864871. DOI: 10.1021/acschemneuro.9b00493.Google Scholar
Hinkle, JT, Graziosi, M, Nayak, SM and Yaden, DB (2024) Adverse events in studies of classic psychedelics: a systematic review and meta-analysis. JAMA Psychiatry 81(12), 12251235. DOI: 10.1001/jamapsychiatry.2024.2546.Google Scholar
Holze, F, Caluori, TV, Vizeli, P and Liechti, ME (2022) Safety pharmacology of acute LSD administration in healthy subjects. Psychopharmacology (Berl) 239(6), 18931905. DOI: 10.1007/s00213-021-05978-6.Google Scholar
Holze, F, Gasser, P, Müller, F, Dolder, PC and Liechti, ME (2023) Lysergic acid diethylamide-assisted therapy in patients with anxiety with and without a life-threatening illness: a randomized, double-blind, placebo-controlled phase II study. Biological Psychiatry 93(3), 215223. DOI: 10.1016/j.biopsych.2022.08.025.Google Scholar
Holze, F, Singh, N, Liechti, ME and D’Souza, DC (2024) Serotonergic psychedelics: a comparative review of efficacy, safety, pharmacokinetics, and binding profile. Biological Psychiatry: Cognitive Neuroscience and Neuroimaging 9(5), 472489. DOI: 10.1016/j.bpsc.2024.01.007.Google Scholar
Huang, J, Pham, M, Panenka, WJ, Honer, WG and Barr, AM (2022) Chronic treatment with psilocybin decreases changes in body weight in a rodent model of obesity. Frontiers in Psychiatry 13, 891512. DOI: 10.3389/fpsyt.2022.891512.Google Scholar
Hutten, NRPW, Mason, NL, Dolder, PC, Theunissen, EL, Holze, F, Liechti, ME, Feilding, A, Ramaekers, JG and Kuypers, KPC (2020a) Mood and cognition after administration of low LSD doses in healthy volunteers: a placebo controlled dose-effect finding study. European Neuropsychopharmacology 41, 8191. DOI: 10.1016/j.euroneuro.2020.10.002.Google Scholar
Hutten, NRPW, Mason, NL, Dolder, PC, Theunissen, EL, Holze, F, Liechti, ME, Varghese, N, Eckert, A, Feilding, A, Ramaekers, JG and Kuypers, KPC (2020b) Low doses of LSD acutely increase BDNF blood plasma levels in healthy volunteers. ACS Pharmacology & Translational Science 4(2), 461466. DOI: 10.1021/acsptsci.0c00099.Google Scholar
Hysek, CM, Schmid, Y, Simmler, LD, Domes, G, Heinrichs, M, Eisenegger, C, Preller, KH, Quednow, BB and Liechti, ME (2014) MDMA enhances emotional empathy and prosocial behavior. Social Cognitive and Affective Neuroscience 9(11), 16451652. DOI: 10.1093/scan/nst161.Google Scholar
Inserra, A, Campanale, A, Rezai, T, Romualdi, P and Rubino, T (2024) Epigenetic mechanisms of rapid-acting antidepressants. Translational Psychiatry 14(1), 359. DOI: 10.1038/s41398-024-03055-y.Google Scholar
Inserra, A, De Gregorio, D and Gobbi, G (2021) Psychedelics in psychiatry: neuroplastic, immunomodulatory, and neurotransmitter mechanisms. Pharmacological Reviews 73(1), 202277. DOI: 10.1124/pharmrev.120.000056.Google Scholar
Jardim, AV, Jardim, DV, Chaves, BR, Steglich, M, Ot’alora, GM, Mithoefer, MC, da Silveira, DX, Tófoli, LF, Ribeiro, S, Matthews, R, Doblin, R and Schenberg, EE (2021) 3,4-methylenedioxymethamphetamine (MDMA)-assisted psychotherapy for victims of sexual abuse with severe post-traumatic stress disorder: an open label pilot study in Brazil. Brazilian Journal of Psychiatry 43(2), 181185. DOI: 10.1590/1516-4446-2020-0980.Google Scholar
Jerome, L, Feduccia, AA, Wang, JB, Hamilton, S, Yazar-Klosinski, B, Emerson, A, Mithoefer, MC and Doblin, R (2020) Long-term follow-up outcomes of MDMA-assisted psychotherapy for treatment of PTSD: a longitudinal pooled analysis of six phase 2 trials. Psychopharmacology (Berl) 237(8), 24852497. DOI: 10.1007/s00213-020-05548-2.Google Scholar
Johansen, PØ. and Krebs, TS (2015) Psychedelics not linked to mental health problems or suicidal behavior: a population study. Journal of Psychopharmacology 29(3), 270279. DOI: 10.1177/0269881114568039.Google Scholar
Johnson, M, Richards, W and Griffiths, R (2008) Human hallucinogen research: guidelines for safety. Journal of Psychopharmacology 22(6), 603620. DOI: 10.1177/0269881108093587.Google Scholar
Johnson, MW, Garcia-Romeu, A, Cosimano, MP and Griffiths, RR (2014) Pilot study of the 5-HT2AR agonist psilocybin in the treatment of tobacco addiction. Journal of Psychopharmacology 28(11), 983992. DOI: 10.1177/0269881114548296.Google Scholar
Johnson, MW, Garcia-Romeu, A and Griffiths, RR (2017) Long-term follow-up of psilocybin-facilitated smoking cessation. American Journal of Drug and Alcohol Abuse 43(1), 5560. DOI: 10.3109/00952990.2016.1170135.Google Scholar
Johnson, MW, Griffiths, RR, Hendricks, PS and Henningfield, JE (2018) The abuse potential of medical psilocybin according to the 8 factors of the controlled substances act. Neuropharmacology 142, 143166. DOI: 10.1016/j.neuropharm.2018.05.012.Google Scholar
Johnson, MW, Hendricks, PS, Barrett, FS and Griffiths, RR (2019) Classic psychedelics: an integrative review of epidemiology, therapeutics, mystical experience, and brain network function. Pharmacology & Therapeutics 197, 83102. DOI: 10.1016/j.pharmthera.2018.11.010.Google Scholar
Kamilar-Britt, P and Bedi, G (2015) The prosocial effects of 3,4-methylenedioxymethamphetamine (MDMA): controlled studies in humans and laboratory animals. Neuroscience & Biobehavioral Reviews 57, 433446. DOI: 10.1016/j.neubiorev.2015.08.016.Google Scholar
Kim, K, Che, T, Panova, O, DiBerto, JF, Lyu, J, Krumm, BE, Wacker, D, Robertson, MJ, Seven, AB, Nichols, DE, Shoichet, BK, Skiniotis, G and Roth, BL (2020) Structure of a hallucinogen-activated gq-coupled 5-HT2A serotonin receptor. Cell 182(6), 15741588.e19. DOI: 10.1016/j.cell.2020.08.024.Google Scholar
Klaiber, A, Humbert-Droz, M, Ley, L, Schmid, Y and Liechti, ME (2024) Safety pharmacology of acute mescaline administration in healthy participants. British Journal of Clinical Pharmacology. DOI: 10.1111/bcp.16349.Google Scholar
Knudsen, GM (2023) Sustained effects of single doses of classical psychedelics in humans. Neuropsychopharmacology 48(1), 145150. DOI: 10.1038/s41386-022-01361-x.Google Scholar
Kopra, EI, Penttinen, J, Rucker, JJ and Copeland, CS (2025) Psychedelic-Related deaths in England, Wales and Northern Ireland (1997-2022). Progress in Neuro-Psychopharmacology & Biological Psychiatry 136, 111177. DOI: 10.1016/j.pnpbp.2024.111177.Google Scholar
Korthuis, PT, Hoffman, K, Wilson-Poe, AR, Luoma, JB, Bazinet, A, Pertl, K, Morgan, DL, Cook, RR, Bielavitz, S, Myers, R, Wolf, RC, McCarty, D and Stauffer, CS (2024) Developing the open psychedelic evaluation nexus consensus measures for assessment of supervised psilocybin services: an e-delphi study. Journal of Psychopharmacology 38(8), 761768. DOI: 10.1177/02698811241257839.Google Scholar
Kraehenmann, R, Preller, KH, Scheidegger, M, Pokorny, T, Bosch, OG, Seifritz, E and Vollenweider, FX (2015) Psilocybin-induced decrease in amygdala reactivity correlates with enhanced positive mood in healthy volunteers. Biological Psychiatry 78(8), 572581. DOI: 10.1016/j.biopsych.2014.04.010.Google Scholar
Laabi, S, LeMmon, C, Vogel, C, Chacon, M and Jimenez, VM Jr (2024) 2024Deciphering psilocybin: cytotoxicity, anti-inflammatory effects, and mechanistic insights. International Immunopharmacology 130, 111753. DOI: 10.1016/j.intimp.2024.111753.Google Scholar
Lake, S and Lucas, P (2024) Co-Use of psychedelics with other substances: Findings from the global psychedelic survey. Journal of Psychopharmacology 28, 2698811241292956. DOI: 10.1177/02698811241292956.Google Scholar
Lebedev, AV, Kaelen, M, Lövdén, M, Nilsson, J, Feilding, A, Nutt, DJ and Carhart-Harris, RL (2016) LSD-induced entropic brain activity predicts subsequent personality change. Human Brain Mapping 37(9), 32033213. DOI: 10.1002/hbm.23234.Google Scholar
Lehmann, ED, Thom, CH and Croft, DN (1995) Delayed severe rhabdomyolysis after taking ‘ecstasy’. Postgraduate Medical Journal 71(833), 186187. DOI: 10.1136/pgmj.71.833.186-a.Google Scholar
Lewis, EC, Jaeger, A, Girn, M, Omene, E, Brendle, M and Argento, E (2024) Exploring psychedelic-assisted therapy in the treatment of functional seizures: a review of underlying mechanisms and associated brain networks. Journal of Psychopharmacology 38(5), 407416. DOI: 10.1177/02698811241248395.Google Scholar
Liao, C, Dua, AN, Wojtasiewicz, C, Liston, C and Kwan, AC (2025) Structural neural plasticity evoked by rapid-acting antidepressant interventions. Nature Reviews Neurosciences 26(2), 101114. DOI: 10.1038/s41583-024-00876-0.Google Scholar
Lima da Cruz, RV, Moulin, TC, Petiz, LL and Leão, RN (2018) A single dose of 5-MeO-DMT stimulates cell proliferation, neuronal survivability, morphological and functional changes in adult mice ventral dentate gyrus. Frontiers in Molecular Neuroscience 11, 312. DOI: 10.3389/fnmol.2018.00312.Google Scholar
Liu, F, Wu, J, Gong, Y, Wang, P, Zhu, L, Tong, L, Chen, X, Ling, Y and Huang, C (2017a) Harmine produces antidepressant-like effects via restoration of astrocytic functions. Progress in Neuro-Psychopharmacology & Biological Psychiatry 79(Pt B), 258267. DOI: 10.1016/j.pnpbp.2017.06.012.Google Scholar
Liu, X, Li, M, Tan, S, Wang, C, Fan, S and Huang, C (2017b) Harmine is an inflammatory inhibitor through the suppression of NF-κB signaling. Biochemical and Biophysical Research Communications 489(3), 332338. DOI: 10.1016/j.bbrc.2017.05.126.Google Scholar
Low, ZXB, Ng, WS, Lim, ESY, Goh, BH and Kumari, Y (2025) The immunomodulatory effects of classical psychedelics: a systematic review of preclinical studies. Progress in Neuro-Psychopharmacology & Biological Psychiatry 136, 111139. DOI: 10.1016/j.pnpbp.2024.111139.Google Scholar
Lukasiewicz, K, Baker, JJ, Zuo, Y and Lu, J (2021) Serotonergic psychedelics in neural plasticity. Frontiers in Molecular Neuroscience 14, 748359. DOI: 10.3389/fnmol.2021.748359.Google Scholar
Luoma, JB, Chwyl, C, Bathje, GJ, Davis, AK and Lancelotta, R (2020) A meta-analysis of placebo-controlled trials of psychedelic-assisted therapy. Journal of Psychoactive Drugs 52(4), 289299. DOI: 10.1080/02791072.2020.1769878.Google Scholar
Ly, C, Greb, AC, Cameron, LP, Wong, JM, Barragan, EV, Wilson, PC, Burbach, KF, Soltanzadeh Zarandi, S, Sood, A, Paddy, MR, Duim, WC, Dennis, MY, McAllister, AK, Ori-McKenney, KM, Gray, JA and Olson, DE (2018) Psychedelics promote structural and functional neural plasticity. Cell Reports 23(11), 31703182. DOI: 10.1016/j.celrep.2018.05.022.Google Scholar
Ma, Y, Li, W, Yao, Q, Liu, Y, Yu, J, Zang, L, Wang, S, Zhou, L, Wen, S, Luo, Y, Li, W and Niu, X (2024) Harmine ameliorates CCl4-induced acute liver injury through suppression of autophagy and inflammation. International Immunopharmacology 129, 111538. DOI: 10.1016/j.intimp.2024.111538.Google Scholar
Madsen, MK, Fisher, PM, Burmester, D, Dyssegaard, A, Stenbæk, DS, Kristiansen, S, Johansen, SS, Lehel, S, Linnet, K, Svarer, C, Erritzoe, D, Ozenne, B and Knudsen, GM (2019) Psychedelic effects of psilocybin correlate with serotonin 2A receptor occupancy and plasma psilocin levels. Neuropsychopharmacology 44(7), 13281334. DOI: 10.1038/s41386-019-0324-9.Google Scholar
Marí-Sanchis, A, Burgos-Balmaseda, J and Hidalgo-Borrajo, R (2022) Eating disorders in sport. Update and proposal for an integrated approach. Endocrinology, Diabetes and Nutrition (Engl Ed) 69(2), 131143. DOI: 10.1016/j.endien.2022.02.016.Google Scholar
Marton, S, González, B, Rodríguez-Bottero, S, Miquel, E, Martínez-Palma, L, Pazos, M, Prieto, JP, Rodríguez, P, Sames, D, Seoane, G, Scorza, C, Cassina, P and Carrera, I (2019) 2019Ibogaine administration modifies GDNF and BDNF expression in brain regions involved in Mesocorticolimbic and nigral dopaminergic circuits. Frontiers in Pharmacology 10, 193. DOI: 10.3389/fphar.2019.00193.Google Scholar
Mason, NL, Kuypers, KPC, Müller, F, Reckweg, J, Tse, DHY, Toennes, SW, Hutten, NRPW, Jansen, JFA, Stiers, P, Feilding, A and Ramaekers, JG (2020) Me, myself, bye: regional alterations in glutamate and the experience of ego dissolution with psilocybin. Neuropsychopharmacology 45(12), 20032011. DOI: 10.1038/s41386-020-0718-8.Google Scholar
Mason, NL, Szabo, A, Kuypers, KPC, Mallaroni, PA, de la Torre Fornell, R, Reckweg, JT, Tse, DHY, Hutten, NRPW, Feilding, A and Ramaekers, JG (2023) Psilocybin induces acute and persisting alterations in immune status in healthy volunteers: an experimental, placebo-controlled study. Brain, Behavior, and Immunity 114, 299310. DOI: 10.1016/j.bbi.2023.09.004.Google Scholar
McClure-Begley, TD and Roth, BL (2022) The promises and perils of psychedelic pharmacology for psychiatry. Nature Reviews Drug Discovery 21(6), 463473. DOI: 10.1038/s41573-022-00421-7.Google Scholar
McDonald, C, Losty, C and MacCarthy, R (2023) An investigation of the psychological status of amateur athletes before and after a triathlon competition. European Journal of Sport Sciences 2(3), 1420. DOI: 10.24018/ejsport.2023.2.3.83.Google Scholar
McIntyre, RS, Alsuwaidan, M, Baune, BT, Berk, M, Demyttenaere, K, Goldberg, JF, Gorwood, P, Ho, R, Kasper, S, Kennedy, SH, Ly-Uson, J, Mansur, RB, McAllister-Williams, RH, Murrough, JW, Nemeroff, CB, Nierenberg, AA, Rosenblat, JD, Sanacora, G, Schatzberg, AF, Shelton, R, Stahl, SM, Trivedi, MH, Vieta, E, Vinberg, M, Williams, N, Young, AH and Maj, M (2023) Treatment-resistant depression: definition, prevalence, detection, management, and investigational interventions. World Psychiatry 22(3), 394412. DOI: 10.1002/wps.21120.Google Scholar
Melani, A, Bonaso, M, Biso, L, Zucchini, B, Conversano, C and Scarselli, M (2025) Uncovering psychedelics: from neural circuits to therapeutic applications. Pharmaceuticals (Basel) 18(1), 130. DOI: 10.3390/ph18010130.Google Scholar
Mendes, FR, Costa, CDS, Wiltenburg, VD, Morales-Lima, G, Fernandes, JB and Filev, R (2022) Classic and non-classic psychedelics for substance use disorder: a review of their historic, past and current research. Addiction Neuroscience 3, 100025. DOI: 10.1016/j.addicn.2022.100025.Google Scholar
Mertens, LJ, Wall, MB, Roseman, L, Demetriou, L, Nutt, DJ and Carhart-Harris, RL (2020) Therapeutic mechanisms of psilocybin: changes in amygdala and prefrontal functional connectivity during emotional processing after psilocybin for treatment-resistant depression. Journal of Psychopharmacology 34(2), 167180. DOI: 10.1177/0269881119895520.Google Scholar
Mitchell, JM and Anderson, BT (2024) Psychedelic therapies reconsidered: compounds, clinical indications, and cautious optimism. Neuropsychopharmacology 49(1), 96103. DOI: 10.1038/s41386-023-01656-7.Google Scholar
Mitchell, JM, Bogenschutz, M, Lilienstein, A, Harrison, C, Kleiman, S, Parker-Guilbert, K, Ot’alora, GM, Garas, W, Paleos, C, Gorman, I, Nicholas, C, Mithoefer, M, Carlin, S, Poulter, B, Mithoefer, A, Quevedo, S, Wells, G, Klaire, SS, van der Kolk, B, Tzarfaty, K, Amiaz, R, Worthy, R, Shannon, S, Woolley, JD, Marta, C, Gelfand, Y, Hapke, E, Amar, S, Wallach, Y, Brown, R, Hamilton, S, Wang, JB, Coker, A, Matthews, R, de Boer, A, Yazar-Klosinski, B, Emerson, A and Doblin, R (2021) MDMA-assisted therapy for severe PTSD: a randomized, double-blind, placebo-controlled phase 3 study. Nature Medicine 27(6), 10251033. DOI: 10.1038/s41591-021-01336-3.Google Scholar
Mitchell, JM, Ot’alora, GM, van der Kolk, B, Shannon, S, Bogenschutz, M, Gelfand, Y, Paleos, C, Nicholas, CR, Quevedo, S, Balliett, B, Hamilton, S, Mithoefer, M, Kleiman, S, Parker-Guilbert, K, Tzarfaty, K, Harrison, C, de Boer, A, Doblin, R Yazar-Klosinski, B and MAPP2 Study Collaborator Group (2023) MDMA-assisted therapy for moderate to severe PTSD: a randomized, placebo-controlled phase 3 trial. Nature Medicine 29(10), 24732480. DOI: 10.1038/s41591-023-02565-4 .Google Scholar
Mithoefer, MC, Feduccia, AA, Jerome, L, Mithoefer, A, Wagner, M, Walsh, Z, Hamilton, S, Yazar-Klosinski, B, Emerson, A and Doblin, R (2019) MDMA-assisted psychotherapy for treatment of PTSD: study design and rationale for phase 3 trials based on pooled analysis of six phase 2 randomized controlled trials. Psychopharmacology (Berl) 236(9), 27352745. DOI: 10.1007/s00213-019-05249-5.Google Scholar
Mithoefer, MC, Mithoefer, AT, Feduccia, AA, Jerome, L, Wagner, M, Wymer, J, Holland, J, Hamilton, S, Yazar-Klosinski, B, Emerson, A and Doblin, R (2018) 3,4-methylenedioxymethamphetamine (MDMA)-assisted psychotherapy for post-traumatic stress disorder in military veterans, firefighters, and police officers: a randomised, double-blind, dose-response, phase 2 clinical trial. Lancet Psychiatry 5(6), 486497. DOI: 10.1016/S2215-0366(18)30135-4.Google Scholar
Mithoefer, MC, Wagner, MT, Mithoefer, AT, Jerome, L and Doblin, R (2011) The safety and efficacy of {+/-}3,4-methylenedioxymethamphetamine-assisted psychotherapy in subjects with chronic, treatment-resistant posttraumatic stress disorder: the first randomized controlled pilot study. Journal of Psychopharmacology 25(4), 439452. DOI: 10.1177/0269881110378371.Google Scholar
Mithoefer, MC, Wagner, MT, Mithoefer, AT, Jerome, L, Martin, SF, Yazar-Klosinski, B, Michel, Y, Brewerton, TD and Doblin, R (2013) Durability of improvement in post-traumatic stress disorder symptoms and absence of harmful effects or drug dependency after 3,4-methylenedioxymethamphetamine-assisted psychotherapy: a prospective long-term follow-up study. Journal of Psychopharmacology 27(1), 2839. DOI: 10.1177/0269881112456611.Google Scholar
Moliner, R, Girych, M, Brunello, CA, Kovaleva, V, Biojone, C, Enkavi, G, Antenucci, L, Kot, EF, Goncharuk, SA, Kaurinkoski, K, Kuutti, M, Fred, SM, Elsilä, LV, Sakson, S, Cannarozzo, C, Diniz, CRAF, Seiffert, N, Rubiolo, A, Haapaniemi, H, Meshi, E, Nagaeva, E, Öhman, T, Róg, T, Kankuri, E, Vilar, M, Varjosalo, M, Korpi, ER, Permi, P, Mineev, KS, Saarma, M, Vattulainen, I, Casarotto, PC and Castrén, E (2023) Psychedelics promote plasticity by directly binding to BDNF receptor trkB. Nature Neuroscience 26(6), 10321041. DOI: 10.1038/s41593-023-01316-5.Google Scholar
Morales-Garcia, JA, Calleja-Conde, J, Lopez-Moreno, JA, Alonso-Gil, S, Sanz-SanCristobal, M, Riba, J and Perez-Castillo, A (2020) N, N-dimethyltryptamine compound found in the hallucinogenic tea ayahuasca, regulates adult neurogenesis in vitro and in vivo. Translational Psychiatry 10(1), 331. DOI: 10.1038/s41398-020-01011-0.Google Scholar
Morris, AD (2015) Improving pharmaceutical care for athletes: a proposed assessment tool and useful resources. Canadian Pharmacists Journal 148(6), 305307. DOI: 10.1177/1715163515607309.Google Scholar
Munier, JJ, Pank, JT, Severino, A, Wang, H, Zhang, P, Vergnes, L and Reue, K (2022) Simultaneous monitoring of mouse grip strength, force profile, and cumulative force profile distinguishes muscle physiology following surgical, pharmacologic and diet interventions. Scientific Reports 12(1), 16428. DOI: 10.1038/s41598-022-20665-y.Google Scholar
Murphy, RJ, Muthukumaraswamy, S and de Wit, H (2024) Microdosing psychedelics: current evidence from controlled studies. Biological Psychiatry: Cognitive Neuroscience and Neuroimaging 9(5), 500511. DOI: 10.1016/j.bpsc.2024.01.002.Google Scholar
Nardai, S, László, M, Szabó, A, Alpár, A, Hanics, J, Zahola, P, Merkely, B, Frecska, E and Nagy, Z (2020) N, N-dimethyltryptamine reduces infarct size and improves functional recovery following transient focal brain ischemia in rats. Experimental Neurology 327, 113245. DOI: 10.1016/j.expneurol.2020.113245.Google Scholar
Nau, F Jr, Miller, J, Saravia, J, Ahlert, T, Yu, B, Happel, KI, Cormier, SA and Nichols, CD (2015) Serotonin 5-HT2 receptor activation prevents allergic asthma in a mouse model. American Journal of Physiology-Lung Cellular and Molecular Physiology 308(2), L1918. DOI: 10.1152/ajplung.00138.2013.Google Scholar
Nau, F Jr, Yu, B, Martin, D and Nichols, CD (2013) Serotonin 5-HT2A receptor activation blocks TNF-α mediated inflammation in vivo. PLoS One 8(10), e75426. DOI: 10.1371/journal.pone.0075426.Google Scholar
Neumann, J, Dhein, S, Kirchhefer, U, Hofmann, B and Gergs, U (2024) Effects of hallucinogenic drugs on the human heart. Frontiers in Pharmacology 15, 1334218. DOI: 10.3389/fphar.2024.1334218.Google Scholar
Nicholas, CR, Wang, JB, Coker, A, Mitchell, JM, Klaire, SS, Yazar-Klosinski, B, Emerson, A, Brown, RT and Doblin, R (2022) The effects of MDMA-assisted therapy on alcohol and substance use in a phase 3 trial for treatment of severe PTSD. Drug and Alcohol Dependence 233, 109356. DOI: 10.1016/j.drugalcdep.2022.109356.Google Scholar
Nichols, DE (2016) Psychedelics. Pharmacological Reviews 68(2), 264355. DOI: 10.1124/pr.115.011478.Google Scholar
Niu, X, Yao, Q, Li, W, Zang, L, Li, W, Zhao, J, Liu, F and Zhi, W (2019) Harmine mitigates LPS-induced acute kidney injury through inhibition of the TLR4-NF-κB/NLRP3 inflammasome signalling pathway in mice. European Journal of Pharmacology 849, 160169. DOI: 10.1016/j.ejphar.2019.01.062.Google Scholar
Nkadimeng, SM, Steinmann, CML and Eloff, JN (2021) 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, 37293738. DOI: 10.2147/JIR.S317182.Google Scholar
Nutt, D and Carhart-Harris, R (2021) The current status of psychedelics in psychiatry. JAMA Psychiatry 78(2), 121122. DOI: 10.1001/jamapsychiatry.2020.2171.Google Scholar
Nutt, D, Crome, I and Young, AH (2024) Is it now time to prepare psychiatry for a psychedelic future? British Journal of Psychiatry 225(2), 308310. DOI: 10.1192/bjp.2024.76.Google Scholar
Odland, AU, Kristensen, JL and Andreasen, JT (2022) Animal behavior in psychedelic research. Pharmacological Reviews 74(4), 11761205. DOI: 10.1124/pharmrev.122.000590.Google Scholar
Oehen, P, Traber, R, Widmer, V and Schnyder, U (2013) A randomized, controlled pilot study of MDMA (± 3,4-methylenedioxymethamphetamine)-assisted psychotherapy for treatment of resistant, chronic post-traumatic stress disorder (PTSD). Journal of Psychopharmacology 27(1), 4052. DOI: 10.1177/0269881112464827.Google Scholar
Olson, DE (2022) Biochemical mechanisms underlying psychedelic-induced neuroplasticity. Biochemistry 61(3), 127136. DOI: 10.1021/acs.biochem.1c00812.Google Scholar
Osmond, H (1957) A review of the clinical effects of psychotomimetic agents. Annals of the New York Academy of Sciences 66(3), 418434. DOI: 10.1111/j.1749-6632.1957.tb40738.x.Google Scholar
Ot’alora, GM, Grigsby, J, Poulter, B, Van Derveer, JW 3rd, Giron, SG, Jerome, L, Feduccia, AA, Hamilton, S, Yazar-Klosinski, B, Emerson, A, Mithoefer, MC and Doblin, R (2018) 3,4-methylenedioxymethamphetamine-assisted psychotherapy for treatment of chronic posttraumatic stress disorder: a randomized phase 2 controlled trial. Journal of Psychopharmacology 32(12), 12951307. DOI: 10.1177/0269881118806297.Google Scholar
Pagni, BA, Petridis, PD, Podrebarac, SK, Grinband, J, Claus, ED and Bogenschutz, MP (2024) Psilocybin-induced changes in neural reactivity to alcohol and emotional cues in patients with alcohol use disorder: an fMRI pilot study. Scientific Reports 14(1), 3159. DOI: 10.1038/s41598-024-52967-8.Google Scholar
Palhano-Fontes, F, Andrade, KC, Tofoli, LF, Santos, AC, Crippa, JA, Hallak, JE, Ribeiro, S and de Araujo, DB (2015) The psychedelic state induced by ayahuasca modulates the activity and connectivity of the default mode network. PLoS One 10(2), e0118143. DOI: 10.1371/journal.pone.0118143.Google Scholar
Palhano-Fontes, F, Barreto, D, Onias, H, Andrade, KC, Novaes, MM, Pessoa, JA, Mota-Rolim, SA, Osório, FL, Sanches, R, Dos Santos, RG, Tófoli, LF, de Oliveira Silveira, G, Yonamine, M, Riba, J, Santos, FR, Silva-Junior, AA, Alchieri, JC, Galvão-Coelho, NL, Lobão-Soares, B, Hallak, JEC, Arcoverde, E, Maia-de-Oliveira, JP and Araújo, DB (2019) Rapid antidepressant effects of the psychedelic ayahuasca in treatment-resistant depression: a randomized placebo-controlled trial. Psychological Medicine 49(4), 655663. DOI: 10.1017/S0033291718001356.Google Scholar
Peck, SK, Shao, S, Gruen, T, Yang, K, Babakanian, A, Trim, J, Finn, DM and Kaye, WH (2023) Psilocybin therapy for females with anorexia nervosa: a phase 1, open-label feasibility study. Nature Medicine 29(8), 19471953. DOI: 10.1038/s41591-023-02455-9.Google Scholar
Peill, J, Marguilho, M, Erritzoe, D, Barba, T, Greenway, KT, Rosas, F, Timmermann, C and Carhart-Harris, R (2024) Psychedelics and the ‘inner healer’: myth or mechanism? Journal of Psychopharmacology 38(5), 417424. DOI: 10.1177/02698811241239206.Google Scholar
Perkins, D, Pagni, BA, Sarris, J, Barbosa, PCR and Chenhall, R (2022) Changes in mental health, wellbeing and personality following ayahuasca consumption: results of a naturalistic longitudinal study. Frontiers in Pharmacology 13, 884703. DOI: 10.3389/fphar.2022.884703.Google Scholar
Polito, V and Stevenson, RJ (2019) A systematic study of microdosing psychedelics. PLoS One 14(2), e0211023. DOI: 10.1371/journal.pone.0211023.Google Scholar
Ponte, L, Jerome, L, Hamilton, S, Mithoefer, MC, Yazar-Klosinski, BB, Vermetten, E and Feduccia, AA (2021) Sleep quality improvements after MDMA-assisted psychotherapy for the treatment of posttraumatic stress disorder. Journal of Traumatic Stress 34(4), 851863. DOI: 10.1002/jts.22696.Google Scholar
Preller, KH, Burt, JB, Ji, JL, Schleifer, CH, Adkinson, BD, Stämpfli, P, Seifritz, E, Repovs, G, Krystal, JH, Murray, JD, Vollenweider, FX and Anticevic, A (2018) Changes in global and thalamic brain connectivity in LSD-induced altered states of consciousness are attributable to the 5-HT2A receptor. Elife 7, e35082. DOI: 10.7554/eLife.35082.Google Scholar
Preller, KH, Duerler, P, Burt, JB, Ji, JL, Adkinson, B, Stämpfli, P, Seifritz, E, Repovš, G, Krystal, JH, Murray, JD, Anticevic, A and Vollenweider, FX (2020) Psilocybin induces time-dependent changes in global functional connectivity. Biological Psychiatry 88(2), 197207. DOI: 10.1016/j.biopsych.2019.12.027.Google Scholar
Rakoczy, RJ, Runge, GN, Sen, AK, Sandoval, O, Wells, HG, Nguyen, Q, Roberts, BR, Sciortino, JH, Gibbons, WJ Jr, Friedberg, LM, Jones, JA and McMurray, MS (2024) Pharmacological and behavioural effects of tryptamines present in psilocybin-containing mushrooms. British Journal of Pharmacology 181(19), 36273641. DOI: 10.1111/bph.16466.Google Scholar
Ramaekers, JG, Hutten, N, Mason, NL, Dolder, P, Theunissen, EL, Holze, F, Liechti, ME, Feilding, A and Kuypers, KP (2021) A low dose of lysergic acid diethylamide decreases pain perception in healthy volunteers. Journal of Psychopharmacology 35(4), 398405. DOI: 10.1177/0269881120940937.Google Scholar
Raote, I, Bhattacharya, A and Panicker, MM (2007) Serotonin 2A (5-HT2A) receptor function: ligand-dependent mechanisms and pathways. In Chattopadhyay, A (ed), Serotonin receptors in neurobiology. Boca Raton (FL): CRC Press/Taylor & Francis, Chapter 6, https://www.ncbi.nlm.nih.gov/books/NBK1853/ Google Scholar
Ray, TS (2010) Psychedelics and the human receptorome. PLoS One 5(2), e9019. DOI: 10.1371/journal.pone.0009019.Google Scholar
Reardon, CL (2016) The sports psychiatrist and psychiatric medication. International Review of Psychiatry 28(6), 606613. DOI: 10.1080/09540261.2016.1190691.Google Scholar
Reardon, CL and Creado, S (2016) Psychiatric medication preferences of sports psychiatrists. Physician and Sportsmedicine 44(4), 397402. DOI: 10.1080/00913847.2016.1216719.Google Scholar
Reardon, CL, Hainline, B, Aron, CM, Baron, D, Baum, AL, Bindra, A, Budgett, R, Campriani, N, Castaldelli-Maia, JM, Currie, A, Derevensky, JL, Glick, ID, Gorczynski, P, Gouttebarge, V, Grandner, MA, Han, DH, McDuff, D, Mountjoy, M, Polat, A, Purcell, R, Putukian, M, Rice, S, Sills, A, Stull, T, Swartz, L, Zhu, LJ and Engebretsen, L (2019) Mental health in elite athletes: International Olympic Committee consensus statement (2019). British Journal of Sports Medicine 53(11), 667699. DOI: 10.1136/bjsports-2019-100715.Google Scholar
Reiff, CM, Richman, EE, Nemeroff, CB, Carpenter, LL, Widge, AS, Rodriguez, CI, Kalin, NH and McDonald, WM (2020) The work group on biomarkers and novel treatments, a division of the American Psychiatric Association Council of Research, 2020 psychedelics and psychedelic-assisted psychotherapy. American Journal of Psychiatry 177(5), 391410. DOI: 10.1176/appi.ajp.2019.19010035.Google Scholar
Rhee, TG, Davoudian, PA, Sanacora, G and Wilkinson, ST (2023) Psychedelic renaissance: revitalized potential therapies for psychiatric disorders. Drug Discovery Today 28(12), 103818. DOI: 10.1016/j.drudis.2023.103818.Google Scholar
Riba, J, Valle, M, Urbano, G, Yritia, M, Morte, A and Barbanoj, MJ (2003) Human pharmacology of ayahuasca: subjective and cardiovascular effects, monoamine metabolite excretion, and pharmacokinetics. Journal of Pharmacology and Experimental Therapeutics 306(1), 7383. DOI: 10.1124/jpet.103.049882.Google Scholar
Romeo, B, Kervadec, E, Fauvel, B, Strika-Bruneau, L, Amirouche, A, Verroust, V, Piolino, P and Benyamina, A (2024) Safety and risk assessment of psychedelic psychotherapy: a meta-analysis and systematic review. Psychiatry Research 335, 115880. DOI: 10.1016/j.psychres.2024.115880.Google Scholar
Roseman, L, Demetriou, L, Wall, MB, Nutt, DJ and Carhart-Harris, RL (2018) Increased amygdala responses to emotional faces after psilocybin for treatment-resistant depression. Neuropharmacology 142, 263269. DOI: 10.1016/j.neuropharm.2017.12.041.Google Scholar
Ross, S, Bossis, A, Guss, J, Agin-Liebes, G, Malone, T, Cohen, B, Mennenga, SE, Belser, A, Kalliontzi, K, Babb, J, Su, Z, Corby, P and Schmidt, BL (2016) Rapid and sustained symptom reduction following psilocybin treatment for anxiety and depression in patients with life-threatening cancer: a randomized controlled trial. Journal of Psychopharmacology 30(12), 11651180. DOI: 10.1177/0269881116675512.Google Scholar
Rusyniak, DE, Tandy, SL, Hekmatyar, SK, Mills, E, Smith, DJ, Bansal, N, MacLellan, D, Harper, ME and Sprague, JE (2005) The role of mitochondrial uncoupling in 3,4-methylenedioxymethamphetamine-mediated skeletal muscle hyperthermia and rhabdomyolysis. Journal of Pharmacology and Experimental Therapeutics 313(2), 629639. DOI: 10.1124/jpet.104.079236.Google Scholar
Sabé, M, Sulstarova, A, Glangetas, A, De Pieri, M, Mallet, L, Curtis, L, Richard-Lepouriel, H, Penzenstadler, L, Seragnoli, F, Thorens, G, Zullino, D, Preller, K, Böge, K, Leucht, S, Correll, CU, Solmi, M, Kaiser, S and Kirschner, M (2025) Reconsidering evidence for psychedelic-induced psychosis: an overview of reviews, a systematic review, and meta-analysis of human studies. Molecular Psychiatry 30(3), 12231255. DOI:10.1038/s41380-024-02800-5.Google Scholar
Sanches, RF, de Lima Osório, F, Dos Santos, RG, Macedo, LR, Maia-de-Oliveira, JP, Wichert-Ana, L, de Araujo, DB, Riba, J, Crippa, JA and Hallak, JE (2016) Antidepressant effects of a single dose of ayahuasca in patients with recurrent depression: a SPECT study. Journal of Clinical Psychopharmacology 36(1), 7781. DOI: 10.1097/JCP.0000000000000436.Google Scholar
Sarparast, A, Thomas, K, Malcolm, B and Stauffer, CS (2022) Drug-drug interactions between psychiatric medications and MDMA or psilocybin: a systematic review. Psychopharmacology (Berl) 239(6), 19451976. DOI: 10.1007/s00213-022-06083-y.Google Scholar
Schlag, AK, Aday, J, Salam, I, Neill, JC and Nutt, DJ (2022) Adverse effects of psychedelics: from anecdotes and misinformation to systematic science. Journal of Psychopharmacology 36(3), 258272. DOI: 10.1177/02698811211069100.Google Scholar
Schmid, Y and Liechti, ME (2018) Long-lasting subjective effects of LSD in normal subjects. Psychopharmacology (Berl) 235(2), 535545. DOI: 10.1007/s00213-017-4733-3.Google Scholar
Schneier, FR, Feusner, J, Wheaton, MG, Gomez, GJ, Cornejo, G, Naraindas, AM and Hellerstein, DJ (2023) Pilot study of single-dose psilocybin for serotonin reuptake inhibitor-resistant body dysmorphic disorder. Journal of Psychiatric Research 161, 364370. DOI: 10.1016/j.jpsychires.2023.03.031.Google Scholar
Screaton, GR, Singer, M, Cairns, HS, Thrasher, A, Sarner, M and Cohen, SL (1992) Hyperpyrexia and rhabdomyolysis after MDMA (“ecstasy”) abuse. Lancet 339(8794), 677678. DOI: 10.1016/0140-6736(92)90834-p.Google Scholar
Shafiee, A, Arabzadeh Bahri, R, Rafiei, MA, Esmaeilpur Abianeh, F, Razmara, P, Jafarabady, K and Amini, MJ (2024) The effect of psychedelics on the level of brain-derived neurotrophic factor: a systematic review and meta-analysis. Journal of Psychopharmacology 38(5), 425431. DOI: 10.1177/02698811241234247.Google Scholar
Shahraki, MR and Irani, M (2014) The effects of ecstasy on liver function tests, blood glucose, and lipids profile of male rats. International Journal of High Risk Behaviors and Addiction 3(4), e21076. DOI: 10.5812/ijhrba.21076.Google Scholar
Shakir, J, Pedicini, M, Bullock, BC, Hoen, PW, Macias, LK, Freiman, J, Pletnikov, MV, Tamashiro, KLK and Cordner, ZA (2024) Effects of psilocybin on body weight, body composition, and metabolites in male and female mice. Physiology & Behavior 284, 114627. DOI: 10.1016/j.physbeh.2024.114627.Google Scholar
Shan, J, Khelashvili, G, Mondal, S, Mehler, EL and Weinstein, H (2012) Ligand-dependent conformations and dynamics of the serotonin 5-HT(2A) receptor determine its activation and membrane-driven oligomerization properties. PLoS Computational Biology 8(4), e1002473. DOI: 10.1371/journal.pcbi.1002473.Google Scholar
Shao, LX, Liao, C, Gregg, I, Davoudian, PA, Savalia, NK, Delagarza, K and Kwan, AC (2021) Psilocybin induces rapid and persistent growth of dendritic spines in frontal cortex in vivo. Neuron 109(16), 25352544.e4. DOI: 10.1016/j.neuron.2021.06.008.Google Scholar
Shen, L, Lv, X, Yang, X, Deng, S, Liu, L, Zhou, J, Zhu, Y and Ma, H (2022) Bufotenines-loaded liposome exerts anti-inflammatory, analgesic effects and reduce gastrointestinal toxicity through altering lipid and bufotenines metabolism. Biomedicine & Pharmacotherapy 153, 113492. DOI: 10.1016/j.biopha.2022.113492.Google Scholar
Siegel, JS, Subramanian, S, Perry, D, Kay, BP, Gordon, EM, Laumann, TO, Reneau, TR, Metcalf, NV, Chacko, RV, Gratton, C, Horan, C, Krimmel, SR, Shimony, JS, Schweiger, JA, Wong, DF, Bender, DA, Scheidter, KM, Whiting, FI, Padawer-Curry, JA, Shinohara, RT, Chen, Y, Moser, J, Yacoub, E, Nelson, SM, Vizioli, L, Fair, DA, Lenze, EJ, Carhart-Harris, R, Raison, CL, Raichle, ME, Snyder, AZ, Nicol, GE and Dosenbach, NUF (2024) Psilocybin desynchronizes the human brain. Nature 632(8023), 131138. DOI: 10.1038/s41586-024-07624-5.Google Scholar
Simon, MW, Olsen, HA, Hoyte, CO, Black, JC, Reynolds, KM, Dart, RC and Monte, AA (2024) Clinical effects of psychedelic substances reported to United States poison centers: 2012 to 2022. Annals of Emergency Medicine 84(6), 605618. DOI: 10.1016/j.annemergmed.2024.06.025.Google Scholar
Simonsson, O, Osika, W, Carhart-Harris, R and Hendricks, PS (2021) Associations between lifetime classic psychedelic use and cardiometabolic diseases. Scientific Reports 11(1), 14427. DOI: 10.1038/s41598-021-93787-4.Google Scholar
Singleton, SP, Wang, JB, Mithoefer, M, Hanlon, C, George, MS, Mithoefer, A, Mithoefer, O, Coker, AR, Yazar-Klosinski, B, Emerson, A, Doblin, R and Kuceyeski, A (2023) Altered brain activity and functional connectivity after MDMA-assisted therapy for post-traumatic stress disorder. Frontiers in Psychiatry 13, 947622. DOI: 10.3389/fpsyt.2022.947622.Google Scholar
Skosnik, PD, Sloshower, J, Safi-Aghdam, H, Pathania, S, Syed, S, Pittman, B and D’Souza, DC (2023) Sub-acute effects of psilocybin on EEG correlates of neural plasticity in major depression: relationship to symptoms. Journal of Psychopharmacology 37(7), 687697. DOI: 10.1177/02698811231179800.Google Scholar
Slattery, DA and Cryan, JF (2012) Using the rat forced swim test to assess antidepressant-like activity in rodents. Nature Protocols 7(6), 10091014. DOI: 10.1038/nprot.2012.044.Google Scholar
Sloshower, J, Skosnik, PD, Safi-Aghdam, H, Pathania, S, Syed, S, Pittman, B and D’Souza, DC (2023) Psilocybin-assisted therapy for major depressive disorder: an exploratory placebo-controlled, fixed-order trial. Journal of Psychopharmacology 37(7), 698706. DOI: 10.1177/02698811231154852.Google Scholar
Sloshower, J, Zeifman, RJ, Guss, J, Krause, R, Safi-Aghdam, H, Pathania, S, Pittman, B and D’Souza, DC (2024) Psychological flexibility as a mechanism of change in psilocybin-assisted therapy for major depression: results from an exploratory placebo-controlled trial. Scientific Reports 14(1), 8833. DOI: 10.1038/s41598-024-58318-x.Google Scholar
Smith, A, Buadze, A, Colangelo, J and Liebrenz, M (2023) A review of mental health issues in high-performance and elite-level cycling. International Journal of Sports Medicine 44(14), 10341042. DOI: 10.1055/a-2145-6355.Google Scholar
Soto-Angona, Ó., Fortea, A, Fortea, L, Martínez-Ramírez, M, Santamarina, E, López, FJG, Knudsen, GM and Ona, G (2024) Do classic psychedelics increase the risk of seizures? A scoping review. European Neuropsychopharmacology 85, 3542. DOI: 10.1016/j.euroneuro.2024.05.002.Google Scholar
Soto-Montenegro, ML, Vaquero, JJ, Arango, C, Ricaurte, G, García-Barreno, P and Desco, M (2007) Effects of MDMA on blood glucose levels and brain glucose metabolism. European Journal of Nuclear Medicine and Molecular Imaging 34(6), 916925. DOI: 10.1007/s00259-006-0262-8.Google Scholar
Sprague, JE, Brutcher, RE, Mills, EM, Caden, D and Rusyniak, DE (2004) Attenuation of 3,4-methylenedioxymethamphetamine (MDMA, ecstasy)-induced rhabdomyolysis with alpha1- plus beta3-adrenoreceptor antagonists. British Journal of Pharmacology 142(4), 667670. DOI: 10.1038/sj.bjp.0705823.Google Scholar
Sprague, JE, Moze, P, Caden, D, Rusyniak, DE, Holmes, C, Goldstein, DS and Mills, EM (2005) Carvedilol reverses hyperthermia and attenuates rhabdomyolysis induced by 3,4-methylenedioxymethamphetamine (MDMA, ecstasy) in an animal model. Critical Care Medicine 33(6), 13111316. DOI: 10.1097/01.ccm.0000165969.29002.70.Google Scholar
Stankevicius, D, Ferraz-de-Paula, V, Ribeiro, A, Pinheiro, ML, Ligeiro de Oliveira, AP, Damazo, AS, Lapachinske, SF, Moreau, RL, Tavares de Lima, W and Palermo-Neto, J (2012) 3,4-methylenedioxymethamphetamine (ecstasy) decreases inflammation and airway reactivity in a murine model of asthma. Neuroimmunomodulation 19(4), 209219. DOI: 10.1159/000334098.Google Scholar
Stoliker, D, Novelli, L, Vollenweider, FX, Egan, GF, Preller, KH and Razi, A (2024) Neural mechanisms of resting-state networks and the amygdala underlying the cognitive and emotional effects of psilocybin. Biological Psychiatry 96(1), 5766. DOI: 10.1016/j.biopsych.2024.01.002.Google Scholar
Stone, MB, Yaseen, ZS, Miller, BJ, Richardville, K, Kalaria, SN and Kirsch, I (2022) Response to acute monotherapy for major depressive disorder in randomized, placebo controlled trials submitted to the US Food and Drug Administration: individual participant data analysis. British Medical Journal 378, e067606. DOI: 10.1136/bmj-2021-067606.Google Scholar
Strand, NH, Whitney, M, Johnson, B, Dunn, T, Attanti, S, Maloney, J, Misra, L, Gomez, D, Viswanath, O, Emami, E and Leathem, J (2025) Pain and perception: exploring psychedelics as novel therapeutic agents in chronic pain management. Current Pain and Headache Reports 29(1), 15. DOI: 10.1007/s11916-024-01353-0.Google Scholar
Sue, YM, Lee, YL and Huang, JJ (2002) Acute hyponatremia, seizure, and rhabdomyolysis after ecstasy use. Journal of Toxicology: Clinical Toxicology 40(7), 931932. DOI: 10.1081/clt-120016964.Google Scholar
Sun, J, Yang, X, Zhang, Y, Zhang, W, Lu, J, Hu, Q, Liu, R, Zhou, C and Chen, C (2019) Salvinorin a attenuates early brain injury through PI3K/Akt pathway after subarachnoid hemorrhage in rat. Brain Research 1719, 6470. DOI: 10.1016/j.brainres.2019.05.026.Google Scholar
Tabaa, MME, Tabaa, MME, Rashad, E, Elballal, MS, and Elazazy, O (2024) Harmine alleviated STZ-induced rat diabetic nephropathy: a potential role via regulating AMPK/Nrf2 pathway and deactivating ataxia-telangiectasia mutated (ATM) signaling. International Immunopharmacology 132, 111954. DOI: 10.1016/j.intimp.2024.111954.Google Scholar
Thomas, K (2024) Toxicology and pharmacological interactions of classic psychedelics, Current topics in behavioral Neurosciences. https://doi.org/10.1007/7854_2024_508.Google Scholar
Thompson, C and Szabo, A (2020) Psychedelics as a novel approach to treating autoimmune conditions. Immunology Letters 228, 4554. DOI: 10.1016/j.imlet.2020.10.001.Google Scholar
Thuany, M, Viljoen, C, Gomes, TN, Knechtle, B and Scheer, V (2023) Mental health in ultra-endurance runners: a systematic review. Sports Medicine 53(10), 18911904. DOI: 10.1007/s40279-023-01890-5.Google Scholar
Tso, JV and Pelliccia, A (2022) Psychiatric medications and cardiovascular performance: uncommon depressing side effects. Journals of the American College of Cardiology Case Reports 4(20), 13411343. DOI: 10.1016/j.jaccas.2022.08.004.Google Scholar
Vamvakopoulou, IA and Nutt, DJ (2024) Psychedelics: from cave art to 21st-century medicine for addiction. European Addiction Research 30(5), 302320. DOI: 10.1159/000540062.Google Scholar
van der Kolk, BA, Wang, JB, Yehuda, R, Bedrosian, L, Coker, AR, Harrison, C, Mithoefer, M, Yazar-Klosinki, B, Emerson, A and Doblin, R (2024). Effects of MDMA-assisted therapy for PTSD on self-experience. PLoS One 19(1), e0295926. DOI: 10.1371/journal.pone.0295926.Google Scholar
van Oorsouw, K, Toennes, SW and Ramaekers, JG (2022) Therapeutic effect of an ayahuasca analogue in clinically depressed patients: a longitudinal observational study. Psychopharmacology (Berl) 239(6), 18391852. DOI: 10.1007/s00213-021-06046-9.Google Scholar
Vanden Eede, H, Montenij, LJ, Touw, DJ and Norris, EM (2012) Rhabdomyolysis in MDMA intoxication: a rapid and underestimated killer. “Clean” ecstasy, a safe party drug? Journal of Emergency Medicine 42(6), 655658. DOI: 10.1016/j.jemermed.2009.04.057.Google Scholar
VanderZwaag, B, Garcia-Romeu, A and Garcia-Barrera, MA (2024) Exploring psychedelic use in athletes and their attitudes toward psilocybin-assisted therapy in concussion recovery. Therapeutic Advances in Psychopharmacology 14, 20451253241264812. DOI: 10.1177/20451253241264812.Google Scholar
Vargas, MV, Dunlap, LE, Dong, C, Carter, SJ, Tombari, RJ, Jami, SA, Cameron, LP, Patel, SD, Hennessey, JJ, Saeger, HN, McCorvy, JD, Gray, JA, Tian, L and Olson, DE (2023) Psychedelics promote neuroplasticity through the activation of intracellular 5-HT2A receptors. Science 379(6633), 700706. DOI: 10.1126/science.adf0435.Google Scholar
Vollenweider, FX and Preller, KH (2020) Psychedelic drugs: neurobiology and potential for treatment of psychiatric disorders. Nature Reviews Neurosciences 21(11), 611624. DOI: 10.1038/s41583-020-0367-2.Google Scholar
Wallach, J, Cao, AB, Calkins, MM, Heim, AJ, Lanham, JK, Bonniwell, EM, Hennessey, JJ, Bock, HA, Anderson, EI, Sherwood, AM, Morris, H, de Klein, R, Klein, AK, Cuccurazzu, B, Gamrat, J, Fannana, T, Zauhar, R, Halberstadt, AL and McCorvy, JD (2023) Identification of 5-HT2A receptor signaling pathways associated with psychedelic potential. Nature Communications 14(1), 8221. DOI: 10.1038/s41467-023-44016-1.Google Scholar
Wang, J, Xu, D, Shen, L, Zhou, J, Lv, X, Ma, H, Li, N, Wu, Q and Duan, J (2021a) Anti-inflammatory and analgesic actions of bufotenine through inhibiting lipid metabolism pathway. Biomedicine & Pharmacotherapy 140, 111749. DOI: 10.1016/j.biopha.2021.111749.Google Scholar
Wang, L, Wang, Q, Wang, W, Ge, G, Xu, N, Zheng, D, Jiang, S, Zhao, G, Xu, Y, Wang, Y, Zhu, R and Geng, D (2021b) Harmine alleviates titanium particle-induced inflammatory bone destruction by immunomodulatory effect on the macrophage polarization and subsequent osteogenic differentiation. Frontiers in Immunology 12, 657687. DOI: 10.3389/fimmu.2021.657687.Google Scholar
Werle, I and Bertoglio, LJ (2024) Psychedelics: a review of their effects on recalled aversive memories and fear/anxiety expression in rodents. Neuroscience & Biobehavioral Reviews 167, 105899. DOI: 10.1016/j.neubiorev.2024.105899.Google Scholar
Werle, I, Nascimento, LMM, Dos Santos, ALA, Soares, LA, Dos Santos, RG, Hallak, JEC and Bertoglio, LJ (2024) Ayahuasca-enhanced extinction of fear behaviour: role of infralimbic cortex 5-HT2A and 5-HT1A receptors. British Journal of Pharmacology 181(11), 16711689. DOI: 10.1111/bph.16315.Google Scholar
White, SR, Obradovic, T, Imel, KM and Wheaton, MJ (1996) The effects of methylenedioxymethamphetamine (MDMA, “Ecstasy”) on monoaminergic neurotransmission in the central nervous system. Progress in Neurobiology 49(5), 455479. DOI: 10.1016/0301-0082(96)00027-5.Google Scholar
Wittmann, M, Carter, O, Hasler, F, Cahn, BR, Grimberg, U, Spring, P, Hell, D, Flohr, H and Vollenweider, FX (2007) Effects of psilocybin on time perception and temporal control of behaviour in humans. Journal of Psychopharmacology 21(1), 5064. DOI: 10.1177/0269881106065859.Google Scholar
Wolfson, PE, Andries, J, Feduccia, AA, Jerome, L, Wang, JB, Williams, E, Carlin, SC, Sola, E, Hamilton, S, Yazar-Klosinski, B, Emerson, A, Mithoefer, MC and Doblin, R (2020) MDMA-assisted psychotherapy for treatment of anxiety and other psychological distress related to life-threatening illnesses: a randomized pilot study. Scientific Reports 10(1), 20442. DOI: 10.1038/s41598-020-75706-1.Google Scholar
Wsół, A (2023) Cardiovascular safety of psychedelic medicine: current status and future directions. Pharmacological Reports 75(6), 13621380. DOI: 10.1007/s43440-023-00539-4.Google Scholar
Xin, J, Ma, X, Chen, W, Zhou, W, Dong, H, Wang, Z and Ji, F (2021) Regulation of blood-brain barrier permeability by salvinorin a via alleviating endoplasmic reticulum stress in brain endothelial cell after ischemia stroke. Neurochemistry International 149, 105093. DOI: 10.1016/j.neuint.2021.105093.Google Scholar
Yanakieva, S, Polychroni, N, Family, N, Williams, LTJ, Luke, DP and Terhune, DB (2019) The effects of microdose LSD on time perception: a randomized, double-blind, placebo-controlled trial. Psychopharmacology (Berl) 236(4), 11591170. DOI: 10.1007/s00213-018-5119-x.Google Scholar
Yao, Y, Guo, D, Lu, TS, Liu, FL, Huang, SH, Diao, MQ, Li, SX, Zhang, XJ, Kosten, TR, Shi, J, Bao, YP, Lu, L and Han, Y (2024) Efficacy and safety of psychedelics for the treatment of mental disorders: a systematic review and meta-analysis. Psychiatry Research 335, 115886. DOI: 10.1016/j.psychres.2024.115886.Google Scholar
Yerubandi, A, Thomas, JE, Bhuiya, NMMA, Harrington, C, Villa Zapata, L and Caballero, J (2024) Acute adverse effects of therapeutic doses of psilocybin: a systematic review and meta-analysis. JAMA Network Open 7(4), e245960. DOI: 10.1001/jamanetworkopen.2024.5960.Google Scholar
Yousefi, P, Lietz, MP, O’Higgins, FJ, Rippe, RCA, Hasler, G, van Elk, M and Enriquez-Geppert, S (2025) Acute effects of psilocybin on attention and executive functioning in healthy volunteers: a systematic review and multilevel meta-analysis. Psychopharmacology (Berl). DOI: 10.1007/s00213-024-06742-2.Google Scholar
Yu, B, Becnel, J, Zerfaoui, M, Rohatgi, R, Boulares, AH and Nichols, CD (2008) Serotonin 5-hydroxytryptamine(2A) receptor activation suppresses tumor necrosis factor-alpha-induced inflammation with extraordinary potency. Journal of Pharmacology and Experimental Therapeutics 327(2), 316323. DOI: 10.1124/jpet.108.143461.Google Scholar
Zanikov, T, Gerasymchuk, M, Ghasemi Gojani, E, Robinson, GI, Asghari, S, Groves, A, Haselhorst, L, Nandakumar, S, Stahl, C, Cameron, M, Li, D, Rodriguez-Juarez, R, Snelling, A, Hudson, D, Fiselier, A, Kovalchuk, O and Kovalchuk, I (2023) The effect of combined treatment of psilocybin and eugenol on lipopolysaccharide-induced brain inflammation in mice. Molecules 28(6), 2624. DOI: 10.3390/molecules28062624.Google Scholar
Zaretsky, TG, Jagodnik, KM, Barsic, R, Antonio, JH, Bonanno, PA, MacLeod, C, Pierce, C, Carney, H, Morrison, MT, Saylor, C, Danias, G, Lepow, L and Yehuda, R (2024) The psychedelic future of post-traumatic stress disorder treatment. Current Neuropharmacology 22(4), 636735. DOI: 10.2174/1570159X22666231027111147.Google Scholar
Zeifman, RJ, Singhal, N, Dos Santos, RG, Sanches, RF, de Lima Osório, F, Hallak, JEC and Weissman, CR (2021) Rapid and sustained decreases in suicidality following a single dose of ayahuasca among individuals with recurrent major depressive disorder: results from an open-label trial. Psychopharmacology (Berl) 238(2), 453459. DOI: 10.1007/s00213-020-05692-9.Google Scholar
Zeifman, RJ, Wagner, AC, Monson, CM and Carhart-Harris, RL (2023) How does psilocybin therapy work? an exploration of experiential avoidance as a putative mechanism of change. Journal of Affective Disorders Reports 334, 100112. DOI: 10.1016/j.jad.2023.04.105.Google Scholar
Zeng, Y, Chourpiliadis, C, Hammar, N, Seitz, C, Valdimarsdóttir, UA, Fang, F, Song, H and Wei, D (2024) Inflammatory biomarkers and risk of psychiatric disorders. JAMA Psychiatry 81(11), 11181129. DOI: 10.1001/jamapsychiatry.2024.2185.Google Scholar
Zheng, ZH, Lin, XC, Lu, Y, Cao, SR, Liu, XK, Lin, D, Yang, FH, Zhang, YB, Tu, JL, Pan, BX, Hu, P and Zhang, WH (2023) Harmine exerts anxiolytic effects by regulating neuroinflammation and neuronal plasticity in the basolateral amygdala. International Immunopharmacology 119, 110208. DOI: 10.1016/j.intimp.2023.110208.Google Scholar
Zhong, Z, Tao, Y and Yang, H (2015) Treatment with harmine ameliorates functional impairment and neuronal death following traumatic brain injury. Molecular Medicine Reports 12(6), 79857991. DOI: 10.3892/mmr.2015.4437.Google Scholar
Figure 0

Table 1. Effects of single or repeated administration of psilocybin, LSD, ayahuasca, DMT, or MDMA on the mental health of individuals diagnosed with selected psychiatric disorders

Figure 1

Figure 1. An overview of the current landscape of psychedelics and athletic performance.

Figure 2

Figure 2. Helpful behavioural and physiological responses in rodents for inferring the physical effects of psychedelic drugs in humans.