Hostname: page-component-54dcc4c588-dbm8p Total loading time: 0 Render date: 2025-09-29T04:46:23.672Z Has data issue: false hasContentIssue false

Narrating the psychoneuroimmunomodulatory properties of serotonin 5-HT2A receptor psychedelics from a transdiagnostic perspective

Published online by Cambridge University Press:  25 July 2025

Guillaume Thuery
Affiliation:
Psychedelic Research Group, Tallaght University Hospital and Trinity College Dublin, Dublin, Ireland Department of Psychiatry, School of Medicine, Trinity College Dublin, Dublin, Ireland Neuropsychopharmacology Research Group, Trinity College Institute of Neuroscience, Dublin, Ireland
Christopher Sheridan
Affiliation:
Psychedelic Research Group, Tallaght University Hospital and Trinity College Dublin, Dublin, Ireland Department of Psychiatry, School of Medicine, Trinity College Dublin, Dublin, Ireland Neuropsychopharmacology Research Group, Trinity College Institute of Neuroscience, Dublin, Ireland
Patricia Iusan
Affiliation:
Psychedelic Research Group, Tallaght University Hospital and Trinity College Dublin, Dublin, Ireland Neuropsychopharmacology Research Group, Trinity College Institute of Neuroscience, Dublin, Ireland School of Pharmacy and Pharmaceutical Sciences, Trinity College Dublin, Dublin, Ireland
Gurjot Brar
Affiliation:
Psychedelic Research Group, Tallaght University Hospital and Trinity College Dublin, Dublin, Ireland Department of Psychiatry, School of Medicine, Trinity College Dublin, Dublin, Ireland
Kathryn Ledden
Affiliation:
Psychedelic Research Group, Tallaght University Hospital and Trinity College Dublin, Dublin, Ireland Department of Psychiatry, School of Medicine, Trinity College Dublin, Dublin, Ireland
Aoife Freyne
Affiliation:
Psychedelic Research Group, Tallaght University Hospital and Trinity College Dublin, Dublin, Ireland
John R. Kelly*
Affiliation:
Psychedelic Research Group, Tallaght University Hospital and Trinity College Dublin, Dublin, Ireland Department of Psychiatry, School of Medicine, Trinity College Dublin, Dublin, Ireland
Andrew Harkin*
Affiliation:
Psychedelic Research Group, Tallaght University Hospital and Trinity College Dublin, Dublin, Ireland Neuropsychopharmacology Research Group, Trinity College Institute of Neuroscience, Dublin, Ireland School of Pharmacy and Pharmaceutical Sciences, Trinity College Dublin, Dublin, Ireland
*
Corresponding authors: John R. Kelly, Andrew Harkin; Emails: kellyjr@tcd.ie; aharkin@tcd.ie
Corresponding authors: John R. Kelly, Andrew Harkin; Emails: kellyjr@tcd.ie; aharkin@tcd.ie
Rights & Permissions [Opens in a new window]

Abstract

Objective:

By synthesising findings from both clinical and preclinical research, this review aims to provide an understanding of the interplay between 5-HT2A receptor psychedelics and the immune system and considers how their immunomodulatory effects associate with neuronal and behavioural changes.

Methods:

A PubMed literature search covering the past 30 years was conducted using keywords such as ‘5-HT2A receptor’, ‘psychedelics’, ‘immune system’, and ‘HPA axis’. Studies were included if they addressed the effects of 5-HT2AR psychedelics on immune function, neuroimmune interactions, or HPA axis involvement. This narrative review synthesises evidence highlighting the bi-directional effects of 5-HT2AR psychedelics between the immune and nervous systems, identified through this search process.

Results:

Preclinical and clinical studies report that 5-HT2AR psychedelics have some direct immunomodulatory properties with downregulation of gene regulators like NF-κB, and reduced cytokine expression such as TNF-α, IL-6, and IL-1β at a central and peripheral level, accompanied by modulation of corticotrophin releasing hormone (CRH), adrenocorticotrophic hormone (ACTH), and cortisol. Direct immunomodulatory effects are mediated by pathways involving serotonin receptors, the Sigma-1 receptor, and the TrkB receptor. Immunomodulation is further mediated indirectly via the HPA axis.

Conclusion:

Further studies will determine the molecular and cellular mechanisms underlying these immunomodulatory effects. There is growing interest in the potential of 5-HT2AR psychedelics for treating a range of mental health and brain disorders. In keeping with their immunomodulatory actions, the likely modulation of brain glia and glial-neuronal interaction remains to be determined, representing a promising direction of further research on the therapeutic potential of 5-HT2AR psychedelics.

Information

Type
Review Article
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

Summations

  • Preclinical in vitro and in vivo studies indicate 5-HT2AR psychedelics have immunomodulatory properties.

  • Immunomodulatory effects are mediated by pathways involving serotonin receptors, the Sigma-1 receptor (SIG-1R), and the TrkB receptor.

  • Further investigation is warranted to understand their influence on glial cells, glial-neuronal interactions, and neuroinflammatory processes.

Considerations

  • Investigation with consistent experimental protocols is warranted to elucidate underlying mechanisms.

  • Additional well-designed clinical investigations will be crucial to definitively assess the impact of psychedelics on the human immune system.

  • Combined with existing post-mortem imaging techniques in animals, imaging modalities like MRI scans allow researchers to directly correlate functional connectivity and other MRI measures with neuroplasticity, glial activation, neuroinflammation, and neuronal cell death to further elucidate the immunoregulatory properties of psychedelics with impacts on the nervous system.

Introduction

Research on serotonin 5-hydroxytryptamine (5-HT)-2A receptor (5-HT2AR) psychedelic drugs, namely lysergic acid diethylamide (LSD), psilocybin, or N,N-dimethyltryptamine (N,N-DMT), has experienced a resurgence in recent years. This has been focused on their therapeutic effects for the treatment of various mental disorders (Griffiths et al., Reference Griffiths, Johnson, Carducci, Umbricht, Richards, Richards, Cosimano and Klinedinst2016; Carhart-Harris et al., Reference Carhart-Harris, Roseman, Bolstridge, Demetriou, Pannekoek, Wall, Tanner, Kaelen, Mcgonigle, Murphy, Leech, Curran and Nutt2017, Reference Carhart-Harris, Bolstridge, Day, Rucker, Watts, Erritzoe, Kaelen, Giribaldi, Bloomfield, Pilling, Rickard, Forbes, Feilding, Taylor, Curran and Nutt2018, Reference Carhart-Harris, Giribaldi, Watts, Baker-Jones, Murphy-Beiner, Murphy, Martell, Blemings, Erritzoe and Nutt2021; Mertens et al., Reference Mertens, Wall, Roseman, Demetriou, Nutt and Carhart-Harris2020; Doss et al., Reference Doss, Považan, Rosenberg, Sepeda, Davis, Finan, Smith, Pekar, Barker, Griffiths and Barrett2021 Raison et al., Reference Raison, Sanacora, Woolley, Heinzerling, Dunlop, Brown, Kakar, Hassman, Trivedi, Robison, Gukasyan, Nayak, Hu, O’donnell, Kelmendi, Sloshower, Penn, Bradley, Kelly, Mletzko, Nicholas, Hutson, Tarpley, Utzinger, Lenoch, Warchol, Gapasin, Davis, Nelson-Douthit, Wilson, Brown, Linton, Ross and Griffiths2023; Goodwin et al., Reference Goodwin, Aaronson, Alvarez, Atli, Bennett, Croal, Debattista, Dunlop, Feifel, Hellerstein, Husain, Kelly, Lennard-Jones, Licht, Marwood, Mistry, Páleníček, Redjep, Repantis, Schoevers, Septimus, Simmons, Soares, Somers, Stansfield, Stuart, Tadley, Thiara, Tsai, Wahba, Williams, Winzer, Young, Young, Zisook and Malievskaia2023a, c), with hundreds of registered clinical trials underway.

Researchers and biotechnology companies are exploring how psychedelic drugs may be used in a clinical setting as an additional treatment modality to address the growing mental health challenges that have become more pronounced in the wake of the COVID-19 pandemic (Akil et al., Reference Akil, Brenner, Kandel, Kendler, King, Scolnick, Watson and Zoghbi2010; Nissen et al., Reference Nissen, Hojgaard and Thomsen2020; Jones et al., Reference Jones, Mitra and Bhuiyan2021; Oliveira et al., Reference Oliveira, Butini, Pauletto, Lehmkuhl, Stefani, Bolan, Guerra, Dick, De Luca Canto and Massignan2022; Moncrieff et al., Reference Moncrieff, Cooper, Stockmann, Amendola, Hengartner and Horowitz2023).

Most clinical and preclinical studies are focusing on how these drugs can induce changes in neurocircuitry and neuroplasticity in the short- and long-term (Preller et al., Reference Preller, Burt, Ji, Schleifer, Adkinson, Stampfli, Seifritz, Repovs, Krystal, Murray, Vollenweider and Anticevic2018; Shao et al., Reference Shao, Liao, Gregg, Davoudian, Savalia, Delagarza and Kwan2021, 2024; Raval et al., Reference Raval, Johansen, Donovan, Ros, Ozenne, Hansen and Knudsen2021; Grieco et al., Reference Grieco, Castrén, Knudsen, Kwan, Olson, Zuo, Holmes and Xu2022; Inserra et al., Reference Inserra, Giorgini, Lacroix, Bertazzo, Choo, Markopolous, Grant, Abolghasemi, De Gregorio, Flamand, Rogers, Comai, Silvestri, Gobbi and Di Marzo2023; Funk et al., Reference Funk, Araujo, Slassi, Lanthier, Atkinson, Feng, Lau, Le and Higgins2024), with what seems to be promising therapeutic effects in the context of mental health (Castren and Antila, Reference Castren and Antila2017; Olson, Reference Olson2018; Grieco et al., Reference Grieco, Castrén, Knudsen, Kwan, Olson, Zuo, Holmes and Xu2022).

Evidence is also emerging to indicate that these drugs affect the immune system and peripheral areas including the gut and vascular systems. The immune system is a complex network of different organs, cell types, and chemical mediators which may be directly or indirectly modulated by psychedelics. The numerous interactions between organs, cells, and soluble mediators have been extensively reviewed elsewhere (Carpenter and O’Neill, Reference Carpenter and O’Neill2024; Delves et al., Reference Delves, Martin, Burton and Roitt2017; Marshall et al., Reference Marshall, Warrington, Watson and Kim2018).

Gut-immune-brain interactions further complicate our understanding of how psychedelics cause psychological effects. Although research on psychedelics for mental health disorders is advancing rapidly, exploring their impact on the immune and neuroendocrine systems in humans is also showing promise. Studies to date of 5-HT2AR psychedelics exploring their impact on bi-directional links between the immune and central nervous system (CNS), and their psychological effects have yielded mixed results, prompting a review of their psychoneuroimmunological properties.

A literature search was performed using PubMed, focusing on articles published within the last 30 years. Relevant keywords and phrases included combinations such as ‘5-HT2A receptor’, ‘psychedelics’, ‘immune system’, ‘HPA axis’, and related terms. The search strategy aimed to capture studies investigating both the acute and long-term effects of 5-HT2AR psychedelics on immune function, as well as their indirect effects mediated through the nervous system and the hypothalamic-pituitary-adrenal (HPA) axis. Articles were initially screened by title and abstract for relevance to the review’s focus. Full-text articles were subsequently reviewed if they met the inclusion criteria, which required that studies addressed the effects of 5-HT2AR psychedelics on immune parameters, neuroimmune interactions, or HPA axis involvement. A summary of the in vitro and in vivo studies having measured the immunomodulatory properties of 5-HT psychedelics can be found in Tables 1 and 2 respectively.

Peripheral immune system signalling to the brain

The relationship between the peripheral immune system, the CNS, and brain resident glial cells is an ongoing area of research. Peripheral immune cells at the choroid plexus (CP), blood brain barrier (BBB), and meninges participate in the transport of antigens from the CNS to lymph nodes via the cerebrospinal fluid (CSF) (Rustenhoven and Kipnis, Reference Rustenhoven and Kipnis2022). Activation of the immune system in the periphery signals in turn to the CNS. Disruption of the CP and BBB may allow immune cells to enter the brain, while immune signalling molecules like cytokines and chemotactic factors cross into the CNS via cerebrovascular endothelial transporters (Millett et al., Reference Millett, Burdick and Kubicki2022). The infiltration of immune system mediators from the periphery into the CNS triggers the mobilisation of glial cells in the brain.

Viral infections, for instance the severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2), may trigger sustained peripheral inflammatory responses with production of interleukin (IL)-6, tumour necrosis factor (TNF)-α, adipokines, chemo-attractants, and reactive oxygen species (ROS) which can disrupt BBB permeability (Brundin et al., Reference Brundin, Nath and Beckham2020). These effects are particularly relevant given the prevalence of long-COVID, which often involves persistent psychological and neurological symptoms after infection (Wong et al., Reference Wong, Devason, Umana, Cox, Dohnalova, Litichevskiy, Perla, Lundgren, Etwebi, Izzo, Kim, Tetlak, Descamps, Park, Wisser, Mcknight, Pardy, Kim, Blank, Patel, Thum, Mason, Beltra, Michieletto, Ngiow, Miller, Liou, Madhu, Dmitrieva-Posocco, Huber, Hewins, Petucci, Chu, Baraniecki-Zwil, Giron, Baxter, Greenplate, Kearns, Montone, Litzky, Feldman and Henao-Mejia2023; Greene et al., Reference Greene, Connolly, Brennan, Laffan, O’keeffe, Zaporojan, O’callaghan, Thomson, Connolly, Argue, Meaney, Martin-Loeches, Long, Cheallaigh, Conlon, Doherty and Campbell2024). Furthermore, gut microbiota disruption and life stress, signal to the brain via the immune system which impact on behaviour and symptoms mediated through the CNS (Zhang et al., Reference Zhang, Xiao, Mao and Xia2023b).

Beyond innate immune mechanisms, disruption of BBB permeability can also allow circulating autoantibodies from the adaptive immune system to enter the brain parenchyma. Under normal conditions, the BBB prevents brain-reactive antibodies from causing brain pathology. However, under pathological conditions, these antibodies can penetrate the CNS and bind to various neuronal and non-neuronal targets (Diamond et al., Reference Diamond, Honig, Mader, Brimberg and Volpe2013). These autoantibodies can modulate microglial activation and function, directly affect neural signalling by interacting with neurotransmitter receptors, and contribute to neuroinflammation and tissue damage. For instance, in systemic lupus erythematosus, autoantibodies that cross-react with neuronal N-methyl-D-aspartate (NMDA) receptors can lead to neurocognitive dysfunction, while in neuromyelitis optica, antibodies targeting astrocytic aquaporin-4 cause astrocyte damage (Mader et al., Reference Mader, Brimberg and Diamond2017). Recent perspectives have transformed our understanding of brain immunity from viewing the brain as isolated to recognising the complex bi-directional communication between the CNS and the immune system through various interfaces and compartments at the brain’s borders (Castellani et al., Reference Castellani, Croese, Peralta Ramos and Schwartz2023), with important implications for neurological and psychiatric disorders.

Glial compartments

Astrocytes, microglia, and oligodendrocytes support neuronal function in the CNS and facilitate the connection between the CNS and the immune system. Astrocytes support neuronal function and maintain BBB integrity but can also contribute to neuroinflammation through cytokine release (Cekanaviciute and Buckwalter, Reference Cekanaviciute and Buckwalter2016). Microglia are the primary immune effector cells within the CNS and play a pivotal role in response to the presence of pathogens. Activated microglia can adopt a variety of phenotypes, displaying either predominantly pro-inflammatory (M1) or anti-inflammatory (M2) characteristics. However, their activation states are not strictly limited to these two categories and are more complex than a simple M1/M2 distinction (Gao et al., Reference Gao, Jiang, Tan and Chen2023; Wang et al., Reference Wang, He and Zhang2023). Microglia release a variety of factors such as cytokines which can target neuronal cells (Ransohoff and Brown, Reference Ransohoff and Brown2012) and influence synaptic plasticity and other CNS functions (Werneburg et al., Reference Werneburg, Feinberg, Johnson and Schafer2017; Cornell et al., Reference Cornell, Salinas, Huang and Zhou2022). The relationship between microglia and astrocytes is important in the immune response of the brain, as they communicate bidirectionally (Norden et al., Reference Norden, Fenn, Dugan and Godbout2014; Bhusal et al., Reference Bhusal, Afridi, Lee and Suk2023). Glial cells play a role in maintaining CNS homeostasis through their gatekeeper function at the BBB, the expression of various factors can impact the function of this barrier, which is important in the context of inflammatory insults (Alvarez et al., Reference Alvarez, Katayama and Prat2013). Glial cells can also signal to BBB endothelial cells to increase monocyte trafficking into the CNS (Weber et al., Reference Weber, Godbout and Sheridan2017).

CNS signalling to the immune system

The bi-directional relationship between the CNS and the immune system has been well reviewed (Maier, Reference Maier2003; Wrona, Reference Katoh, Soga, Nara, Tamagawa-Mineoka, Nin, Kotani, Masuda and Kishimoto2006; Kamimura et al., Reference Kamimura, Tanaka, Hasebe and Murakami2020; Gentile et al., Reference Gentile, D’acquisto and Leposavic2021). Efferent sympathetic and parasympathetic innervation of all lymphoid organs allows the CNS to release noradrenaline or acetylcholine and influence immune functions such as thymocyte maturation, T-cell development, and cytokine release (Kavelaars, Reference Kavelaars2002; Leposavic et al., Reference Leposavic, Pilipovic and Perisic2011; Sundman and Olofsson, Reference Sundman and Olofsson2014; Carnevale et al., Reference Carnevale, Pallante, Fardella, Fardella, Iacobucci, Federici, Cifelli, de Lucia and Lembo2014; Dubeykovskaya et al., Reference Dubeykovskaya, Y., Chen, Worthley, Renz, Urbanska, Hayakawa, T., Westphalen, Dubeykovskiy, Chen, Friedman, Asfaha, Nagar, Tailor, Muthupalani, Fox, Kitajewski and Wang2016; Chen et al., Reference Chen, Barnoud and Scheiermann2021; Francelin et al., Reference Francelin, Veneziani, Farias, Mendes-Da-Cruz and Savino2021).

In addition to these sympathetic and vagal influences, the hypothalamic-pituitary-adrenal (HPA) axis introduces an indirect communication pathway between the CNS and the immune system. Acute and chronic environmental stress activates the HPA axis, leading to the release of glucocorticoids into the blood (Bellavance and Rivest, Reference Bellavance and Rivest2014). Cortisol is a known immunosuppressor and leads to anti-inflammatory effects via mitogen-activated kinase (MAPK1)-dependent and nuclear factor kappa B (NF-κB)-dependent pathways (Coutinho and Chapman, Reference coutinho and Chapman2011; Zefferino et al., Reference Zefferino, Di Gioia and Conese2021). However, chronic stress can dysregulate the HPA axis, cause glucocorticoid resistance, and decreased expression of glucocorticoid receptors, contributing to immune dysregulation (Cohen et al., Reference Cohen, Janicki-Deverts, Doyle, Miller, Frank, Rabin and Turner2012; Silverman and Sternberg, Reference Silverman and Sternberg2012; Lam et al., Reference Lam, Chen, Chiang and Miller2022).

A focus on serotonin and 5-HT2AR in neuroimmunomodulation

There is a total of fourteen known 5-HT receptors classified into seven families (5-HT1R–5-HT7R) based on their structural and functional properties. These have different downstream cellular effects, leading to increased or decreased cellular levels of cyclic adenosine monophosphate (cAMP), inositol triphosphate (IP3), and diacylglycerol (DAG), producing inhibitory or excitatory neuromodulatory responses (Frazer and Hensler, Reference Frazer, Hensler, Gj, Bw, Rw, Sk and Md1999).

5-HT2AR agonists

Psychedelics have a wide range of binding profiles with the 5-HT receptor family (Ray, Reference Ray2010) (Figure 1). The 5-HT2AR is of particular interest in the field of psychedelic research as it is largely associated with the psychotropic effects of psychedelics (Nichols, Reference Nichols2004; Preller et al., Reference Preller, Burt, Ji, Schleifer, Adkinson, Stampfli, Seifritz, Repovs, Krystal, Murray, Vollenweider and Anticevic2018; Madsen et al., Reference Madsen, Fisher, Burmester, Dyssegaard, Stenbaek, Kristiansen, Johansen, Lehel, Linnet, Svarer, Erritzoe, Ozenne and Knudsen2019; Shao et al., Reference Shao, Liao, Davoudian, Savalia, Jiang, Wojtasiewicz, Tan, Nothnagel, Liu, Woodburn, Bilash, Kim, Che and Kwan2025). This receptor is widely distributed throughout the cortex, whereas moderate to low levels of expression can be found in the limbic system, including the amygdala and the hippocampus, which plays a crucial role in emotional processing, memory formation, and behavioural regulation (Saulin et al., Reference Saulin, Savli and Lanzenberger2012).

Psychedelics can induce different downstream effects after activation of the 5-HT2AR. Psychedelics display biased agonism, also known as functional selectivity, referring to the ability of different ligands to selectively stabilise distinct receptor conformations, leading to the activation of specific signalling pathways over others (Inserra et al., Reference Inserra, De Gregorio and Gobbi2021).

In the context of 5-HT receptors, psychedelics can induce alternative 5-HT receptor conformations, leading to altered ligand affinity recruitment of intracellular effector proteins, including β-arrestins, which mediate receptor desensitisation, internalisation, and signalling pathways independent of G proteins. For example, LSD induces a receptor conformational change that will preferentially recruit β-arrestin. Interestingly, psychedelic-related effects induced by LSD appear to be mediated by β-arrestin (Rodriguiz et al., Reference Rodriguiz, Nadkarni, Means, Pogorelov, Chiu, Roth and Wetsel2021) unlike the full 5-HT2AR agonist 2,5-dimethoxy-4-iodoamphetamine [(R)-DOI] (Schmid et al., Reference Schmid, Raehal and Bohn2008).

A recent meta-analysis found that there are no significant differences in selectivity between N,N-DMT, LSD, and psilocin, relative to the 5-HT1AR, but they reported that LSD induced a significantly higher formation of inositol phosphate (Shinozuka et al., Reference Shinozuka, Jerotic, Mediano, Zhao, Preller, Carhart-Harris and Kringelbach2024).

The 5-HT2CR has been shown to be involved in the head-twitch response (HTR), a commonly used measure for a hallucinogenic-like experience in rodents. It remains unclear exactly how this receptor contributes to this behavioural response as some studies have found that antagonists increase – and agonists attenuate – the HTR (Canal et al., Reference Canal, Booth and Morgan2013; Erkizia-Santamaria et al., Reference Erkizia-Santamaria, Alles-Pascual, Horrillo, Meana and Ortega2022), whereas some studies have found that its activation can induce this behaviour (Custodio et al., Reference Custodio, Ortiz, Lee, Sayson, Kim, Lee, Kim, Cheong and Kim2023).

Psychedelics mediate their effects through 5-HT2A receptors, to distinct signalling and functional outcomes that occur depending on the localisation of receptor activation (Olson, Reference Olson2021). Extracellular 5-HT2A activation by psychedelics like psilocin initiates Gq/11-PLC signalling, which exceeds 70% efficacy threshold to induce hallucinations and HTR, while sub-threshold agonists (e.g., lisuride) lack psychedelic effects (de Vos et al., Reference De Vos, Mason and Kuypers2021; 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). β-Arrestin2 recruitment via cell-surface receptors does not correlate with psychedelic potential or with the psychedelic experience and may instead promote receptor downregulation (Schmid and Bohn, Reference Schmid and Bohn2010).

Lipophilic psychedelics (e.g., DMT) activate intracellular 5-HT2A pools, triggering neuroplasticity via transglutaminase 2 (TGM2)-mediated serotonylation of Rac1, which enhances dendritic arborisation and synaptogenesis (Ly et al., Reference Ly, Greb, Cameron, Wong, Barragan, Wilson, Burbach, Soltanzadeh Zarandi, Sood, Paddy, Duim, Dennis, Mcallister, Ori-Mckenney, Gray and Olson2018; de la Fuente Revenga et al., Reference de la Fuente Revenga, Zhu, Guevara, Naler, Saunders, Zhou, Toneatti, Sierra, Wolstenholme, Beardsley and Huntley2021). This intracellular signalling may contribute to the sustained therapeutic effects observed in clinical trials for the treatment of depression, independent of hallucinogenic activity (Vargas et al., Reference Vargas, Dunlap, Dong, Carter, Tombari, Jami, Cameron, Patel, Hennessey, Saeger, Mccorvy, Gray, Tian and Olson2023). Emerging strategies aim to decouple neuroplasticity from psychedelic effects by targeting intracellular receptors, using specific β-arrestin modulators or designing Gq-sub-threshold agonists, offering a pathway for non-hallucinogenic therapeutics (Dunlap et al., Reference Dunlap, Azinfar, C., Cameron, Viswanathan, Tombari, Myers-Turnbull, Taylor, Grodzki, Lein, Kokel and Olson2020; 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).

β2-arrestin biased signalling modulates MAPK pathways such as ERK1/2, JNK, and p38, influencing immune cell activity through scaffold-mediated regulation. β-arrestins form complexes with ERK1/2, sequestering it in the cytosol and preventing nuclear translocation, altering transcription, and dampening pro-inflammatory cytokine production (Sharma and Parameswaran, Reference Sharma and Parameswaran2015). In macrophages, β-arrestin2 inhibits TLR2/ERK1/2 signalling, reducing TNF-α expression (Fan, Reference Fan2014). β-arrestin1 promotes CD4+ T-cell survival by upregulating Bcl2, while β-arrestin2 suppresses NK cell cytotoxicity by interacting with inhibitory receptors (Crepieux et al., Reference Crepieux, Poupon, Langonne-Gallay, Reiter, Delgado, Schaefer, Bourquard, Serrano and Kiel2017). β-arrestin2 also negatively regulates NF-κB by stabilising IκBα, limiting inflammatory cytokine release in sepsis models (Fan, Reference Fan2014). β-arrestin appears to act in a modulatory fashion in immune regulation, balancing pro- and anti-inflammatory responses through modulation of MAPK activity and crosstalk with PRR signalling (Sharma and Parameswaran, Reference Sharma and Parameswaran2015).

Certain immune cells, including monocytes, macrophages, dendritic cells, and T cells, express high levels of 5-HT2A receptors making them functionally sensitive to co-ligation by psychedelics that target 5-HT receptors and activate β-arrestin signalling through crosstalk with PRR pathways such as TLRs, and can influence key signalling cascades like NF-κB and MAPK. β-arrestin-biased agonists at 5-HT2ARs can further fine-tune these effects, potentially reducing pro-inflammatory responses (Nau et al., Reference Nau, Yu, Martin and Nichols2013; Szabo, Reference Szabo2015; Flanagan and Nichols, Reference Flanagan and Nichols2018).

5-HT2AR and regulation of brain glia

Brain glia, and specifically microglia, have been shown to express various forms of 5-HT receptors including the 5-HT2AR (Krabbe et al., Reference Krabbe, Matyash, Pannasch, Mamer, Boddeke and Kettenmann2012; Glebov et al., Reference Glebov, Lochner, Jabs, Lau, Merkel, Schloss, Steinhauser and Walter2015) and 5-HT2BR influencing microglial development (Kolodziejczak et al., Reference Kolodziejczak, Bechade, Gervasi, Irinopoulou, Banas, Cordier, Rebsam, Roumier and Maroteaux2015; Turkin et al., Reference Turkin, Tuchina and Klempin2021). Although the exact functions of these remain unclear, studies to date suggest that activation of these 5-HT2AR promotes the release of microglia-derived vesicles known as exosomes containing a variety of proteins and ribonucleic acids (RNA) (Glebov et al., Reference Glebov, Lochner, Jabs, Lau, Merkel, Schloss, Steinhauser and Walter2015). 5-HT2AR influences the dynamic extensions of microglia which play an important role in surveillance and maintenance functions within the CNS (Krabbe et al., Reference Krabbe, Matyash, Pannasch, Mamer, Boddeke and Kettenmann2012). Microglia may play a role in the neuropharmacological and therapeutic effects of various drugs, including dissociative NMDA receptor psychedelics like ketamine and 5-HT2AR psychedelics (VanderZwaag et al., Reference Vanderzwaag, Halvorson, Dolhan, Simoncicova, Ben-Azu and Tremblay2023).

Astrocytes express multiple forms of 5-HT receptors, including the 5-HT2AR (Hagberg et al., Reference Hagberg, Blomstrand, Nilsson, Tamir and Hansson1998; Hirst et al., Reference Hirst, Cheung, Rattray, Price and Wilkin1998; Cohen et al., Reference Cohen, Bouchelet, Olivier, Villemure, Ball, Stanimirovic and Hamel1999; Maxishima et al., Reference Maxishima, Shiga, Shutoh, Hamada, Maeshima and Okado2001; Kong et al., Reference Kong, Peng, Chen, Yu and Hertz2002; Verkhratsky et al., Reference Verkhratsky, Parpura, Scuderi and Li2021). Although more research is required, 5-HT2AR seems to play a role in 5-HT-driven astrocytic calcium signalling with a possible role in synaptic plasticity (Jalonen et al., Reference Jalonen, Margraf, Wielt, Charniga, Linne and Kimelberg1997; Hagberg et al., Reference Hagberg, Blomstrand, Nilsson, Tamir and Hansson1998; Gonzalez-Arias et al., Reference Gonzalez-Arias, Sanchez-Ruiz, Esparza, Sanchez-Puelles, Arancibia, Ramirez-Franco, Gobbo, Kirchhoff and Perea2023). An immunocytochemical study found increased 5-HT2AR expression in astrocytes within the prefrontal cortex of Alzheimer’s disease patients and the caudate nucleus of Huntington’s disease patients (Wu et al., 1999). Astrocytes also express the serotonin transporter (SERT) (Fitzgerald et al., Reference Fitzgerald, Kaplinsky and Kimelberg1990; Bel et al., Reference Bel, Figueras, Vilaro, Sunol and Artigas1997), enabling them to regulate the extracellular availability of 5-HT (Edmondson et al., Reference Edmondson, Binda and Mattevi2007).

Oligodendrocytes form the myelin sheath around neuronal axons and support neuronal plasticity (Jang et al., Reference Jang, Gould, Xu, Kim and Kim2019). Both in vitro and in vivo studies demonstrate that these cells express the 5-HT2AR, with elevated serotonin levels altering myelination via this receptor (Simpson et al., Reference Simpson, Weaver, De Villers-Sidani, Lu, Cai, Pang, Rodriguez-Porcel, Paul, Merzenich and Lin2011; Fan et al., Reference Fan, Bhatt, Tien, Zheng, Simpson, Lin, Cai, Kumar and Pang2015). Psilocybin elicits an increase in cellular activation indicated by expression of the immediate early gene c-Fos in neurons and in oligodendrocytes (Funk et al., Reference Funk, Araujo, Slassi, Lanthier, Atkinson, Feng, Lau, Le and Higgins2024). This increase was found in 10–20% of neurons and 25% of oligodendrocytes in the medial prefrontal cortex (mPFC), basolateral amygdala, and the dorsal raphe nucleus of male rats.

Effect of 5-HT on the immune system

In the periphery, most 5-HT production occurs in enterochromaffin cells located in the gut. Gut-derived 5-HT modulates gastrointestinal functions and immune cells in or near the gut epithelium. It can also enter the bloodstream, where platelets absorb it via SERT (Cloutier et al., Reference Cloutier, Allaeys, Marcoux, Machlus, Mailhot, Zufferey, Levesque, Becker, Tessandier, Melki, Zhi, Poirier, Rondina, Italiano, Flamand, Mckenzie, Cote, Nieswandt, Khan, Flick, Newman, Lacroix, Fortin and Boilard2018).

5-HT is known to play a versatile role within the immune system influencing macrophage and monocyte activity, dendritic cell maturation, and natural killer (NK) cell cytotoxicity (Herr et al., Reference Herr, Bode and Duerschmied2017; Roumier et al., Reference Roumier, Béchade, Maroteaux and Pilowsky2019). By binding to various 5-HT receptors, 5-HT can either promote or suppress immune responses via the modulation of cytokine production and release. 5-HT receptors are abundantly expressed by immune cells [see Hodo et al. (Reference Hodo, de Aquino, Shimamoto and Shanker2020) for a thorough review on expression patterns and functions of each receptor]. Figure 2 summarises the expression pattern for 5-HT receptors in peripheral immune cells and associated functions.

Figure 1. Heat map visualisation of serotonergic receptor binding profiles. Yellow cells with crosses indicate known binding activity of ligands (rows) to specific 5-HT receptors (columns), while dark purple cells indicate no reported binding. Data compiled from: Nichols (Reference Nichols2004); Kitson (Reference Kitson2007); Keiser et al. (Reference Keiser, Setola, Irwin, Laggner, Abbas, Hufeisen, Jensen, Kuijer, Matos, Tran, Whaley, Glennon, Hert, Thomas, Edwards, Shoichet and Roth2009); Besnard et al. (Reference Besnard, Ruda, Setola, Abecassis, Rodriguiz, Huang, Norval, Sassano, Shin, Webster, Simeons, Stojanovski, Prat, Seidah, Constam, Bickerton, Read, Wetsel, Gilbert, Roth and Hopkins2012); Rickli et al. (Reference Rickli, Luethi, Reinisch, Buchy, Hoener and Liechti2015); Wsol (2023); Hatzipantelis and Olson (Reference Hatzipantelis and Olson2024); Ippolito et al. (Reference Ippolito, Vasudevan, Hurley, Gilmour, Westhorpe, Churchill and Sharp2024). 2C-(x) refers to the family of 2,5-dimethoxy-phenethylamine analogues. Note that binding affinity varies based on pharmacological method, cell type, and experimental conditions. The psychoactive drug screening programme (PDSP) has been a primary source for standardised binding data (Ki values) for many of these compounds, as reviewed in Alexander et al. (Reference Alexander, Anderson, Baxter, Claydon, Rucker and Robinson2024); Hatzipantelis and Olson (Reference Hatzipantelis and Olson2024).

Previous in vitro studies demonstrate that exogenous 5-HT regulates the release of pro- and anti-inflammatory cytokines and chemokines (e.g., TNF-α) through 5-HT3R, 5-HT4R, and 5-HT7R activation in lipopolysaccharide (LPS)-stimulated peripheral blood mononuclear cell (PBMC) cultures and whole blood (Kubera et al., Reference Kubera, Kenis, Bosmans, Scharpe and Maes2000; Cloez-Tayarani et al., Reference Cloez-Tayarani, Petit-Bertron, Venters and Cavaillon2003; Durk et al., Reference Durk, Panther, Muller, Sorichter, Ferrari, Pizzirani, Di Virgilio, Myrtek, Norgauer and Idzko2005; Kubera et al., Reference Kubera, Maes, Kenis, Kim and Lason2005). Exogenous 5-HT also promoted anti-inflammatory mechanisms in splenocytes, lymph nodes, and PBMC cultures (Toh and Miossec, Reference Toh and Miossec2007; Chabbi-Achengli et al., Reference Chabbi-Achengli, Coman, Collet, Callebert, Corcelli, Lin, Rignault, Dy, De Vernejoul and Cote2016; Sacramento et al., Reference Sacramento, Monteiro, Dias, Kasahara, Ferreira, Hygino, Wing, Andrade, Rueda, Sales, Vasconcelos and Bento2018). In contrast, exogenous 5-HT impaired the ability of mouse-derived dendritic cells (DCs) to induce Type 1 regulatory (T-reg1) cells and reduced expression of the anti-inflammatory cytokine IL-10 (Liao et al., Reference Liao, Tao, Wang, Wu, Yao, Yang, Huang, Liu, Yang and Yang2023).

In vitro evidence also suggests that eosinophils can be modulated by 5-HT. Indeed, treatment with 5-HT caused human eosinophils to roll onto vascular cell adhesion molecule (Vcam)-1, a process important for immune cell migration (Kang et al., Reference Kang, Ha, Bahaie, Hosseinkhani, Ge, Blumenthal, Rao and Sriramarao2013). This effect was found to be driven by 5-HT2AR activation and was coupled with an increase in intracellular calcium (Ca2+) levels and with distinct changes in the cytoskeleton and cell shape.

These observations suggest that 5-HT has different cell-specific effects on the immune system, and that these immunomodulatory effects are dependent on various 5-HT receptors.

Immunomodulatory effects have also been observed with commonly prescribed selective 5-HT reuptake inhibitors (SSRIs) and 5-HT and noradrenaline reuptake inhibitors (SNRIs) in whole blood (Diamond et al., Reference Diamond, Kelly and Connor2006), PBMC (Taler et al., Reference Taler, Gil-Ad, Lomnitski, Korov, Baharav, Bar, Zolokov and Weizman2007), and microglial cultures (Horikawa et al., Reference Horikawa, Kato, Mizoguchi, Monji, Seki, Ohkuri, Gotoh, Yonaha, Ueda, Hashioka and Kanba2010; Tynan et al., Reference Tynan, Weidenhofer, Hinwood, Cairns, Day and Walker2012; Liu et al., Reference Liu, Zou, Wang, Zhu, Lai, Zhou, Chen, Zhang and Zhu2014), and in in vivo immune stimulation paradigms (Pellegrino and Bayer, Reference Pellegrino and Bayer2000; Dong et al., Reference Dong, Zhang, Yao, Ren, Yang, Ma, Han, Saito and Hashimoto2016; Tomaz et al., Reference Tomaz, Chaves Filho, Cordeiro, Juca, Soares, Barroso, Cristino, Jiang, Teixeira, De Lucena and Macedo2020).

Other mechanisms by which SSRIs might modulate the immune system include signalling pathways such as extracellular signal-regulated-protein kinase (Erk) and p38 MAPK cascades (Russo-Neustadt et al., Reference Russo-Neustadt, Alejandre, Garcia, Ivy and Chen2004; Mercier et al., Reference Mercier, Lennon, Renouf, Dessouroux, Ramauge, Courtin and Pierre2004; Chilmonczyk et al., Reference Chilmonczyk, Bojarski, Pilc and Sylte2017), cAMP production (Zhou et al., Reference Zhou, Ma, Yeung, Wong, Tsim, So, Lam and Chung2016), membrane-associated lipid rafts (Singh et al., Reference Singh, Wray, Schappi and Rasenick2018), glucocorticoid receptors (Pariante et al., Reference Pariante, Makoff, Lovestone, Feroli, Heyden, Miller and Kerwin2001; Antonioli et al., Reference Antonioli, Rybka and Carvalho2012; Gobin et al., Reference Gobin, Van Steendam, Denys and Deforce2014), the brain derived neurotrophic factor (BDNF) (Wang et al., Reference Wang, Wang, Wu, Huang and Yang2022), and the Sigma-1 receptor (Nguyen et al., Reference Nguyen, Lucke-Wold, Mookerjee, Cavendish, Robson, Scandinaro and Matsumoto2015; Hashimoto, Reference Hashimoto2015; Rosen et al., Reference Rosen, Seki, Fernandez-Castaneda, Beiter, Eccles, Woodfolk and Gaultier2019; Salaciak and Pytka, Reference Salaciak and Pytka2022), as well as directly modulating the vagal nerve (Ondicova et al., Reference Ondicova, Tillinger, Pecenak and Mravec2019). This suggests that pathways independent of the serotonergic system may be involved in the immunomodulatory effects of SSRIs.

Finally, classical psychedelics moderately influence platelet function and immune responses via serotonin 5-HT2A receptor pathways on platelets and other immune cells but lack significant clotting risks (Szabo, Reference Szabo2015). In contrast, MDMA can markedly disrupt hemostasis, triggering coagulopathies like thrombocytopenia and disseminated intravascular coagulation (DIC) through serotonin syndrome and rhabdomyolysis, highlighting critical safety concerns in therapeutic use (Szabo, Reference Szabo2015; Doyle et al., Reference Doyle, Meyer, Breen and Hunt2020).

While preclinical models provide valuable insights into 5-HT’s immunomodulatory potential, there are critical interspecies differences in receptor expression patterns and pharmacological responses that are crucial to keep in mind. For example, the 5-HT6R is abundantly expressed in human and rat striatal regions, however it shows negligible expression in mouse brain tissue (Hirst et al., Reference Hirst, Abrahamsen, Blaney, Calver, Aloj, Price and Medhurst2003; Kirkpatrick, Reference Kirkpatrick2004). Receptor structural variations render many compounds targeting 5-HT6R ineffective in murine models (Hirst et al., Reference Hirst, Abrahamsen, Blaney, Calver, Aloj, Price and Medhurst2003; Kirkpatrick, Reference Kirkpatrick2004). These variations are relevant for studying serotonergic psychedelics, as demonstrated by Haberzettl et al. (Reference Haberzettl, Bert, Fink and Fox2013) in their systematic analysis of serotonin syndrome models. Their work revealed that monoamine oxidase (MAO)-A knockout mice exhibit greater sensitivity to 5-HT-enhancing drugs compared to wild-type strains, whereas human MAO polymorphisms show more nuanced clinical manifestations (Haberzettl et al., Reference Haberzettl, Bert, Fink and Fox2013; Chiew and Isbister, Reference Chiew and Isbister2024). This difference originates not only from receptor diversity but also from differences in systemic 5-HT homeostasis and storage mechanisms (Mossner and Lesch, Reference Mossner and Lesch1998). When considering neuroimmune interactions, physiological disparities complicate translation, particularly given that psychedelics like LSD demonstrate species-specific binding kinetics at the 5-HT2AR critical for both psychoactive and immunomodulatory effects (Szabo, Reference Szabo2015; Canal, Reference Canal2018). These findings underscore the necessity of validating preclinical observations in human cell systems (Figure 2).

Figure 2. Sankey diagram depicting 5-HT receptor expression in peripheral immune cells and their associated cellular functions. Function-specific references are provided for each cell type. Second messengers and their targets are identified for specific receptors in certain immune cells: 5-HT 2A R in Eos is associated to increased intracellular Ca2+ and to the activation of ROCK, MAPK, PI3K, PKC, and Calmodulin (Boehme et al., Reference Boehme, Lio, Sikora, Pandit, Lavrador, Rao and Sriramarao2004; Kang et al., Reference Kang, Ha, Bahaie, Hosseinkhani, Ge, Blumenthal, Rao and Sriramarao2013), 5-HT 1A R in both T- and B-cells has been associated to NF-κB translocation (Abdouh et al., Reference Abdouh, Albert, Drobetsky, Filep and Kouassi2004), 5-HT 7 R in T-cells has been found associated to the activation of ERK1/2 and translocation of NF-κB (León-Ponte et al., Reference León-Ponte, Ahern and O’connell2007), 5-HT 2C R in macrophages has been associated to increased intracellular Ca2+ (Mikulski et al., Reference Mikulski, Zaslona, Cakarova, Hartmann, Wilhelm, Tecott, Lohmeyer and Kummer2010), and 5-HT 4 R and 5-HT 7 R in dendritic cells have been associated to increases in cAMP levels (Muller et al., Reference Muller, Durk, Blumenthal, Grimm, Cicko, Panther, Sorichter, Herouy, Di Virgilio, Ferrari, Norgauer and Idzko2009).

Effect of the immune system on serotonergic transmission

Systemic inflammation has consistently been demonstrated to influence serotonergic signalling in the brain. Systemic LPS injection had a wide range of effects on the brain including increased reuptake of 5-HT from the synaptic cleft via SERT in the mPFC in mice (van Heesch et al., Reference Van Heesch, Prins, Konsman, Korte-Bouws, Westphal, Rybka, Olivier, Kraneveld and Korte2014), reduced 5-HT concentrations in the hippocampus in mice (Zhao et al., Reference Zhao, Cao, Liu, Li, Xu, Liu, Zhang, Yang, Yi, Xu, Fan and Ma2019) and in the anteroventral preoptic region in rats (Mota et al., Reference Mota, Rodrigues-Santos, Fernandez, Carolino, Antunes-Rodrigues, Anselmo-Franci and Branco2017), increased 5-HT2AR mRNA levels in the PFC and hippocampus in mice (Couch et al., Reference Couch, Xie, lundberg, Sharp and Anthony2015), increased the functional response to (R)-DOI (Couch et al., Reference Couch, Xie, lundberg, Sharp and Anthony2015), and increased 5-HT turnover in the PFC and hippocampus in mice (Swiergiel and Dunn, Reference Swiergiel and Dunn2006).

In rats treated with the immune stimulus and toll-like receptor (TLR)-3 agonist polyinosinic:polycytidylic acid [Poly(I:C)], levels of messenger RNA (mRNA) coding for SERT increased in the cortex, cerebellum, medial preoptic area, and paraventricular hypothalamic nucleus, which in turn was associated with a decrease in extracellular 5-HT concentrations as measured by microdialysis (Katafuchi et al., Reference Katafuchi, Kondo, Take and Yoshimura2005).

In zebrafish, intra-cerebroventricular (ICV) microinjection of IL-4 suppressed 5-HT production, whereas ICV microinjection of 5-HT suppressed neurogenesis in periventricular neurons via a neuron-glia interaction involving the induction of BDNF (Bhattarai et al., Reference Bhattarai, Cosacak, Mashkaryan, Demir, Popova, Govindarajan, Brandt, zhang, Chang, Ampatzis and Kizil2020).

Systemic LPS injection also leads to induction of the kynurenine pathway in the hippocampus by increasing the expression of the indoleamine-2,3-dioxygenase (IDO) enzyme involved in tryptophan metabolism, leading to an increase in the production of kynurenine from tryptophan (Zhao et al., Reference Zhao, Cao, Liu, Li, Xu, Liu, Zhang, Yang, Yi, Xu, Fan and Ma2019; Marx et al., Reference Marx, Mcguinness, Rocks, Ruusunen, Cleminson, Walker, Gomes-Da-Costa, Lane, Sanches, Diaz, Tseng, Lin, Berk, Clarke, O’neil, Jacka, Stubbs, Carvalho, Quevedo, Soares and Fernandes2021). As tryptophan is an essential amino acid required for the biosynthesis of 5-HT, it has been proposed that IDO induction may limit the availability of tryptophan for 5-HT biosynthesis.

5-HT2AR psychedelic modulation of immune responses in isolated systems

Effects of 5-HT2AR psychedelics on peripheral blood mononuclear cells

In LPS or Poly(I:C)-stimulated primary monocyte-derived dendritic cells (moDC), N,N-DMT, and the methoxylated derivative 5-methoxy-N,N-dimethyltryptamine (5-MeO-DMT) inhibit the production of pro-inflammatory cytokines IL-1β, IL-6, and TNF-α and the chemokine IL-8, while increasing the secretion of the anti-inflammatory cytokine IL-10 (Szabo et al., Reference Szabo, Kovacs, Frecska and Rajnavolgyi2014). This treatment also reduced the amount of induced T helper (Th)-1 and Th17 effector T-cells versus vehicle in a SIG-1R-dependent manner. Considering that both compounds are endogenous ligands of SIG-1R, the authors propose that activation of the SIG-1R in peripheral immune cells with psychedelics like N,N-DMT may trigger a unique cluster of differentiation (CD)4+ T-cell response upon viral or bacterial stimulation.

In CD4+ and CD8+ T-cell cultures stimulated with concanavalin A (ConA), a lectin mitogen, (R)-DOI was found to provoke a stimulatory response with an increase in the production of cytokines interferon (IFN)-γ and IL-2 (Inoue et al., Reference Inoue, Okazaki, Kitazono, Mizushima, Omata and Ozaki2011). This effect was reversed by the 5-HT2AR antagonist sarpogrelate hydrochloride. This study found higher doses of (R)-DOI to enhance IL-2 and IFN-γ production, indicating a link between 5-HT2AR activity and IFN-γ production. The use of ConA, known to induce a distinct anti-cancer T-cell response, might explain the observed difference compared to other studies (Wiersma, Reference Wiersma and Chouaib2020).

In primary cultures of CD4+ and CD8+ T-cells stimulated with anti-human CD3, a widely used T-cell stimulant, levels of TNF-α were unaffected by LSD, N,N-DMT, psilocin, or mescaline (Rudin et al., Reference Rudin, Areesanan, Liechti and Grundemann2023). The authors suggest that an initial anti-inflammatory cortisol release upon administration of psychedelics may be responsible for the effects measured in previous investigations (Dos Santos et al., Reference Dos Santos, Valle, Bouso, Nomdedéu, Rodríguez-Espinosa, McIlhenny, Barker, Barbanoj and Riba2011) which examined blood and serum samples following administration of psychedelics. Furthermore, cell cultures in this study were treated for 24 to 72 hours which may miss the initial window of immunomodulatory action. Notably, the peak anti-proliferative effects of ayahuasca – a psychoactive decoction containing N,N-DMT and monoamine oxidase inhibitors – on CD3+, CD4+, and CD8+ cell populations begin to diminish two hours after administration (Dos Santos et al., Reference Dos Santos, Valle, Bouso, Nomdedéu, Rodríguez-Espinosa, McIlhenny, Barker, Barbanoj and Riba2011).

In cultured RAW 264.7 macrophages, Nkadimeng et al. (Reference Nkadimeng, Nabatanzi, Steinmann and Eloff2020) demonstrated that psilocybin-containing mushroom extracts significantly reduced the expression levels of IL-1β following LPS stimulation. Similarly, Oppong-Damoah et al. (Reference Oppong-Damoah, Curry, Blough, Rice and Murnane2019) observed that macrophages stimulated with ethanol produce NO, an effect that was reduced by (R)-DOI.

Ghasemi Gojani et al. (Reference Ghasemi Gojani, Wang, Li, Kovalchuk and Kovalchuk2024) found that a concentration of 15 μM psilocybin was sufficient to inhibit NLRP3 inflammasome activation and downregulate inflammatory-regulating transcription factors in LPS-stimulated human monocytes. Both 15 and 10 μM doses resulted in the downregulation of NF-κB, Tyrosine Kinase 2 (TYK2), Signal Transducer and Activator of Transcription (STAT)-1, and STAT3 activation – key regulators of IL-1β, IL-6, TNF-α, and cyclooxygenase (COX)-2. This was accompanied by a reduction of COX-2, Pro-TNFα, IL-1β, IL-6, and Pro-IL-1β levels, and suppression of IL-1β release. Interestingly, the 5 μM dose stimulated an increase in the levels of these transcription factors. Higher doses of psilocybin did not alter total NF-κB levels but prevented its nuclear translocation, thereby inhibiting transcription of the regulated genes. In the cases of IL-6 and COX-2, protein levels – but not mRNA levels – were reduced, suggesting that psilocybin may modulate post-translational processing or modifications. This study also found that the LPS-stimulation decreased protein levels of 5-HT2A and 5-HT2B receptors while psilocybin attenuated this downregulation in LPS-stimulated cells, upregulating 5-HT2A receptors above baseline at the highest dose. In unstimulated cells, psilocybin reduced 5-HT2A receptors in a dose-dependent fashion. This research emphasises the immune-modifying capabilities of psilocybin, while also revealing distinct outcomes when applied to inflamed versus non-inflamed biological systems.

Psilocybin inducing opposing immunoregulatory effects at low compared to higher doses has also been observed in resting macrophages, with lower doses inducing higher levels of TNF-α (Laabi et al., Reference Laabi, LeMmon, Vogel, Chacon and Jimenez2024). Whereas in LPS-stimulated and classically activated macrophages, post-treatment with psilocin, but not psilocybin, produced anti-inflammatory-like effects, reducing levels of TNFα, and increasing levels of IL-10 (Laabi et al., Reference Laabi, LeMmon, Vogel, Chacon and Jimenez2024). Peyote extract, containing the phenethylamine psychedelic mescaline, activated nitric oxide (NO) production by murine macrophages (Franco-Molina et al., Reference Franco-Molina, Gomez-Flores, Tamez-Guerra, Tamez-Guerra, Castillo-Leon and Rodriguez-Padilla2003). This study further reported that peyote extract stimulated murine thymic lymphocyte proliferation and induced an increase in mRNA levels of IL-1, IL-6, and IL-8 in human leukocytes.

Tourino et al. (Reference Tourino, De Oliveira, Belle, Knebel, Albuquerque, Dorr, Okada, Migliorini, Soares and Campa2013) aimed to assess the effects of N,N-DMT and tryptamine (TRY) on IDO activity and the subsequent production of kynurenine. They found that both compounds acted as classical non-competitive inhibitors of IDO. N,N-DMT and TRY also increased cytotoxic activity in co-culture assays of A172 glioblastoma cells with PBMCs, suggesting that IDO inhibition by these compounds contributes to a more effective tumour-reactive response by PBMCs. IDO inhibitors have previously been found to produce antidepressant-like properties in animal models of sickness behaviour (O’Farrell and Harkin, Reference O’Farrell and Harkin2017; O’Connor et al., Reference O’Connor, Lawson, André, Moreau, Lestage, Castanon, Kelley and Dantzer2009).

These findings highlight the potential therapeutic application of psychedelics like N,N-DMT in enhancing immune responses against tumours, warranting further investigation into their use in cancer immunotherapy.

Effects of 5-HT2AR psychedelics on microglia

In LPS-stimulated microglial cultures derived from mice, N,N-DMT and psilocybin reduce expression levels of TLR4 (Kozłowska et al., Reference Kozłowska, Klimczak, Wiatr and Figiel2021). This suggests their potential to dampen the inflammatory cascade. Additionally, treatment with N,N-DMT and psilocybin altered microglial morphology, with treated cells exhibiting a more rounded and compact shape compared to controls. Furthermore, both drugs decreased the expression of co-stimulatory T-cell molecule CD80 and NF-κβ protein, suggestive of a decreased ability of presenting antigens to T-cells. Interestingly, treatment with psilocybin, but not N,N-DMT, reduced phagocytosis of healthy neurons by LPS-stimulated microglia, suggesting increased neuroprotection. Additionally, psilocybin upregulated a regulator of microglial phagocytosis and synaptic pruning, Triggering Receptor Expressed on Myeloid cells 2 (TREM2). Mutations in TREM2 have been associated to various neurodegenerative diseases including Alzheimer’s disease and Parkinson’s disease, and its pathway is implicated in anti-inflammatory and neuroprotective functions. If psilocybin upregulates a functional form of TREM2, this could produce potent anti-inflammatory effects, decreasing expression of pro-inflammatory proteins, thus ameliorating disease outcomes.

Proteome analysis of cerebral organoids treated with 5-MeO-DMT found significant alterations in genes associated with long term potentiation, dendritic spine formation, cellular protrusion formation, microtubule and cytoskeletal organisation, and mild activation of T lymphocyte differentiation (Dakic et al., Reference Dakic, Minardi Nascimento, Costa Sartore, Maciel, De Araujo, Ribeiro, Martins-de-Souza and Rehen2017). Following 5-MeO-DMT treatment, they also observed downregulation in protein expression of pathways associated to nuclear factor of activated T-cells (NFAT) and NF-κB signalling via TLR- and Gq-coupled receptors.

In TNF-α and IFN-γ stimulated C6-glioma cells, (R)-DOI produced a dose-dependent inhibition of cytokine-induced NO levels (Miller and Gonzalez, Reference Miller and Gonzalez1998). This effect was not seen when (R)-DOI was added more than 2 hours after the immune stimulation was added. This suggests that (R)-DOI suppression of NO occurs at a transcription regulation level.

Another study looking at NO release in LPS and IFN-γ stimulated BV-2 murine microglia showed that pre-treatment with psilocin reduced NO release. This effect was dependent on the 5-HT2AR, as inhibiting it with the antagonists cyproheptadine and risperidone prevents the reduction in NO release seen by psilocin (Wiens et al., Reference Wiens, Brooks, Riar, Greuel, Lindhout and Klegeris2024). Additionally, this study demonstrated that psilocin could reduce levels of ROS in human microglia-like cells primed with LPS and stimulated with the bacterial peptide N-formyl-Met-Leu-Phe (fMLP) (Wiens et al., Reference Wiens, Brooks, Riar, Greuel, Lindhout and Klegeris2024). Increases in ROS have been seen in clinical studies and preclinical models of neuropsychiatric disorders like major depressive disorder (MDD) and are also believed to play a role in the pathogenesis of neurodegenerative diseases (Simpson and Oliver, Reference Simpson and Oliver2020; Rossetti et al., Reference Rossetti, Paladini, Riva and Molteni2020). The capability of psychedelics to reduce the production of ROS might be one of the mechanisms with which they modulate the inflammatory response.

Although the research reviewed so far in cell cultures or organoids indicate immunomodulatory properties of some 5-HT2AR psychedelics, there is no clear consensus on whether psychedelics are pro- or anti-inflammatory. Additional research is warranted to definitively establish whether psychedelics can directly affect PBMC proliferation and function, microglial activation, and the production and release of cytokines in vivo and the mechanisms underlying such effects.

Immune organs and psychedelics

In LPS-stimulated mouse splenic cell cultures, low concentrations of LSD enhanced both baseline and IL-2-augmented NK cell function, but higher doses suppressed the NK response (House et al., Reference House, Thomas and Bhargava1994). These high LSD doses also led to suppression of B-cell function, macrophage function, reduced numbers of cytotoxic lymphocytes, and a reduction in Th1 cell IL-2 production.

In rat primary aortic smooth muscle cells treated with TNF-α, pre-treatment and co-treatment with (R)-DOI for 24 hours inhibited the TNF-α-induced inflammatory responses in a 5-HT2AR-dependent fashion (Yu et al., Reference Yu, Becnel, Zerfaoui, Rohatgi, Boulares and Nichols2008). (R)-DOI potently suppressed the expression of key inflammatory mediators, notably the cytokine IL-6, the intracellular adhesion molecules ICAM-1 and VCAM-1, important components of atherosclerotic plaque formation, compared to TNF-α stimulated cells alone. Notably, (R)-DOI by itself did not elicit any response. Furthermore, the authors showed similar inhibitory effects on TNF-α induced inflammation with other 5-HT2AR agonists, including a phenethylamine [(4-bromo-3,6-dimethoxybenzocyclobuten-1-yl) methylamine (2C-BCB)], and two indolealkylamines, lysergic acid 2,4-dimethylazetidide (LA-SS-Az) and LSD, suggesting a broader class effect.

Psilocybin was found to reduce expression levels of TNFα, IFN-γ, IL-6 and IL-8, in a human 3D epi-Intestinal tissue model treated with TFN-α/IFN-γ, providing additional evidence for the anti-inflammatory properties of psychedelics (Robinson et al., Reference Robinson, Li, Wang, Rahman, Gerasymchuk, Hudson, Kovalchuk and Kovalchuk2023).

The studies mentioned so far have used a range of concentrations of psychedelics. The clinically relevant psychedelics psilocybin, N,N-DMT, 5-MeO-DMT, and LSD were all used in supraphysiological concentrations (House et al., Reference House, Thomas and Bhargava1994; Kozłowska et al., Reference Kozłowska, Klimczak, Wiatr and Figiel2021; Szabo et al., Reference Szabo, Kovacs, Frecska and Rajnavolgyi2014; Rudin et al., Reference Rudin, Areesanan, Liechti and Grundemann2023), meaning that the doses used may have exceeded circulating concentrations as observed in clinical trials of psychedelics for the treatment of MDD (Ross et al., Reference Ross, Bossis, Guss, Agin-Liebes, Malone, Cohen, Mennenga, Belser, Kalliontzi, Babb, Su, Corby and Schmidt2016; Carhart-Harris et al., Reference Carhart-Harris, Roseman, Bolstridge, Demetriou, Pannekoek, Wall, Tanner, Kaelen, Mcgonigle, Murphy, Leech, Curran and Nutt2017; Carhart-Harris et al., Reference Carhart-Harris, Bolstridge, Day, Rucker, Watts, Erritzoe, Kaelen, Giribaldi, Bloomfield, Pilling, Rickard, Forbes, Feilding, Taylor, Curran and Nutt2018; Doss et al., Reference Doss, Považan, Rosenberg, Sepeda, Davis, Finan, Smith, Pekar, Barker, Griffiths and Barrett2021; Carhart-Harris et al., Reference Carhart-Harris, Giribaldi, Watts, Baker-Jones, Murphy-Beiner, Murphy, Martell, Blemings, Erritzoe and Nutt2021; Goodwin et al., Reference Goodwin, Aaronson, Alvarez, Arden, Baker, Bennett, Bird, Blom, Brennan, Brusch, Burke, Campbell-Coker, Carhart-Harris, Cattell, Daniel, Debattista, Dunlop, Eisen, Feifel, Forbes, Haumann, Hellerstein, Hoppe, Husain, Jelen, Kamphuis, Kawasaki, Kelly, Key, Kishon, Knatz Peck, Knight, Koolen, Lean, Licht, Maples-Keller, Mars, Marwood, Mcelhiney, Miller, Mirow, Mistry, Mletzko-Crowe, Modlin, Nielsen, Nielson, Offerhaus, O’keane, Palenicek, Printz, Rademaker, Van and Malievskaia2022, Reference Goodwin, Aaronson, Alvarez, Atli, Bennett, Croal, Debattista, Dunlop, Feifel, Hellerstein, Husain, Kelly, Lennard-Jones, Licht, Marwood, Mistry, Páleníček, Redjep, Repantis, Schoevers, Septimus, Simmons, Soares, Somers, Stansfield, Stuart, Tadley, Thiara, Tsai, Wahba, Williams, Winzer, Young, Young, Zisook and Malievskaia2023a, Reference Goodwin, Croal, Feifel, Kelly, Marwood, Mistry, O’keane, Peck, Simmons, Sisa, Stansfield, Tsai, Williams and Malievskaia2023c;Raison et al., Reference Raison, Sanacora, Woolley, Heinzerling, Dunlop, Brown, Kakar, Hassman, Trivedi, Robison, Gukasyan, Nayak, Hu, O’donnell, Kelmendi, Sloshower, Penn, Bradley, Kelly, Mletzko, Nicholas, Hutson, Tarpley, Utzinger, Lenoch, Warchol, Gapasin, Davis, Nelson-Douthit, Wilson, Brown, Linton, Ross and Griffiths2023).

Receptors mediating the immunomodulatory effects of 5-HT2AR psychedelics

Although the involvement of the 5-HT2AR in the immunomodulatory effects of psychedelics has not yet been fully elucidated, the studies explored so far have reported anti-inflammatory effects which appear to be 5-HT2AR-dependent (Yu et al., Reference Yu, Becnel, Zerfaoui, Rohatgi, Boulares and Nichols2008; Nau et al., Reference Nau, Yu, Martin and Nichols2013). The 5-HT2AR is likely the major binding site which is most closely associated with the anti-inflammatory effects observed in preclinical studies. However, 5-HT2AR psychedelics have been reported to bind other receptors.

Sigma-1 receptor

Alongside N,N-DMT, psilocybin and the non-classical psychedelic ibogaine are hypothesised to bind Sig-1R in the CNS and on circulating immune cells (Sershen et al., Reference Sershen, Hashim and Lajtha1996; Mason et al., Reference Mason, Szabo, Kuypers, Mallaroni, De La Torre Fornell, Reckweg, Tse, Hutten, Feilding and Ramaekers2023). Sig-1R is highly enriched at the mitochondria-associated endoplasmic reticulum (ER) membrane and is expressed in immune cells (Zhang et al., Reference Zhang, Li, Tao, Qin, Chen, Yang, Chen, Liu, Dai and Zhen2023a), and throughout the CNS (Shi et al., Reference Shi, Chen, Chen, Yang, Yue, Zhang and Chen2021). It is believed to exert neuroprotective effects by suppressing ER stress, regulating Ca2+ influx into mitochondria and adenosine triphosphate (ATP) synthesis, preventing excitotoxicity and oxidative stress by regulating key ER membrane proteins and downstream transcription factors NF-κB and X-box binding protein 1 (XBP-1) (Hayashi and Su, Reference Hayashi and Su2007; Ho et al., Reference Ho, Xu and Thibault2018; Hayashi, Reference Hayashi2019). Additionally, Sig-1R plays crucial roles in neuronal differentiation, cell survival, and immune modulation, promoting anti-inflammatory actions and BDNF secretion, as well as regulating neuroplasticity and glial reactivity in rodent models (Peviani et al., Reference Peviani, Salvaneschi, Bontempi, Petese, Manzo, Rossi, Salmona, Collina, Bigini and Curti2014; Ruiz-Cantero et al., Reference Ruiz-Cantero, Gonzalez-Cano, Tejada, Santos-Caballero, Perazzoli, Nieto and Cobos2021).

Overexpression of astrocytic Sig-1R using an adeno-associated virus in cultured primary astrocytes was found to attenuate LPS-driven IL-1β, TNF-α, and inducible nitric oxide synthase (iNOS) production, while increasing BDNF production and reducing astrocyte and microglial activation. Overexpression of astrocytic Sig-1R in a mouse model also reduced LPS-induced depressive-like behaviour and improved memory function (Guo et al., Reference Guo, Gao, Gao, Jia, Ni, Han and Wang2021).

Macrophages and microglia activation states have been organised in a spectrum based on protein expression ranging from an M1 pro-inflammatory state to an M2 anti-inflammatory state. Activation of Sig-1R promotes the microglial M2 state while inhibiting the M1 state, and promotes astrocytic glial fibrillary acidic protein (GFAP) expression and BDNF secretion in response to inflammatory stimuli (Jia et al., Reference Jia, Cheng, Wang and Zhen2018).

It has been reported that Sig-1R activation suppresses the ability of microglia to rearrange their actin cytoskeleton, migrate, and release cytokines in response to ATP, monocyte chemoattractant protein 1 (MCP-1), and LPS in primary glial cultures (Hall et al., Reference Hall, Herrera, Ajmo, Cuevas and Pennypacker2009). In the same study, it was observed that stimulation of Sig-1R suppressed both transient and sustained intracellular Ca2+ elevations associated with the microglial response to these activators. Furthermore Sig-1R activation suppressed membrane ruffling, preventing microglial migration and stress-induced actin reorganisation in the cell, in a Ca2+-independent manner, suggesting that interactions between microglia and the Sig-1R may be multifaceted.

Shen et al. (Reference Shen, Wang, Chou, Liou, Yen, Wang and Liao2008) assessed treatment with the Sig-1R agonist dimemorfan in an inflammatory ischaemic stroke model in rats. They observed inhibited expression of MCP-1 and IL-1β, decreased neutrophil infiltration, decreased activation of p38 MAPK, NF-κB, and STAT1, and decreased expression of neuronal and inducible NOS in the cortex. These changes were further attributed to decreased extracellular glutamate accumulation. Rather than directly modulating microglia, Sig-1R might be influencing microglial function indirectly by regulating glutamate concentrations extrasynaptically.

Tropomyosin receptor kinase B receptor

5-HT2AR psychedelics might also influence the immune system by interacting with Tropomyosin receptor kinase B (TrkB) receptors, to which BDNF and other neurotrophins can bind. Neurotrophins are signalling molecules primarily found in the peripheral and CNS and are primarily stored in platelets with platelet concentrations reaching up to 1000 times the concentrations found in neurons (Boukhatem et al., Reference Boukhatem, Fleury, Welman, Le Blanc, Thys, Freson, Best, Wurdinger, Allen and Lordkipanidze2021). TrkB mediates processes such as synaptogenesis, neuroplasticity, apoptosis, mammalian target of rapamycin (mTOR) signalling pathway activation, and phospholipase C gamma (PLC-γ1) activity (Colle et al., Reference Colle, Deflesselle, Martin, David, Hardy, Taranu, Falissard, Verstuyft and Corruble2015; Zhang et al., Reference Zhang, Yao and Hashimoto2016).

It has also been implicated in the regulation of the immune system. Subsets of T-cells, macrophages, and DCs express TrkB receptors (Ciriaco et al., Reference Ciriaco, Dall’aglio, Hannestad, Huerta, Laura, Germana and Vega1996; De Santi et al., Reference De Santi, Annunziata, Sessa and Bramanti2009; Kozlov et al., Reference Kozlov, Grechko, Chegodaev, Wu and Orekhov2020). BDNF is produced by CD4+ and CD8+ T-cells, B-cells, and monocytes, and has been shown to promote neuronal survival in vitro (Kerschensteiner et al., Reference Kerschensteiner, Gallmeier, Behrens, Leal, Misgeld, Klinkert, Kolbeck, Hoppe, Oropeza-Wekerle, Bartke, Stadelmann, Lassmann, Wekerle and Hohlfeld1999). Reduced TrkB signalling and BDNF signalling in the brain are observed in response to LPS administration and Poly(I:C) administration in rodents (Gibney et al., Reference Gibney, Mcguinness, Prendergast, Harkin and Connor2013), as well as increased depressive-like behaviour, which can be rescued by administration of a TrkB agonist (Zhang et al., Reference Zhang, Wu, Fujita, Yao, Ren, Yang, Li, Shirayama and Hashimoto2014).

Psilocybin and LSD directly bind to the TrkB receptor with 1000-fold higher affinity than SSRIs (Moliner et al., Reference Moliner, Girych, Brunello, Kovaleva, Biojone, Enkavi, Antenucci, Kot, Goncharuk, Kaurinkoski, Kuutti, Fred, Elsila, Sakson, Cannarozzo, Diniz, Seiffert, Rubiolo, Haapaniemi, Meshi, Nagaeva, Ohman, Rog, Kankuri, Vilar, Varjosalo, Korpi, Permi, Mineev, Saarma, Vattulainen, Casarotto and Castren2023). Psychedelics increase the neuronal surface retention of TrkB and promote BDNF downstream signalling (Moliner et al., Reference Moliner, Girych, Brunello, Kovaleva, Biojone, Enkavi, Antenucci, Kot, Goncharuk, Kaurinkoski, Kuutti, Fred, Elsila, Sakson, Cannarozzo, Diniz, Seiffert, Rubiolo, Haapaniemi, Meshi, Nagaeva, Ohman, Rog, Kankuri, Vilar, Varjosalo, Korpi, Permi, Mineev, Saarma, Vattulainen, Casarotto and Castren2023). This binding increases TrkB interaction with PLC-γ1, a critical regulator of NF-κB, MAPK/ERK signalling, calcium homeostasis via IP3, and activated T-cell signalling, via the NFAT family of transcription factors (Tao et al., Reference Tao, Han, Wang, Wang, Zhang, Liu, Fan, Liu, Liu, Guo, Lee, Aksentijevich and Zhou2023).

While the exact mechanisms are unclear, preliminary studies suggest TrkB might be a key player in how 5-HT2AR psychedelics influence the immune system. Psychedelics bind to TrkB with greater affinity than traditional antidepressants, influencing cellular signalling pathways, potentially affecting immune cell function. Further research is needed to fully understand these effects.

Psychedelic modulation of immune responses in whole systems

Isolated systems cannot fully capture the interactions between different tissues and organs within a whole organism. Preclinical studies in animals and clinical studies are crucial to determine whether 5-HT2AR psychedelics directly impact the immune system or exert an indirect immunomodulatory effect mediated by the CNS, the HPA or sympathetic-adrenal-medullary (SAM) axes, or other cellular pathways. These in vivo whole system investigations provide insights into how psychedelics interact with the immune system and shed light on the translational potential of 5-HT2AR psychedelics for immune modulation. This section reviews recent studies assessing the immune system’s response to 5-HT2AR psychedelics conducted in animal models and in human clinical trials.

Animal models

Preclinical models are not perfect representations of human diseases; however, they offer many advantages to elucidate the mechanisms underlying the effect of psychedelics on the CNS and the immune system. These advantages include a controlled environment to isolate specific factors, the accessibility of cells and tissues at different life stages to track progression, genetic manipulation, and the means to test experimental drugs on bi-directional immune-nervous system interactions. There have been several studies investigating the effect of psilocybin, (R)-DOI, LSD, ayahuasca, and novel psychedelics, on immune responses and mediators in vivo, in healthy animals as well as in disease models.

In an exploratory study, Bove and Mokler (Reference Bove and Mokler2022) administered psilocybin to healthy female rats and measured pro- and anti-inflammatory markers in peripheral serum. Psilocybin induced the release of pro-inflammatory factors IL-1β, TNF-α, IL-13, MCP-1, and C-X-C motif chemokine ligand 10 (CXCL10), and anti-inflammatory factors IFN-γ, IL-10, granulocyte colony-stimulating factor (G-CSF), into the serum, consistent with a generalised activation of the immune system. This difference was persistent seven days after psilocybin injection, however, as the authors note, high variability in the results limit the conclusions that can be drawn from this study. In support of this, Custodio et al. (Reference Custodio, Ortiz, Lee, Sayson, Kim, Lee, Kim, Cheong and Kim2023) reported neurotoxic properties of (R)-DOI and other 5-HT2CR agonists in healthy mice, with increased expression of ionised calcium binding adaptor molecule 1 (Iba1), IL-6, and TNF-α, however, these effects were reported at very high doses (30 mg/kg).

LSD, administered to healthy mice, was found to reduce hippocampal levels of kynurenine, altering the kynurenine/tryptophan and the kynurenine/5-HT ratios significantly, without altering levels of hippocampal 5-HT (Inserra et al., Reference Inserra, Giorgini, Lacroix, Bertazzo, Choo, Markopolous, Grant, Abolghasemi, De Gregorio, Flamand, Rogers, Comai, Silvestri, Gobbi and Di Marzo2023). This highlights psychedelics’ potential therapeutic role in disorders with a dysregulated kynurenine pathway, which is discussed in detail later. The same group also observed modulation of endocannabinoid-related metabolites in the hippocampus following LSD administration. These effects were correlated with increased sociability (Inserra et al., Reference Inserra, Giorgini, Lacroix, Bertazzo, Choo, Markopolous, Grant, Abolghasemi, De Gregorio, Flamand, Rogers, Comai, Silvestri, Gobbi and Di Marzo2023).

In the context of disease models, 5-HT2AR psychedelics have consistently been reported to suppress the immune response.

Mice injected with sheep erythrocytes – activating both T and B lymphocytes (McAllister et al., Reference Mcallister, Apgar, Leung, Rickert and Jellusova2017) – and subsequently treated with (R)-DOI exhibited a marked suppression of the immune response (Davydova et al., Reference Davydova, Cheido, Gevorgyan and Idova2010). This was evidenced by a significant decrease in CD8+ T-cells, both in peripheral blood and the spleen. Notably, administration of ketanserin, a 5-HT2AR antagonist, produced the opposite effect. These findings strongly support the involvement of 5-HT2AR in mechanisms leading to immunosuppression.

Nichols and colleagues have been able to demonstrate anti-inflammatory properties for psychedelics specifically via 5-HT2 receptor sub-type activation using various animal models. For example, a rapid immune response can be induced after systemic administration of exogenous TNF-α, with an increase in circulating IL-6 and VCAM-1. Administration of (R)-DOI efficiently blocks these effects of TNF-α, with reduced IL-6 and VCAM-1 expression in the aortic arch and small intestine (Nau et al., Reference Nau, Yu, Martin and Nichols2013). The mechanism underlying this potent anti-inflammatory response appears to be the activation of 5-HT2AR.

Ovalbumin (OVA) induced acute allergic asthma in mice provides a model to study immune-modulatory properties of experimental compounds. Using this model, (R)-DOI was found to attenuate various asthma parameters in response to a non-selective muscarinic receptor agonist methacholine, including elevated airway hyperresponsiveness and pulmonary inflammation (Flanagan et al., Reference Flanagan, Sebastian, Battaglia, Foster, Cormier and Nichols2019a). Interestingly, the authors measured a reduction in expression of certain pro-inflammatory cytokines such as IL-15 and IL-9, but an increase in expression of other pro-inflammatory cytokines IL-13 and IL-33 which are known contributors to a persistent chronic asthma state. In a later study using the OVA model, (R)-DOI was found to prevent OVA-induced increases in mRNA levels for pro-inflammatory cytokines IL-5, IL-6, TNF-α and IL-1β (Flanagan et al., Reference Flanagan, Billac, Landry, Sebastian, Cormier and Nichols2021). The authors identified 2,5-dimethoxyphenethylamine (2C-H) from 21 different 5-HT2A agonists as the key pharmacophore mediating effective anti-inflammatory properties. This may be a useful starting point for future drug development focusing on anti-inflammatory action.

More recently, Flanagan et al. (Reference Flanagan, Foster, Galbato, Lum, Louie, Song, Halberstadt, Billac and Nichols2024) aimed to identify key structural components of 5-HT2AR agonists mediating their anti-inflammatory effects focusing on their ability to suppress Arginase-1 (Arg1) expression in peripheral tissues. Arg1 catalyses the conversion of L-arginine into L-ornithine and urea. Upregulation of Arg1 has been shown to contribute to inflammation and airway obstruction (North et al., Reference North, Khanna, Marsden, Grasemann and Scott2009), whereas in the absence of Arg1 activity, L-arginine is instead converted into NO via NOS activity, contributing to relaxation of bronchial smooth muscle and inhibition of inflammation (Cloots et al., Reference Cloots, Sankaranarayanan, De Theije, Poynter, Terwindt, Van Dijk, Hakvoort, Lamers and Kohler2013). Despite having similar in vitro activity on 5-HT2AR and similar behavioural potency, the novel agonist (R)-2,5-dimethoxy-4-trifluoromethylamphetamine [(R)-DOTFM] did not exhibit anti-inflammatory properties like (R)-DOI. Only (R)-DOI led to significant reductions in levels of Arg1, IL-6, and CXCL10. The authors argue that the different effects of (R)-DOTFM and (R)-DOI may originate from differences in receptor stabilisation and conformation which could lead to separate downstream effectors and pathways being recruited. These experiments are the first to study and identify differences in functional selectivity of 5-HT2AR agonists in peripheral tissues, indicating molecular and cellular sensitivities underlying anti-inflammatory properties of serotonergic 5-HT2AR psychedelics. This may inform future studies to identify novel anti-inflammatory compounds devoid of subjective psychedelic effects.

There are additional animal models that have provided insights into the anti-inflammatory effects of psychedelics. In a mouse model of cardiovascular disease [Apolipoprotein E (ApoE)-/- on a high fat diet], continuous systemic infusion of low-dose (R)-DOI resulted in significant reductions in mRNA expression of pro-inflammatory markers, including IL-6, TNF-α and CXCL10 (Flanagan et al., Reference Flanagan, Sebastian, Battaglia, Foster, Maillet and Nichols2019b). (R)-DOI treatment normalised glucose homeostasis and reduced circulating cholesterol. Although this study is not directly comparable, research in human vascular smooth cell cultures found that serotonin increased IL-6 production through 5-HT2AR, suggesting that psychedelics and serotonin may activate different downstream cellular pathways (Ito et al., Reference Ito, Ikeda, Shimpo, Yamamoto and Shimada2000).

Psilocybin has been shown to have anti-inflammatory properties in different models of peripheral inflammation. In mice systemically injected with LPS, psilocybin treatment, pre- or post-administration of LPS, was found to reduce expression of IL-6 and TNF-α cytokines in homogenised brain tissue and in peripheral blood (Zanikov et al., Reference Zanikov, Gerasymchuk, Ghasemi Gojani, Robinson, Asghari, Groves, Haselhorst, Nandakumar, Stahl, Cameron, Li, Rodriguez-Juarez, Snelling, Hudson, Fiselier, Kovalchuk and Kovalchuk2023). Inflammation can also be induced via the gut-brain-axis by orally administering dextran sulphate sodium (DSS) to mice or rats to induce acute colitis. This has been shown to lead to immune responses in the gastrointestinal tract as well as in the CNS (Dempsey et al., Reference Dempsey, Abautret-Daly, Docherty, Medina and Harkin2019). In a recent study using this model, post-treatment with psilocybin and eugenol – a positive allosteric modulator of the gamma-aminobutyric acid (GABA)-A receptor – reduced expression of pro-inflammatory cytokines and markers IL-1β, IL-6, and COX-2 in the brain (Zanikov et al., Reference Zanikov, Gerasymchuk, Robinson, Gojani, Asghari, Groves, Cameron, Rodriguez-Juarez, Snelling, Hudson, Fiselier, Kovalchuk and Kovalchuk2024). These results demonstrate a clear immunomodulatory property of psilocybin in the context of induced systemic inflammation.

Streptozotocin (STZ), a glucosamine–nitrosourea compound leading to an insulin-resistant brain state, has been previously used to induce an Alzheimer’s disease model in the rat as intraperitoneal or intracerebroventricular injection of STZ leads to production of amyloid-beta (Kadhim et al., Reference Kadhim, Al-Mumen, Nahi and Hamidi2022). Using this model, Afshar et al. (Reference Afshar, Shahidi, Rohani, Soleimani Asl and Komaki2019) reported that both a selective antagonist of 5-HT1AR (NAD-299) and a selective agonist of 5-HT2AR (TCB-2) reduced oxidative stress in the hippocampus and provided neuroprotection. In support of this, a separate study using an Aβ-induced mouse model of AD found that N,N-DMT alleviated astrocytic activation and astrogliosis in the hippocampus and dentate gyrus measured via GFAP immunostaining (Borbély et al., Reference Borbély, Varga, Szögi, Schuster, Bozsó, Penke and Fülöp2022).

N,N-DMT administration also produced anti-inflammatory and pro-neurotrophic effects in a rat ischaemic brain injury model. Pretreatment with N,N-DMT led to lower expression of both mRNA and protein levels of a key activator of the apoptotic cascade (Apoptotic Protease Activating Factor 1 – APAF-1), and higher BNDF expression in this model, as well as a reduction in TNF-α, IL-1β, IL-6 and an increase in IL-10 levels (Nardai et al., Reference Nardai, László, Szabó, Alpár, Hanics, Zahola, Merkely, Frecska and Nagy2020).

Szabo et al. (Reference Szabo, Varga, Dvoracsko, Farkas, Kormoczi, Berkecz, Kecskes, Menyhart, Frank, Hantosi, Cozzi, Frecska, Tomboly, Krizbai, Bari and Farkas2021) also demonstrated a neuroprotective effect in the ischaemic rat brain following N,N-DMT administration, with a reduction in the number of apoptotic cells and improved astrocytic survival. However, they demonstrated this effect to be mediated by Sig-1R activation. In a more recent study, pre-treatment with psilocybin reduced brain infarction and neurological deficits following induced stroke in rats, whereas post-treatment also led to downregulation of Iba1 (Yu et al., Reference Yu, Wu, Wang, Bae, Chianelli, Bambakidis and Wang2024). These effects were attenuated by the BDNF inhibitor and TrkB antagonist ANA12.

A recent study used caecal ligation and puncture to model sepsis in rats and assess how ayahuasca might produce anti-inflammatory and neuroprotective effects (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). Ayahuasca pre-treatment increased levels of IL-4 and BDNF in the cortex, while enhancing neutrophil activation and decreasing nitric oxide signalling. At a behavioural level, ayahuasca reduced anxiety-like measures in behavioural tests, suggesting that it can prevent sepsis-induced neuroinflammatory and oxidative stress, and reduce anxiety-like behaviour.

Overall, these results suggest that modulation of serotonin receptors, and possibly (SIG-1R), offer protection against neuroinflammation induced by various disease salient stimuli, highlighting the potential role of the serotonergic system and the Sig-1R in inflammation associated neurodegenerative processes.

Studies have also found that psychedelics modulate the expression of immunological factors reported to be increased in stress associated behavioural paradigms. Kelley et al. (Reference Kelley, Venable, Destouni, Billac, Ebenezer, Stadler, Nichols, Barker and Francis2022) measured an increased expression of IL-1β and its receptor IL1r1 in a rat model for post-traumatic stress disorder in the prefrontal cortex. They found that IL1r1 was significantly reduced after treatment with N,N-DMT, or a combination of N,N-DMT and harmaline, as well as TLR4, TLR6, and TLR7 (Kelley et al., Reference Kelley, Venable, Destouni, Billac, Ebenezer, Stadler, Nichols, Barker and Francis2022). In response to repeated social aggression, stressed mice exhibit elevated cytokine gene expression and increased TNFα levels in plasma and CSF (Krupp et al., Reference Krupp, Yaeger, Ledesma, Withanage, Gale, Howe, Allen, Sathyanesan, Newton and Summers2024). A single administration of (R)-DOI after stress induction reduced plasma and limbic brain levels of TNFα and promoted escape, a dynamic coping strategy, indicating anxiolytic effects.

This emerging evidence suggests potent immunomodulatory properties of classical psychedelics, particularly (R)-DOI and psilocybin. In vivo studies in both mice and rats have demonstrated that 5-HT2AR agonists can influence responses in immune-challenge models via a suppression of factors associated with the inflammatory response system (Nau et al., Reference Nau, Yu, Martin and Nichols2013; Flanagan et al., Reference Flanagan, Sebastian, Battaglia, Foster, Cormier and Nichols2019a, b, Reference Flanagan, Billac, Landry, Sebastian, Cormier and Nichols2021; Nardai et al., Reference Nardai, László, Szabó, Alpár, Hanics, Zahola, Merkely, Frecska and Nagy2020; Zanikov et al., Reference Zanikov, Gerasymchuk, Ghasemi Gojani, Robinson, Asghari, Groves, Haselhorst, Nandakumar, Stahl, Cameron, Li, Rodriguez-Juarez, Snelling, Hudson, Fiselier, Kovalchuk and Kovalchuk2023), a decrease in CD8+ T-cells proportion (Davydova et al., Reference Davydova, Cheido, Gevorgyan and Idova2010), and a reduction in oxidative stress (Afshar et al., Reference Afshar, Shahidi, Rohani, Soleimani Asl and Komaki2019; Szabo et al., Reference Szabo, Varga, Dvoracsko, Farkas, Kormoczi, Berkecz, Kecskes, Menyhart, Frank, Hantosi, Cozzi, Frecska, Tomboly, Krizbai, Bari and Farkas2021). Interestingly, as referred to earlier, psilocybin led to a general activation of the immune system with a release of pro- and anti-inflammatory cytokines in healthy unstimulated rats, suggesting possibly different responses based on activation state (Bove and Mokler, Reference Bove and Mokler2022). Similarly, (R)-DOI treatment has been associated with reduced response induced in the OVA model, such as a significant reduction in mucus production and pulmonary inflammation (Flanagan et al., Reference Flanagan, Sebastian, Battaglia, Foster, Cormier and Nichols2019a), and normalised glucose homeostasis in the ApoE-/- model, in mice (Flanagan et al., Reference Flanagan, Sebastian, Battaglia, Foster, Maillet and Nichols2019b).

According to these preclinical in vivo studies, psychedelics appear to have a dual effect on the immune system. Under normal conditions, without external triggers, psychedelics tend to stimulate immune system activity. However, when the immune system is already activated by external factors, such as with LPS administration, psychedelics seem to inhibit the immune response to these triggers. This suggests that psychedelics may have a regulatory effect on immune function, potentially stimulating or suppressing immune activity depending on the body’s current state.

Human studies

Following administration of a high dose of psilocybin in healthy volunteers, Hasler et al. (Reference Hasler, Grimberg, Benz, Huber and Vollenweider2004) observed a significant rise in thyroid–stimulating hormone (TSH), prolactin, adrenocorticotropic hormone (ACTH), and cortisol. Interestingly, this increased hormonal release wasn’t associated with heightened anxiety. The authors suggest this observation aligns with known effects of 5-HT2 receptor stimulation, which can trigger the HPA axis, leading to elevated ACTH and cortisol levels. Given that cortisol is known for its anti-inflammatory properties (Rhen and Cidlowski, Reference Rhen and Cidlowski2005), it might be expected that elevated cortisol levels could influence inflammatory markers. Conversely, Burmester et al. (Reference Burmester, Madsen, Szabo, Aripaka, Stenbaek, Frokjaer, Elfving, Mikkelsen, Knudsen and Fisher2023) reported no change in circulating inflammatory markers [C-reactive protein (CRP) and TNF-α] after a single psilocybin dose in 16 healthy participants. Similarly, four weeks after a psilocybin dose, a transient increase in peripheral cytokine production was reported. However, this increase was not consistent across different patient populations which included healthy participants, patients with depression, anxiety, and cancers of various types (DiRenzo et al., Reference DiRenzo, Barrett, Perin, Darrah, Christopher-Stine and Griffiths2024).

Studies on N,N-DMT, a key component of ayahuasca, indicate its potential ability to acutely influence neuroendocrine and immune markers. After N,N-DMT administration using freeze-dried ayahuasca capsules, Dos Santos et al. (Reference Dos Santos, Valle, Bouso, Nomdedéu, Rodríguez-Espinosa, McIlhenny, Barker, Barbanoj and Riba2011) observed decreased CD4+ and CD3+ cell populations in healthy individuals and those with treatment-resistant depression (TRD), increased proportion of NK cells, and increased prolactin and cortisol levels. Notably, this modulatory effect was transient, with peak effects occurring two hours after intake and returning to baseline within 24 hours. A larger study by Galvao et al. (Reference Galvao, De Almeida, Silva, Freire, Palhano-Fontes, Onias, Arcoverde, Maia-De-Oliveira, De Araujo, Lobao-Soares and Galvao-Coelho2018) demonstrated a rise in salivary cortisol levels in both healthy controls and patients with TRD during the ayahuasca session compared to placebo, returning to baseline within 48 hours. Interestingly, subsequent research revealed a significant correlation between greater reductions in CRP and lower depressive symptoms at 48 hours post-ayahuasca in both patient and control groups (Galvao-Coelho et al., Reference Galvao-Coelho, De Menezes Galvao, De Almeida, Palhano-Fontes, Campos Braga, Lobao Soares, Maia-De-Oliveira, Perkins, Sarris and De Araujo2020). These findings suggest a potential link between ayahuasca’s antidepressant action and its effect on neuroendocrine and immune systems.

Preliminary evidence also indicates that 5-MeO-DMT has immunomodulatory properties. In experienced psychedelic users, a significant decrease in salivary IL-6 and a rise in cortisol was measured after 5-MeO-DMT inhalation (Uthaug et al., Reference Uthaug, Lancelotta, Szabo, Davis, Riba and Ramaekers2020). However, further investigation is required as this study lacked a suitable placebo, had a small sample size, and used variable dosing. 5-MeO-DMT is similarly active at 5-HT2A and 5-HT1A receptors and binds to 5-HT1AR as a biased agonist, similar to the partial agonist and anxiolytic buspirone (Warren et al., Reference Warren, Lankri, Cunningham, Serrano, Parise, Kruegel, Duggan, Zilberg, Capper, Havel, Russo, Sames and Wacker2024).

Table 1. Summary of in vitro studies having measured immunomodulatory properties of 5-HT2AR psychedelics

Table 2. Summary of in vivo studies, both in rodents and humans, having measured immunomodulatory properties of 5-HT2AR psychedelics

Translational application for disorders in humans

Research into the possible therapeutic use of psychedelics has surged in recent years, both in clinical and preclinical research. Landmark clinical trials have found that a single dose of psilocybin is sufficient for a rapid and long-lasting anti-depressant effect in patients with MDD and TRD, even with concomitant SSRI medication (Ross et al., Reference Ross, Bossis, Guss, Agin-Liebes, Malone, Cohen, Mennenga, Belser, Kalliontzi, Babb, Su, Corby and Schmidt2016; Griffiths et al., Reference Griffiths, Johnson, Carducci, Umbricht, Richards, Richards, Cosimano and Klinedinst2016; Carhart-Harris et al., Reference Carhart-Harris, Roseman, Bolstridge, Demetriou, Pannekoek, Wall, Tanner, Kaelen, Mcgonigle, Murphy, Leech, Curran and Nutt2017, Reference Carhart-Harris, Bolstridge, Day, Rucker, Watts, Erritzoe, Kaelen, Giribaldi, Bloomfield, Pilling, Rickard, Forbes, Feilding, Taylor, Curran and Nutt2018, Reference Carhart-Harris, Giribaldi, Watts, Baker-Jones, Murphy-Beiner, Murphy, Martell, Blemings, Erritzoe and Nutt2021; Goodwin et al., Reference Goodwin, Aaronson, Alvarez, Arden, Baker, Bennett, Bird, Blom, Brennan, Brusch, Burke, Campbell-Coker, Carhart-Harris, Cattell, Daniel, Debattista, Dunlop, Eisen, Feifel, Forbes, Haumann, Hellerstein, Hoppe, Husain, Jelen, Kamphuis, Kawasaki, Kelly, Key, Kishon, Knatz Peck, Knight, Koolen, Lean, Licht, Maples-Keller, Mars, Marwood, Mcelhiney, Miller, Mirow, Mistry, Mletzko-Crowe, Modlin, Nielsen, Nielson, Offerhaus, O’keane, Palenicek, Printz, Rademaker, Van and Malievskaia2022, Reference Goodwin, Croal, Feifel, Kelly, Marwood, Mistry, O’keane, Peck, Simmons, Sisa, Stansfield, Tsai, Williams and Malievskaia2023b ; Raison et al., Reference Raison, Sanacora, Woolley, Heinzerling, Dunlop, Brown, Kakar, Hassman, Trivedi, Robison, Gukasyan, Nayak, Hu, O’donnell, Kelmendi, Sloshower, Penn, Bradley, Kelly, Mletzko, Nicholas, Hutson, Tarpley, Utzinger, Lenoch, Warchol, Gapasin, Davis, Nelson-Douthit, Wilson, Brown, Linton, Ross and Griffiths2023). It was recently reported that the intensity of the psychedelic experience is correlated with depression response as measured by the Montgomery–Åsberg Depression Rating Scale (MADRS) (Goodwin et al., Reference Goodwin, Aaronson, Alvarez, Carhart-Harris, Chai-Rees, Croal, Debattista, Dunlop, Feifel, Hellerstein, Husain, Kelly, Kirlic, Licht, Marwood, Meyer, Mistry, Nowakowska, Palenicek, Repantis, Schoevers, Simmons, Somers, Teoh, Tsai, Wahba, Williams, Young, Young, Zisook and Malievskaia2025).

Results reported in the largest phase II double-blind, parallel group, randomised clinical trial so far, suggest that previously reported antidepressant effects may have been inflated. The 25 mg dose of psilocybin had a 37% response rate three weeks after dosing according to the MADRS, compared to 18% with the 1 mg dose (Goodwin et al., Reference Goodwin, Aaronson, Alvarez, Arden, Baker, Bennett, Bird, Blom, Brennan, Brusch, Burke, Campbell-Coker, Carhart-Harris, Cattell, Daniel, Debattista, Dunlop, Eisen, Feifel, Forbes, Haumann, Hellerstein, Hoppe, Husain, Jelen, Kamphuis, Kawasaki, Kelly, Key, Kishon, Knatz Peck, Knight, Koolen, Lean, Licht, Maples-Keller, Mars, Marwood, Mcelhiney, Miller, Mirow, Mistry, Mletzko-Crowe, Modlin, Nielsen, Nielson, Offerhaus, O’keane, Palenicek, Printz, Rademaker, Van and Malievskaia2022). Small sample sizes and other limitations commonly found in psychedelic research have led to larger and more robust phase III clinical trials that are underway at time of writing. However, results so far are promising for treatment of psychiatric illnesses.

Recent evidence suggests that psychedelics and psychedelic-like agents, including ketamine, may serve as novel interventions for patients who do not respond to conventional treatments (Kiraly et al., Reference Kiraly, Horn, Van Dam, Costi, Schwartz, Kim-Schulze, Patel, Hodes, Russo, Merad, Iosifescu, Charney and Murrough2017; Quintanilla et al., Reference Quintanilla, Zarate and Pillai2024). Current research is exploring psilocybin’s potential to treat neurological conditions involving immune system imbalances, such as fibromyalgia and functional neurological disorder (Butler et al., Reference Butler, Bird, Maggio, Durden, Modlin, Campbell-Coker, Edwards, Pick, Millman, Lowery, Bhagavan, Kanaan, Golder, Mildon, Mehta, Rucker and Nicholson2024; Bornemann et al., Reference Bornemann, Close, Ahmad, Barba, Godfrey, Macdonald, Erritzoe, Nutt and Carhart-Harris2024). The question remains as to which patients will see the most beneficial effects from psychedelics, and whether there may be biomarkers that could be used to predict responses from psychedelic-assisted therapy. As explored in this review, these biomarkers could include immune, hormonal, neuronal, or even microbiota parameters (Kelly et al., Reference Kelly, Clarke, Harkin, Corr, Galvin, Pradeep, Cryan, O’keane and Dinan2023). Future biomarker detection could use less invasive methods, such as wearable device technology for example.

Additionally, the optimal treatment regimen for psychedelic therapy remains undetermined. Although major clinical studies have used either one or two dosing sessions, the potential for periodic retreatment – perhaps every six to twelve months – for those who have previously gone through psychedelic-assisted therapy needs to be further investigated. In any case, psychedelic-assisted therapy will inherently be different from conventional antidepressant treatments that require daily consumption and often produce persistent side effects. Additionally, microdosing of psychedelics may offer similar beneficial effects without the acute subjective effects, although further double-blind research is required to validate this regimen (Kinderlehrer, Reference Kinderlehrer2025).

The immune system and the CNS maintain a dynamic interplay mediated through immune cells and organs, and the HPA axis (Bellavance and Rivest, Reference Bellavance and Rivest2014; Malek et al., Reference Malek, Ebadzadeh, Safabakhsh, Razavi and Zaringhalam2015). Dysregulation of communication networks between these systems is implicated in a spectrum of mental health disorders, including depression (Arteaga-Henriquez et al., Reference Arteaga-Henriquez, Simon, Burger, Weidinger, Wijkhuijs, Arolt, Birkenhager, Musil, Muller and Drexhage2019; Lynall et al., Reference Lynall, Turner, Bhatti, Cavanagh, de Boer, Mondelli, Jones, Drevets, Cowen, Harrison and Pariante2020; Lamers et al., Reference Lamers, Milaneschi, Vinkers, Schoevers, Giltay and Penninx2020; Zeng et al., Reference Zeng, Chourpiliadis, Hammar, Seitz, Valdimarsdottir, Fang, Song and Wei2024; Jarkas et al., Reference Jarkas, Villeneuve, Daneshmend, Villeneuve and Mcquaid2024; Hagenberg et al., Reference Hagenberg, Be, Group, Bruckl, Erhart, Kopf-Beck, Kodel, Rehawi, Roh-Karamihalev, Sauer, Yusupov, Rex-Haffner, Spoormaker, Samann, Binder and Knauer-Arloth2025; Penninx et al., Reference Penninx, Lamers, Jansen, Berk, Khandaker, De Picker and Milaneschi2025). A meta-analysis conducted by Osimo et al. (Reference Osimo, Baxter, Lewis, Jones and Khandaker2019) revealed that approximately 25% of individuals diagnosed with depression exhibit signs of low-grade inflammation. In lower-middle-income countries, the prevalence of elevated plasma CRP could be higher, with 87% of participants with treatment-resistant depression displaying low-grade inflammation (Fellows et al., Reference Fellows, Jones, Hodsoll, Husain, Khoso, Young, Chaudhry and Husain2024). The authors suggest that targeting inflammatory symptoms individually may improve treatment outcomes in this cohort.

According to a recent meta-analysis, CRP was higher in females but not males with depression when compared to healthy controls; however, the sex effect did not reach significance (Jarkas et al., Reference Jarkas, Villeneuve, Daneshmend, Villeneuve and Mcquaid2024). When disregarding sex, CRP levels were increased slightly when compared to controls, but again this result did not reach significance. However, a recent study revealed that the presence of elevated CRP levels does not necessarily correlate with increased depression severity, indicating a complex relationship between inflammation and depressive symptoms (Suneson et al., Reference Suneson, Grudet, Ventorp, Malm, Asp, Westrin and Lindqvist2023).

Elevated inflammatory markers among patients with depression have been well-documented in the literature, with particular emphasis on cytokines IL-1β, IL-6, and TNF-α, which have been associated with exacerbated depressive symptoms (Dantzer et al., Reference Dantzer, O’Connor, Freund, Johnson and Kelley2008; Suneson et al., Reference Suneson, Grudet, Ventorp, Malm, Asp, Westrin and Lindqvist2023; Hassamal, Reference Hassamal2023). Cytokines can influence the secretory activity of the HPA axis, further intensifying inflammatory responses (Turnbull and Rivier, Reference Turnbull and Rivier1999; Schiepers et al., Reference Schiepers, Wichers and Maes2005). In addition, current therapeutic treatments for depression have demonstrated anti-inflammatory effects, although findings have been inconsistent (Horikawa et al., Reference Horikawa, Kato, Mizoguchi, Monji, Seki, Ohkuri, Gotoh, Yonaha, Ueda, Hashioka and Kanba2010; Tynan et al., Reference Tynan, Weidenhofer, Hinwood, Cairns, Day and Walker2012; Wang et al., Reference Wang, Wang, Liu, Qiao, Baldwin and Hou2019; Strawbridge et al., Reference Strawbridge, Izurieta, Day, Tee, Young, Tong, Young and Cleare2023), and the number of failed treatment trials for MDD has been associated with levels of inflammatory markers (Haroon et al., Reference Haroon, Daguanno, Woolwine, Goldsmith, Baer, Wommack, Felger and Miller2018), hinting at an interaction between the immune system and antidepressant efficacy. Further work is required to understand the potential relevance of chronic low-level inflammation to the serotonergic system in a significant subset of the depressed population. Given the role of 5-HT in both the central nervous and immune systems, 5-HT2AR psychedelics present a potential avenue for modulating central and immune system responses.

Clinical trials have increasingly pointed to significant alterations in brain functional connectivity and the default mode network following psychedelic administration, as observed through magnetic resonance imaging (MRI) (Carhart-Harris et al., Reference Carhart-Harris, Roseman, Bolstridge, Demetriou, Pannekoek, Wall, Tanner, Kaelen, Mcgonigle, Murphy, Leech, Curran and Nutt2017; Mertens et al., Reference Mertens, Wall, Roseman, Demetriou, Nutt and Carhart-Harris2020; Doss et al., Reference Doss, Považan, Rosenberg, Sepeda, Davis, Finan, Smith, Pekar, Barker, Griffiths and Barrett2021). Animal models and in vitro studies have also suggested enduring neuroplastic effects, including enhanced synaptogenesis (Ly et al., Reference Ly, Greb, Cameron, Wong, Barragan, Wilson, Burbach, Soltanzadeh Zarandi, Sood, Paddy, Duim, Dennis, Mcallister, Ori-Mckenney, Gray and Olson2018; Raval et al., Reference Raval, Johansen, Donovan, Ros, Ozenne, Hansen and Knudsen2021; Moliner et al., Reference Moliner, Girych, Brunello, Kovaleva, Biojone, Enkavi, Antenucci, Kot, Goncharuk, Kaurinkoski, Kuutti, Fred, Elsila, Sakson, Cannarozzo, Diniz, Seiffert, Rubiolo, Haapaniemi, Meshi, Nagaeva, Ohman, Rog, Kankuri, Vilar, Varjosalo, Korpi, Permi, Mineev, Saarma, Vattulainen, Casarotto and Castren2023; Purple et al., Reference Purple, Gupta, Thomas, Golden, Froudist-Walsh and Jones2024; Schmidt et al., Reference Schmidt, Hoffrichter, Davoudi, Horschitz, Lau, Meinhardt, Spanagel, Ladewig, Köhr and Koch2024; Duque et al., Reference Duque, Chen, Hsu, Narayan, Rymbek, Begum, Saher, Cohen, Olson, Li, Prober, Bergles, Fishman, Engert and Ahrens2024). In an LPS-induced mouse depressive-like model (R)-ketamine and the non-hallucinogenic LSD analogue lisuride led to an antidepressant-like effect in the forced-swim test, a commonly used preclinical test of antidepressant activity (Qu et al., Reference Qu, Chang, Ma, Wan and Hashimoto2023). Interestingly, (R)-DOI by itself had no effect on the LPS-induced depressive-like behaviour in this test. Both (R)-ketamine and lisuride prevented the LPS-induced decrease in dendritic spine density in the prelimbic area of the mPFC, the CA3 and the dentate gyrus regions of the hippocampus, suggesting a potential for preserving neuronal connectivity within the hippocampus.

Liu et al. (Reference Liu, Ma, Qu, Wan, Xu, Zhao, Murayama and Hashimoto2023) assessed the effect of pre-treatment with (R)-ketamine, (R)-DOI, and lisuride, in two different models of depressive-like behaviour in mice, either using systemic LPS administration or chronic restraint stress (CRS). In both models, an increase in the immobility time in the FST, and a decrease in the sucrose preference test, were measured. Interestingly, (R)-ketamine, but not (R)-DOI or lisuride, had a significant antidepressant-like effect in both models. However, pre-administration of (R)-DOI, lisuride, and (R)-ketamine led to decreased circulating plasma IL-6 levels in the LPS model. In both models, (R)-ketamine was the only pre-treatment that blocked the reduction in postsynaptic density-95 (PSD-95) expression, a protein critical for synaptic plasticity.

Additionally, as 5-HT2AR is expressed in the hypothalamic paraventricular nucleus, psychedelics may directly trigger ACTH release, which in turn drives cortisol release from the adrenal glands (Zhang et al., Reference Zhang, Damjanoska, Carrasco, Dudas, D’souza, Tetzlaff, Garcia, Hanley, Scripathirathan, Petersen, Gray, Battaglia, Muma and Van De Kar2002). This effect has been reported in preliminary studies in humans with increased ACTH and cortisol levels after psychedelic treatment, indicative of HPA axis activation (Hasler et al., Reference Hasler, Grimberg, Benz, Huber and Vollenweider2004; Galvao et al., Reference Galvao, De Almeida, Silva, Freire, Palhano-Fontes, Onias, Arcoverde, Maia-De-Oliveira, De Araujo, Lobao-Soares and Galvao-Coelho2018; Galvao-Coelho et al., Reference Galvao-Coelho, De Menezes Galvao, De Almeida, Palhano-Fontes, Campos Braga, Lobao Soares, Maia-De-Oliveira, Perkins, Sarris and De Araujo2020; Uthaug et al., Reference Uthaug, Lancelotta, Szabo, Davis, Riba and Ramaekers2020). Modulation of the HPA axis may contribute to central and immune system responses to psychedelics (Schindler et al., Reference Schindler, Wallace, Sloshower and D’souza2018; Johnston et al., Reference Johnston, Kadriu, Allen, Gilbert, Henter and Zarate2023). Recent results in healthy mice suggest that the psilocybin-induced short- and long-term anxiolytic effects result from psilocybin-induced glucocorticoid release (Jones et al., Reference Jones, Zahid, Grady, Sultan, Zheng, Razidlo, Banks and Wenthur2023). Indeed, the anxiolytic effect of psilocybin was blunted by pre-treatment with a glucocorticoid receptor antagonist. In mice, psilocybin was found to increase corticosterone production in both males and female mice (Farinha-Ferreira et al., Reference Farinha-Ferreira, Miranda-Lourenco, Galipeau, Lenkei and Sebastiao2025) producing anxiolytic-like effects. These results support the idea that the HPA axis may be directly involved with the anxiolytic effects observed with psychedelics.

Peripheral inflammation and chronic stress alter tryptophan metabolism via kynurenine pathway induction, leading to the production of neuroactive metabolites (Barone, Reference Barone2019; Castro-Portuguez and Sutphin, Reference Castro-Portuguez and Sutphin2020; Salminen et al., Reference Salminen, Kaarniranta and Kauppinen2020; Brown et al., Reference Brown, Huang and Newell2021), where the induction of IDO, the rate-limiting enzyme in the kynurenine pathway, plays a key role (O’Connor et al., Reference O’Connor, Lawson, André, Moreau, Lestage, Castanon, Kelley and Dantzer2009). However, there is little evidence to indicate that the 5-HT2AR influences the kynurenine pathway. Inserra et al. (Reference Inserra, Giorgini, Lacroix, Bertazzo, Choo, Markopolous, Grant, Abolghasemi, De Gregorio, Flamand, Rogers, Comai, Silvestri, Gobbi and Di Marzo2023) found that LSD decreased hippocampal levels of kynurenine and endocannabinoid related metabolites in the mouse brain. These results suggest that 5-HT2AR-mediated central effects may influence metabolites from the kynurenine pathway, and that psychedelics may modulate kynurenine metabolism. Further studies are required to determine the extent to which the kynurenine pathway is involved in the biological effects of psychedelics.

Rijsketic et al. (Reference Rijsketic, Casey, Barbosa, Zhang, Hietamies, Ramirez-Ovalle, Pomrenze, Halpern, Williams, Malenka and Heifets2023) examined the impact of environmental context on psilocybin-induced neural activity in mice by measuring immediate early gene expression. Both the environmental context and psilocybin elicited independent brain-wide neural responses, but there was little synergistic interaction between the two (Rijsketic et al., Reference Rijsketic, Casey, Barbosa, Zhang, Hietamies, Ramirez-Ovalle, Pomrenze, Halpern, Williams, Malenka and Heifets2023).

Using (R)-DOI, a recent study was able to isolate and stimulate psychedelic-responsive neurons located in the mPFC of mice. Stimulating these mimicked the anxiolytic effects of (R)-DOI without inducing the HTR, providing evidence that the anxiolytic and hallucinogenic effects of psychedelics could be dissociated (Muir et al., Reference Muir, Lin, Aarrestad, Daniels, Ma, Tian, Olson and Kim2024).

Future directions

Neuroinflammation, along with neuronal atrophy and death, significantly contributes to the development of many disorders, including depression (Dantzer et al., Reference Dantzer, O’Connor, Freund, Johnson and Kelley2008; Osimo et al., Reference Osimo, Baxter, Lewis, Jones and Khandaker2019; Suneson et al., Reference Suneson, Grudet, Ventorp, Malm, Asp, Westrin and Lindqvist2023), stroke (Candelario-Jalil et al., Reference Candelario-Jalil, Dijkhuizen and Magnus2022), neurodevelopmental disorders (Han et al., Reference Han, Patel, Jones and Dale2021), and neurodegenerative diseases (Teleanu et al., Reference Teleanu, Niculescu, Lungu, Radu, Vladacenco, Roza, Costachescu, Grumezescu and Teleanu2022; Singh, Reference Singh2022). Additionally, the BBB is frequently compromised in various neurodegenerative conditions (Ruan et al., Reference Ruan, Zhang, Huang, Sun, Hou, Zhao and Wang2022; Sulimai et al., Reference Sulimai, Brown and Lominadze2023; Hang et al., Reference Hang, Zhou, Xing, Wen and Du2023; Bruno et al., Reference Bruno, Bonomi, Ricci, Di Donna, Mercuri, Koch, Martorana and Motta2024). A recent study found that patients with long-COVID-associated brain fog exhibited persistent systemic inflammation with BBB disruption (Greene et al., Reference Greene, Connolly, Brennan, Laffan, O’keeffe, Zaporojan, O’callaghan, Thomson, Connolly, Argue, Meaney, Martin-Loeches, Long, Cheallaigh, Conlon, Doherty and Campbell2024). Additionally, it was reported that long-COVID was associated with 5-HT deficiency, possibly attributed to viral-induced inhibition of intestinal amino acid absorption (Wong et al., Reference Wong, Devason, Umana, Cox, Dohnalova, Litichevskiy, Perla, Lundgren, Etwebi, Izzo, Kim, Tetlak, Descamps, Park, Wisser, Mcknight, Pardy, Kim, Blank, Patel, Thum, Mason, Beltra, Michieletto, Ngiow, Miller, Liou, Madhu, Dmitrieva-Posocco, Huber, Hewins, Petucci, Chu, Baraniecki-Zwil, Giron, Baxter, Greenplate, Kearns, Montone, Litzky, Feldman and Henao-Mejia2023). These studies indicate a direct link between a viral challenge on the immune system and its likely long-term effects on the CNS.

Measuring neuroinflammation, BBB permeability, and neuronal death is challenging in both animal models and patients. However, neuroimaging techniques, such as positron emission tomography (PET), MRI, and computed tomography (CT) scans, can be used in preclinical and clinical studies to assess these measures. Recent advancements in MRI modalities have enabled researchers to measure enlarged perivascular spaces, BBB permeability, cerebral perfusion, and neuroinflammation (Rowsthorn et al., Reference Rowsthorn, Pham, Nazem-Zadeh, Law, Pase and Harding2023; Kim et al., Reference Kim, Carreira Figueiredo, Simmons, Randall, Rojo Gonzalez, Wood, Ranieri, Sureda-Gibert, Howes, Pariante, Nima, Pasternak, Dell’acqua, Turkheimer and Cash2023). Advances in PET now allow the measurement of tracers associated with neuroinflammation (Masdeu et al., Reference Masdeu, Pascual and Fujita2022).

These imaging techniques can also be deployed to evaluate the effects of psychedelics. Although MRI scans have been utilised in clinical trials for psychedelics in depressed and healthy participants (Sanches et al., Reference Sanches, De Lima Osorio, Dos Santos, Macedo, Maia-De-Oliveira, Wichert-Ana, De Araujo, Riba, Crippa and Hallak2016; Carhart-Harris et al., Reference Carhart-Harris, Roseman, Bolstridge, Demetriou, Pannekoek, Wall, Tanner, Kaelen, Mcgonigle, Murphy, Leech, Curran and Nutt2017; Mertens et al., Reference Mertens, Wall, Roseman, Demetriou, Nutt and Carhart-Harris2020; Doss et al., Reference Doss, Považan, Rosenberg, Sepeda, Davis, Finan, Smith, Pekar, Barker, Griffiths and Barrett2021; Shinozuka et al., Reference Shinozuka, Jerotic, Mediano, Zhao, Preller, Carhart-Harris and Kringelbach2024). A recent study even demonstrated, in a small sample size, that treatment response can be predicted using functional connectivity (Copa et al., Reference Copa, Erritzoe, Giribaldi, Nutt, Carhart-Harris and Tagliazucchi2024). However, neuroinflammation has not yet been assessed in this context.

A significant advantage of these imaging methods is that they can be applied similarly in animal models. MRI and other imaging modalities may provide imaging markers to assist in monitoring the long-term effects of psychedelics in animal models and patients. This approach can help ensure the safety and efficacy of these treatments in clinical practice and optimise therapeutic outcomes.

As previously highlighted, potency and affinity values vary greatly between 5-HT2AR psychedelics. There is a lack of pre-clinical and clinical studies that directly compare their effects on the immune and CNS. Exploring their possible differences could help researchers and clinicians understand the receptors and cellular pathways involved in the immunomodulatory and antidepressant properties of these compounds. Additionally, studies discussed in this review draw attention to the lack of direct comparison between unstimulated and stimulated immune systems, whether in isolated cells or whole biological systems. This type of study would be valuable understanding how psychedelics are immunoregulatory, and not simply anti- or pro-inflammatory.

Conclusion

In vitro and in vivo studies have investigated the effects of 5-HT2AR psychedelics at supraphysiological concentrations and have shown immune-modulating properties of these compounds. Receptors beyond the serotonergic family, such as the TrkB receptor, may play a key role in mediating the immunomodulatory effects. However, well-powered clinical studies are lacking, making it challenging to evaluate the immunomodulatory properties of psychedelics in humans – both in healthy individuals and those with psychiatric disorders. Further research is necessary to elucidate these effects and their connection to therapeutic outcomes. MRI and other brain imaging techniques offer a valuable translational tool to bridge the gap between preclinical and clinical studies and to advance our understanding of the effects of psychedelics in the brain.

Financial support

JRK has consulted for Clerkenwell Health and has received grant funding from the Health Research Board (ILP-POR-2022-030, KTA-2024-002, DIFA-2023-005).

Competing interests

JRK is principal investigator (Ireland) on COMPASS, GH and Transcend Therapeutics sponsored clinical trials in Dublin, Ireland.

Footnotes

*

Ex aequo co-senior and corresponding authorship.

References

Abdouh, M, Albert, PR, Drobetsky, E, Filep, JG and Kouassi, E (2004) 5-HT1A-mediated promotion of mitogen-activated T and B cell survival and proliferation is associated with increased translocation of NF-kappaB to the nucleus. Brain Behavior and Immunity 18, 2434.Google Scholar
Afshar, S, Shahidi, S, Rohani, AH, Soleimani Asl, S, Komaki, A (2019) Protective effects of 5-HT(1A) receptor antagonist and 5-HT(2A) receptor agonist on the biochemical and histological features in a rat model of Alzheimer’s disease. Journal of Chemical Neuroanatomy 96, 140147.Google Scholar
Akil, H, Brenner, S, Kandel, E, Kendler, KS, King, MC, Scolnick, E, Watson, JD and Zoghbi, HY (2010) Medicine. The future of psychiatric research: genomes and neural circuits. Science 327, 15801581.Google Scholar
Alexander, L, Anderson, D, Baxter, L, Claydon, M, Rucker, J and Robinson, ESJ (2024) Preclinical models for evaluating psychedelics in the treatment of major depressive disorder. British Journal of Pharmacology. doi: 10.1111/bph.17370. Epub ahead of print.Google Scholar
Alvarez, JI, Katayama, T and Prat, A(2013) Glial influence on the blood brain barrier. Glia 61, 19391958.Google Scholar
Antonioli, M, Rybka, J and Carvalho, LA (2012) Neuroimmune endocrine effects of antidepressants. Neuropsychiatric Disease and Treatment 8, 6583.Google Scholar
Arteaga-Henriquez, G, Simon, MS, Burger, B, Weidinger, E, Wijkhuijs, A, Arolt, V, Birkenhager, TK, Musil, R, Muller, N and Drexhage, HA (2019) Low-grade inflammation as a predictor of antidepressant and anti-inflammatory therapy response in MDD patients: a systematic review of the literature in combination with an analysis of experimental data collected in the EU-MOODINFLAME consortium. Frontiers in Psychiatry 10, 458.Google Scholar
Arzt, E, Costas, M, Finkielman, S and Nahmod, VE (1991) Serotonin inhibition of tumor necrosis factor-alpha synthesis by human monocytes. Life Sciences 48, 25572562. doi: 10.1016/0024-3205(91)90612-f.Google Scholar
Bahr, FS, Muller, FE, Weber, N, Aguapong, W, Neumueller, S, Benen, N, Ricke-Hoch, M and Ponimaskin, E (2024) The role of serotonin receptor 7 signalling in macrophages for the inflammatory response after myocardial infarction. Cardiovascular Research 120, cvae088.041. doi: 10.1093/cvr/cvae088.041.Google Scholar
Bahr, FS, Muller, FE, Kasten, M, Benen, N, Sieve, I, Scherr, M, Falk, CS, Hilfiker-Kleiner, D, Ricke-Hoch, M and Ponimaskin, E (2025) Serotonin receptor 5-HT7 modulates inflammatory-associated functions of macrophages. Cellular and Molecular Life Sciences 82, 51.Google Scholar
Barone, P (2019) The ‘Yin’ and the ‘Yang’ of the kynurenine pathway: excitotoxicity and neuroprotection imbalance in stress-induced disorders. Behavioural Pharmacology 30, 163186.Google Scholar
Bel, N, Figueras, G, Vilaro, MT, Sunol, C and Artigas, F (1997) Antidepressant drugs inhibit a glial 5-hydroxytryptamine transporter in rat brain. European Journal of Neuroscience 9, 17281738.Google Scholar
Bellavance, MA and Rivest, S (2014) The HPA - immune axis and the immunomodulatory actions of glucocorticoids in the brain. Frontiers in Immunology 5, 136.Google Scholar
Besnard, J, Ruda, GF, Setola, V, Abecassis, K, Rodriguiz, RM, Huang, XP, Norval, S, Sassano, MF, Shin, AI, Webster, LA, Simeons, FR, Stojanovski, L, Prat, A, Seidah, NG, Constam, DB, Bickerton, GR, Read, KD, Wetsel, WC, Gilbert, IH, Roth, BL and Hopkins, AL (2012) Automated design of ligands to polypharmacological profiles. Nature 492, 215220.Google Scholar
Bhattarai, P, Cosacak, MI, Mashkaryan, V, Demir, S, Popova, SD, Govindarajan, N, Brandt, K, zhang, Y, Chang, W, Ampatzis, K and Kizil, C (2020) Neuron-glia interaction through serotonin-BDNF-NGFR axis enables regenerative neurogenesis in Alzheimer’s model of adult zebrafish brain. PLos Biology 18, e3000585.Google Scholar
Bhusal, A, Afridi, R, Lee, WH and Suk, K (2023) Bidirectional communication between microglia and astrocytes in neuroinflammation. Current Neuropharmacology 21, 20202029.Google Scholar
Boehme, SA, Lio, FM, Sikora, L, Pandit, TS, Lavrador, K, Rao, SP and Sriramarao, P (2004) Cutting edge: serotonin is a chemotactic factor for eosinophils and functions additively with eotaxin. Journal of Immunology 173, 35993603.Google Scholar
Borbély, E, Varga, V, Szögi, T, Schuster, I, Bozsó, Z, Penke, B and Fülöp, L (2022) Impact of two neuronal sigma-1 receptor modulators, PRE084 and DMT, on neurogenesis and neuroinflammation in an Aβ1-42-injected, wild-type mouse model of AD. International Journal of Molecular Sciences [Online] 23, 2514.Google Scholar
Bornemann, J, Close, JB, Ahmad, K, Barba, T, Godfrey, K, Macdonald, L, Erritzoe, D, Nutt, D and Carhart-Harris, R (2024) Study protocol for “Psilocybin in patients with fibromyalgia: brain biomarkers of action”. Frontiers in Psychiatry 15, 1320780.Google Scholar
Boukhatem, I, Fleury, S, Welman, M, Le Blanc, J, Thys, C, Freson, K, Best, MG, Wurdinger, T, Allen, BG and Lordkipanidze, M (2021) The brain-derived neurotrophic factor prompts platelet aggregation and secretion. Blood Advances 5, 35683580.Google Scholar
Bove, GM and Mokler, DJ (2022) Effects of a single dose of psilocybin on cytokines, chemokines and leptin in rat serum. Journal of Psychedelic Studies 6, 4.Google Scholar
Brown, SJ, Huang, XF and Newell, KA (2021) The kynurenine pathway in major depression: what we know and where to next. Neuroscience & Biobehavioral Reviews 127, 917927.Google Scholar
Brundin, P, Nath, A and Beckham, JD (2020) Is COVID-19 a perfect storm for Parkinson’s disease? Trends in Neurosciences 43, 931933.Google Scholar
Bruno, M, Bonomi, CG, Ricci, F, Di Donna, MG, Mercuri, NB, Koch, G, Martorana, A and Motta, C (2024) Blood-brain barrier permeability is associated with different neuroinflammatory profiles in Alzheimer’s disease. European Journal of Neurology 31, e16095.Google Scholar
Burmester, DR, Madsen, MK, Szabo, A, Aripaka, SS, Stenbaek, DS, Frokjaer, VG, Elfving, B, Mikkelsen, JD, Knudsen, GM and Fisher, PM (2023) Subacute effects of a single dose of psilocybin on biomarkers of inflammation in healthy humans: An open-label preliminary investigation. Comprehensive Psychoneuroendocrinology 13, 100163.Google Scholar
Butler, M, Bird, C, Maggio, C, Durden, A, Modlin, N, Campbell-Coker, K, Edwards, M, Pick, S, Millman, LSM, Lowery, E, Bhagavan, C, Kanaan, R, Golder, D, Mildon, B, Mehta, M, Rucker, J and Nicholson, TR (2024) Probing the functional magnetic resonance imaging response to psilocybin in functional neurological disorder (PsiFUND): study protocol [version 1; peer review: 1 approved, 3 approved with reservations]. Wellcome Open Research 9, 401.Google Scholar
Canal, CE (2018) Serotonergic psychedelics: experimental approaches for assessing mechanisms of action. Handbook of Experimental Pharmacology 252, 227260.Google Scholar
Canal, CE, Booth, RG and Morgan, D (2013) Support for 5-HT2C receptor functional selectivity in vivo utilizing structurally diverse, selective 5-HT2C receptor ligands and the 2,5-dimethoxy-4-iodoamphetamine elicited head-twitch response model. Neuropharmacology 70, 112121.Google Scholar
Candelario-Jalil, E, Dijkhuizen, RM and Magnus, T (2022) Neuroinflammation, stroke, blood-brain barrier dysfunction, and imaging modalities. Stroke 53, 14731486.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, 14021411.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 (Berl) 235, 399408.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, 13187.Google Scholar
Carnevale, D, Pallante, F, Fardella, V, Fardella, S, Iacobucci, R, Federici, M, Cifelli, G, de Lucia, M and Lembo, G (2014) The angiogenic factor PlGF mediates a neuroimmune interaction in the spleen to allow the onset of hypertension. Immunity 41, 737752.Google Scholar
Carpenter, S, O’Neill, LAJ (2024) From periphery to center stage: 50 years of advancements in innate immunity. Cell 187, 20302051.Google Scholar
Castellani, G, Croese, T, Peralta Ramos, JM and Schwartz, M (2023) Transforming the understanding of brain immunity. Science 380, eabo7649.Google Scholar
Castren, E and Antila, H (2017) Neuronal plasticity and neurotrophic factors in drug responses. Molecular Psychiatry 22, 10851095.Google Scholar
Castro-Portuguez, R and Sutphin, GL (2020) Kynurenine pathway, NAD(+) synthesis, and mitochondrial function: targeting tryptophan metabolism to promote longevity and healthspan. Experimental Gerontology 132, 110841.Google Scholar
Cekanaviciute, E and Buckwalter, MS (2016) Astrocytes: integrative regulators of neuroinflammation in stroke and other neurological diseases. Neurotherapeutics 13, 685701.Google Scholar
Chabbi-Achengli, Y, Coman, T, Collet, C, Callebert, J, Corcelli, M, Lin, H, Rignault, R, Dy, M, De Vernejoul, MC and Cote, F (2016) Serotonin is involved in autoimmune arthritis through Th17 immunity and bone resorption. American Journal of Pathology 186, 927937.Google Scholar
Chen, C-S, Barnoud, C and Scheiermann, C (2021) Peripheral neurotransmitters in the immune system. Current Opinion in Physiology 19, 7379.Google Scholar
Chiew, AA-O and Isbister, GA-O (2024) Management of serotonin syndrome (toxicity). British Journal of Clinical Pharmacology 90, 3369.Google Scholar
Chilmonczyk, Z, Bojarski, AJ, Pilc, A and Sylte, I (2017) Serotonin transporter and receptor ligands with antidepressant activity as neuroprotective and proapoptotic agents. Pharmacological Reports 69, 469478.Google Scholar
Ciriaco, E, Dall’aglio, C, Hannestad, J, Huerta, JJ, Laura, R, Germana, G and Vega, JA (1996) Localization of trk neurotrophin receptor-like proteins in avian primary lymphoid organs (thymus and bursa of Fabricius). Journal of Neuroimmunology 69, 7383.Google Scholar
Cloez-Tayarani, I, Petit-Bertron, AF, Venters, HD and Cavaillon, JM (2003) Differential effect of serotonin on cytokine production in lipopolysaccharide-stimulated human peripheral blood mononuclear cells: involvement of 5-hydroxytryptamine2A receptors. International Immunology 15, 233240.Google Scholar
Cloots, RH, Sankaranarayanan, S, De Theije, CC, Poynter, ME, Terwindt, E, Van Dijk, P, Hakvoort, TB, Lamers, WH and Kohler, SE (2013) Ablation of Arg1 in hematopoietic cells improves respiratory function of lung parenchyma, but not that of larger airways or inflammation in asthmatic mice. American Journal of Physiology-Lung Cellular and Molecular Physiology 305, L364L376 Google Scholar
Cloutier, N, Allaeys, I, Marcoux, G, Machlus, KR, Mailhot, B, Zufferey, A, Levesque, T, Becker, Y, Tessandier, N, Melki, I, Zhi, H, Poirier, G, Rondina, MT, Italiano, JE, Flamand, L, Mckenzie, SE, Cote, F, Nieswandt, B, Khan, WI, Flick, MJ, Newman, PJ, Lacroix, S, Fortin, PR and Boilard, E (2018) Platelets release pathogenic serotonin and return to circulation after immune complex-mediated sequestration. Proceedings of the National Academy of Sciences 115, E1550E1559.Google Scholar
Cohen, S, Janicki-Deverts, D, Doyle, WJ, Miller, GE, Frank, E, Rabin, BS and Turner, RB (2012) Chronic stress, glucocorticoid receptor resistance, inflammation, and disease risk. Proceedings of the National Academy of Sciences 109, 59955999.Google Scholar
Cohen, Z, Bouchelet, I, Olivier, A, Villemure, JG, Ball, R, Stanimirovic, DB and Hamel, E (1999) Multiple microvascular and astroglial 5-hydroxytryptamine receptor subtypes in human brain: molecular and pharmacologic characterization. Journal of Cerebral Blood Flow & Metabolism 19, 908917.Google Scholar
Colle, R, Deflesselle, E, Martin, S, David, DJ, Hardy, P, Taranu, A, Falissard, B, Verstuyft, C and Corruble, E (2015) BDNF/TRKB/P75NTR polymorphisms and their consequences on antidepressant efficacy in depressed patients. Pharmacogenomics 16, 9971013.Google Scholar
Copa, D, Erritzoe, D, Giribaldi, B, Nutt, D, Carhart-Harris, R and Tagliazucchi, E (2024) Predicting the outcome of psilocybin treatment for depression from baseline fMRI functional connectivity. Journal of Affective Disorders 353, 6069.Google Scholar
Cornell, J, Salinas, S, Huang, HY and Zhou, M (2022) Microglia regulation of synaptic plasticity and learning and memory. Neural Regeneration Research 17, 705716.Google Scholar
Couch, Y, Xie, Q, lundberg, L, Sharp, T and Anthony, DC (2015) A model of post-infection fatigue is associated with increased TNF and 5-HT2A receptor expression in mice. PLoS One 10, e0130643.Google Scholar
coutinho, AE and Chapman, KE (2011) The anti-inflammatory and immunosuppressive effects of glucocorticoids, recent developments and mechanistic insights. Molecular and Cellular Endocrinology 335, 213.Google Scholar
Crepieux, P, Poupon, A, Langonne-Gallay, N, Reiter, E, Delgado, J, Schaefer, MH, Bourquard, T, Serrano, L and Kiel, C (2017) A comprehensive view of the beta-arrestinome. Frontiers in Endocrinology 8, 32.Google Scholar
Custodio, RJP, Ortiz, DM, Lee, HJ, Sayson, LV, Kim, M, Lee, YS, Kim, KM, Cheong, JH and Kim, HJ (2023) Serotonin 2C receptors are also important in head-twitch responses in male mice. Psychopharmacology (Berl) 242, 1585.Google Scholar
Dakic, V, Minardi Nascimento, J, Costa Sartore, R, Maciel, RM, De Araujo, DB, Ribeiro, S, Martins-de-Souza, D and Rehen, SK (2017) Short term changes in the proteome of human cerebral organoids induced by 5-MeO-DMT. Scientific Reports 7, 12863.Google Scholar
Dantzer, R, O’Connor, JC, Freund, GG, Johnson, RW, Kelley, KW (2008) From inflammation to sickness and depression: when the immune system subjugates the brain. Nature Reviews Neuroscience 9, 4656.Google Scholar
Davydova, SM, Cheido, MA, Gevorgyan, MM and Idova, GV (2010) Effects of 5-HT2A receptor stimulation and blocking on immune response. Bulletin of Experimental Biology and Medicine 150, 219221.Google Scholar
de Camargo, RW, Joaquim, L, Machado, RS, de Souza Ramos, S, da Rosa, LR, de Novais Junior, Lério R, Mathias, K, Maximiano, L, Strickert, YR, Nord, R, Gava, ML, Scarpari, E, Martins, HM, Lins, EMF, Chaves, Jéssica S, da Silva, LE, de Oliveira, MP, da Silva, MR, Fernandes, BB, Tiscoski, Adal Bó, Piacentini, Nália, Santos, FP, Inserra, A, Bobinski, F, Rezin, GT, Yonamine, M, Petronilho, Fícia, de Bitencourt, RM (2024) Ayahuasca pretreatment prevents sepsis-induced anxiety-like behavior, neuroinflammation, and oxidative stress, and increases brain-derived neurotrophic factor. Molecular Neurobiology 62(5), 56955719.Google Scholar
de la Fuente Revenga, M, Zhu, B, Guevara, CA, Naler, LB, Saunders, JM, Zhou, Z, Toneatti, R, Sierra, S, Wolstenholme, JT, Beardsley, PM and Huntley, GW (2021) Prolonged epigenomic and synaptic plasticity alterations following single exposure to a psychedelic in mice. Cell Reports 37, 109836.Google Scholar
De Santi, L, Annunziata, P, Sessa, E and Bramanti, P (2009) Brain-derived neurotrophic factor and TrkB receptor in experimental autoimmune encephalomyelitis and multiple sclerosis. Journal of the Neurological Sciences 287, 1726.Google Scholar
De Vos, CM, Mason, NL and Kuypers, KP (2021) Psychedelics and neuroplasticity: a systematic review unraveling the biological underpinnings of psychedelics. Frontiers in Psychiatry 12, 724606.Google Scholar
Delves, PJ, Martin, SJ, Burton, DR and Roitt, IM (2017) Roitt’s essential immunology. Chichester: West Sussex; Hoboken: John Wiley & Sons, Inc Google Scholar
Dempsey, E, Abautret-Daly, A, Docherty, NG, Medina, C and Harkin, A (2019) Persistent central inflammation and region specific cellular activation accompany depression- and anxiety-like behaviours during the resolution phase of experimental colitis. Brain, Behavior, and Immunity 80, 616632.Google Scholar
Diamond, B, Honig, G, Mader, S, Brimberg, L and Volpe, BT (2013) Brain-reactive antibodies and disease. Annual Review of Immunology 31, 345385.Google Scholar
Diamond, M, Kelly, JP and Connor, TJ (2006) Antidepressants suppress production of the Th1 cytokine interferon-gamma, independent of monoamine transporter blockade. European Neuropsychopharmacology 16, 481490.Google Scholar
DiRenzo, D, Barrett, FS, Perin, J, Darrah, E, Christopher-Stine, L and Griffiths, RR (2024) Impact of psilocybin on peripheral cytokine production. Psychedelic Medicine (New Rochelle) 2, 109115.Google Scholar
Domínguez-Soto, Á., Usategui, A, Casas-Engel, MD, Simón-Fuentes, M, Nieto, C, Cuevas, VD, Vega, MA, Luis Pablos, J and Corbí, Á.L (2017) Serotonin drives the acquisition of a profibrotic and anti-inflammatory gene profile through the 5-HT7R-PKA signaling axis. Scientific Reports 7, 14761.Google Scholar
Dong, C, Zhang, JC, Yao, W, Ren, Q, Yang, C, Ma, M, Han, M, Saito, R and Hashimoto, K (2016) Effects of escitalopram, R-citalopram, and reboxetine on serum levels of tumor necrosis factor-alpha, interleukin-10, and depression-like behavior in mice after lipopolysaccharide administration. Pharmacology Biochemistry and Behavior 144, 712.Google Scholar
Dos Santos, RG, Valle, M, Bouso, JC, Nomdedéu, JF, Rodríguez-Espinosa, J, McIlhenny, EH, Barker, SA, Barbanoj, MJ and Riba, J (2011) Autonomic, neuroendocrine, and immunological effects of ayahuasca: a comparative study with d-amphetamine. Journal of Clinical Psychopharmacology 31, 717726.Google Scholar
Doss, MK, Považan, M, Rosenberg, MD, Sepeda, ND, Davis, AK, Finan, PH, Smith, GS, Pekar, JJ, Barker, PB, Griffiths, RR and Barrett, FS (2021) Psilocybin therapy increases cognitive and neural flexibility in patients with major depressive disorder. Translational Psychiatry 11, 574.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, 829834.Google Scholar
Dubeykovskaya, Z, Y., SI, Chen, X, Worthley, DL, Renz, BW, Urbanska, AM, Hayakawa, Y, T., XU, Westphalen, CB, Dubeykovskiy, A, Chen, D, Friedman, RA, Asfaha, S, Nagar, K, Tailor, Y, Muthupalani, S, Fox, JG, Kitajewski, J and Wang, TC (2016) Neural innervation stimulates splenic TFF2 to arrest myeloid cell expansion and cancer. Nature Communications 7, 10517.Google Scholar
Dunlap, LE, Azinfar, A, C., LY, Cameron, LP, Viswanathan, J, Tombari, RJ, Myers-Turnbull, D, Taylor, JC, Grodzki, AC, Lein, PJ, Kokel, D and Olson, DE (2020) Identification of psychoplastogenic N,N-dimethylaminoisotryptamine (isoDMT) analogues through structure-activity relationship studies. Journal of Medicinal Chemistry 63, 11421155.Google Scholar
Duque, M, Chen, AB, Hsu, E, Narayan, S, Rymbek, A, Begum, S, Saher, G, Cohen, AE, Olson, DE, Li, Y, Prober, DA, Bergles, DE, Fishman, MC, Engert, F and Ahrens, MB (2024) Ketamine induces plasticity in a norepinephrine-astroglial circuit to promote behavioral perseverance. Neuron 113, 426443.e5. doi: 10.1016/j.neuron.2024.11.011.Google Scholar
Durk, T, Panther, E, Muller, T, Sorichter, S, Ferrari, D, Pizzirani, C, Di Virgilio, F, Myrtek, D, Norgauer, J and Idzko, M (2005) 5-Hydroxytryptamine modulates cytokine and chemokine production in LPS-primed human monocytes via stimulation of different 5-HTR subtypes. International Immunology 17, 599606.Google Scholar
Edmondson, DE, Binda, C and Mattevi, A (2007) Structural insights into the mechanism of amine oxidation by monoamine oxidases A and B. Archives of Biochemistry and Biophysics 464, 269276.Google Scholar
Erkizia-Santamaria, I, Alles-Pascual, R, Horrillo, I, Meana, JJ and Ortega, JE (2022) Serotonin 5-HT(2A), 5-HT(2c) and 5-HT(1A) receptor involvement in the acute effects of psilocybin in mice. In vitro pharmacological profile and modulation of thermoregulation and head-twich response. Biomedicine & Pharmacotherapy 154, 113612.Google Scholar
Fan, H (2014) Beta-arrestins 1 and 2 are critical regulators of inflammation. Innate Immunity 20, 451460.Google Scholar
Fan, LW, Bhatt, A, Tien, LT, Zheng, B, Simpson, KL, Lin, RC, Cai, Z, Kumar, P and Pang, Y (2015) Exposure to serotonin adversely affects oligodendrocyte development and myelination in vitro . Journal of Neurochemistry 133, 532543.Google Scholar
Farinha-Ferreira, M, Miranda-Lourenco, C, Galipeau, C, Lenkei, Z and Sebastiao, AM (2025) Concurrent stress modulates the acute and post-acute effects of psilocybin in a sex-dependent manner. Neuropharmacology 266, 110280.Google Scholar
Fellows, E, Jones, BDM, Hodsoll, J, Husain, N, Khoso, AB, Young, AH, Chaudhry, IB and Husain, MI (2024) Associations between C-reactive protein and individual symptoms of depression in a lower-middle income country. BJPsych Open 10, e169.Google Scholar
Fitzgerald, LW, Kaplinsky, L and Kimelberg, HK (1990) Serotonin metabolism by monoamine oxidase in rat primary astrocyte cultures. Journal of Neurochemistry 55, 20082014.Google Scholar
Flanagan, TW, Billac, GB, Landry, AN, Sebastian, MN, Cormier, SA and Nichols, CD (2021) Structure-activity relationship analysis of psychedelics in a rat model of asthma reveals the anti-inflammatory pharmacophore. ACS Pharmacology & Translational Science 4, 488502.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, 478492.Google Scholar
Flanagan, TW and Nichols, CD (2018) Psychedelics as anti-inflammatory agents. International Review of Psychiatry 30, 363375.Google Scholar
Flanagan, TW, Sebastian, MN, Battaglia, DM, Foster, TP, Cormier, SA and Nichols, CD (2019a) 5-HT(2) receptor activation alleviates airway inflammation and structural remodeling in a chronic mouse asthma model. Life Science Part 1 Physiology & Pharmacology 236, 116790.Google Scholar
Flanagan, TW, Sebastian, MN, Battaglia, DM, Foster, TP, Maillet, EL and Nichols, CD (2019b) Activation of 5-HT(2) receptors reduces inflammation in vascular tissue and cholesterol levels in high-fat diet-fed apolipoprotein E knockout mice. Scientific Reports 9, 13444.Google Scholar
Francelin, C, Veneziani, LP, Farias, ADS, Mendes-Da-Cruz, DA and Savino, W (2021) Neurotransmitters modulate intrathymic T-cell development. Frontiers in Cell and Developmental Biology 9, 668067.Google Scholar
Franco-Molina, M, Gomez-Flores, R, Tamez-Guerra, P, Tamez-Guerra, R, Castillo-Leon, L and Rodriguez-Padilla, C (2003) In vitro immunopotentiating properties and tumour cell toxicity induced by Lophophora williamsii (peyote) cactus methanolic extract. Phytotherapy Research 17, 10761081.Google Scholar
Frazer, A and Hensler, J (1999) Chapter 13: serotonin receptors. In Gj, S, Bw, A, Rw, A, Sk, F and Md, U (ed), Basic neurochemistry: molecular, cellular, and medical aspects. Philadelphia: Lippincott-Raven Google Scholar
Freire-Garabal, M, Nunez, MJ, Balboa, J, Lopez-Delgado, P, Gallego, R, Garcia-Caballero, T, Fernandez-Roel, MD, Brenlla, J and Rey-Mendez, M (2003) Serotonin upregulates the activity of phagocytosis through 5-HT1A receptors. British Journal of Pharmacology 139, 457463. doi: 10.1038/sj.bjp.0705188.Google Scholar
Funk, D, Araujo, J, Slassi, M, Lanthier, J, Atkinson, J, Feng, D, Lau, W, Le, A and Higgins, GA (2024) Effect of a single psilocybin treatment on FOS protein expression in male rat brain. Neuroscience 539, 111.Google Scholar
Galvao-Coelho, NL, De Menezes Galvao, AC, De Almeida, RN, Palhano-Fontes, F, Campos Braga, I, Lobao 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, 11251133.Google Scholar
Galvao, ACM, De Almeida, RN, Silva, E, Freire, FAM, Palhano-Fontes, F, Onias, H, Arcoverde, E, Maia-De-Oliveira, JP, De Araujo, DB, Lobao-Soares, B and Galvao-Coelho, NL (2018) Cortisol modulation by ayahuasca in patients with treatment resistant depression and healthy controls. Frontiers in Psychiatry 9, 185.Google Scholar
Gao, C, Jiang, J, Tan, Y and Chen, S (2023) Microglia in neurodegenerative diseases: mechanism and potential therapeutic targets. Signal Transduction and Targeted Therapy 8, 359.Google Scholar
Gentile, A, D’acquisto, F and Leposavic, G (2021) Editorial: the bidirectional communication between neurons and immune cells in the development of psychiatric, neurological and immune-mediated disorders. Frontiers in Immunology 12, 781151.Google Scholar
Ghasemi Gojani, E, Wang, B, Li, D, Kovalchuk, O and Kovalchuk, I (2024) The effects of psilocybin on lipopolysaccharide-induced inflammation in THP-1 human macrophages. Psychoactives [Online] 3, 4864.Google Scholar
Gibney, SM, Mcguinness, B, Prendergast, C, Harkin, A and Connor, TJ (2013) Poly I: C-induced activation of the immune response is accompanied by depression and anxiety-like behaviours, kynurenine pathway activation and reduced BDNF expression. Brain Behavior and Immunity 28, 170181.Google Scholar
Glebov, K, Lochner, M, Jabs, R, Lau, T, Merkel, O, Schloss, P, Steinhauser, C and Walter, J (2015) Serotonin stimulates secretion of exosomes from microglia cells. Glia 63, 626634.Google Scholar
Gobin, V, Van Steendam, K, Denys, D and Deforce, D (2014) Selective serotonin reuptake inhibitors as a novel class of immunosuppressants. International Immunopharmacology 20, 148156.Google Scholar
Gonzalez-Arias, C, Sanchez-Ruiz, A, Esparza, J, Sanchez-Puelles, C, Arancibia, L, Ramirez-Franco, J, Gobbo, D, Kirchhoff, F and Perea, G (2023) Dysfunctional serotonergic neuron-astrocyte signaling in depressive-like states. Molecular Psychiatry 28, 38563873.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, Palenicek, T, Printz, D, Rademaker, MC, Van, Reemst and Malievskaia, E (2022) Single-dose psilocybin for a treatment-resistant episode of major depression. New England Journal of Medicine 387, 16371648.Google Scholar
Goodwin, GM, Aaronson, ST, Alvarez, O, Atli, M, Bennett, JC, Croal, M, Debattista, C, Dunlop, BW, Feifel, D, Hellerstein, DJ, Husain, MI, Kelly, JR, Lennard-Jones, MR, Licht, RW, Marwood, L, Mistry, S, Páleníček, T, Redjep, O, Repantis, D, Schoevers, RA, Septimus, B, Simmons, HJ, Soares, JC, Somers, M, Stansfield, SC, Stuart, JR, Tadley, HH, Thiara, NK, Tsai, J, Wahba, M, Williams, S, Winzer, RI, Young, AH, Young, MB, Zisook, S and Malievskaia, E (2023a) Single-dose psilocybin for a treatment-resistant episode of major depression: impact on patient-reported depression severity, anxiety, function, and quality of life. Journal of Affective Disorders 327, 120127.Google Scholar
Goodwin, GM, Aaronson, ST, Alvarez, O, Carhart-Harris, R, Chai-Rees, J, Croal, M, Debattista, C, Dunlop, BW, Feifel, D, Hellerstein, DJ, Husain, MI, Kelly, JR, Kirlic, N, Licht, RW, Marwood, L, Meyer, TD, Mistry, S, Nowakowska, A, Palenicek, T, Repantis, D, Schoevers, RA, Simmons, H, Somers, M, Teoh, E, Tsai, J, Wahba, M, Williams, S, Young, AH, Young, MB, Zisook, S and Malievskaia, E (2025) The role of the psychedelic experience in psilocybin treatment for treatment-resistant depression. Journal of Affective Disorders 372, 523532.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 (2023b) Psilocybin for treatment resistant depression in patients taking a concomitant SSRI medication. Neuropsychopharmacology 48, 14921499.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 (2023c) Psilocybin for treatment resistant depression in patients taking a concomitant SSRI medication. Neuropsychopharmacology 48, 14921499.Google Scholar
Greene, C, Connolly, R, Brennan, D, Laffan, A, O’keeffe, E, Zaporojan, L, O’callaghan, J, Thomson, B, Connolly, E, Argue, R, Meaney, JFM, Martin-Loeches, I, Long, A, Cheallaigh, CN, Conlon, N, Doherty, CP and Campbell, M (2024) Blood-brain barrier disruption and sustained systemic inflammation in individuals with long COVID-associated cognitive impairment. Nature Neuroscience 27, 421432.Google Scholar
Grieco, SF, Castrén, E, Knudsen, GM, Kwan, AC, Olson, DE, Zuo, Y, Holmes, TC and Xu, X (2022) Psychedelics and neural plasticity: therapeutic implications. The Journal of Neuroscience 42, 84398449.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, 11811197.Google Scholar
Guo, L, Gao, T, Gao, C, Jia, X, Ni, J, Han, C and Wang, Y (2021) Stimulation of astrocytic sigma-1 receptor is sufficient to ameliorate inflammation- induced depression. Behavioural Brain Research 410, 113344.Google Scholar
Haberzettl, R, Bert, B, Fink, H and Fox, MA (2013) Animal models of the serotonin syndrome: a systematic review. Behavioural Brain Research 256, 328345.Google Scholar
Hagberg, GB, Blomstrand, F, Nilsson, M, Tamir, H and Hansson, E (1998) Stimulation of 5-HT2A receptors on astrocytes in primary culture opens voltage-independent Ca2+ channels. Neurochemistry International 32, 153162.Google Scholar
Hagenberg, J, Be, CSG, Group, OS, Bruckl, TM, Erhart, M, Kopf-Beck, J, Kodel, M, Rehawi, G, Roh-Karamihalev, S, Sauer, S, Yusupov, N, Rex-Haffner, M, Spoormaker, VI, Samann, P, Binder, E and Knauer-Arloth, J (2025) Dissecting depression symptoms: multi-omics clustering uncovers immune-related subgroups and cell-type specific dysregulation. Brain Behavior and Immunity 123, 353369.Google Scholar
Hall, AA, Herrera, Y, Ajmo, CT Jr., Cuevas, J and Pennypacker, KR (2009) Sigma receptors suppress multiple aspects of microglial activation. Glia 57, 744754.Google Scholar
Han, VX, Patel, S, Jones, HF and Dale, RC (2021) Maternal immune activation and neuroinflammation in human neurodevelopmental disorders. Nature Reviews Neurology 17, 564579.Google Scholar
Hang, Z, Zhou, L, Xing, C, Wen, Y and Du, H (2023) The blood-brain barrier, a key bridge to treat neurodegenerative diseases. Ageing Research Reviews 91, 102070.Google Scholar
Haroon, E, Daguanno, AW, Woolwine, BJ, Goldsmith, DR, Baer, WM, Wommack, EC, Felger, JC and Miller, AH (2018) Antidepressant treatment resistance is associated with increased inflammatory markers in patients with major depressive disorder. Psychoneuroendocrinology 95, 4349.Google Scholar
Hashimoto, K (2015) Activation of sigma-1 receptor chaperone in the treatment of neuropsychiatric diseases and its clinical implication. Journal of Pharmacological Sciences 127, 69.Google Scholar
Hasler, F, Grimberg, U, Benz, MA, Huber, T and Vollenweider, FX (2004) Acute psychological and physiological effects of psilocybin in healthy humans: a double-blind, placebo-controlled dose-effect study. Psychopharmacology (Berl) 172, 145156.Google Scholar
Hassamal, S (2023) Chronic stress, neuroinflammation, and depression: an overview of pathophysiological mechanisms and emerging anti-inflammatories. Frontiers in Psychiatry 14, 1130989.Google Scholar
Hatzipantelis, CJ and Olson, DE (2024) The effects of psychedelics on neuronal physiology. Annual Review of Physiology 86, 2747.Google Scholar
Hayashi, T (2019) The sigma-1 receptor in cellular stress signaling. Frontiers in Neuroscience 13, 733.Google Scholar
Hayashi, T and Su, TP (2007) Sigma-1 receptor chaperones at the ER-mitochondrion interface regulate Ca(2+) signaling and cell survival. Cell 131, 596610.Google Scholar
Herr, N, Bode, C and Duerschmied, D (2017) The effects of serotonin in immune cells. Frontiers in Cardiovascular Medicine 4, 48.Google Scholar
Hirst, WD, Abrahamsen, B, Blaney, FE, Calver, AR, Aloj, L, Price, GW and Medhurst, AD (2003) Differences in the central nervous system distribution and pharmacology of the mouse 5-hydroxytryptamine-6 receptor compared with rat and human receptors investigated by radioligand binding, site-directed mutagenesis, and molecular modeling. Molecular Pharmacology 64, 12951308.Google Scholar
Hirst, WD, Cheung, NY, Rattray, M, Price, GW and Wilkin, GP (1998) Cultured astrocytes express messenger RNA for multiple serotonin receptor subtypes, without functional coupling of 5-HT1 receptor subtypes to adenylyl cyclase. Molecular Brain Research 61, 9099.Google Scholar
Ho, N, Xu, C and Thibault, G (2018) From the unfolded protein response to metabolic diseases - lipids under the spotlight. Journal of Cell Science 131, jcs199307.Google Scholar
Hodo, TW, de Aquino, MTP, Shimamoto, A and Shanker, A (2020) Critical neurotransmitters in the neuroimmune network. Frontiers in Immunology 11, 1869.Google Scholar
Holst, K, Guseva, D, Schindler, S, Sixt, M, Braun, A, Chopra, H, Pabst, O and Ponimaskin, E (2015) The serotonin receptor 5-HT(7)R regulates the morphology and migratory properties of dendritic cells. Journal of Cell Science 128, 28662880.Google Scholar
Horikawa, H, Kato, TA, Mizoguchi, Y, Monji, A, Seki, Y, Ohkuri, T, Gotoh, L, Yonaha, M, Ueda, T, Hashioka, S and Kanba, S (2010) Inhibitory effects of SSRIs on IFN-gamma induced microglial activation through the regulation of intracellular calcium. Progress in Neuro-Psychopharmacology and Biological Psychiatry 34, 13061316.Google Scholar
House, RV, Thomas, PT and Bhargava, HN (1994) Immunological consequences of in vitro exposure to lysergic acid diethylamide (LSD). Immunopharmacology and Immunotoxicology 16, 2340.Google Scholar
Idzko, M, Panther, E, Stratz, C, Muller, T, Bayer, H, Zissel, G, Durk, T, Sorichter, S, Di Virgilio, F, Geissler, M, Fiebich, B, Herouy, Y, Elsner, P, Norgauer, J and Ferrari, D (2004) The serotoninergic receptors of human dendritic cells: identification and coupling to cytokine release. Journal of Immunology 172, 60116019.Google Scholar
Iken, K, Chheng, S, Fargin, A, Goulet, AC and Kouassi, E (1995) Serotonin upregulates mitogen-stimulated B lymphocyte proliferation through 5-HT1A receptors. Cellular Immunology 163, 19.Google Scholar
Inoue, M, Okazaki, T, Kitazono, T, Mizushima, M, Omata, M and Ozaki, S (2011) Regulation of antigen-specific CTL and Th1 cell activation through 5-hydroxytryptamine 2A receptor. International Immunopharmacology 11, 6773.Google Scholar
Inserra, A, De Gregorio, D and Gobbi, G (2021) Psychedelics in psychiatry: neuroplastic, immunomodulatory, and neurotransmitter mechanisms. Pharmacological Reviews 73, 202277.Google Scholar
Inserra, A, Giorgini, G, Lacroix, S, Bertazzo, A, Choo, J, Markopolous, A, Grant, E, Abolghasemi, A, De Gregorio, D, Flamand, N, Rogers, G, Comai, S, Silvestri, C, Gobbi, G and Di Marzo, V (2023) Effects of repeated lysergic acid diethylamide (LSD) on the mouse brain endocannabinoidome and gut microbiome. British Journal of Pharmacology 180, 721739.Google Scholar
Ippolito, A, Vasudevan, S, Hurley, S, Gilmour, G, Westhorpe, F, Churchill, G and Sharp, T (2024) Increased 5-HT2A receptor signalling efficacy differentiates serotonergic psychedelics from non-psychedelics. BioRxiv. doi: 10.1101/2024.06.13.594677.Google Scholar
Ito, T, Ikeda, U, Shimpo, M, Yamamoto, K and Shimada, K (2000) Serotonin increases Interleukin-6 synthesis in human vascular smooth muscle cells. Circulation 102, 25222527.Google Scholar
Jalonen, TO, Margraf, RR, Wielt, DB, Charniga, CJ, Linne, ML and Kimelberg, HK (1997) Serotonin induces inward potassium and calcium currents in rat cortical astrocytes. Brain Research 758, 6982.Google Scholar
Jang, M, Gould, E, Xu, J, Kim, EJ and Kim, JH (2019) Oligodendrocytes regulate presynaptic properties and neurotransmission through BDNF signaling in the mouse brainstem. Elife 8, e42156.Google Scholar
Jarkas, DA, Villeneuve, AH, Daneshmend, AZB, Villeneuve, PJ and Mcquaid, RJ (2024) Sex differences in the inflammation-depression link: a systematic review and meta-analysis. Brain Behavior and Immunity 121, 257268.Google Scholar
Jia, J, Cheng, J, Wang, C and Zhen, X (2018) Sigma-1 receptor-modulated neuroinflammation in neurological diseases. Frontiers in Cellular Neuroscience 12, 314.Google Scholar
Johnston, JN, Kadriu, B, Allen, J, Gilbert, JR, Henter, ID, Zarate, CA Jr. (2023) Ketamine and serotonergic psychedelics: An update on the mechanisms and biosignatures underlying rapid-acting antidepressant treatment. Neuropharmacology 226, 109422.Google Scholar
Jones, EAK, Mitra, AK and Bhuiyan, AR (2021) Impact of COVID-19 on mental health in adolescents: a systematic review. International Journal of Environmental Research and Public Health 18, 2470.Google Scholar
Jones, NT, Zahid, Z, Grady, SM, Sultan, ZW, Zheng, Z, Razidlo, J, Banks, MI and Wenthur, CJ (2023) Transient elevation of plasma glucocorticoids supports psilocybin-induced anxiolysis in mice. ACS Pharmacology & Translational Science 6, 12211231.Google Scholar
Kadhim, HJ, Al-Mumen, H, Nahi, HH and Hamidi, SM (2022) Streptozotocin-induced alzheimer’s disease investigation by one-dimensional plasmonic grating chip. Scientific Reports 12, 21878.Google Scholar
Kamimura, D, Tanaka, Y, Hasebe, R and Murakami, M (2020) Bidirectional communication between neural and immune systems. International Immunology 32, 693701.Google Scholar
Kang, BN, Ha, SG, Bahaie, NS, Hosseinkhani, MR, Ge, XN, Blumenthal, MN, Rao, SP and Sriramarao, P (2013) Regulation of serotonin-induced trafficking and migration of eosinophils. PLoS One 8, e54840.Google Scholar
Katafuchi, T, Kondo, T, Take, S and Yoshimura, M (2005) Enhanced expression of brain interferon-alpha and serotonin transporter in immunologically induced fatigue in rats. European Journal of Neuroscience 22, 28172826.Google Scholar
Katoh, N, Soga, F, Nara, T, Tamagawa-Mineoka, R, Nin, M, Kotani, H, Masuda, K and Kishimoto, S (2006) Effect of serotonin on the differentiation of human monocytes into dendritic cells. Clinical & Experimental Immunology 146, 354361. doi: 10.1111/j.1365-2249.2006.03197.x.Google Scholar
Kavelaars, A (2002) Regulated expression of alpha-1 adrenergic receptors in the immune system. Brain Behavior and Immunity 16, 799807.Google Scholar
Keiser, MJ, Setola, V, Irwin, JJ, Laggner, C, Abbas, AI, Hufeisen, SJ, Jensen, NH, Kuijer, MB, Matos, RC, Tran, TB, Whaley, R, Glennon, RA, Hert, J, Thomas, KL, Edwards, DD, Shoichet, BK and Roth, BL (2009) Predicting new molecular targets for known drugs. Nature 462, 175181.Google Scholar
Kelley, DP, Venable, K, Destouni, A, Billac, G, Ebenezer, P, Stadler, K, Nichols, C, Barker, S and Francis, J (2022) Pharmahuasca and DMT rescue ROS production and differentially expressed genes observed after predator and psychosocial stress: relevance to human PTSD. ACS Chemical Neuroscience 13, 257274.Google Scholar
Kelly, JR, Clarke, G, Harkin, A, Corr, SC, Galvin, S, Pradeep, V, Cryan, JF, O’keane, V and Dinan, TG (2023) Seeking the psilocybiome: psychedelics meet the microbiota-gut-brain axis. International Journal of Clinical and Health Psychology 23, 100349.Google Scholar
Kerschensteiner, M, Gallmeier, E, Behrens, L, Leal, VV, Misgeld, T, Klinkert, WE, Kolbeck, R, Hoppe, E, Oropeza-Wekerle, RL, Bartke, I, Stadelmann, C, Lassmann, H, Wekerle, H and Hohlfeld, R (1999) Activated human T cells, B cells, and monocytes produce brain-derived neurotrophic factor in vitro and in inflammatory brain lesions: a neuroprotective role of inflammation? Journal of Experimental Medicine 189, 865870.Google Scholar
Kim, E, Carreira Figueiredo, I, Simmons, C, Randall, K, Rojo Gonzalez, L, Wood, T, Ranieri, B, Sureda-Gibert, P, Howes, O, Pariante, C, Nima, C, Pasternak, O, Dell’acqua, F, Turkheimer, F and Cash, D (2023) Mapping acute neuroinflammation in vivo with diffusion-MRI in rats given a systemic lipopolysaccharide challenge. Brain Behavior and Immunity 113, 289301.Google Scholar
Kinderlehrer, DA (2025) Mushrooms, microdosing, and mental illness: the effect of psilocybin on neurotransmitters, neuroinflammation, and neuroplasticity. Neuropsychiatric Disease and Treatment 21, 141155.Google Scholar
Kiraly, DD, Horn, SR, Van Dam, NT, Costi, S, Schwartz, J, Kim-Schulze, S, Patel, M, Hodes, GE, Russo, SJ, Merad, M, Iosifescu, DV, Charney, DS and Murrough, JW (2017) Altered peripheral immune profiles in treatment-resistant depression: response to ketamine and prediction of treatment outcome. Translational Psychiatry 7, e1065.Google Scholar
Kirkpatrick, P (2004) Of mice and men and rats. Nature Reviews Drug Discovery 3, 1515.Google Scholar
Kitson, SL (2007) 5-hydroxytryptamine (5-HT) receptor ligands. Current Pharmaceutical Design 13, 26212637.Google Scholar
Kolodziejczak, M, Bechade, C, Gervasi, N, Irinopoulou, T, Banas, SM, Cordier, C, Rebsam, A, Roumier, A and Maroteaux, L (2015) Serotonin modulates developmental microglia via 5-HT2B receptors: potential implication during synaptic refinement of retinogeniculate projections. ACS Chemical Neuroscience 6, 12191230.Google Scholar
Kong, EK, Peng, L, Chen, Y, Yu, AC and Hertz, L (2002) Up-regulation of 5-HT2B receptor density and receptor-mediated glycogenolysis in mouse astrocytes by long-term fluoxetine administration. Neurochemical Research 27, 113120.Google Scholar
Koupaei, MTS, Ahangari, G and Samanguiei, S (2010) Inflammatory mediator serotonin receptor gene (5-HTR3A) expression changes on human peripheral blood lymphocytes in rheumatoid arthritis. European Journal of Inflammation 8, 8388.Google Scholar
Kozlov, EM, Grechko, AV, Chegodaev, YS, Wu, WK and Orekhov, AN (2020) Contribution of Neurotrophins to the immune system regulation and possible connection to alcohol addiction. Biology (Basel) 9, 63.Google Scholar
Kozłowska, U, Klimczak, A, Wiatr, K and Figiel, M (2021). The DMT and psilocin treatment changes CD11B+ activated microglia immunological phenotype. BioRxiv.Google Scholar
Krabbe, G, Matyash, V, Pannasch, U, Mamer, L, Boddeke, HW and Kettenmann, H (2012) Activation of serotonin receptors promotes microglial injury-induced motility but attenuates phagocytic activity. Brain Behavior and Immunity 26, 419428.Google Scholar
Krupp, KT, Yaeger, JDW, Ledesma, LJ, Withanage, MHH, Gale, JJ, Howe, CB, Allen, TJ, Sathyanesan, M, Newton, SS and Summers, CH (2024) Single administration of a psychedelic [(R)-DOI] influences coping strategies to an escapable social stress. Neuropharmacology 252, 109949.Google Scholar
Kubera, M, Kenis, G, Bosmans, E, Scharpe, S and Maes, M (2000) Effects of serotonin and serotonergic agonists and antagonists on the production of interferon-gamma and interleukin-10. Neuropsychopharmacology 23, 8998.Google Scholar
Kubera, M, Maes, M, Kenis, G, Kim, YK and Lason, W (2005) Effects of serotonin and serotonergic agonists and antagonists on the production of tumor necrosis factor alpha and interleukin-6. Psychiatry Research 134, 251258.Google Scholar
Kushnir-Sukhov, NM, Gilfillan, AM, Coleman, JW, Brown, JM, Bruening, S, Toth, M and Metcalfe, DD (2006) 5-hydroxytryptamine induces mast cell adhesion and migration. Journal of Immunology 177, 64226432.Google Scholar
Laabi, S, LeMmon, C, Vogel, C, Chacon, M, Jimenez, VM Jr. (2024) Deciphering psilocybin: cytotoxicity, anti-inflammatory effects, and mechanistic insights. International Immunopharmacology 130, 111753.Google Scholar
Lam, PH, Chen, E, Chiang, JJ and Miller, GE (2022) Socioeconomic disadvantage, chronic stress, and proinflammatory phenotype: an integrative data analysis across the lifecourse. PNAS Nexus 1, pgac219.Google Scholar
Lamers, F, Milaneschi, Y, Vinkers, CH, Schoevers, RA, Giltay, EJ and Penninx, B (2020) Depression profilers and immuno-metabolic dysregulation: longitudinal results from the NESDA study. Brain Behavior and Immunity 88, 174183.Google Scholar
León-Ponte, M, Ahern, GP and O’connell, PJ (2007) Serotonin provides an accessory signal to enhance T-cell activation by signaling through the 5-HT7 receptor. Blood 109, 31393146.Google Scholar
Leposavic, G, Pilipovic, I and Perisic, M (2011) Cellular and nerve fibre catecholaminergic thymic network: steroid hormone dependent activity. Physiological Research 60, S7182.Google Scholar
Liao, Y, Tao, S, Wang, S, Wu, G, Yao, W, Yang, L, Huang, Q, Liu, Y, Yang, G and Yang, P (2023) 5-HT modulates the properties of dendritic cells to interfere with the development of type 1 regulating T cells. Molecular Immunology 160, 161167.Google Scholar
Liu, G, Ma, L, Qu, Y, Wan, X, Xu, D, Zhao, M, Murayama, R and Hashimoto, K (2023) Prophylactic effects of arketamine, but not hallucinogenic psychedelic DOI nor non-hallucinogenic psychedelic analog lisuride, in lipopolysaccharide-treated mice and mice exposed to chronic restrain stress. Pharmacology Biochemistry and Behavior 233, 173659.Google Scholar
Liu, RP, Zou, M, Wang, JY, Zhu, JJ, Lai, JM, Zhou, LL, Chen, SF, Zhang, X and Zhu, JH (2014) Paroxetine ameliorates lipopolysaccharide-induced microglia activation via differential regulation of MAPK signaling. Journal of neuroinflammation 11, 47.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, 31703182.Google Scholar
Lynall, ME, Turner, L, Bhatti, J, Cavanagh, J, de Boer, P, Mondelli, V, Jones, D, Drevets, WC, Cowen, P, Harrison, NA and Pariante, CM (2020) Peripheral blood cell-stratified subgroups of inflamed depression. Biological Psychiatry 88, 185196.Google Scholar
Mader, S, Brimberg, L and Diamond, B (2017) The role of brain-reactive autoantibodies in brain pathology and cognitive impairment. Frontiers in Immunology 8, 1101.Google Scholar
Madsen, MK, Fisher, PM, Burmester, D, Dyssegaard, A, Stenbaek, 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, 13281334.Google Scholar
Magrini, E, Szabò, I, Doni, A, Cibella, J and Viola, A (2011) Serotonin-mediated tuning of human helper T cell responsiveness to the chemokine Cxcl12. PLoS One 6, e22482.Google Scholar
Maier, SF (2003) Bi-directional immune-brain communication: implications for understanding stress, pain, and cognition. Brain Behavior and Immunity 17, 6985.Google Scholar
Malek, H, Ebadzadeh, MM, Safabakhsh, R, Razavi, A and Zaringhalam, J (2015) Dynamics of the HPA axis and inflammatory cytokines: insights from mathematical modeling. Computers in Biology and Medicine 67, 112.Google Scholar
Marshall, JS, Warrington, R, Watson, W and Kim, HL (2018) An introduction to immunology and immunopathology. Allergy, Asthma & Clinical Immunology 14, 49.Google Scholar
Marx, W, Mcguinness, AJ, Rocks, T, Ruusunen, A, Cleminson, J, Walker, AJ, Gomes-Da-Costa, S, Lane, M, Sanches, M, Diaz, AP, Tseng, PT, Lin, PY, Berk, M, Clarke, G, O’neil, A, Jacka, F, Stubbs, B, Carvalho, AF, Quevedo, J, Soares, JC and Fernandes, BS (2021) The kynurenine pathway in major depressive disorder, bipolar disorder, and schizophrenia: a meta-analysis of 101 studies. Molecular Psychiatry 26, 41584178.Google Scholar
Masdeu, JC, Pascual, B and Fujita, M (2022) Imaging neuroinflammation in neurodegenerative disorders. Journal of Nuclear Medicine 63, 45S52S.Google Scholar
Mason, NL, Szabo, A, Kuypers, KPC, Mallaroni, PA, De La Torre Fornell, R, Reckweg, JT, Tse, DHY, Hutten, N, 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.Google Scholar
Maxishima, M, Shiga, T, Shutoh, F, Hamada, S, Maeshima, T and Okado, N (2001) Serotonin 2A receptor-like immunoreactivity is detected in astrocytes but not in oligodendrocytes of rat spinal cord. Brain Research 889, 270273.Google Scholar
Mcallister, EJ, Apgar, JR, Leung, CR, Rickert, RC and Jellusova, J (2017) New methods to analyze B cell immune responses to thymus-dependent antigen sheep red blood cells. Journal of Immunology 199, 29983003.Google Scholar
Mercier, G, Lennon, AM, Renouf, B, Dessouroux, A, Ramauge, M, Courtin, F and Pierre, M (2004) Map kinase activation by fluoxetine and its relation to gene expression in cultured rat astrocytes. Journal of Molecular Neuroscience 24, 207216.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, 167180.Google Scholar
Mikulski, Z, Zaslona, Z, Cakarova, L, Hartmann, P, Wilhelm, J, Tecott, LH, Lohmeyer, J and Kummer, W (2010) Serotonin activates murine alveolar macrophages through 5-HT2C receptors. American Journal of Physiology-Lung Cellular and Molecular Physiology 299, L27280.Google Scholar
Miller, KJ and Gonzalez, HA (1998) Serotonin 5-HT2A receptor activation inhibits cytokine-stimulated inducible nitric oxide synthase in C6 glioma cells. Annals of the New York Academy of Sciences 861, 169173.Google Scholar
Millett, CE, Burdick, KE and Kubicki, MR (2022) The effects of peripheral inflammation on the brain-a neuroimaging perspective. Harvard Review of Psychiatry 30, 5458.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, Elsila, LV, Sakson, S, Cannarozzo, C, Diniz, C, Seiffert, N, Rubiolo, A, Haapaniemi, H, Meshi, E, Nagaeva, E, Ohman, T, Rog, T, Kankuri, E, Vilar, M, Varjosalo, M, Korpi, ER, Permi, P, Mineev, KS, Saarma, M, Vattulainen, I, Casarotto, PC and Castren, E (2023) Psychedelics promote plasticity by directly binding to BDNF receptor TrkB. Nature Neuroscience 26, 10321041.Google Scholar
Moncrieff, J, Cooper, RE, Stockmann, T, Amendola, S, Hengartner, MP and Horowitz, MA (2023) The serotonin theory of depression: a systematic umbrella review of the evidence. Molecular Psychiatry 28, 32433256.Google Scholar
Mossner, R and Lesch, KP (1998) Role of serotonin in the immune system and in neuroimmune interactions. Brain Behavior and Immunity 12, 249271.Google Scholar
Mota, CMD, Rodrigues-Santos, C, Fernandez, RAR, Carolino, ROG, Antunes-Rodrigues, J, Anselmo-Franci, JA and Branco, LGS (2017) Central serotonin attenuates LPS-induced systemic inflammation. Brain Behavior and Immunity 66, 372381.Google Scholar
Muir, J, Lin, S, Aarrestad, IK, Daniels, HR, Ma, J, Tian, L, Olson, DE and Kim, CK (2024) Isolation of psychedelic-responsive neurons underlying anxiolytic behavioral states. Science 386, 802810.Google Scholar
Muller, T, Durk, T, Blumenthal, B, Grimm, M, Cicko, S, Panther, E, Sorichter, S, Herouy, Y, Di Virgilio, F, Ferrari, D, Norgauer, J and Idzko, M (2009) 5-hydroxytryptamine modulates migration, cytokine and chemokine release and T-cell priming capacity of dendritic cells in vitro and in vivo . PLoS One 4, e6453.Google Scholar
Nakamura, K, Sato, T, Ohashi, A, Tsurui, H and Hasegawa, H (2008) Role of a serotonin precursor in development of gut microvilli. The American Journal of Pathology 172, 333–344. doi: 10.2353/ajpath.2008.070358.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.Google Scholar
Nau, F Jr., Yu, B, Martin, D and Nichols, CD (2013) Serotonin 5-HT2A receptor activation blocks TNF-alpha mediated inflammation in vivo . PLoS One 8, e75426.Google Scholar
Nguyen, L, Lucke-Wold, BP, Mookerjee, SA, Cavendish, JZ, Robson, MJ, Scandinaro, AL and Matsumoto, RR (2015) Role of sigma-1 receptors in neurodegenerative diseases. Journal of Pharmacological Sciences 127, 1729.Google Scholar
Nichols, DE (2004)Hallucinogens. Pharmacology & Therapeutics 101, 131181 Google Scholar
Nissen, JB, Hojgaard, D and Thomsen, PH (2020) The immediate effect of COVID-19 pandemic on children and adolescents with obsessive compulsive disorder. BMC Psychiatry 20, 511.Google Scholar
Nkadimeng, SM, Nabatanzi, A, Steinmann, CML and Eloff, JN (2020) Phytochemical, cytotoxicity, antioxidant and anti-inflammatory effects of Psilocybe natalensis magic mushroom. Plants (Basel) 9, 1127.Google Scholar
Norden, DM, Fenn, AM, Dugan, A and Godbout, JP (2014) Tgfbeta produced by Il-10 redirected astrocytes attenuates microglial activation. Glia 62, 881895.Google Scholar
North, ML, Khanna, N, Marsden, PA, Grasemann, H and Scott, JA (2009) Functionally important role for arginase 1 in the airway hyperresponsiveness of asthma. American Journal of Physiology-Lung Cellular and Molecular Physiology 296, L91120.Google Scholar
O’Connor, J C, Lawson, M A, André, C, Moreau, M, Lestage, J, Castanon, N, Kelley, K W, Dantzer, R (2009) Lipopolysaccharide-induced depressive-like behavior is mediated by indoleamine 2,3-dioxygenase activation in mice. Molecular Psychiatry 14, 511522.Google Scholar
O’Farrell, K, Harkin, A (2017) Stress-related regulation of the kynurenine pathway: relevance to neuropsychiatric and degenerative disorders. Neuropharmacology 112, 307323.Google Scholar
Oliveira, JMD, Butini, L, Pauletto, P, Lehmkuhl, KM, Stefani, CM, Bolan, M, Guerra, E, Dick, B, De Luca Canto, G and Massignan, C (2022) Mental health effects prevalence in children and adolescents during the COVID-19 pandemic: a systematic review. Worldviews on Evidence-Based Nursing 19, 130137.Google Scholar
Olson, DE (2018) Psychoplastogens: a promising class of plasticity-promoting neurotherapeutics. Journal of Experimental Neuroscience 12, 1179069518800508.Google Scholar
Olson, DE (2021) The subjective effects of psychedelics may not be necessary for their enduring therapeutic effects. ACS Pharmacology & Translational Science 4, 563567.Google Scholar
Ondicova, K, Tillinger, A, Pecenak, J and Mravec, B (2019) The vagus nerve role in antidepressants action: efferent vagal pathways participate in peripheral anti-inflammatory effect of fluoxetine. Neurochemistry International 125, 4756.Google Scholar
Oppong-Damoah, A, Curry, KE, Blough, BE, Rice, KC and Murnane, KS (2019) Effects of the synthetic psychedelic 2,5-dimethoxy-4-iodoamphetamine (Doi) on ethanol consumption and place conditioning in male mice. Psychopharmacology 236, 35673578.Google Scholar
Osimo, EF, Baxter, LJ, Lewis, G, Jones, PB and Khandaker, GM (2019) Prevalence of low-grade inflammation in depression: a systematic review and meta-analysis of CRP levels. Psychological Medicine 49, 19581970.Google Scholar
Pariante, CM, Makoff, A, Lovestone, S, Feroli, S, Heyden, A, Miller, AH and Kerwin, RW (2001) Antidepressants enhance glucocorticoid receptor function in vitro by modulating the membrane steroid transporters. British Journal of Pharmacology 134, 13351343.Google Scholar
Pellegrino, TC and Bayer, BM (2000) Specific serotonin reuptake inhibitor-induced decreases in lymphocyte activity require endogenous serotonin release. Neuroimmunomodulation 8, 179187.Google Scholar
Penninx, B, Lamers, F, Jansen, R, Berk, M, Khandaker, GM, De Picker, L and Milaneschi, Y (2025) Immuno-metabolic depression: from concept to implementation. The Lancet Regional Health 48, 101166.Google Scholar
Peviani, M, Salvaneschi, E, Bontempi, L, Petese, A, Manzo, A, Rossi, D, Salmona, M, Collina, S, Bigini, P and Curti, D (2014) Neuroprotective effects of the Sigma-1 receptor (S1R) agonist PRE-084, in a mouse model of motor neuron disease not linked to SOD1 mutation. Neurobiology of Disease 62, 218232.Google Scholar
Preller, KH, Burt, JB, Ji, JL, Schleifer, CH, Adkinson, BD, Stampfli, 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.Google Scholar
Purple, RJ, Gupta, R, Thomas, CW, Golden, CT, Froudist-Walsh, S and Jones, MW (2024) Short- and long-term reconfiguration of rat prefrontal cortical networks following single doses of psilocybin. BioRxiv. doi: 10.1101/2024.12.10.627734.Google Scholar
Qu, Y, Chang, L, Ma, L, Wan, X and Hashimoto, K (2023) Rapid antidepressant-like effect of non-hallucinogenic psychedelic analog lisuride, but not hallucinogenic psychedelic in lipopolysaccharide-treated mice. Pharmacology Biochemistry and Behavior 222, 173500.Google Scholar
Quintanilla, B, Zarate, CA Jr. and Pillai, A (2024) Ketamine’s mechanism of action with an emphasis on neuroimmune regulation: can the complement system complement ketamine’s antidepressant effects? Molecular Psychiatry 29, 28492858.Google Scholar
Raison, CL, Sanacora, G, Woolley, J, Heinzerling, K, Dunlop, BW, Brown, RT, Kakar, R, Hassman, M, Trivedi, RP, Robison, R, Gukasyan, N, Nayak, SM, Hu, X, O’donnell, KC, Kelmendi, B, Sloshower, J, Penn, AD, Bradley, E, Kelly, DF, Mletzko, T, Nicholas, CR, Hutson, PR, Tarpley, G, Utzinger, M, Lenoch, K, Warchol, K, Gapasin, T, Davis, MC, Nelson-Douthit, C, Wilson, S, Brown, C, Linton, W, Ross, S and Griffiths, RR (2023) Single-dose psilocybin treatment for major depressive disorder: a randomized clinical trial. JAMA 330, 843853.Google Scholar
Ransohoff, RM and Brown, MA (2012) Innate immunity in the central nervous system. Journal of Clinical Investigation 122, 11641171.Google Scholar
Raval, NR, Johansen, A, Donovan, LL, Ros, NF, Ozenne, B, Hansen, HD and Knudsen, GM (2021) A single dose of psilocybin increases synaptic density and decreases 5-HT(2A) receptor density in the pig brain. International Journal of Molecular Sciences 22, 835.Google Scholar
Ray, TS (2010) Psychedelics and the human receptorome. PLoS One 5, e9019.Google Scholar
Rhen, T and Cidlowski, JA (2005) Antiinflammatory action of glucocorticoids--new mechanisms for old drugs. New England Journal of Medicine 353, 17111723.Google Scholar
Rickli, A, Luethi, D, Reinisch, J, Buchy, D, Hoener, MC and Liechti, ME (2015) Receptor interaction profiles of novel N-2-methoxybenzyl (NBOMe) derivatives of 2,5-dimethoxy-substituted phenethylamines (2C drugs). Neuropharmacology 99, 546553.Google Scholar
Rijsketic, DR, Casey, AB, Barbosa, DAN, Zhang, X, Hietamies, TM, Ramirez-Ovalle, G, Pomrenze, MB, Halpern, CH, Williams, LM, Malenka, RC and Heifets, BD (2023) Unraveling the synergistic effects of psilocybin and environment on brain-wide immediate early gene expression in mice. Neuropsychopharmacology 48, 17981807.Google Scholar
Rinaldi, A, Chiaravalli, AM, Mian, M, Zucca, E, Tibiletti, MG, Capella, C and Bertoni, F (2010) Serotonin receptor 3A expression in normal and neoplastic B cells. Pathobiology 77, 129135.Google Scholar
Robinson, GI, Li, D, Wang, B, Rahman, T, Gerasymchuk, M, Hudson, D, Kovalchuk, O and Kovalchuk, I (2023) Psilocybin and eugenol reduce inflammation in human 3D epiintestinal tissue. Life (Basel) 13, 2345.Google Scholar
Rodriguiz, RM, Nadkarni, V, Means, CR, Pogorelov, VM, Chiu, YT, Roth, BL and Wetsel, WC (2021) LSD-stimulated behaviors in mice require beta-arrestin 2 but not beta-arrestin 1. Scientific Reports 11, 17690.Google Scholar
Rosen, DA, Seki, SM, Fernandez-Castaneda, A, Beiter, RM, Eccles, JD, Woodfolk, JA and Gaultier, A (2019) Modulation of the sigma-1 receptor-IRE1 pathway is beneficial in preclinical models of inflammation and sepsis. Science Translational Medicine 11, eaau5266.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, 11651180.Google Scholar
Rossetti, AC, Paladini, MS, Riva, MA and Molteni, R (2020) Oxidation-reduction mechanisms in psychiatric disorders: a novel target for pharmacological intervention. Pharmacology & Therapeutics 210, 107520.Google Scholar
Roumier, A, Béchade, C and Maroteaux, L (2019) Chapter 10 - serotonin and the immune system. In Pilowsky, PM (ed), Serotonin. Boston: Academic Press.Google Scholar
Rowsthorn, E, Pham, W, Nazem-Zadeh, MR, Law, M, Pase, MP and Harding, IH (2023) Imaging the neurovascular unit in health and neurodegeneration: a scoping review of interdependencies between MRI measures. Fluids and Barriers of the CNS 20, 97.Google Scholar
Ruan, Z, Zhang, D, Huang, R, Sun, W, Hou, L, Zhao, J and Wang, Q (2022) Microglial activation damages dopaminergic neurons through MMP-2/-9-mediated increase of blood-brain barrier permeability in a parkinsons disease mouse model. International Journal of Molecular Sciences 23, 2793.Google Scholar
Rudin, D, Areesanan, A, Liechti, ME and Grundemann, C (2023) Classic psychedelics do not affect T cell and monocyte immune responses. Frontiers in Psychiatry 14, 1042440.Google Scholar
Ruiz-Cantero, MC, Gonzalez-Cano, R, Tejada, MA, Santos-Caballero, M, Perazzoli, G, Nieto, FR and Cobos, EJ (2021) Sigma-1 receptor: a drug target for the modulation of neuroimmune and neuroglial interactions during chronic pain. Pharmacological Research 163, 105339.Google Scholar
Russo-Neustadt, AA, Alejandre, H, Garcia, C, Ivy, AS and Chen, MJ (2004) Hippocampal brain-derived neurotrophic factor expression following treatment with reboxetine, citalopram, and physical exercise. Neuropsychopharmacology 29, 21892199.Google Scholar
Rustenhoven, J and Kipnis, J (2022) Brain borders at the central stage of neuroimmunology. Nature 612, 417429.Google Scholar
Sacramento, PM, Monteiro, C, Dias, ASO, Kasahara, TM, Ferreira, TB, Hygino, J, Wing, AC, Andrade, RM, Rueda, F, Sales, MC, Vasconcelos, CC and Bento, CAM (2018) Serotonin decreases the production of Th1/Th17 cytokines and elevates the frequency of regulatory CD4(+) T-cell subsets in multiple sclerosis patients. European Journal of Immunology 48, 13761388.Google Scholar
Salaciak, K and Pytka, K (2022) Revisiting the sigma-1 receptor as a biological target to treat affective and cognitive disorders. Neuroscience & Biobehavioral Reviews 132, 11141136.Google Scholar
Salminen, A, Kaarniranta, K and Kauppinen, A (2020) ER stress activates immunosuppressive network: implications for aging and alzheimer’s disease. Journal of Molecular Medicine 98, 633650.Google Scholar
Sanches, RF, De Lima Osorio, 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, 7781.Google Scholar
Saulin, A, Savli, M and Lanzenberger, R (2012) Serotonin and molecular neuroimaging in humans using PET. Amino Acids 42, 20392057.Google Scholar
Schiepers, OJ, Wichers, MC and Maes, M (2005) Cytokines and major depression. Progress in Neuro-Psychopharmacology and Biological Psychiatry 29, 201217.Google Scholar
Schindler, EAD, Wallace, RM, Sloshower, JA and D’souza, DC (2018) Neuroendocrine associations underlying the persistent therapeutic effects of classic serotonergic psychedelics. Frontiers in Pharmacology 9, 177.Google Scholar
Schmid, CL and Bohn, LM (2010) Serotonin, but not N-methyltryptamines, activates the serotonin 2A receptor via a ss-arrestin2/Src/Akt signaling complex in vivo . Journal of Neuroscience 30, 1351313524.Google Scholar
Schmid, CL, Raehal, KM and Bohn, LM (2008) Agonist-directed signaling of the serotonin 2A receptor depends on beta-arrestin-2 interactions in vivo . Proceedings of the National Academy of Sciences 105, 10791084.Google Scholar
Schmidt, M, Hoffrichter, A, Davoudi, M, Horschitz, S, Lau, T, Meinhardt, M, Spanagel, R, Ladewig, J, Köhr, G and Koch, P (2024) Psilocin fosters neuroplasticity in IPSC-derived human cortical neurons.Google Scholar
Sershen, H, Hashim, A and Lajtha, A (1996) The effect of ibogaine on sigma- and NMDA-receptor-mediated release of [3H]dopamine. Brain Research Bulletin 40, 6367.Google Scholar
Shao, LX, Liao, C, Davoudian, PA, Savalia, NK, Jiang, Q, Wojtasiewicz, C, Tan, D, Nothnagel, JD, Liu, RJ, Woodburn, SC, Bilash, OM, Kim, H, Che, A and Kwan, AC (2025) Psilocybin’s lasting action requires pyramidal cell types and 5-HT2A receptors. Nature 642, 411420. doi: 10.1038/s41586-025-08813-6.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, 25352544 e4.Google Scholar
Sharma, D and Parameswaran, N (2015) Multifaceted role of beta-arrestins in inflammation and disease. Genes and Immunity 16, 499513.Google Scholar
Shen, YC, Wang, YH, Chou, YC, Liou, KT, Yen, JC, Wang, WY and Liao, JF (2008) Dimemorfan protects rats against ischemic stroke through activation of sigma-1 receptor-mediated mechanisms by decreasing glutamate accumulation. Journal of Neurochemistry 104, 558572.Google Scholar
Shi, M, Chen, F, Chen, Z, Yang, W, Yue, S, Zhang, J and Chen, X (2021) Sigma-1 receptor: a potential therapeutic target for traumatic brain injury. Frontiers in Cellular Neuroscience 15, 685201.Google Scholar
Shinozuka, K, Jerotic, K, Mediano, P, Zhao, AT, Preller, KH, Carhart-Harris, R and Kringelbach, ML (2024) Synergistic, multi-level understanding of psychedelics: three systematic reviews and meta-analyses of their pharmacology, neuroimaging and phenomenology. Translational Psychiatry 14, 485.Google Scholar
Silverman, MN and Sternberg, EM (2012) Glucocorticoid regulation of inflammation and its functional correlates: from HPA axis to glucocorticoid receptor dysfunction. Annals of the New York Academy of Sciences 1261, 5563.Google Scholar
Simpson, DSA and Oliver, PL (2020) ROS generation in microglia: understanding oxidative stress and inflammation in neurodegenerative disease. Antioxidants (Basel) 9, 743.Google Scholar
Simpson, KL, Weaver, KJ, De Villers-Sidani, E, Lu, JY, Cai, Z, Pang, Y, Rodriguez-Porcel, F, Paul, IA, Merzenich, M and Lin, RC (2011) Perinatal antidepressant exposure alters cortical network function in rodents. Proceedings of the National Academy of Sciences 108, 1846518470.Google Scholar
Singh, D (2022) Astrocytic and microglial cells as the modulators of neuroinflammation in Alzheimer’s disease. Journal of Neuroinflammation 19, 206.Google Scholar
Singh, H, Wray, N, Schappi, JM and Rasenick, MM (2018) Disruption of lipid-raft localized galpha(s)/tubulin complexes by antidepressants: a unique feature of HDAC6 inhibitors, SSRI and tricyclic compounds. Neuropsychopharmacology 43, 14811491.Google Scholar
Strawbridge, R, Izurieta, E, Day, E, Tee, H, Young, K, Tong, CC, Young, AH and Cleare, AJ (2023) Peripheral inflammatory effects of different interventions for treatment-resistant depression: a systematic review. Neuroscience Applied 2, 101014.Google Scholar
Sulimai, N, Brown, J and Lominadze, D (2023) Vascular effects on cerebrovascular permeability and neurodegeneration. Biomolecules 13, 648.Google Scholar
Sundman, E and Olofsson, PS (2014) Neural control of the immune system. Advances in Physiology Education 38, 135139.Google Scholar
Suneson, K, Grudet, C, Ventorp, F, Malm, J, Asp, M, Westrin, A and Lindqvist, D (2023) An inflamed subtype of difficult-to-treat depression. Progress in Neuro-Psychopharmacology and Biological Psychiatry 125, 110763.Google Scholar
Swiergiel, AH and Dunn, AJ (2006) Feeding, exploratory, anxiety- and depression-related behaviors are not altered in interleukin-6-deficient male mice. Behavioural Brain Research 171, 94108.Google Scholar
Szabo, A (2015) Psychedelics and immunomodulation: novel approaches and therapeutic opportunities. Frontiers in Immunology 6, 358.Google Scholar
Szabo, A, Gogolak, P, Koncz, G, Foldvari, Z, Pazmandi, K, Miltner, N, Poliska, S, Bacsi, A, Djurovic, S and Rajnavolgyi, E (2018) Immunomodulatory capacity of the serotonin receptor 5-HT2B in a subset of human dendritic cells. Scientific Reports 8, 1765.Google Scholar
Szabo, A, Kovacs, A, Frecska, E and Rajnavolgyi, E (2014) Psychedelic N, N-dimethyltryptamine and 5-methoxy-N, N-dimethyltryptamine modulate innate and adaptive inflammatory responses through the sigma-1 receptor of human monocyte-derived dendritic cells. PLoS One 9, e106533.Google Scholar
Szabo, I, Varga, VE, Dvoracsko, S, Farkas, AE, Kormoczi, T, Berkecz, R, Kecskes, S, Menyhart, A, Frank, R, Hantosi, D, Cozzi, NV, Frecska, E, Tomboly, C, Krizbai, IA, Bari, F and Farkas, E (2021) N, N-dimethyltryptamine attenuates spreading depolarization and restrains neurodegeneration by sigma-1 receptor activation in the ischemic rat brain. Neuropharmacology 192, 108612.Google Scholar
Taler, M, Gil-Ad, I, Lomnitski, L, Korov, I, Baharav, E, Bar, M, Zolokov, A and Weizman, A (2007) Immunomodulatory effect of selective serotonin reuptake inhibitors (SSRIs) on human T lymphocyte function and gene expression. European Neuropsychopharmacology 17, 774780.Google Scholar
Tao, P, Han, X, Wang, Q, Wang, S, Zhang, J, Liu, L, Fan, X, Liu, C, Liu, M, Guo, L, Lee, PY, Aksentijevich, I and Zhou, Q (2023) A gain-of-function variation in PLCG1 causes a new immune dysregulation disease. Journal of Allergy and Clinical Immunology 152, 12921302.Google Scholar
Teleanu, DM, Niculescu, AG, Lungu, Ii, Radu, CI, Vladacenco, O, Roza, E, Costachescu, B, Grumezescu, AM and Teleanu, RI (2022) An overview of oxidative stress, neuroinflammation, and neurodegenerative diseases. International Journal of Molecular Sciences 23, 5938.Google Scholar
Toh, ML and Miossec, P (2007) The role of T cells in rheumatoid arthritis: new subsets and new targets. Current Opinion in Rheumatology 19, 284288.Google Scholar
Tomaz, VS, Chaves Filho, AJM, Cordeiro, RC, Juca, PM, Soares, MVR, Barroso, PN, Cristino, LMF, Jiang, W, Teixeira, AL, De Lucena, DF and Macedo, DS (2020) Antidepressants of different classes cause distinct behavioral and brain pro- and anti-inflammatory changes in mice submitted to an inflammatory model of depression. Journal of Affective Disorders 268, 188200.Google Scholar
Tourino, MC, De Oliveira, EM, Belle, LP, Knebel, FH, Albuquerque, RC, Dorr, FA, Okada, SS, Migliorini, S, Soares, IS and Campa, A (2013) Tryptamine and dimethyltryptamine inhibit indoleamine 2,3 dioxygenase and increase the tumor-reactive effect of peripheral blood mononuclear cells. Cell Biochemistry and Function 31, 361364.Google Scholar
Turkin, A, Tuchina, O and Klempin, F (2021) Microglia function on precursor cells in the adult hippocampus and their responsiveness to serotonin signaling. Frontiers in Cell and Developmental Biology 9, 665739.Google Scholar
Turnbull, AV and Rivier, CL (1999) Regulation of the hypothalamic-pituitary-adrenal axis by cytokines: actions and mechanisms of action. Physiological Reviews 79, 171.Google Scholar
Tynan, RJ, Weidenhofer, J, Hinwood, M, Cairns, MJ, Day, TA and Walker, FR (2012) A comparative examination of the anti-inflammatory effects of SSRI and SNRI antidepressants on LPS stimulated microglia. Brain Behavior and Immunity 26, 469479.Google Scholar
Uthaug, MV, Lancelotta, R, Szabo, A, Davis, AK, Riba, J and Ramaekers, JG (2020) Prospective examination of synthetic 5-methoxy-N, N-dimethyltryptamine inhalation: effects on salivary Il-6, cortisol levels, affect, and non-judgment. Psychopharmacology (Berl) 237, 773785.Google Scholar
Van Heesch, F, Prins, J, Konsman, JP, Korte-Bouws, GA, Westphal, KG, Rybka, J, Olivier, B, Kraneveld, AD and Korte, SM (2014) Lipopolysaccharide increases degradation of central monoamines: an in vivo microdialysis study in the nucleus accumbens and medial prefrontal cortex of mice. European Journal of Pharmacology 725, 5563.Google Scholar
Vanderzwaag, J, Halvorson, T, Dolhan, K, Simoncicova, E, Ben-Azu, B and Tremblay, ME (2023) The missing piece? A case for microglia’s prominent role in the therapeutic action of anesthetics, ketamine, and psychedelics. Neurochemical Research 48, 11291166.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, 700706.Google Scholar
Verkhratsky, A, Parpura, V, Scuderi, C and Li, B (2021) Astroglial serotonin receptors as the central target of classic antidepressants. Advances in Neurobiology 26, 317347.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-HT(2A) receptor signaling pathways associated with psychedelic potential. Nature Communications 14, 8221.Google Scholar
Wang, J, He, W and Zhang, J (2023) A richer and more diverse future for microglia phenotypes. Heliyon 9, e14713.Google Scholar
Wang, L, Wang, R, Liu, L, Qiao, D, Baldwin, DS and Hou, R (2019) Effects of SSRIs on peripheral inflammatory markers in patients with major depressive disorder: a systematic review and meta-analysis. Brain Behavior and Immunity 79, 2438.Google Scholar
Wang, X, Wang, K, Wu, X, Huang, W and Yang, L (2022) Role of the Camp-PKA-CREB-BDNF pathway in abnormal behaviours of serotonin transporter knockout mice. Behavioural Brain Research 419, 113681.Google Scholar
Warren, AL, Lankri, D, Cunningham, MJ, Serrano, IC, Parise, LF, Kruegel, AC, Duggan, P, Zilberg, G, Capper, MJ, Havel, V, Russo, SJ, Sames, D and Wacker, D (2024) Structural pharmacology and therapeutic potential of 5-methoxytryptamines. Nature 630, 237246. doi: 10.1038/s41586-024-07403-2.Google Scholar
Weber, MD, Godbout, JP and Sheridan, JF (2017) Repeated social defeat, neuroinflammation, and behavior: monocytes carry the signal. Neuropsychopharmacology 42, 4661.Google Scholar
Werneburg, S, Feinberg, PA, Johnson, KM and Schafer, DP (2017) A microglia-cytokine axis to modulate synaptic connectivity and function. Current Opinion in Neurobiology 47, 138145.Google Scholar
Wiens, KR, Brooks, NAH, Riar, I, Greuel, BK, Lindhout, IA and Klegeris, A (2024) Psilocin, the psychoactive metabolite of psilocybin, modulates select neuroimmune functions of microglial cells in a 5-HT(2) receptor-dependent manner. Molecules 29.Google Scholar
Wiersma, VR (2020) Chapter 4 - lectins as modulators of autophagy in cancer immunotherapy. In Chouaib, S (ed), Autophagy in immune response: impact on cancer immunotherapy. Academic Press.Google Scholar
Wong, AC, Devason, AS, Umana, IC, Cox, TO, Dohnalova, L, Litichevskiy, L, Perla, J, Lundgren, P, Etwebi, Z, Izzo, LT, Kim, J, Tetlak, M, Descamps, HC, Park, SL, Wisser, S, Mcknight, AD, Pardy, RD, Kim, J, Blank, N, Patel, S, Thum, K, Mason, S, Beltra, JC, Michieletto, MF, Ngiow, SF, Miller, BM, Liou, MJ, Madhu, B, Dmitrieva-Posocco, O, Huber, AS, Hewins, P, Petucci, C, Chu, CP, Baraniecki-Zwil, G, Giron, LB, Baxter, AE, Greenplate, AR, Kearns, C, Montone, K, Litzky, LA, Feldman, M and Henao-Mejia, J (2023) Serotonin reduction in post-acute sequelae of viral infection. Cell 186, 48514867.Google Scholar
Wrona, D (2006) Neural-immune interactions: an integrative view of the bidirectional relationship between the brain and immune systems. Journal of Neuroimmunology 172, 3858.Google Scholar
Wsol, A (2023) Cardiovascular safety of psychedelic medicine: current status and future directions. Pharmacological Reports 75, 13621380.Google Scholar
Wu, C, Singh, SK, Dias, P, Kumar, S and Mann, DM (1999) Activated astrocytes display increased 5-ht2a receptor expression in pathological states. Experimental Neurology 158, 529533.Google Scholar
Yin, J, Albert, RH, Tretiakova, AP and Jameson, BA (2006) 5-ht(1B) receptors play a prominent role in the proliferation of T-lymphocytes. Journal of Neuroimmunology 181, 6881.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, 316323.Google Scholar
Yu, S-J, Wu, K-J, Wang, Y-S, Bae, E, Chianelli, F, Bambakidis, N and Wang, Y (2024) Neuroprotective effects of psilocybin in a rat model of stroke. BMC Neuroscience 25, 49.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.Google Scholar
Zanikov, T, Gerasymchuk, M, Robinson, GI, Gojani, EG, Asghari, S, Groves, A, Cameron, M, Rodriguez-Juarez, R, Snelling, A, Hudson, D, Fiselier, A, Kovalchuk, O and Kovalchuk, I (2024) Psilocybin and eugenol prevent dss-induced neuroinflammation in mice. Biocatalysis and Agricultural Biotechnology 56, 103033.Google Scholar
Zefferino, R, Di Gioia, S and Conese, M (2021) Molecular links between endocrine, nervous and immune system during chronic stress. Brain and Behavior 11, e01960.Google Scholar
Zeng, Y, Chourpiliadis, C, Hammar, N, Seitz, C, Valdimarsdottir, UA, Fang, F, Song, H and Wei, D (2024) Inflammatory biomarkers and risk of psychiatric disorders. JAMA Psychiatry 81, 11181129.Google Scholar
Zhang, G, Li, Q, Tao, W, Qin, P, Chen, J, Yang, H, Chen, J, Liu, H, Dai, Q and Zhen, X (2023a) Sigma-1 receptor-regulated efferocytosis by infiltrating circulating macrophages/microglial cells protects against neuronal impairments and promotes functional recovery in cerebral ischemic stroke. Theranostics 13, 543559.Google Scholar
Zhang, JC, Wu, J, Fujita, Y, Yao, W, Ren, Q, Yang, C, Li, SX, Shirayama, Y and Hashimoto, K (2014) Antidepressant effects of TrkB ligands on depression-like behavior and dendritic changes in mice after inflammation. The International Journal of Neuropsychopharmacology 18.Google Scholar
Zhang, JC, Yao, W and Hashimoto, K (2016) Brain-derived neurotrophic factor (BDNF)-trkB signaling in inflammation-related depression and potential therapeutic targets. Current Neuropharmacology 14, 721731.Google Scholar
Zhang, W, Xiao, D, Mao, Q and Xia, H (2023b) Role of neuroinflammation in neurodegeneration development. Signal Transduction and Targeted Therapy 8, 267.Google Scholar
Zhang, Y, Damjanoska, KJ, Carrasco, GA, Dudas, B, D’souza, DN, Tetzlaff, J, Garcia, F, Hanley, NR, Scripathirathan, K, Petersen, BR, Gray, TS, Battaglia, G, Muma, NA and Van De Kar, LD (2002) Evidence that 5-ht2A receptors in the hypothalamic paraventricular nucleus mediate neuroendocrine responses to (-)DOI. Journal of Neuroscience 22, 96359642.Google Scholar
Zhao, X, Cao, F, Liu, Q, Li, X, Xu, G, Liu, G, Zhang, Y, Yang, X, Yi, S, Xu, F, Fan, K and Ma, J (2019) Behavioral, inflammatory and neurochemical disturbances in LPS and UCMS-induced mouse models of depression. Behavioural Brain Research 364, 494502.Google Scholar
Zhou, L, Ma, SL, Yeung, PK, Wong, YH, Tsim, KW, So, KF, Lam, LC and Chung, SK (2016) Anxiety and depression with neurogenesis defects in exchange protein directly activated by cAMP 2-deficient mice are ameliorated by a selective serotonin reuptake inhibitor, Prozac. Translational Psychiatry 6, e881.Google Scholar
Figure 0

Figure 1. Heat map visualisation of serotonergic receptor binding profiles. Yellow cells with crosses indicate known binding activity of ligands (rows) to specific 5-HT receptors (columns), while dark purple cells indicate no reported binding. Data compiled from: Nichols (2004); Kitson (2007); Keiser et al. (2009); Besnard et al. (2012); Rickli et al. (2015); Wsol (2023); Hatzipantelis and Olson (2024); Ippolito et al. (2024). 2C-(x) refers to the family of 2,5-dimethoxy-phenethylamine analogues. Note that binding affinity varies based on pharmacological method, cell type, and experimental conditions. The psychoactive drug screening programme (PDSP) has been a primary source for standardised binding data (Ki values) for many of these compounds, as reviewed in Alexander et al. (2024); Hatzipantelis and Olson (2024).

Figure 1

Figure 2. Sankey diagram depicting 5-HT receptor expression in peripheral immune cells and their associated cellular functions. Function-specific references are provided for each cell type. Second messengers and their targets are identified for specific receptors in certain immune cells: 5-HT2AR in Eos is associated to increased intracellular Ca2+ and to the activation of ROCK, MAPK, PI3K, PKC, and Calmodulin (Boehme et al., 2004; Kang et al., 2013), 5-HT1AR in both T- and B-cells has been associated to NF-κB translocation (Abdouh et al., 2004), 5-HT7R in T-cells has been found associated to the activation of ERK1/2 and translocation of NF-κB (León-Ponte et al., 2007), 5-HT2CR in macrophages has been associated to increased intracellular Ca2+ (Mikulski et al., 2010), and 5-HT4R and 5-HT7R in dendritic cells have been associated to increases in cAMP levels (Muller et al., 2009).

Figure 2

Table 1. Summary of in vitro studies having measured immunomodulatory properties of 5-HT2AR psychedelics

Figure 3

Table 2. Summary of in vivo studies, both in rodents and humans, having measured immunomodulatory properties of 5-HT2AR psychedelics