Volume 9 Issue 5, December 2023, pp. 28-37

LAY SUMMARY

Concussion is a type of acquired brain injury that is common in the military, as well as among civilians and contact sport athletes, and is defined by a transient impairment in mental function. Nevertheless, concussion presents a considerable health burden, and a small minority of people suffer from continued impairment. Repetitive sub-concussive head injury is a known risk factor for neurodegenerative disease, including dementias. Concussions are difficult to treat because symptom profiles vary, but psychedelic therapies may help address some of the neurological issues that occur after brain injury. Classic psychedelics show promise as an emerging pharmacological intervention because they appear to help the brain to rewire, and they have anti-inflammatory effects. Nutraceutical interventions are widely available, cost-effective, and well tolerated, and they could also support recovery when combined with psychedelic compounds. Here, studies presenting classical psychedelics and nutraceuticals that may be combined with psychedelics as therapeutic strategies for the treatment of concussions and persistent symptoms are discussed.

Mild traumatic brain injury (mTBI), also referred to as concussion, is an acquired brain injury that is common in the military. mTBI results in alterations to brain function that are underpinned by a cascade of neuropathological and neurometabolic events that can result in excitotoxicity, oxidative stress, edema, neuroinflammation, and cell death. To date, traditional pharmaceutical therapies have been used with limited success to treat mTBI, leading to interest in the potential for psychedelic and nutraceutical therapies to alleviate the deleterious pathological sequelae that follow neurotrauma. An accumulating body of evidence supports research into the use of classic psychedelics and nutraceutical interventions for treating mTBI and concussion. This article highlights pre-clinical and clinical studies that show classic psychedelics are promising pharmacological interventions for mTBI because of their potent neuroplastic properties — including synaptogenesis and large-scale re-wiring, anti-inflammatory effects, and modulatory properties — on neural excitation and inhibition. The addition of nutraceutical interventions could also hasten recovery and improve general functioning along the spectrum from mild acute injury to long-term chronic issues (e.g., persistent post-concussive symptoms) and, potentially, to other forms of neurodegenerative disorders after neurotrauma and repetitive head injury.

Le traumatisme craniocérébral léger (TCCL), également appelé commotion cérébrale, est une lésion cérébrale acquise, fréquemment observée chez les militaires. Le TCCL entraine des modifications de la fonction cérébrale qui sont renforcées par une cascade d’évènements neuropathologiques et neurométaboliques pouvant entrainer une excitotoxicité, un stress oxydatif, un œdème, une neuroinflammation et la mort cellulaire. À ce jour, le succès des thérapies pharmaceutiques classiques à traiter les TCCL demeure limité, ce qui suscite de l’intérêt pour le potentiel des thérapies psychotropes et nutraceutiques à soulager les séquelles pathologiques délétères qui suivent un neurotraumatisme. Des données toujours plus nombreuses encouragent la recherche sur l’utilisation des psychotropes et des interventions nutraceutiques dans le traitement des traumatismes craniocérébraux légers et des commotions cérébrales. Cet article signale des études précliniques et cliniques qui montrent que les psychotropes constituent des interventions pharmacologiques prometteuses pour les TCCL, en raison de leurs puissantes propriétés neuroplastiques – synaptogenèse et recâblage à grande échelle, effets antiinflammatoires et propriétés modulatrices, entre autres – agissant sur l’excitation et l’inhibition neuronales. Des interventions nutraceutiques complémentaires pourraient également accélérer le rétablissement et améliorer le fonctionnement général, dans le spectre allant des blessures aigües légères aux problèmes chroniques à long terme (p. ex. les symptômes postcommotionnels persistants) et, potentiellement, pour d’autres formes de troubles neurodégénératifs suivant un neurotraumatisme ou un traumatisme crânien répété.

Traumatic brain injury (TBI) is a major cause of death and disability worldwide, and it is common in the military,1 often associated with falls, assaults, collisions, contact sports, whiplash, or an overpressure shockwave from a blast. The severities of TBI cover a broad continuum from mild, to moderate, to severe, with 80% to 95% being mild TBIs (mTBIs), commonly referred to as concussion. After the initial primary mechanical injury, a cascade of neurochemical and neurometabolic events occur in the brain, called secondary injury (Figure 1). Secondary injury is a potential therapeutic target.

Figure 1. Primary traumatic injury typically occurs as a result of a direct impact to the brain from biomechanical forces

Note: This momentum causes the brain to collide with the inner skull (coup and contracoup injury), creating localised and distributed damage through compression or stretching, ranging from regional contusions to diffuse axonal injuries caused by the deformation and shearing of axons. In the secondary injury cascade, neuroactive molecules and immune response cause neuronal dysfunction and an enduring state of inflammation. This secondary neurochemical cascade can be more damaging than the primary insult and may be a potential target of therapeutic intervention with psychedelic-based drugs. Ameliorating secondary injury processes may mitigate enduring structural and functional damage to the brain, as well as neurodegenerative and psychiatric disorders. Image created in http://biorender.com.

Repeated concussions or sub-concussions are concerning for military personnel involved in training exercises and combat duty because they may precipitate deleterious neurobiological responses that can lead to pronounced neurofunctional changes.2,3 The intense physical training routines of military personnel place high metabolic demands on the brain and body. In tandem with stress placed on the brain from physical activity, concussions and sub-concussions cause neurometabolic crises as a result of increased energy demands.3

Although most individuals recover from a concussion within 14 days, a significant proportion of individuals — up to 30% — experience persistent cognitive and behavioural impairment, termed persistent post-concussive symptoms.4 Moreover, a single injury or the cumulative effects of repeated concussions pose a risk factor for secondary neuropsychiatric illness.4 Cognitive impairments and mood shift after mTBI have negative implications for quality of life and, in a military context, significantly impair fighting and operational readiness.

Emerging research and clinical trials using psychedelic-assisted psychotherapy led to initiation of larger-scale Phase 2 and 3 clinical trials for major depressive disorder,57 a common comorbidity among people with chronic mTBI. Moreover, in this article, the authors propose that the neurobiological actions of psychedelics could address pathologies associated with neurological disorders, including mTBI.

Pre-clinical and clinical research with classic psychedelic compounds

Classic psychedelics include psilocybin, the prodrug form of psilocin, N,N-dimethyltryptamine (DMT), 5-MeO-DMT, mescaline, and lysergic acid diethylamide (LSD). Non-classic psychedelics are synthetic derivatives of these classic compounds and include 2,5-Dimethoxy-4-iodoamphetamine (DOI) and 4-Bromo-2,5-dimethoxyphenethylamine, as well as dissociative drugs such as ketamine and ibogaine.

The G-protein-coupled serotonin (5-hydroxytryptamine) 2A (5-HT2A) receptor is the common target of psychedelic compounds to elicit hallucinogenic effects. The 5-HT2A receptor is widely expressed in cortical brain regions and is also found in endocrine, endothelial, immune, and muscle tissue, thus providing multiple pharmacological targets.8 Psychedelic drugs also activate other serotonin receptor subtypes, including 5-HT1A, 5-HT2B, and 5-HT2C.

Brain injuries can alter 5-HT2A receptor signalling in the brain, as shown in a recent pre-clinical study in mice in which blast-induced closed-head mTBI increased cortical 5-HT2A receptor sensitivity and ex vivo radioligand binding, corresponding with social behaviour deficits that were reversed by repeated administration of DOI.9 However, if 5-HT2A receptor sensitivity is chronically altered, patients with mTBI may have different psychedelic dosing requirements to reach therapeutic efficacy.

Psychedelic drugs can promote neuroplasticity

Psychedelic drugs have been shown to have neurobiological effects that include altering neuronal structure,10 brain activity,11 and functional connectivity,12,13 which may provide novel approaches for those experiencing prolonged effects of concussion. Pre-clinical studies using in vitro and in vivo models show that psychedelic drugs interact with neurogenic pathways, such as tropomyosin receptor kinase B and the mammalian target of rapamycin (mTOR), in a manner equivalent to brain-derived neurotrophic factor (BDNF), which is key for the repair, maintenance, and survival of neurons.10

The plasticity-enhancing effects of psychedelic drugs may provide therapeutic avenues for people with neurodegenerative conditions,10,14 including mTBI (Figure 2). Pre-clinical studies show that DMT can increase dendritic spine density and hippocampal neurogenesis and reduce cytokine expression, mediated in part by its affinity for sigma-1 receptors.10,15 However, currently no clear evidence exists that physiological doses of DMT in humans lead to concentrations in the brain high enough to engage sigma-1 receptors. In addition, a single psilocybin treatment has been shown to rapidly increase dendritic spine density in the mouse prefrontal cortex, an effect that was observed 24 hours after treatment and persisted for a month.16 Psychedelic-evoked spineogenesis is proposed to result from indirect modulation of synaptic architecture by elevated glutamate levels acting through BDNF/tropomyosin receptor kinase B and mTOR signalling, leading to protein synthesis and synapse formation.17 mTBI is associated with the loss of functional N-methyl-D-aspartate (NMDA) receptors, which persists for weeks after initial injury;18 therefore, the elevation of glutamate by psychedelics may restore aspects of structural and functional plasticity. In addition, one study established an association between ayahuasca-induced increases in BDNF and decreased depression symptoms in people with major depressive disorder.19

Figure 2. Psychedelic compounds can elicit both neuroplastic and anti-inflammatory effects that may attenuate or counter neuropathological events caused by mild traumatic brain injury.

Note: Psychedelic drugs can increase protein and gene expression of neurotrophic factors, including brain-derived neurotrophic factor, glial-cell-derived neurotrophic factor, and nerve growth factor, as well as increasing expression of immediate early genes such as c-Fos, early growth response protein 1/2, and homer1, which can alter synaptic plasticity. Reduced gene and protein expression of pro-inflammatory cytokines include interleukin (IL)-1β, IL-6, and tumour necrosis factor-α, and increased anti-inflammatory cytokines include IL-10 and transforming growth factor-β. Immunomodulatory hormones include prolactin, cortisol, and prolactin. Image created in http://biorender.com.

Psychedelic drugs can decrease inflammation

The initial oxidative stress and neuroinflammation that follow a TBI play a beneficial role in recovery; however, chronic activation of microglia affects neuronal regeneration, including neurogenesis.20 The strain on mitochondrial capacity to scavenge reactive oxygen species (ROS) that results in heightened oxidative stress is a key contributor to neuron damage and death after trauma.21 As shown in Figure 2, psychedelics might elicit neuroprotective activity by decreasing apoptotic protease activating factor 1 and pro-inflammatory cytokines (e.g., interleukin [IL]-1 β, IL-6) while up-regulating the expression of neurogenic and anti-inflammatory factors (e.g., BDNF, glial-derived neurotrophic factor, IL-10).22

The anti-inflammatory capacity of psychedelics on mouse neural tissue in vivo has recently been shown after pre-treatment with psilocybin (0.88 mg/kg), which decreased lipopolysaccharide (LPS)-induced messenger RNA (mRNA) expression of IL-6, cyclooxygenase-2, and tumour necrosis factor (TNF)-α, and post-treatment psilocybin reduced LPS-induced mRNA expression of IL-6 and TNF-α.23 In vitro, DOI has been shown to decrease inflammation markers in tissue and cell cultures stimulated with TNF-α,24,25 which activates nuclear factor kappa B (NF-κB) signalling cascades to evoke inflammation.

The translatability from rodent cells to human immune cells is still in question, because a recent study demonstrated that the 5-HT2A partial agonists LSD, psilocin, DMT, and mescaline did not attenuate lipopolysaccharide (LPS)-triggered NF-κB induction in human monocytes.26 However, studies examining the anti-inflammatory effects of the full 5-HT2A agonist R-DOI on non-human cells have used TNF-α as an inflammatory stimulus,24,25 which induces inflammation via different pathways to LPS.

Gastrointestinal (GI) dysfunction is one of several complications among TBI patients that may contribute to systemic inflammation (Figure 3). Neurotrauma can damage gut mucosa and alter gut microbiome composition.27,28 Because of the expression of serotonin receptors in the gut, orally ingested psychedelic drugs may affect the GI tract, and a recent study showed that repeated LSD treatment altered gut microbiome composition in mice.29 In smooth muscle cell culture models of inflammation, psychedelic drugs have potent anti-inflammatory effects,24 and, as such, development of low-dose psychedelics to treat peripheral inflammation may be harnessed as a therapeutic approach in mTBI, as well as in other psychiatric and GI disorders.

Figure 3. How TBI can cause systemic inflammation and alter gastrointestinal tract function and how psychedelics may diminish these effects

Note: Altered gut microbiome composition can follow brain injury, including increases in populations of pathological microbes, such as Helicobacter, Campylobacter, and Escherichia coli. This can influence neurological health via the gut-microbiome-brain axis — changing neurotransmission, neuroplasticity, microglial activation, and behaviour. Accordingly, therapeutic alterations to the gut microbiota, or attenuating microglia priming or reduced systemic inflammation by psychedelics, may temper the neuropsychiatric consequences of mild TBI. Red arrows indicate the bidirectional gut-microbiome-brain axis. Image created in http://biorender.com.

TBI = traumatic brain injury.

Psychedelics may restore brain network connectivity disrupted by traumatic brain injury

After mTBI, damage to neurons and synaptic loss result in reduced neural dynamics and functional complexity.30 An objective measure of neural network dynamics can be calculated from resting-state functional imaging with electroencephalography or magnetoencephalography (MEG). It has been shown that neural complexity is reduced across multiple brain regions among military Veterans who experienced mTBIs relative to healthy controls.31 In addition, performance of motor responses, visual perception, and memory correlated with functional complexity, and, as such, the decreased repertoire of neural states measured in MEG signals in TBI is likely the result of injured neurons and damaged network connectivity.32 The acute administration of psilocybin, LSD, or ketamine increased spontaneous neural complexity measured by MEG,32 and psilocybin globally increased brain network integration measured with functional MRI.33 This observation suggests that psychedelic therapies may restore the impaired dynamic neural repertoire and improve cognitive flexibility;34 however, this needs to be tested with patients with mTBI, and the durability of such brain state changes is as yet unknown.

Entourage effects in naturally derived psychedelics

The term entourage effect refers to the synergistic interaction of two or more co-administered compounds. In the case of the psychedelic preparation ayahuasca, β-carboline derivative harmala alkaloids are monoamine oxidase inhibitors that allow DMT to enter the systemic circulation and central nervous system but may also enhance neuroplastogenic effects.35 Harmine, tetrahydroharmine, and harmaline, the three main alkaloids, and the harmine metabolite harmol stimulate adult neurogenesis in vitro and increase neural stem cell proliferation, migration, and differentiation into adult neurons,36 and harmine and harmaline possess antioxidant properties.36 Pharmahuasca, a form of DMT co-dosed with defined quantities of specific β-carbolines, may be used to reduce the confounds of heterogeneous ayahuasca preparations.

Psilocybin, baeocystin, norbaeocystin, and aeruginascin are tryptamines present in psilocybe mushrooms. Although less studied than psilocybin and psilocin, in vitro assessment of baeocystin, norbaeocystin, and aeruginascin shows that they display affinity for 5-HT2A and 5-HT1A in mouse brain tissue and exhibit agonist efficacy in assays examining 5-HT2A-mediated calcium mobilization and β-arrestin 2 recruitment while not evoking head-twitch response in mice in vivo,37 a proxy for 5-HT2A-induced hallucinogenic effects. Moreover, botanical psilocybe extracts were 10 times as potent as synthetic psilocin at inducing head-twitch responses in mice,38 further suggesting synergistic effects. Although clinical trials can use both synthetic and naturally derived psilocybin, whole psilocybe extracts may have some advantages because they contain a spectrum of tryptamines, although these require further characterization and likely must be delivered in defined quantities, similar to pharmahuasca in clinical studies.

Nutraceutical compounds

Non-psychedelic functional mushrooms fall into the category of nutraceutical compounds — food or dietary supplements with potential physiological benefits. Lion’s mane (Hericium erinaceus) and reishi (Ganoderma lucidum) have a long history of use in traditional Eastern medicines and are becoming recognized as nutraceutical approaches to neurological health through neuroprotective and anti-inflammatory mechanisms. The putative neuroregenerative mechanism of H. erinaceus has been examined pre-clinically, and it has recently been shown that the bioactive compounds N-dephenylethyl isohericerin and hericene A promote extensive axon outgrowth and neurite branching, enhanced hippocampal memory, and increased neurotrophin expression and downstream signalling.39

Extracts from G. lucidum have been shown to downregulate pro-inflammatory cytokine expression in microglia cell culture via attenuation of NF-κB and mitogen-activated protein kinase signalling pathways after stimulation with lipopolysaccharide.40 Ganoderma microsporum (another species of Ganoderma mushrooms native to Taiwan) supplementation has been explored in a penetrating cortical damage model of TBI in rats that reduced astrogliosis, increased anti-oxidative superoxide dismutase type 1 expression, and improved spatial memory deficits.41

Synergistic interactions between psychedelic and non-psychedelic compounds

The combination of functional mushrooms, particularly lion’s mane and niacin with sub-perceptual micro-doses of psilocybin or LSD, which refers to the administration of doses equal to approximately 10% of the hallucinogenic doses used recreationally, is a growing practice in individuals. The micro-dosing literature remains early and exploratory, with clear limitations on existing study designs, such as lack of randomized placebo-controlled trials,42 and therapeutic margins of receptor occupancy have yet to be determined. Citizen science studies of micro-dosing have shown some mental health benefits in humans, as well as improvements in psychomotor performance among older adults.43 Further research, including the examination of objective biomarkers of inflammation and neuroplasticity in humans and rodents, is needed to establish whether there are synergistic effects of low doses of psilocybin combined with lion’s mane or niacin.

N-acetyl-cysteine (NAC) has been shown to have antioxidant and neurovascular-protective effects after TBI.44 NAC administration has been shown to protect against oxidative injury mediated by ROS and to reduce inflammatory markers in a rat model of TBI,45 as well as to enhance cognitive recovery.46 In a human study, blast-exposed military personnel who received NAC supplementation within 24 hours of TBI exposure had a greater resolution of symptoms than controls who received placebo.47 The potential synergy between NAC and psilocybin is currently being investigated in a Phase 1 clinical trial for posttraumatic stress disorder.

Conclusions, limitations, and future research

To be an effective moderator of neurological outcomes at multiple points along the mTBI continuum, a therapeutic intervention needs to restore cognitive reserve, attenuate or modify signalling inflammatory cascades in the brain and other organ systems, such as the GI tract, and support neural compensation and repair. Moreover, the experimental models of mTBI differ greatly from the biomechanics of mTBIs experienced by humans (Figure 4). If pre-clinical studies do translate to humans, the neuroplasticity-enhancing and anti-inflammatory capacity of psychedelic compounds may provide a two-pronged approach to therapies for mTBI.

Figure 4. Preclinical models of TBI can inform about the neurological, morphological, biochemical, and behavioural outcomes of injury in a controlled and replicable setting, but there are translational limitations.

Note: Rodents have small, lissencephalic brains with less cerebrospinal fluid and white matter than the large, gyrencephalic human brains; thus, the biomechanics of TBIs differ substantially. The consequences of the controlled experimental injury mechanisms used in rodent studies (e.g., fluid percussion injury, closed cortical impact, weight drop) differ from the heterogeneous injury mechanisms in human TBIs, so success with interventions in rodents might not always be translatable to humans. Nonetheless, rodent models can help reveal therapeutic mechanisms and provide proof-of-concept information to drive clinical investigations.

TBI = traumatic brain injury; mTBI = mild traumatic brain injury.

Whether psychedelic compounds can address mTBI neuropathologies in humans is currently unknown, and currently no clinical trials are investigating psychedelic compounds with TBI as a direct indication. Moreover, the therapeutic parameters of 5-HT2A receptor occupancy and duration of activation are still to be determined, and if both 5-HT2A and NMDA receptor sensitivity are altered by mTBI, this may alter the therapeutic dose and increase potential side effects of psychedelic-based compounds. Nevertheless, pre-clinical data that suggest psychedelics could potentially ameliorate some of the negative neurobiological effects of brain injury are promising.

The authors have nothing to disclose.

Conceptualization: AC Reichelt and BT Dunkley

Methodology: AC Reichelt

Investigation: AC Reichelt and BT Dunkley

Writing — Original Draft: AC Reichelt, E Vermetten, and BT Dunkley

Writing — Review & Editing: AC Reichelt, E Vermetten, and BT Dunkley

Ethics approval was not required for this article.

N/A

N/A

N/A

No funding was received for this article.

This article has been peer reviewed.

1. Rigg JL, Mooney SR. Concussions and the military: issues specific to service members. PM R. 2011;3(10 Suppl 2):S3806. https://doi.org/10.1016/j.pmrj.2011.08.005. Medline:22035680 Google Scholar
2. Garber BG, Rusu C, Zamorski MA. Deployment-related mild traumatic brain injury, mental health problems, and post-concussive symptoms in Canadian Armed Forces personnel. BMC Psychiatry. 2014;14:325. https://doi.org/10.1186/s12888-014-0325-5. Medline:25410348 Google Scholar
3. MacFarlane MP, Glenn TC. Neurochemical cascade of concussion. Brain Inj. 2015;29(2):13953. https://doi.org/10.3109/02699052.2014.965208. Medline:25587743 Google Scholar
4. Permenter CM, Fernández-de Thomas RJ, Sherman AL. Postconcussive syndrome. In: StatPearls. Treasure Island (FL): StatPearls Publishing; 2021. Google Scholar
5. Carhart-Harris R, Giribaldi B, Watts R, et al. Trial of psilocybin versus escitalopram for depression. N Engl J Med. 2021;384(15):140211. https://doi.org/10.1056/nejmoa2032994. Medline:33852780 Google Scholar
6. Davis AK, Barrett FS, May DG, et al. Effects of psilocybin-assisted therapy on major depressive disorder: a randomized clinical trial. JAMA Psychiatry. 2021;78(5):4819. https://doi.org/10.1001/jamapsychiatry.2020.3285. Medline:33146667 Google Scholar
7. Goodwin GM, Aaronson ST, Alvarez O, et al. Single-dose psilocybin for a treatment-resistant episode of major depression. N Engl J Med. 2022;387(18):163748. Google Scholar
8. Flanagan TW, Nichols CD. Psychedelics and anti-inflammatory activity in animal models. Curr Top Behav Neurosci. 2022;56:22945. Google Scholar
9. Collins SM, O’Connell CJ, Reeder EL, et al. Altered serotonin 2A (5-HT(2A)) receptor signaling underlies mild TBI-elicited deficits in social dominance. Front Pharmacol. 2022;13:930346. https://doi.org/10.3389/fphar.2022.930346. Medline:35910378 Google Scholar
10. Ly C, Greb AC, Cameron LP, et al. Psychedelics promote structural and functional neural plasticity. Cell Rep. 2018;23(11):317082. https://doi.org/10.1016/j.celrep.2018.05.022. Medline:29898390 Google Scholar
11. Schenberg EE, Alexandre JF, Filev R, et al. Acute biphasic effects of ayahuasca. PLoS One. 2015;10(9):e0137202. https://doi.org/10.1371/journal.pone.0137202. Medline:26421727 Google Scholar
12. Barnett L, Muthukumaraswamy SD, Carhart-Harris RL, Seth AK. Decreased directed functional connectivity in the psychedelic state. Neuroimage. 2020;209:116462. https://doi.org/10.1016/j.neuroimage.2019.116462. Medline:31857204 Google Scholar
13. Tagliazucchi E, Roseman L, Kaelen M, et al. Increased global functional connectivity correlates with LSD-induced ego dissolution. Curr Biol. 2016;26(8):104350. https://doi.org/10.1016/j.cub.2016.02.010. Medline:27085214 Google Scholar
14. Cameron LP, Tombari RJ, Lu J, et al. A non-hallucinogenic psychedelic analogue with therapeutic potential. Nature. 2021;589(7842):4749. https://doi.org/10.1038/s41586-020-3008-z. Medline:33299186 Google Scholar
15. Szabo I, Varga VE, Dvoracsko S, et al. N,N-dimethyltryptamine attenuates spreading depolarization and restrains neurodegeneration by sigma-1 receptor activation in the ischemic rat brain. Neuropharmacology. 2021;192:108612. https://doi.org/10.1016/j.neuropharm.2021.108612. Medline:34023338 Google Scholar
16. Shao LX, Liao C, Gregg I, et al. Psilocybin induces rapid and persistent growth of dendritic spines in frontal cortex in vivo. Neuron. 2021;109(16):253544 e4. https://doi.org/10.1016/j.neuron.2021.06.008. Medline:34228959 Google Scholar
17. Kozlowska U, Nichols C, Wiatr K, Figiel M. From psychiatry to neurology: psychedelics as prospective therapeutics for neurodegenerative disorders. J Neurochem. 2022;162(1):89108. https://doi.org/10.1111/jnc.15509. Medline:34519052 Google Scholar
18. Biegon A, Fry PA, Paden CM, et al. Dynamic changes in N-methyl-d-aspartate receptors after closed head injury in mice: implications for treatment of neurological and cognitive deficits. Proc Natl Acad Sciences U S A. 2004;101(14):511722. https://doi.org/10.1073/pnas.0305741101. Medline:15044697 Google Scholar
19. de Almeida RN, Galvao ACM, da Silva FS, et al. Modulation of serum brain-derived neurotrophic factor by a single dose of ayahuasca: observation from a randomized controlled trial. Front Psychol. 2019;10:1234. https://doi.org/10.3389/fpsyg.2019.01234. Medline:31231276 Google Scholar
20. Morganti-Kossmann MC, Rancan M, Stahel PF, Kossmann T. Inflammatory response in acute traumatic brain injury: a double-edged sword. Curr Opin Crit Care. 2002;8(2):1015. https://doi.org/10.1097/00075198-200204000-00002. Medline:12386508 Google Scholar
21. Bhatti J, Nascimento B, Akhtar U, et al. Systematic review of human and animal studies examining the efficacy and safety of N-acetylcysteine (NAC) and N-acetylcysteine amide (NACA) in traumatic brain injury: impact on neurofunctional outcome and biomarkers of oxidative stress and inflammation. Front Neurol. 2018;8:744. https://doi.org/10.3389/fneur.2017.00744. Medline:29387038 Google Scholar
22. Nardai S, Laszlo M, Szabo A, et al. N,N-dimethyltryptamine reduces infarct size and improves functional recovery following transient focal brain ischemia in rats. Exp Neurol. 2020;327:113245. https://doi.org/10.1016/j.expneurol.2020.113245. Medline:32067950 Google Scholar
23. Zanikov T, Gerasymchuk M, Ghasemi Gojani E, et al. The effect of combined treatment of psilocybin and eugenol on lipopolysaccharide-induced brain inflammation in mice. Molecules. 2023;28(6):2624. https://doi.org/10.3390/molecules28062624. Medline:36985596 Google Scholar
24. Nau F, Jr., Yu B, Martin D, Nichols CD. Serotonin 5-HT2A receptor activation blocks TNF-alpha mediated inflammation in vivo. PLoS One. 2013;8(10):e75426. https://doi.org/10.1371/journal.pone.0075426. Medline:24098382 Google Scholar
25. Yu B, Becnel J, Zerfaoui M, et al. Serotonin 5-hydroxytryptamine(2A) receptor activation suppresses tumor necrosis factor-alpha-induced inflammation with extraordinary potency. J Pharmacol Exp Ther. 2008;327(2):31623. https://doi.org/10.1124/jpet.108.143461. Medline:18708586 Google Scholar
26. Rudin D, Areesanan A, Liechti ME, Gründemann C. Classic psychedelics do not affect T cell and monocyte immune responses. Front Psychiatry. 2023;14:1042440. https://doi.org/10.3389/fpsyt.2023.1042440. Medline:36741125 Google Scholar
27. Hang CH, Shi JX, Li JS, et al. Alterations of intestinal mucosa structure and barrier function following traumatic brain injury in rats. World J Gastroenterol. 2003;9(12):277681. https://doi.org/10.3748/wjg.v9.i12.2776. Medline:14669332 Google Scholar
28. Treangen TJ, Wagner J, Burns MP, Villapol S. Traumatic brain injury in mice induces acute bacterial dysbiosis within the fecal microbiome. Front Immunol. 2018;9:2757. https://doi.org/10.3389/fimmu.2018.02757. Medline:30546361 Google Scholar
29. Inserra A, Giorgini G, Lacroix S, et al. Effects of repeated lysergic acid diethylamide (LSD) on the mouse brain endocannabinoidome and gut microbiome. Br J Pharmacol. 2023;180(6):72139. https://doi.org/10.1111/bph.15977. Medline:36316276 Google Scholar
30. Zhang J, Safar K, Emami Z, et al. Local and large-scale beta oscillatory dysfunction in males with mild traumatic brain injury. J Neurophysiol. 2020;124(6):194858. https://doi.org/10.1152/jn.00333.2020. Medline:33052746 Google Scholar
31. Luo Q, Xu D, Roskos T, et al. Complexity analysis of resting state magneto-encephalography activity in traumatic brain injury patients. J Neurotrauma. 2013;30(20):17029. https://doi.org/10.1089/neu.2012.2679. Medline:23692211 Google Scholar
32. Schartner MM, Carhart-Harris RL, Barrett AB, et al. Increased spontaneous MEG signal diversity for psychoactive doses of ketamine, LSD and psilocybin. Sci Rep. 2017;7:46421. https://doi.org/10.1038/srep46421. Medline:28422113 Google Scholar
33. Daws RE, Timmermann C, Giribaldi B, et al. Increased global integration in the brain after psilocybin therapy for depression. Nat Med. 2022;28(4):84451. https://doi.org/10.1038/s41591-022-01744-z. Medline:35411074 Google Scholar
34. Doss MK, Považan M, Rosenberg MD, et al. Psilocybin therapy increases cognitive and neural flexibility in patients with major depressive disorder. Transl Psychiatry. 2021;11(1):574. https://doi.org/10.1038/s41398-021-01706-y. Medline:34750350 Google Scholar
35. Morales-Garcia JA, de la Fuente Revenga M, Alonso-Gil S, et al. The alkaloids of Banisteriopsis caapi, the plant source of the Amazonian hallucinogen ayahuasca, stimulate adult neurogenesis in vitro. Sci Rep. 2017;7(1):5309. https://doi.org/10.1038/s41598-017-05407-9. Medline:28706205 Google Scholar
36. Li S-P, Wang Y-W, Qi S-L, et al. Analogous β-carboline alkaloids harmaline and harmine ameliorate scopolamine-induced cognition dysfunction by attenuating acetylcholinesterase activity, oxidative stress, and inflammation in mice. Front Pharmacol. 2018;9:346. https://doi.org/10.3389/fphar.2018.00346. Medline:29755345 Google Scholar
37. Glatfelter GC, Pottie E, Partilla JS, et al. Structure-activity relationships for psilocybin, baeocystin, aeruginascin, and related analogues to produce pharmacological effects in mice. ACS Pharmacol Transl Sci. 2022;5(11):118196. https://doi.org/10.1021/acsptsci.2c00177. Medline:36407948 Google Scholar
38. Zhuk O, Jasicka-Misiak I, Poliwoda A, et al. Research on acute toxicity and the behavioral effects of methanolic extract from psilocybin mushrooms and psilocin in mice. Toxins (Basel). 2015;7(4):101829. https://doi.org/10.3390/toxins7041018. Medline:25826052 Google Scholar
39. Martínez-Mármol R, Chai Y, Conroy JN, et al. Hericerin derivatives activates a pan-neurotrophic pathway in central hippocampal neurons converging to ERK1/2 signaling enhancing spatial memory. J Neurochemistry. 2023;165(6):791808. https://doi.org/10.1111/jnc.15767. Medline:36660878 Google Scholar
40. Hilliard A, Mendonca P, Soliman KFA. Involvement of NFkB and MAPK signaling pathways in the preventive effects of Ganoderma lucidum on the inflammation of BV-2 microglial cells induced by LPS. J Neuroimmunol. 2020;345:577269. https://doi.org/10.1016/j.jneuroim.2020.577269. Medline:32480240 Google Scholar
41. Chao MW, Liao CW, Lin CH, Tseng CY. Immunomodulatory protein from Ganoderma microsporum protects against oxidative damages and cognitive impairments after traumatic brain injury. Mol Cell Neurosci. 2022;120:103735. https://doi.org/10.1016/j.mcn.2022.103735. Medline:35562037 Google Scholar
42. Szigeti B, Kartner L, Blemings A, et al. Self-blinding citizen science to explore psychedelic microdosing. Elife. 2021;10:e62878. https://doi.org/10.7554/elife.62878. Medline:33648632 Google Scholar
43. Rootman JM, Kiraga M, Kryskow P, et al. Psilocybin microdosers demonstrate greater observed improvements in mood and mental health at one month relative to non-microdosing controls. Sci Rep. 2022;12(1):11091. https://doi.org/10.1038/s41598-022-14512-3. Medline:35773270 Google Scholar
44. Pandya JD, Readnower RD, Patel SP, et al. N-acetylcysteine amide confers neuroprotection, improves bioenergetics and behavioral outcome following TBI. Exp Neurol. 2014;257:10613. https://doi.org/10.1016/j.expneurol.2014.04.020. Medline:24792639 Google Scholar
45. Şenol N, Nazıroğlu M, Yürüker V. N-acetylcysteine and selenium modulate oxidative stress, antioxidant vitamin and cytokine values in traumatic brain injury-induced rats. Neurochem Res. 2014;39(4):68592. https://doi.org/10.1007/s11064-014-1255-9. Medline:24519543 Google Scholar
46. Eakin K, Baratz-Goldstein R, Pick CG, et al. Efficacy of N-acetyl cysteine in traumatic brain injury. PloS One. 2014;9(4):e90617. https://doi.org/10.1371/journal.pone.0090617. Medline:24740427 Google Scholar
47. Hoffer ME, Balaban C, Slade MD, et al. Amelioration of acute sequelae of blast induced mild traumatic brain injury by N-acetyl cysteine: a double-blind, placebo controlled study. PloS One. 2013;8(1):e54163. https://doi.org/10.1371/journal.pone.0054163. Medline:23372680 Google Scholar