Antidepressant Effects of Ayahuasca in Humans: History
Please note this is an old version of this entry, which may differ significantly from the current revision.

Ayahuasca is a psychedelic preparation usually made by the decoction of Banisteriopsis caapi and Psychotria viridis, or Diplopterys cabrerana, plants endemic to the Amazonian Basin where the brew is traditionally used in ritualistic contexts. B. caapi is known to contain a class of substances called β-carbolines or harmala alkaloids, mainly harmine, tetrahydroharmine (THH), and harmaline. These substances are known to selectively and reversibly inhibit the enzyme monoamine oxidase type A (MAO-A), which is believed to be their main mechanism of action. On the other hand, P. viridis is a source of dimethyltryptamine (DMT), a serotoninergic psychedelic belonging to the same pharmacological class of substances as lysergic acid diethylamide (LSD) and psilocybin. The main mechanism of action for DMT and related psychedelic substances is widely accepted to be agonism at the serotonin receptors 5-HT1A,2A,2C, with the 2A subtype being the primary molecular target and its activation dose-dependently related to the psychoactive effects these substances cause. 

  • ayahuasca
  • DMT
  • antidepressant
  • psychedelic

1. A Brief Review of the Current Evidence for the Antidepressant Effects of Ayahuasca in Humans

As with other classical psychedelics such as psilocybin and LSD, published research regarding the therapeutic effects of ayahuasca has grown steadily in the last two decades. Although preliminary and still lacking further and more thorough evaluation, results so far have been promising, especially regarding the antidepressant and anxiolytic effects the brew seems to exert. Grob et al. (1996) [1] performed the first observational study with frequent ritual ayahuasca practitioners that reported possible antidepressant effects of the brew. Similar results have been demonstrated in observational studies in the following years by other groups [2][3][4][5][6]. The first preliminary open-label clinical trial investigating the antidepressant effects of ayahuasca was published by our group in 2015 [7], which was followed by a complementary study that expanded the sample in 2016 [8]. The first randomized, placebo-controlled trial (RCT) investigating the antidepressant effects of ayahuasca replicated the positive results found in the preliminary clinical trial [9]. Nevertheless, small sample sizes, single-dose administration, short time assessments, and alkaloid dose standardization are limitations that still need to be addressed in future research on the topic. Table 1 below summarizes the current evidence regarding ayahuasca’s antidepressant effects in depressed patients, which was basically derived from two clinical trials (one open and one placebo-controlled).
Table 1. Clinical trials that investigated ayahuasca’s potential for the treatment of depression and related research.

BDNF: Brain-Derived Neurotrophic Factor; BPRS: Brief Psychiatric Rating Scale; CADDS: Dissociative States Rating Scale; Administered by the Clinician; HAM-D: Hamilton Depression Rating Scale; HRS: Hallucinogen Rating Scale; MADRS: Montgomery–Asberg Depression Scale; MADRS-SI: Montgomery-Asberg Depression Scale suicidal intent subscale; MEQ30: Mystical Experiences Questionnaire; YMRS: Clinician-Administered Dissociative States Rating Scale. 1 Preliminary study; 2 Definitive study including patients from the preliminary study; 3 Original study. All other studies that share the same design were derived from this experiment (randomized controlled trial).

2. Safety and Tolerability of Ayahuasca Administration

Although there is no formal report of deaths caused by ayahuasca intake, the lack of regulation over its production and consumption in some countries can result in medical tourism scenarios [14]. Excessive optimism of psychedelics’ therapeutic properties portraited in mediatic coverage can mask the consequences of reckless use, namely interaction with other psychoactive substances (such as Selective Serotonin Reuptake Inhibitors (SSRIs)) and with previous medical conditions, which can produce lifelong health impairments [14][15].
Ayahuasca consumption at reported doses in observational studies and clinical trials causes a wide variety of effects, of which some are desired, some are unwanted, and/or can be considered adverse events (AEs). Regarding randomized, blinded (single or double), and placebo-controlled trials, a recent review evaluated the occurrence of adverse events following ayahuasca administration to healthy volunteers and treatment-resistant depressive patients in 11 distinct trials (n = 108 ayahuasca administrations) [16]. On one hand, most common AEs reported were gastrointestinal malaise, nausea, vomiting, headaches, and mild-to-moderate transient increases in heart rate and blood pressure [16]. These AEs were all expected and known to occur with ayahuasca and other psychedelic administration such as LSD and psilocybin [16][17]. On the other hand, there were more clinically significant reports of anxiety, confusion, emotional distress, depersonalization, and dysphoric state manifestations after ayahuasca administration, although these are much less common. However, there are currently no reports of psychotic states, lasting AEs, trial dropouts, or the need for medical interventions in any case, even in more significant situations. All reported AEs were transient and resolved on their own with researcher’s psychological support [16].
Regarding the occurrence of more significant adverse events in clinical trials with ayahuasca performed by our group, there is a detailed report of the two instances where these effects had to be managed by our research team, how the situation was resolved, and an initial nine step guideline on managing this kind of occurrence [18]. Overall, it seems that as with other psychedelic administration, a cautious and detailed selection of the volunteers who are allowed to participate in the trials and a supportive setting constructed by researchers has been demonstrated to be enough to reduce and manage the occurrence of AEs in clinical trials. Nevertheless, there is still the need for further research with bigger samples to confirm the preliminary findings we have so far for the occurrence of both positive and negative effects.

3. Ayahuasca’s Alkaloid Content

The concentrations of psychoactive alkaloids in ayahuasca vary greatly due to the lack of standardization in the quantity and quality of the plants used in its production, the region where it is produced, cultural aspects related to its use, and the desired final concentration of the drink [19]. Previous studies that quantified the alkaloid levels present in diverse ayahuasca samples reported a wide range of concentrations. For example, the content of alkaloids reported by McKenna et al. (1984) [20] ranged from 0.15 mg/mL of harmine, 0.05 mg/mL of THH, and 0.125 mg/mL of DMT, to 4.67 mg/mL of harmine, 1.60 mg/mL of THH, 0.41 mg/mL of harmaline, and 0.60 mg/mL of DMT in a dose. Callaway (2005) [21] measured the levels of alkaloids in 29 samples of ayahuasca from different religions where it is consumed. Again, a wide variation in alkaloid levels was demonstrated in the different samples, with DMT ranging from 0 to 14.15 mg/mL, harmine from 0.45 to 22.85 mg/mL, THH from 0.48 to 23.8 mg/mL, and harmaline from <0.01 to 0.9 mg/mL [21]. Santos et al. (2017) [22] validated a solid-phase extraction technique to quantify the alkaloids of 20 ayahuasca samples, where the levels of DMT, harmine, harmaline, THH, tryptamine, and harmalol were evaluated in concentrations ranging from 0.3 to 36,7 mg/ml. Souza et al. (2019) [23] quantified DMT, harmine, THH, and harmaline in 38 ayahuasca samples. DMT was reported in concentrations from 0.62 to 3.4 mg/mL, harmine from 4.14 to 18.16 mg/mL, THH from 4.02 to 30.88 mg/mL, and for harmaline from 0.4 to 3.92 mg/mL. Table 2 below summarizes the data for studies that evaluated alkaloid levels in at least eight ayahuasca samples.
Table 2. Ayahuasca’s alkaloid concentration reported in studies with at least 8 different samples.
DMT: N,N-Dimethyltryptamine; THH: Tetrahydroharmine. 1 Individual values for each analysis not specified by the authors. The values presented represent the minimum and maximum of all analyses.
Even across clinical trials with healthy and depressed patients, there is still a lack of standardization of the dosage administered to participants. Table 3 below illustrates this variation.
Table 3. Ayahuasca’s alkaloid concentrations and dosages from clinical trials with ayahuasca.
DMT: N, N-Dimethyltryptamine; NA: Not Analyzed; ND: Not Detected; THH: Tetrahydroharmine. 1 Administration of three different doses; 2 Administration of two different doses; 3 Administration of two equal doses in four hours apart.
This variation in alkaloid administration in investigations where ayahuasca is used is one of the major challenges associated with its possible use as an antidepressant. Although clinical research with ayahuasca is still in its infancy, the necessity for standardizing dosages given in different trials is becoming more apparent. In the current state, it is difficult to directly compare results amongst the published research. Furthermore, with the current published data it is very challenging to determine the optimal DMT to β-carboline ratio and dosages that maximize the therapeutic effects while also minimizing the occurrence of AEs. This and other challenges associated with standardized ayahuasca medicinal use (and other psychedelics) go beyond the main focus of this article and have been thoroughly discussed elsewhere [16][17][18][33].

3. Pharmacokinetics of Ayahuasca

The human pharmacokinetics of ayahuasca were evaluated for the first time in a study by Callaway et al. (1999) [34] after the administration of 2 mL/kg of ayahuasca (with alkaloid concentrations: harmine 1.70 mg/mL, harmaline 0.20 mg/mL, THH 1.07 mg/mL, and DMT 0.24 mg/mL). In this study, the following values of maximum concentration (CMAX) and time to reach CMAX (TMAX) were found in blood analysis: DMT—CMAX 15.8 ± 4.4 ng/mL, TMAX 107.5 ± 32.5 min; harmine—CMAX 114.8 ± 61.7 ng/mL, TMAX 102.0 ± 58.3 min; THH—CMAX 91.0 ± 22.0 ng/mL, TMAX 174.0 ± 39.6 min; harmaline—CMAX 6.3 ± 3.1 ng/mL, TMAX 145.0 ± 66.9 min. Another investigation looking at plasma alkaloid levels after administration of a low dose (standardized at 0.6 mg/kg DMT) and a high dose (standardized at 0.85 mg/kg DMT) of lyophilized ayahuasca was performed by Riba et al. (2003) [27]. In this study, CMAX values were found for DMT of 12.14 ng/mL and 17.44 ng/mL for low and high doses, respectively, with TMAX of 1.5 hours after administration for both doses. Dos Santos et al. (2011) [29] measured plasma DMT levels after administration of placebo, one dose and two consecutive doses of ayahuasca four hours apart, at a standardized dose of 0.75 mg/kg DMT. The mean ± SD of maximum concentration values was 13.97 ± 9.35 ng/mL for ayahuasca preceded by placebo and 32.57 ± 20.96 ng/mL for ayahuasca preceded by ayahuasca, suggesting a non-linear increase in DMT levels after administration of two consecutive doses of ayahuasca, likely due to prolonged peripheral MAO-A inhibition [29]. Another study by the same research group demonstrated a comparison with a single dose of ayahuasca (standardized at 1 mg/kg of DMT) and 20 mg of dextroamphetamine [30]. The mean ± SD of maximum plasma DMT concentration values was 11.8 ± 6.4 ng/mL. The median time at which CMAX was reached was 1.8 hours (range 1 to 4.5 hours) after administration [30].
According to these data, we can estimate the nM levels of the alkaloids at CMAX. Considering a single ayahuasca administration, the mean CMAX for DMT of the cited studies where CMAX was measured was (15.8 + 12.14 + 17.44 + 13.97 + 11.8)/5 = 14.23 ng/mL. The molar mass of DMT is 188.274, which results in a 75.58 nM mean maximum plasma concentration. At first this concentration appears to be lower than the required to interact with certain molecular targets, but there is an evidence-based proposed mechanism through which DMT can reach high local concentrations within neurons of the CNS [35][36]. Briefly, this three-step mechanism starts with the crossing of DMT through the blood-brain barrier (BBB) via uptake across the endothelial plasma membrane. To cross the BBB, DMT is actively transported through the endothelial plasma membrane via Mg2+ and ATP-dependent uptake [37][38][39]. Although we are not aware of an investigation regarding the precise mechanism by which this accomplished, it is possible that the Organic Cation Transporter (OTC) family of transporters is involved, since it can transport other closely related monoamines, such as serotonin [40].
This is corroborated to happen given the results from studies that verified the accumulation of DMT and other tryptamines in the brain after peripheral administration [38][41][42][43]. It has been shown that DMT promptly enters the CNS and is kept there after excretion in urine has stopped (24 hours), being detected up to 7 days after administration [44]. Next, cell-membrane serotonin transporter (SERT) uptakes DMT to inside the neurons, where VMAT2 promotes its sequestration and accumulation within vesicles [36]. Here DMT is protected from MAO degradation and can be stored several days before being released when the correct stimuli are given [44]. The proposed mechanism and investigations that support it are evidence of DMT’s role as an important cellular messenger, since there is a considerable physiological effort and prioritization to transport, accumulate, and store it within the CNS [36]. Furthermore, it shows that DMT must have molecular activity endogenously in a much lower average concentration environment inside the human body, in the absence of exogenous intake [36]. If this molecule is active within this relatively hostile environment to its existence, exogenously administered DMT in tandem with peripheral degradation inhibition provided by MAO-A deactivation from β-carbolines surely augments its concentrations to levels high enough to influence other molecular targets it would otherwise not be capable of influencing. The current evidence for the action of DMT and β-carbolines within these possible targets is discussed with more detail in the next sections.

This entry is adapted from the peer-reviewed paper 10.3390/biom12111618

References

  1. Grob, C.S.; McKenna, D.J.; Callaway, J.C.; Brito, G.S.; Neves, E.S.; Oberlaender, G.; Saide, O.L.; Labigalini, E.; Tacla, C.; Miranda, C.T.; et al. Human Psychopharmacology of Hoasca, a Plant Hallucinogen Used in Ritual Context in Brazil. J. Nerv. Ment. Dis. 1996, 184, 86–94.
  2. Bouso, J.C.; González, D.; Fondevila, S.; Cutchet, M.; Fernández, X.; Ribeiro Barbosa, P.C.; Alcázar-Córcoles, M.; Araújo, W.S.; Barbanoj, M.J.; Fábregas, J.M.; et al. Personality, Psychopathology, Life Attitudes and Neuropsychological Performance among Ritual Users of Ayahuasca: A Longitudinal Study. PLoS ONE 2012, 7, e42421.
  3. Jiménez-Garrido, D.F.; Gómez-Sousa, M.; Ona, G.; Dos Santos, R.G.; Hallak, J.E.C.; Alcázar-Córcoles, M.; Bouso, J.C. Effects of Ayahuasca on Mental Health and Quality of Life in Naïve Users: A Longitudinal and Cross-Sectional Study Combination. Sci. Rep. 2020, 10, 4075.
  4. Uthaug, M.V.; van Oorsouw, K.; Kuypers, K.P.C.; van Boxtel, M.; Broers, N.J.; Mason, N.L.; Toennes, S.W.; Riba, J.; Ramaekers, J.G. Sub-Acute and Long-Term Effects of Ayahuasca on Affect and Cognitive Thinking Style and Their Association with Ego Dissolution. Psychopharmacology 2018, 235, 2979–2989.
  5. Da Silveira, D.X.; Grob, C.S.; de Rios, M.D.; Lopez, E.; Alonso, L.K.; Tacla, C.; Doering-Silveira, E. Ayahuasca in Adolescence: A Preliminary Psychiatric Assessment. J. Psychoact. Drugs 2005, 37, 129–133.
  6. Sarris, J.; Perkins, D.; Cribb, L.; Schubert, V.; Opaleye, E.; Bouso, J.C.; Scheidegger, M.; Aicher, H.; Simonova, H.; Horák, M.; et al. Ayahuasca Use and Reported Effects on Depression and Anxiety Symptoms: An International Cross-Sectional Study of 11,912 Consumers. J. Affect. Disord. Rep. 2021, 4, 100098.
  7. Osório Fde, L.; Sanches, R.F.; Macedo, L.R.; Santos, R.G.; Maia-de-Oliveira, J.P.; Wichert-Ana, L.; Araujo, D.B.; Riba, J.; Crippa, J.A.; Hallak, J.E. Antidepressant Effects of a Single Dose of Ayahuasca in Patients with Recurrent Depression: A Preliminary Report. Braz. J. Psychiatry 2015, 37, 13–20.
  8. Sanches, R.F.; Osório, F.L.; dos Santos, R.G.; Macedo, L.R.H.; Maia-de-Oliveira, J.P.; Wichert-Ana, L.; de Araujo, D.B.; Riba, J.; Crippa, J.A.S.; Hallak, J.E.C. Antidepressant Effects of a Single Dose of Ayahuasca in Patients with Recurrent Depression: A SPECT Study. J. Clin. Psychopharmacol. 2016, 36, 77–81.
  9. Palhano-Fontes, F.; Barreto, D.; Onias, H.; Andrade, K.C.; Novaes, M.M.; Pessoa, J.A.; Mota-Rolim, S.A.; Osório, F.L.; Sanches, R.; Dos Santos, R.G.; et al. Rapid Antidepressant Effects of the Psychedelic Ayahuasca in Treatment-Resistant Depression: A Randomized Placebo-Controlled Trial. Psychol. Med. 2019, 49, 655–663.
  10. Galvão, A.C.M.; de Almeida, R.N.; Silva, E.; Freire, F.A.M.; Palhano-Fontes, F.; Onias, H.; Arcoverde, E.; Maia-de-Oliveira, J.P.; de Araújo, D.B.; Lobão-Soares, B.; et al. Cortisol Modulation by Ayahuasca in Patients With Treatment Resistant Depression and Healthy Controls. Front. Psychiatry 2018, 9, 185.
  11. Zeifman, R.J.; Palhano-Fontes, F.; Hallak, J.; Arcoverde, E.; Maia-Oliveira, J.P.; Araujo, D.B. The Impact of Ayahuasca on Suicidality: Results From a Randomized Controlled Trial. Front. Pharmacol. 2019, 10, 1325.
  12. de Almeida, R.N.; Galvão, A.C.M.; da Silva, F.S.; Silva, E.; Palhano-Fontes, F.; Maia-de-Oliveira, J.P.; de Araújo, L.B.; Lobão-Soares, B.; Galvão-Coelho, N.L. Modulation of Serum Brain-Derived Neurotrophic Factor by a Single Dose of Ayahuasca: Observation From a Randomized Controlled Trial. Front. Psychol. 2019, 10, 1234.
  13. Galvão-Coelho, N.L.; de Menezes Galvão, A.C.; de Almeida, R.N.; Palhano-Fontes, F.; Campos Braga, I.; Lobão Soares, B.; Maia-de-Oliveira, J.P.; Perkins, D.; Sarris, J.; de Araujo, D.B. Changes in Inflammatory Biomarkers Are Related to the Antidepressant Effects of Ayahuasca. J. Psychopharmacol. 2020, 34, 1125–1133.
  14. Bauer, I.L. Ayahuasca: A risk for travellers? Travel Med. Infect. Dis. 2018, 21, 74–76.
  15. Callaway, J.C.; Grob, C.S. Ayahuasca Preparations and Serotonin Reuptake Inhibitors: A Potential Combination for Severe Adverse Interactions. J. Psychoact. Drugs 1998, 30, 367–369.
  16. Rossi, G.N.; Dias, I.C.D.S.; Baker, G.; Bouso Saiz, J.C.; Dursun, S.M.; Hallak, J.E.C.; Dos Santos, R.G. Ayahuasca, a Potentially Rapid Acting Antidepressant: Focus on Safety and Tolerability. Expert Opin. Drug Saf. 2022, 21, 789–801.
  17. Rossi, G.N.; Hallak, J.E.C.; Baker, G.; Dursun, S.M.; Dos Santos, R.G. The Effects of Ketamine and Classic Hallucinogens on Neurotrophic and Inflammatory Markers in Unipolar Treatment-Resistant Depression: A Systematic Review of Clinical Trials. Eur. Arch. Psychiatry Clin. Neurosci. 2022, 1–27.
  18. Rocha, J.M.; Rossi, G.N.; Osório, F.L.; Hallak, J.E.C.; Dos Santos, R.G. Adverse Effects After Ayahuasca Administration in the Clinical Setting. J. Clin. Psychopharmacol. 2022, 42, 321–324.
  19. McKenna, D.J. Clinical Investigations of the Therapeutic Potential of Ayahuasca: Rationale and Regulatory Challenges. Pharmacol. Ther. 2004, 102, 111–129.
  20. McKenna, D.J.; Towers, G.H.; Abbott, F. Monoamine Oxidase Inhibitors in South American Hallucinogenic Plants: Tryptamine and Beta-Carboline Constituents of Ayahuasca. J. Ethnopharmacol. 1984, 10, 195–223.
  21. Callaway, J.C. Various Alkaloid Profiles in Decoctions of Banisteriopsis caapi. J. Psychoact. Drugs 2005, 37, 151–155.
  22. Santos, M.C.; Navickiene, S.; Gaujac, A. Determination of Tryptamines and β-Carbolines in Ayahuasca Beverage Consumed During Brazilian Religious Ceremonies. J. AOAC Int. 2017, 100, 820–824.
  23. Souza, R.C.Z.; Zandonadi, F.S.; Freitas, D.P.; Tófoli, L.F.F.; Sussulini, A. Validation of an Analytical Method for the Determination of the Main Ayahuasca Active Compounds and Application to Real Ayahuasca Samples from Brazil. J. Chromatogr. B 2019, 1124, 197–203.
  24. Santos, B.W.L.; de Oliveira, R.C.; Sonsin-Oliveira, J.; Fagg, C.W.; Barbosa, J.B.F.; Caldas, E.D. Biodiversity of β-Carboline Profile of Banisteriopsis Caapi and Ayahuasca, a Plant and a Brew with Neuropharmacological Potential. Plants 2020, 9, 870.
  25. Kaasik, H.; Souza, R.C.Z.; Zandonadi, F.S.; Tófoli, L.F.; Sussulini, A. Chemical Composition of Traditional and Analog Ayahuasca. J. Psychoact. Drugs 2021, 53, 65–75.
  26. Riba, J.; Rodríguez-Fornells, A.; Urbano, G.; Morte, A.; Antonijoan, R.; Montero, M.; Callaway, J.C.; Barbanoj, M.J. Subjective Effects and Tolerability of the South American Psychoactive Beverage Ayahuasca in Healthy Volunteers. Psychopharmacology 2001, 154, 85–95.
  27. Riba, J.; Valle, M.; Urbano, G.; Yritia, M.; Morte, A.; Barbanoj, M.J. Human Pharmacology of Ayahuasca: Subjective and Cardiovascular Effects, Monoamine Metabolite Excretion, and Pharmacokinetics. J. Pharmacol. Exp. Ther. 2003, 306, 73–83.
  28. Riba, J.; Romero, S.; Grasa, E.; Mena, E.; Carrió, I.; Barbanoj, M.J. Increased Frontal and Paralimbic Activation Following Ayahuasca, the Pan-Amazonian Inebriant. Psychopharmacology 2006, 186, 93–98.
  29. dos Santos, R.G.; Valle, M.; Bouso, J.C.; Nomdedéu, J.F.; Rodríguez-Espinosa, J.; McIlhenny, E.H.; Barker, S.A.; Barbanoj, M.J.; Riba, J. Autonomic, Neuroendocrine, and Immunological Effects of Ayahuasca: A Comparative Study With d-Amphetamine. J. Clin. Psychopharmacol. 2011, 31, 717–726.
  30. Dos Santos, R.G.; Grasa, E.; Valle, M.; Ballester, M.R.; Bouso, J.C.; Nomdedéu, J.F.; Homs, R.; Barbanoj, M.J.; Riba, J. Pharmacology of Ayahuasca Administered in Two Repeated Doses. Psychopharmacology 2012, 219, 1039–1053.
  31. Rocha, J.M.; Rossi, G.N.; de Lima Osório, F.; Bouso, J.C.; de Oliveira Silveira, G.; Yonamine, M.; Campos, A.C.; Bertozi, G.; Cecílio Hallak, J.E.; Dos Santos, R.G. Effects of Ayahuasca on the Recognition of Facial Expressions of Emotions in Naive Healthy Volunteers: A Pilot, Proof-of-Concept, Randomized Controlled Trial. J. Clin. Psychopharmacol. 2021, 41, 267–274.
  32. Dos Santos, R.G.; Osório, F.D.L.; Rocha, J.M.; Rossi, G.N.; Bouso, J.C.; Rodrigues, L.S.; Silveira, G.D.O.; Yonamine, M.; Hallak, J.E.C. Ayahuasca improves self-perception of speech performance in subjects with social anxiety disorder. J. Clin. Psychopharmacol. 2021, 41, 540–550.
  33. Dos Santos, R.G.; Bouso, J.C.; Rocha, J.M.; Rossi, G.N.; Hallak, J.E. The Use of Classic Hallucinogens/Psychedelics in a Therapeutic Context: Healthcare Policy Opportunities and Challenges. Risk Manag. Healthc. Policy 2021, 14, 901–910.
  34. Callaway, J.C.; McKenna, D.J.; Grob, C.S.; Brito, G.S.; Raymon, L.P.; Poland, R.E.; Andrade, E.N.; Andrade, E.O.; Mash, D.C. Pharmacokinetics of Hoasca Alkaloids in Healthy Humans. J. Ethnopharmacol. 1999, 65, 243–256.
  35. Cozzi, N.V.; Gopalakrishnan, A.; Anderson, L.L.; Feih, J.T.; Shulgin, A.T.; Daley, P.F.; Ruoho, A.E. Dimethyltryptamine and Other Hallucinogenic Tryptamines Exhibit Substrate Behavior at the Serotonin Uptake Transporter and the Vesicle Monoamine Transporter. J. Neural Transm. 2009, 116, 1591–1599.
  36. Frecska, E.; Szabo, A.; Winkelman, M.J.; Luna, L.E.; McKenna, D.J. A Possibly Sigma-1 Receptor Mediated Role of Dimethyltryptamine in Tissue Protection, Regeneration, and Immunity. J. Neural Transm. 2013, 120, 1295–1303.
  37. Cohen, I.; Vogel, W.H. Determination and Physiological Disposition of Dimethyltryptamine and Diethyltryptamine in Rat Brain, Liver and Plasma. Biochem. Pharmacol. 1972, 21, 1214–1216.
  38. Barker, S.A.; Beaton, J.M.; Christian, S.T.; Monti, J.A.; Morris, P.E. Comparison of the Brain Levels of N,N-Dimethyltryptamine and Alpha, Alpha, Beta, Beta-Tetradeutero-N-N-Dimethyltryptamine Following Intraperitoneal Injection. The in Vivo Kinetic Isotope Effect. Biochem. Pharmacol. 1982, 31, 2513–2516.
  39. Christian, S.T.; Harrison, R.; Quayle, E.; Pagel, J.; Monti, J. The in Vitro Identification of Dimethyltryptamine (DMT) in Mammalian Brain and Its Characterization as a Possible Endogenous Neuroregulatory Agent. Biochem. Med. 1977, 18, 164–183.
  40. Busch, A.E.; Karbach, U.; Miska, D.; Gorboulev, V.; Akhoundova, A.; Volk, C.; Arndt, P.; Ulzheimer, J.C.; Sonders, M.S.; Baumann, C.; et al. Human Neurons Express the Polyspecific Cation Transporter HOCT2, Which Translocates Monoamine Neurotransmitters, Amantadine, and Memantine. Mol. Pharmacol. 1998, 54, 342–352.
  41. Sitaram, B.R.; Lockett, L.; Talomsin, R.; Blackman, G.L.; McLeod, W.R. In Vivo Metabolism of 5-Methoxy-N,N-Dimethyltryptamine and N,N-Dimethyltryptamine in the Rat. Biochem. Pharmacol. 1987, 36, 1509–1512.
  42. Takahashi, T.; Takahashi, K.; Ido, T.; Yanai, K.; Iwata, R.; Ishiwata, K.; Nozoe, S. 11C-Labeling of Indolealkylamine Alkaloids and the Comparative Study of Their Tissue Distributions. Int. J. Appl. Radiat. Isot. 1985, 36, 965–969.
  43. Yanai, K.; Ido, T.; Ishiwata, K.; Hatazawa, J.; Takahashi, T.; Iwata, R.; Matsuzawa, T. In Vivo Kinetics and Displacement Study of a Carbon-11-Labeled Hallucinogen, N,N-Dimethyltryptamine. Eur. J. Nucl. Med. 1986, 12, 141–146.
  44. Vitale, A.A.; Pomilio, A.B.; Cañellas, C.O.; Vitale, M.G.; Putz, E.M.; Ciprian-Ollivier, J. In Vivo Long-Term Kinetics of Radiolabeled n,n-Dimethyltryptamine and Tryptamine. J. Nucl. Med. 2011, 52, 970–977.
More
This entry is offline, you can click here to edit this entry!
Video Production Service