1
|
Glynos NG, Huels ER, Nelson A, Kim Y, Kennedy RT, Mashour GA, Pal D. Neurochemical and Neurophysiological Effects of Intravenous Administration of N,N-Dimethyltryptamine in Rats. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2025:2024.04.19.589047. [PMID: 38712161 PMCID: PMC11071436 DOI: 10.1101/2024.04.19.589047] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/08/2024]
Abstract
N,N-dimethyltryptamine (DMT) is a serotonergic psychedelic that is being investigated clinically for the treatment of psychiatric disorders. Although the neurophysiological effects of DMT in humans are well-characterized, similar studies in animal models as well as data on the neurochemical effects of DMT are generally lacking, which are critical for mechanistic understanding. In the current study, we combined behavioral analysis, high-density (32-channel) electroencephalography, and ultra-high-performance liquid chromatography-tandem mass spectrometry to simultaneously quantify changes in behavior, cortical neural dynamics, and levels of 17 neurochemicals in medial prefrontal and somatosensory cortices before, during, and after intravenous administration of three different doses of DMT (0.75 mg/kg, 3.75 mg/kg, 7.5 mg/kg) in male and female adult rats. All three doses of DMT produced head twitch response with most twitches observed after the low dose. DMT caused dose-dependent increases in serotonin and dopamine levels in both cortical sites along with a reduction in EEG spectral power in theta (4-10 Hz) and low gamma (25-55 Hz), and increase in power in delta (1-4 Hz), medium gamma (65-115 ), and high gamma (125-155 Hz) bands. Functional connectivity decreased in the delta band and increased across the gamma bands. In addition, we provide the first measurements of endogenous DMT in these cortical sites at levels comparable to serotonin and dopamine, which together with a previous study in occipital cortex, suggests a physiological role for endogenous DMT. This study represents one of the most comprehensive characterizations of psychedelic drug action in rats and the first to be conducted with DMT. Significance Statement N,N-dimethyltryptamine (DMT) is a serotonergic psychedelic with potential as a tool for probing the neurobiology of consciousness and as a therapeutic agent for psychiatric disorders. However, the neurochemical and neurophysiological effects of DMT in rat, a preferred animal model for mechanistic studies, are unclear. We demonstrate that intravenous DMT caused a dose-dependent increase in serotonin and dopamine in medial prefrontal and somatosensory cortices, and simultaneously increased gamma functional connectivity. Similar effects have been shown for other serotonergic and atypical psychedelics, suggesting a shared mechanism of drug action. Additionally, we report DMT during normal wakefulness in two spatially and functionally distinct cortical sites - prefrontal, somatosensory - at levels comparable to those of serotonin and dopamine, supporting a physiological role for endogenous DMT.
Collapse
Affiliation(s)
- Nicolas G. Glynos
- Department of Molecular and Integrative Physiology, University of Michigan, 7744 Medical Science Building II, 1137 East Catherine Street, Ann Arbor, MI 48109-5622, USA
- Department of Anesthesiology, University of Michigan, 7433 Medical Science Building I, 1150 West Medical Center Drive, Ann Arbor, MI 48109-5615, USA
- Center for Consciousness Science, University of Michigan, Ann Arbor, MI 48109-5615, USA
- Michigan Psychedelic Center, University of Michigan, Ann Arbor, MI 48109-5615, USA
| | - Emma R. Huels
- Department of Anesthesiology, University of Michigan, 7433 Medical Science Building I, 1150 West Medical Center Drive, Ann Arbor, MI 48109-5615, USA
- Center for Consciousness Science, University of Michigan, Ann Arbor, MI 48109-5615, USA
- Michigan Psychedelic Center, University of Michigan, Ann Arbor, MI 48109-5615, USA
- Neuroscience Graduate Program, University of Michigan, 4137 Undergraduate Science Building, 204 Washtenaw Avenue, Ann Arbor, MI 48109-2215, USA
| | - Amanda Nelson
- Department of Anesthesiology, University of Michigan, 7433 Medical Science Building I, 1150 West Medical Center Drive, Ann Arbor, MI 48109-5615, USA
| | - Youngsoo Kim
- Department of Chemistry, University of Michigan, 930 North University Avenue, Ann Arbor, MI 48109-1055, USA
| | - Robert T. Kennedy
- Department of Chemistry, University of Michigan, 930 North University Avenue, Ann Arbor, MI 48109-1055, USA
| | - George A. Mashour
- Department of Anesthesiology, University of Michigan, 7433 Medical Science Building I, 1150 West Medical Center Drive, Ann Arbor, MI 48109-5615, USA
- Center for Consciousness Science, University of Michigan, Ann Arbor, MI 48109-5615, USA
- Michigan Psychedelic Center, University of Michigan, Ann Arbor, MI 48109-5615, USA
- Neuroscience Graduate Program, University of Michigan, 4137 Undergraduate Science Building, 204 Washtenaw Avenue, Ann Arbor, MI 48109-2215, USA
| | - Dinesh Pal
- Department of Molecular and Integrative Physiology, University of Michigan, 7744 Medical Science Building II, 1137 East Catherine Street, Ann Arbor, MI 48109-5622, USA
- Department of Anesthesiology, University of Michigan, 7433 Medical Science Building I, 1150 West Medical Center Drive, Ann Arbor, MI 48109-5615, USA
- Center for Consciousness Science, University of Michigan, Ann Arbor, MI 48109-5615, USA
- Michigan Psychedelic Center, University of Michigan, Ann Arbor, MI 48109-5615, USA
- Neuroscience Graduate Program, University of Michigan, 4137 Undergraduate Science Building, 204 Washtenaw Avenue, Ann Arbor, MI 48109-2215, USA
| |
Collapse
|
2
|
Jodo E, Nakazono T, Takahashi K, Suzuki Y, Hoshino KY, Ukon N, Zhao S, Joho T, Takahashi K, Ito H, Eifuku S. Relationship between increased binding potential of possible 5-HT2A receptors in the ventral hippocampus by subchronic phencyclidine and disturbed social interaction in rats: a PET study using 18F-altanserin. Cereb Cortex 2025; 35:bhaf037. [PMID: 40037412 DOI: 10.1093/cercor/bhaf037] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/06/2024] [Revised: 12/09/2024] [Accepted: 01/30/2025] [Indexed: 03/06/2025] Open
Abstract
Subchronic administration of the psychotomimetic drug phencyclidine is known to exacerbate serotonin 5-HT2A receptor-relevant behavioral abnormalities. However, the effects of subchronic phencyclidine on 5-HT2A receptors remain unclear. Here, we investigated the effects of subchronic phencyclidine on the binding potential (BPND) of 5-HT2A receptors in the rat brain using positron emission tomography. Adult male Sprague-Dawley rats received intraperitoneal injection of either phencyclidine (10 mg/kg) or physiological saline once daily, a total of 15 times. positron emission tomography scans were performed twice, before and after drug administration, using 18F-altanserin, a selective 5-HT2A receptor radioactive marker. Two behavioral tests, the sociability test and the social interaction test, were performed before each positron emission tomography scan. The social interaction time was significantly shortened by subchronic phencyclidine. The BPND of the 5-HT2A receptors was significantly increased after subchronic phencyclidine administration in the medial prefrontal cortex, ventral hippocampus, motor cortex, and somatosensory cortex. The BPND change between the pre- and postdrug periods in the ventral hippocampus showed a significant negative correlation (r = 0.73) with that of the social interaction time change. Our results suggest that upregulation of 5-HT2A receptors in the ventral hippocampus may play a role in disturbed social ability and the development of negative symptoms.
Collapse
Affiliation(s)
- Eiichi Jodo
- Department of Systems Neuroscience, Fukushima Medical University School of Medicine, 1 Hikari-ga-oka, Fukushima 960-1295, Japan
| | - Tomoaki Nakazono
- Department of Systems Neuroscience, Fukushima Medical University School of Medicine, 1 Hikari-ga-oka, Fukushima 960-1295, Japan
| | - Kazumi Takahashi
- Department of Systems Neuroscience, Fukushima Medical University School of Medicine, 1 Hikari-ga-oka, Fukushima 960-1295, Japan
| | - Yoshiaki Suzuki
- Department of Systems Neuroscience, Fukushima Medical University School of Medicine, 1 Hikari-ga-oka, Fukushima 960-1295, Japan
| | - Ken-Yo Hoshino
- Ichiyokai Hospital, 15-27 Yashimachou, Fukushima 960-8136, Japan
| | - Naoyuki Ukon
- Advanced Clinical Research Center, Fukushima Medical University, 1 Hikari-ga-oka, Fukushima 960-1295, Japan
| | - Songji Zhao
- Advanced Clinical Research Center, Fukushima Medical University, 1 Hikari-ga-oka, Fukushima 960-1295, Japan
| | - Taiki Joho
- Advanced Clinical Research Center, Fukushima Medical University, 1 Hikari-ga-oka, Fukushima 960-1295, Japan
| | - Kazuhiro Takahashi
- Advanced Clinical Research Center, Fukushima Medical University, 1 Hikari-ga-oka, Fukushima 960-1295, Japan
| | - Hiroshi Ito
- Department of Radiology and Nuclear Medicine, Fukushima Medical University School of Medicine, 1 Hikariga-oka, Fukushima 960-1295, Japan
| | - Satoshi Eifuku
- Department of Systems Neuroscience, Fukushima Medical University School of Medicine, 1 Hikari-ga-oka, Fukushima 960-1295, Japan
| |
Collapse
|
3
|
Sapienza J, Martini F, Comai S, Cavallaro R, Spangaro M, De Gregorio D, Bosia M. Psychedelics and schizophrenia: a double-edged sword. Mol Psychiatry 2025; 30:679-692. [PMID: 39294303 DOI: 10.1038/s41380-024-02743-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/29/2023] [Revised: 08/29/2024] [Accepted: 09/02/2024] [Indexed: 09/20/2024]
Abstract
Psychedelics have shown promising effects in several psychiatric diseases as demonstrated by multiple clinical trials. However, no clinical experiments on patients with schizophrenia have been conducted up to date, except for some old semi-anecdotal studies mainly performed in the time-span '50s-'60s. Notably, these studies reported interesting findings, particularly on the improvement of negative symptoms and social cognition. With no doubts the lack of modern clinical studies is due to the psychomimetic properties of psychedelics, a noteworthy downside that could worsen positive symptoms. However, a rapidly increasing body of evidence has suggested that the mechanisms of action of such compounds partially overlaps with the pathogenic underpinnings of schizophrenia but in an opposite way. These findings suggest that, despite being a controversial issue, the use of psychedelics in the treatment of schizophrenia would be based on a strong biological rationale. Therefore, the aim of our perspective paper is to provide a background on the old experiments with psychedelics performed on patients with schizophrenia, interpreting them in the light of recent molecular findings on their ability to induce neuroplasticity and modulate connectivity, the immune and TAARs systems, neurotransmitters, and neurotropic factors. No systematic approach was adopted in reviewing the evidence given the difficulty to retrieve and interpret old findings. Interestingly, we identified a therapeutic potential of psychedelics in schizophrenia adopting a critical point of view, particularly on negative symptoms and social cognition, and we summarized all the relevant findings. We also identified an eligible subpopulation of chronic patients predominantly burdened by negative symptoms, outlining possible therapeutic strategies which encompass very low doses of psychedelics (microdosing), carefully considering safety and feasibility, to pave the way to future clinical trials.
Collapse
Affiliation(s)
- Jacopo Sapienza
- IRCCS San Raffaele Scientific Institute, Milan, Italy
- Department of Humanities and Life Sciences, University School for Advanced Studies IUSS, Pavia, Italy
| | | | - Stefano Comai
- IRCCS San Raffaele Scientific Institute, Milan, Italy
- Department of Pharmaceutical and Pharmacological Sciences, University of Padua, Padua, Italy
- Department of Psychiatry, McGill University, Montreal, QC, Canada
- Department of Biomedical Sciences, University of Padua, Padua, Italy
| | - Roberto Cavallaro
- IRCCS San Raffaele Scientific Institute, Milan, Italy
- School of medicine, Vita-Salute San Raffaele University, Milan, Italy
| | | | - Danilo De Gregorio
- IRCCS San Raffaele Scientific Institute, Milan, Italy
- School of medicine, Vita-Salute San Raffaele University, Milan, Italy
| | - Marta Bosia
- IRCCS San Raffaele Scientific Institute, Milan, Italy
- School of medicine, Vita-Salute San Raffaele University, Milan, Italy
| |
Collapse
|
4
|
Erkizia-Santamaría I, Horrillo I, Meana JJ, Ortega JE. Clinical and preclinical evidence of psilocybin as antidepressant. A narrative review. Prog Neuropsychopharmacol Biol Psychiatry 2025; 136:111249. [PMID: 39778644 DOI: 10.1016/j.pnpbp.2025.111249] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/12/2024] [Revised: 01/02/2025] [Accepted: 01/03/2025] [Indexed: 01/11/2025]
Abstract
In the rapidly growing field of psychedelic research, psilocybin (and active metabolite psilocin) has been proposed as a promising candidate in the search for novel treatments for neuropsychiatric disorders. Clinical trials have revealed that psilocybin has a large, rapid, and persistent effect in the improvement of symptoms of depression and anxiety. The safety profile is considered favourable, with low toxicity and good tolerance. Several preclinical studies have also been carried out to determine the long-term mechanism of action of this drug. In this sense, preclinical studies in naïve animals as well as in animal models of disease have shown somewhat discrepant results in conventional tests for assessment of depression- and anxiety-like phenotype in response to psilocybin, but overall suggest positive outcomes. Additionally, several valuable assays in rodent models have been developed over the years to elucidate the neurochemical correlates of serotonin 2A receptor (5HT2AR) activation in the brain, primary molecular target of psilocin. This review aims to provide a general overview of the current and most recent literature in the therapeutic potential of psilocybin through a description of clinical trials of psilocybin-assisted psychotherapy, and to showcase the scene in the up-to-date preclinical research. A detailed description of preclinical rodent models and experimental approaches that have been used to study the neurobiological and behavioural actions of psilocybin is provided, and potential therapeutic mechanisms of action are discussed.
Collapse
Affiliation(s)
| | - Igor Horrillo
- Department of Pharmacology, University of the Basque Country UPV/EHU, Leioa, Bizkaia, Spain; Centro de Investigación Biomédica en Red de Salud Mental, Instituto de Salud Carlos III, Spain; Biobizkaia Health Research Institute, Barakaldo, Bizkaia, Spain
| | - J Javier Meana
- Department of Pharmacology, University of the Basque Country UPV/EHU, Leioa, Bizkaia, Spain; Centro de Investigación Biomédica en Red de Salud Mental, Instituto de Salud Carlos III, Spain; Biobizkaia Health Research Institute, Barakaldo, Bizkaia, Spain
| | - Jorge E Ortega
- Department of Pharmacology, University of the Basque Country UPV/EHU, Leioa, Bizkaia, Spain; Centro de Investigación Biomédica en Red de Salud Mental, Instituto de Salud Carlos III, Spain; Biobizkaia Health Research Institute, Barakaldo, Bizkaia, Spain.
| |
Collapse
|
5
|
McQueney AJ, Garcia EJ. Biological sex modulates the efficacy of 2,5-dimethoxy-4-iodoamphetamine (DOI) to mitigate fentanyl demand. Drug Alcohol Depend 2024; 263:112426. [PMID: 39217832 DOI: 10.1016/j.drugalcdep.2024.112426] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/27/2024] [Revised: 08/22/2024] [Accepted: 08/22/2024] [Indexed: 09/04/2024]
Abstract
BACKGROUND Overdose deaths remain high for opioid use disorder, emphasizing the need to pursue innovative therapeutics. Classic psychedelic drugs that engage many monoamine receptors mitigate opioid use. Here, we tested the hypothesis that the preferential serotonin 5-HT2AR agonist, 2,5-dimethoxy-4-iodoamphetamine (DOI) could reduce the demand for fentanyl in a preclinical model of fentanyl self-administration. METHODS Male and female Sprague-Dawley rats (n = 25-29) were implanted with indwelling jugular catheters and allowed to self-administer fentanyl (3.2μg/kg/infusion). Rats progressed to a novel low price twice within-session threshold procedure where rats sampled the lowest price twice before decreasing the dose of fentanyl by a ¼ log every 10minutes across 11 doses. Once stable, rats were pretreated with saline or DOI (0.01, 0.03, 1mg/kg). Fentanyl consumption was analyzed using an exponentiated demand function to extract the dependent variables, Q0 and α. RESULTS Male and female rats acquired fentanyl self-administration in the lowest price twice within-session threshold procedure. DOI dose-dependently altered fentanyl intake such that 5-HT2AR activation decreased Q0 in female rats but increased Q0 in male rats. For demand elasticity, DOI increased α in male rats but did not alter α in female rats. DOI did not alter inactive lever presses or latency. CONCLUSION DOI reduces consumption at minimally constrained costs but did not affect the reinforcement value of fentanyl in female rats. Alternatively, DOI significantly reduced the reinforcement value of fentanyl in male rats. Biological sex alters the therapeutic efficacy of DOI and 5-HT2AR activation sex-dependently alters opioid reinforcement.
Collapse
Affiliation(s)
- Alice J McQueney
- Neuroscience and Behavior, Psychology Department, University of Nebraska at Omaha, Omaha, NE, USA
| | - Erik J Garcia
- Neuroscience and Behavior, Psychology Department, University of Nebraska at Omaha, Omaha, NE, USA.
| |
Collapse
|
6
|
Zahid Z, Sultan ZW, Krause BM, Wenthur CJ, Pearce RA, Banks MI. Divergent Effects of Ketamine and the Serotoninergic Psychedelic 2,5-Dimethoxy-4-Iodoamphetamine on Hippocampal Plasticity and Metaplasticity. PSYCHEDELIC MEDICINE (NEW ROCHELLE, N.Y.) 2024; 2:166-177. [PMID: 39669671 PMCID: PMC11633440 DOI: 10.1089/psymed.2023.0061] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/14/2024]
Abstract
Introduction Serotonergic psychedelics and ketamine produce rapid and long-lasting symptomatic relief in multiple psychiatric disorders. Evidence suggests that despite having distinct molecular targets, both drugs may exert therapeutic benefit via their pro-neuroplastic effects. Following treatment with ketamine or serotonergic psychedelics, patients are reported to be more open to behavioral change, which is leveraged for psychotherapy-assisted reframing of narratives of the self. This period of enhanced behavioral change is postulated to be supported by a post-treatment window of enhanced neural plasticity, but evidence for such 'metaplastic' effects is limited. In this study, we tested for neural plasticity and metaplasticity in murine hippocampus. Methods Brain slices were obtained from C57BL/6J mice 24 hours after treatment (intraperitoneal injection) with saline, ketamine, or the serotonergic psychedelic 2,5-Dimethoxy-4-iodoamphetamine (DOI). Extracellular fiber volleys (FVs) and field excitatory postsynaptic potentials (fEPSPs) were recorded in stratum radiatum of CA1 in response to stimulation of Schaffer collateral fibers before and after induction of short-term and long-term potentiation (STP, LTP). Results Before LTP induction, responses differed across treatment groups (F1,2 = 5.407, p = 0.00665), with fEPSPs enhanced in slices from DOI-treated animals (p = 0.0182), but not ketamine-treated animals (p = 0.9786), compared to saline. There were no treatment effects on LT (F1,2 = 0.6, p = 0.516), but there were on STP (F1,2 =, p = 0.0167), with enhanced STP in DOI-treated (p = 0.0352) but not ketamine-treated (p = 0.9999) animals compared to saline. A presynaptic component to the mechanism for the DOI effects was suggested by (1) significantly enhanced FV amplitudes (F1,2 = 3.17, p = 0.049) in DOI-treated (p = 0.0457) but not ketamine-treated animals compared to saline (p = 0.8677); and (2) enhanced paired pulse ratios (F1,2 = 3.581, p = 0.0339) in slices from DOI-treated (p= 0.0257) but not ketamine-treated animals (p = 0.4845) compared to saline. Conclusions DOI, but not ketamine, induced significant neuroplastic and metaplastic effects at hippocampal CA1 synapses 24 hours after treatment, likely in part via a presynaptic mechanism.
Collapse
Affiliation(s)
- Zarmeen Zahid
- Department of Anesthesiology, University of Wisconsin School of Medicine and Public Health, Madison, WI, 53706
- School of Pharmacy, University of Wisconsin, Madison, WI, 53705
- Neuroscience Training Program, University of Wisconsin School of Medicine and Public Health, Madison, WI, 53706
| | - Ziyad W. Sultan
- Department of Anesthesiology, University of Wisconsin School of Medicine and Public Health, Madison, WI, 53706
| | - Bryan M. Krause
- Department of Anesthesiology, University of Wisconsin School of Medicine and Public Health, Madison, WI, 53706
| | - Cody J. Wenthur
- School of Pharmacy, University of Wisconsin, Madison, WI, 53705
- Transdisciplinary Center for Research in Psychoactive Substances, University of Wisconsin, Madison, WI, 53705
- Neuroscience Training Program, University of Wisconsin School of Medicine and Public Health, Madison, WI, 53706
| | - Robert A. Pearce
- Department of Anesthesiology, University of Wisconsin School of Medicine and Public Health, Madison, WI, 53706
| | - Matthew I. Banks
- Department of Anesthesiology, University of Wisconsin School of Medicine and Public Health, Madison, WI, 53706
- Transdisciplinary Center for Research in Psychoactive Substances, University of Wisconsin, Madison, WI, 53705
- Neuroscience Training Program, University of Wisconsin School of Medicine and Public Health, Madison, WI, 53706
| |
Collapse
|
7
|
Maia JM, de Oliveira BSA, Branco LGS, Soriano RN. Therapeutic potential of psychedelics: History, advancements, and unexplored frontiers. Prog Neuropsychopharmacol Biol Psychiatry 2024; 131:110951. [PMID: 38307161 DOI: 10.1016/j.pnpbp.2024.110951] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/24/2023] [Revised: 12/20/2023] [Accepted: 01/18/2024] [Indexed: 02/04/2024]
Abstract
Psychedelics (serotonergic hallucinogens) are psychoactive substances that can alter perception and mood, and affect cognitive functions. These substances activate 5-HT2A receptors and may exert therapeutic effects. Some of the disorders for which psychedelic-assisted therapy have been studied include depression, addiction, anxiety and post-traumatic stress disorder. Despite the increasing number of studies reporting clinical effectiveness, with fewer negative symptoms and, additionally, minimal side effects, questions remain to be explored in the field of psychedelic medicine. Although progress has been achieved, there is still little understanding of the relationship among human brain and the modulation induced by these drugs. The present article aimed to describe, review and highlight the most promising findings in the literature regarding the (putative) therapeutic effects of psychedelics.
Collapse
Affiliation(s)
- Juliana Marino Maia
- Department of Medicine, Federal University of Juiz de Fora, Governador Valadares, MG 35032-620, Brazil
| | | | - Luiz G S Branco
- Department of Basic and Oral Biology, School of Dentistry of Ribeirão Preto, University of São Paulo, Ribeirão Preto, SP 14040-904, Brazil; Department of Physiology, Medical School of Ribeirão Preto, University of São Paulo, Ribeirão Preto, SP 14040-900, Brazil.
| | - Renato Nery Soriano
- Division of Physiology and Biophysics, Department of Basic Life Sciences, Federal University of Juiz de Fora, Governador Valadares, MG 35020-360, Brazil
| |
Collapse
|
8
|
Sinha JK, Trisal A, Ghosh S, Gupta S, Singh KK, Han SS, Mahapatra M, Abomughaid MM, Abomughayedh AM, Almutary AG, Iqbal D, Bhaskar R, Mishra PC, Jha SK, Jha NK, Singh AK. Psychedelics for alzheimer's disease-related dementia: Unveiling therapeutic possibilities and pathways. Ageing Res Rev 2024; 96:102211. [PMID: 38307424 DOI: 10.1016/j.arr.2024.102211] [Citation(s) in RCA: 6] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/19/2023] [Revised: 01/25/2024] [Accepted: 01/26/2024] [Indexed: 02/04/2024]
Abstract
Psychedelics have traditionally been used for spiritual and recreational purposes, but recent developments in psychotherapy have highlighted their potential as therapeutic agents. These compounds, which act as potent 5-hydroxytryptamine (5HT) agonists, have been recognized for their ability to enhance neural plasticity through the activation of the serotoninergic and glutamatergic systems. However, the implications of these findings for the treatment of neurodegenerative disorders, particularly dementia, have not been fully explored. In recent years, studies have revealed the modulatory and beneficial effects of psychedelics in the context of dementia, specifically Alzheimer's disease (AD)-related dementia, which lacks a definitive cure. Psychedelics such as N,N-dimethyltryptamine (DMT), lysergic acid diethylamide (LSD), and Psilocybin have shown potential in mitigating the effects of this debilitating disease. These compounds not only target neurotransmitter imbalances but also act at the molecular level to modulate signalling pathways in AD, including the brain-derived neurotrophic factor signalling pathway and the subsequent activation of mammalian target of rapamycin and other autophagy regulators. Therefore, the controlled and dose-dependent administration of psychedelics represents a novel therapeutic intervention worth exploring and considering for the development of drugs for the treatment of AD-related dementia. In this article, we critically examined the literature that sheds light on the therapeutic possibilities and pathways of psychedelics for AD-related dementia. While this emerging field of research holds great promise, further studies are necessary to elucidate the long-term safety, efficacy, and optimal treatment protocols. Ultimately, the integration of psychedelics into the current treatment paradigm may provide a transformative approach for addressing the unmet needs of individuals living with AD-related dementia and their caregivers.
Collapse
Affiliation(s)
| | - Anchal Trisal
- Department of Biosciences, Jamia Millia Islamia, New Delhi 110025, India
| | - Shampa Ghosh
- GloNeuro, Sector 107, Vishwakarma Road, Noida 201301, India
| | - Saurabh Gupta
- Department of Biotechnology, GLA University, Mathura, Uttar Pradesh, India
| | - Krishna Kumar Singh
- Symbiosis Centre for Information Technology (SCIT), Rajiv Gandhi InfoTech Park, Hinjawadi, Pune, Maharashtra 411057, India
| | - Sung Soo Han
- School of Chemical Engineering, Yeungnam University, Gyeonsang 38541, the Republic of Korea; Research Institute of Cell Culture, Yeungnam University, 280 Daehak-Ro, Gyeongsan 38541, the Republic of Korea
| | | | - Mosleh Mohammad Abomughaid
- Department of Medical Laboratory Sciences, College of Applied Medical Sciences, University of Bisha, Bisha 61922, Saudi Arabia
| | - Ali M Abomughayedh
- Pharmacy Department, Aseer Central Hospital, Ministry of Health, Saudi Arabia
| | - Abdulmajeed G Almutary
- Department of Biomedical Sciences, College of Health Sciences, Abu Dhabi University, Abu Dhabi P.O. Box 59911, United Arab Emirates
| | - Danish Iqbal
- Department of Health Information Management, College of Applied Medical Sciences, Buraydah Private Colleges, Buraydah 51418, Saudi Arabia
| | - Rakesh Bhaskar
- School of Chemical Engineering, Yeungnam University, Gyeonsang 38541, the Republic of Korea; Research Institute of Cell Culture, Yeungnam University, 280 Daehak-Ro, Gyeongsan 38541, the Republic of Korea.
| | - Prabhu Chandra Mishra
- Department of Biotechnology, School of Engineering and Technology, Sharda University, Greater Noida, India
| | - Saurabh Kumar Jha
- Department of Zoology, Kalindi College, University of Delhi, 110008, India.
| | - Niraj Kumar Jha
- Centre for Global Health Research, Saveetha Medical College, Saveetha Institute of Medical and Technical Sciences, Saveetha University, Chennai, India; Centre of Research Impact and Outcome, Chitkara University, Rajpura 140401, Punjab, India; School of Bioengineering & Biosciences, Lovely Professional University, Phagwara 144411, India; Department of Biotechnology Engineering and Food Technology, Chandigarh University, Mohali, India.
| | - Abhishek Kumar Singh
- Manipal Centre for Biotherapeutics Research, Manipal Academy of Higher Education, Manipal, Karnataka 576104, India.
| |
Collapse
|
9
|
Hatzipantelis CJ, Olson DE. The Effects of Psychedelics on Neuronal Physiology. Annu Rev Physiol 2024; 86:27-47. [PMID: 37931171 PMCID: PMC10922499 DOI: 10.1146/annurev-physiol-042022-020923] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/08/2023]
Abstract
Psychedelics are quite unique among drugs that impact the central nervous system, as a single administration of a psychedelic can both rapidly alter subjective experience in profound ways and produce sustained effects on circuits relevant to mood, fear, reward, and cognitive flexibility. These remarkable properties are a direct result of psychedelics interacting with several key neuroreceptors distributed across the brain. Stimulation of these receptors activates a variety of signaling cascades that ultimately culminate in changes in neuronal structure and function. Here, we describe the effects of psychedelics on neuronal physiology, highlighting their acute effects on serotonergic and glutamatergic neurotransmission as well as their long-lasting effects on structural and functional neuroplasticity in the cortex. We propose that the neurobiological changes leading to the acute and sustained effects of psychedelics might be distinct, which could provide opportunities for engineering compounds with optimized safety and efficacy profiles.
Collapse
Affiliation(s)
- Cassandra J Hatzipantelis
- Institute for Psychedelics and Neurotherapeutics, University of California, Davis, Davis, California, USA;
- Department of Chemistry, University of California, Davis, Davis, California, USA
- Department of Biochemistry and Molecular Medicine, School of Medicine, University of California, Davis, Sacramento, California, USA
| | - David E Olson
- Institute for Psychedelics and Neurotherapeutics, University of California, Davis, Davis, California, USA;
- Department of Chemistry, University of California, Davis, Davis, California, USA
- Department of Biochemistry and Molecular Medicine, School of Medicine, University of California, Davis, Sacramento, California, USA
- Center for Neuroscience, University of California, Davis, Davis, California, USA
| |
Collapse
|
10
|
Fordyce BA, Roth BL. Making Sense of Psychedelics in the CNS. Int J Neuropsychopharmacol 2024; 27:pyae007. [PMID: 38289825 PMCID: PMC10888522 DOI: 10.1093/ijnp/pyae007] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/17/2023] [Accepted: 01/29/2024] [Indexed: 02/01/2024] Open
Abstract
For centuries, ancient lineages have consumed psychedelic compounds from natural sources. In the modern era, scientists have since harnessed the power of computational tools, cellular assays, and behavioral metrics to study how these compounds instigate changes on molecular, cellular, circuit-wide, and system levels. Here, we provide a brief history of psychedelics and their use in science, medicine, and culture. We then outline current techniques for studying psychedelics from a pharmacological perspective. Finally, we address known gaps in the field and potential avenues of further research to broaden our collective understanding of physiological changes induced by psychedelics, the limits of their therapeutic capabilities, and how researchers can improve and inform treatments that are rapidly becoming accessible worldwide.
Collapse
Affiliation(s)
- Blake A Fordyce
- Department of Neuroscience, UNC Chapel Hill Medical School Chapel Hill, North Carolina, USA
| | - Bryan L Roth
- Department of Pharmacology, UNC Chapel Hill Medical School Chapel Hill, North Carolina, USA
| |
Collapse
|
11
|
Wojtas A, Gołembiowska K. Molecular and Medical Aspects of Psychedelics. Int J Mol Sci 2023; 25:241. [PMID: 38203411 PMCID: PMC10778977 DOI: 10.3390/ijms25010241] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/17/2023] [Revised: 12/17/2023] [Accepted: 12/20/2023] [Indexed: 01/12/2024] Open
Abstract
Psychedelics belong to the oldest psychoactive drugs. They arouse recent interest due to their therapeutic applications in the treatment of major depressive disorder, substance use disorder, end-of-life anxiety,= and anxiety symptoms, and obsessive-compulsive disorder. In this review, the current state of preclinical research on the mechanism of action, neurotoxicity, and behavioral impact of psychedelics is summarized. The effect of selective 5-HT2A receptor agonists, 25I- and 25B-NBOMe, after acute and repeated administration is characterized and compared with the effects of a less selective drug, psilocybin. The data show a significant effect of NBOMes on glutamatergic, dopaminergic, serotonergic, and cholinergic neurotransmission in the frontal cortex, striatum, and nucleus accumbens. The increases in extracellular levels of neurotransmitters were not dose-dependent, which most likely resulted from the stimulation of the 5-HT2A receptor and subsequent activation of the 5-HT2C receptors. This effect was also observed in the wet dog shake test and locomotor activity. Chronic administration of NBOMes elicited rapid development of tolerance, genotoxicity, and activation of microglia. Acute treatment with psilocybin affected monoaminergic and aminoacidic neurotransmitters in the frontal cortex, nucleus accumbens, and hippocampus but not in the amygdala. Psilocybin exhibited anxiolytic properties resulting from intensification of GABAergic neurotransmission. The data indicate that NBOMes as selective 5-HT2A agonists exert a significant effect on neurotransmission and behavior of rats while also inducing oxidative DNA damage. In contrast to NBOMes, the effects induced by psilocybin suggest a broader therapeutic index of this drug.
Collapse
Affiliation(s)
| | - Krystyna Gołembiowska
- Unit II, Department of Pharmacology, Maj Institute of Pharmacology Polish Academy of Sciences, 12 Smętna Street, 31-343 Kraków, Poland;
| |
Collapse
|
12
|
Banushi B, Polito V. A Comprehensive Review of the Current Status of the Cellular Neurobiology of Psychedelics. BIOLOGY 2023; 12:1380. [PMID: 37997979 PMCID: PMC10669348 DOI: 10.3390/biology12111380] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/19/2023] [Revised: 10/26/2023] [Accepted: 10/26/2023] [Indexed: 11/25/2023]
Abstract
Psychedelic substances have gained significant attention in recent years for their potential therapeutic effects on various psychiatric disorders. This review delves into the intricate cellular neurobiology of psychedelics, emphasizing their potential therapeutic applications in addressing the global burden of mental illness. It focuses on contemporary research into the pharmacological and molecular mechanisms underlying these substances, particularly the role of 5-HT2A receptor signaling and the promotion of plasticity through the TrkB-BDNF pathway. The review also discusses how psychedelics affect various receptors and pathways and explores their potential as anti-inflammatory agents. Overall, this research represents a significant development in biomedical sciences with the potential to transform mental health treatments.
Collapse
Affiliation(s)
- Blerida Banushi
- Genetics and Genomic Medicine Department, Great Ormond Street Institute of Child Health, University College London, London WC1N 1EH, UK
| | - Vince Polito
- School of Psychological Sciences, Macquarie University, Sydney, NSW 2109, Australia;
| |
Collapse
|
13
|
Bedford P, Hauke DJ, Wang Z, Roth V, Nagy-Huber M, Holze F, Ley L, Vizeli P, Liechti ME, Borgwardt S, Müller F, Diaconescu AO. The effect of lysergic acid diethylamide (LSD) on whole-brain functional and effective connectivity. Neuropsychopharmacology 2023:10.1038/s41386-023-01574-8. [PMID: 37185950 PMCID: PMC10267115 DOI: 10.1038/s41386-023-01574-8] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/14/2022] [Revised: 03/22/2023] [Accepted: 03/23/2023] [Indexed: 05/17/2023]
Abstract
Psychedelics have emerged as promising candidate treatments for various psychiatric conditions, and given their clinical potential, there is a need to identify biomarkers that underlie their effects. Here, we investigate the neural mechanisms of lysergic acid diethylamide (LSD) using regression dynamic causal modelling (rDCM), a novel technique that assesses whole-brain effective connectivity (EC) during resting-state functional magnetic resonance imaging (fMRI). We modelled data from two randomised, placebo-controlled, double-blind, cross-over trials, in which 45 participants were administered 100 μg LSD and placebo in two resting-state fMRI sessions. We compared EC against whole-brain functional connectivity (FC) using classical statistics and machine learning methods. Multivariate analyses of EC parameters revealed predominantly stronger interregional connectivity and reduced self-inhibition under LSD compared to placebo, with the notable exception of weakened interregional connectivity and increased self-inhibition in occipital brain regions as well as subcortical regions. Together, these findings suggests that LSD perturbs the Excitation/Inhibition balance of the brain. Notably, whole-brain EC did not only provide additional mechanistic insight into the effects of LSD on the Excitation/Inhibition balance of the brain, but EC also correlated with global subjective effects of LSD and discriminated experimental conditions in a machine learning-based analysis with high accuracy (91.11%), highlighting the potential of using whole-brain EC to decode or predict subjective effects of LSD in the future.
Collapse
Affiliation(s)
- Peter Bedford
- Krembil Centre for Neuroinformatics, Centre for Addiction and Mental Health (CAMH), Toronto, ON, Canada
| | - Daniel J Hauke
- Centre for Medical Image Computing, Department of Computer Science, University College London, London, UK.
| | - Zheng Wang
- Krembil Centre for Neuroinformatics, Centre for Addiction and Mental Health (CAMH), Toronto, ON, Canada
| | - Volker Roth
- Department of Mathematics and Computer Science, University of Basel, Basel, Switzerland
| | - Monika Nagy-Huber
- Department of Mathematics and Computer Science, University of Basel, Basel, Switzerland
| | - Friederike Holze
- Division of Clinical Pharmacology and Toxicology, Department of Biomedicine and Department of Clinical Research, University Hospital Basel, University of Basel, Basel, Switzerland
| | - Laura Ley
- Division of Clinical Pharmacology and Toxicology, Department of Biomedicine and Department of Clinical Research, University Hospital Basel, University of Basel, Basel, Switzerland
| | - Patrick Vizeli
- Division of Clinical Pharmacology and Toxicology, Department of Biomedicine and Department of Clinical Research, University Hospital Basel, University of Basel, Basel, Switzerland
| | - Matthias E Liechti
- Division of Clinical Pharmacology and Toxicology, Department of Biomedicine and Department of Clinical Research, University Hospital Basel, University of Basel, Basel, Switzerland
| | - Stefan Borgwardt
- Department of Psychiatry and Psychotherapy, Translational Psychiatry, Lübeck, Germany
| | - Felix Müller
- Department of Psychiatry (UPK), University of Basel, Basel, Switzerland
| | - Andreea O Diaconescu
- Krembil Centre for Neuroinformatics, Centre for Addiction and Mental Health (CAMH), Toronto, ON, Canada
- Department of Psychiatry, University of Toronto, Toronto, ON, Canada
- Institute of Medical Sciences, University of Toronto, Toronto, ON, Canada
- Department of Psychology, University of Toronto, Toronto, ON, Canada
| |
Collapse
|
14
|
Sullivan M, Fernandez-Aranda F, Camacho-Barcia L, Harkin A, Macrì S, Mora-Maltas B, Jiménez-Murcia S, O'Leary A, Ottomana AM, Presta M, Slattery D, Scholtz S, Glennon JC. Insulin and Disorders of Behavioural Flexibility. Neurosci Biobehav Rev 2023; 150:105169. [PMID: 37059405 DOI: 10.1016/j.neubiorev.2023.105169] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/30/2022] [Revised: 04/03/2023] [Accepted: 04/10/2023] [Indexed: 04/16/2023]
Abstract
Behavioural inflexibility is a symptom of neuropsychiatric and neurodegenerative disorders such as Obsessive-Compulsive Disorder, Autism Spectrum Disorder and Alzheimer's Disease, encompassing the maintenance of a behaviour even when no longer appropriate. Recent evidence suggests that insulin signalling has roles apart from its regulation of peripheral metabolism and mediates behaviourally-relevant central nervous system (CNS) functions including behavioural flexibility. Indeed, insulin resistance is reported to generate anxious, perseverative phenotypes in animal models, with the Type 2 diabetes medication metformin proving to be beneficial for disorders including Alzheimer's Disease. Structural and functional neuroimaging studies of Type 2 diabetes patients have highlighted aberrant connectivity in regions governing salience detection, attention, inhibition and memory. As currently available therapeutic strategies feature high rates of resistance, there is an urgent need to better understand the complex aetiology of behaviour and develop improved therapeutics. In this review, we explore the circuitry underlying behavioural flexibility, changes in Type 2 diabetes, the role of insulin in CNS outcomes and mechanisms of insulin involvement across disorders of behavioural inflexibility.
Collapse
Affiliation(s)
- Mairéad Sullivan
- Conway Institute of Biomedical and Biomolecular Research, School of Medicine, University College Dublin, Dublin, Ireland.
| | - Fernando Fernandez-Aranda
- Department of Psychiatry, University Hospital of Bellvitge, Barcelona, Spain; Psychoneurobiology of Eating and Addictive Behaviors Group, Neurosciences Program, Bellvitge Biomedical Research Institute (IDIBELL), Barcelona, Spain; CIBER Fisiopatología Obesidad y Nutrición (CIBERobn), Instituto de Salud Carlos III, Barcelona, Spain; Department of Clinical Sciences, School of Medicine and Health Sciences, University of Barcelona, Barcelona, Spain
| | - Lucía Camacho-Barcia
- Department of Psychiatry, University Hospital of Bellvitge, Barcelona, Spain; Psychoneurobiology of Eating and Addictive Behaviors Group, Neurosciences Program, Bellvitge Biomedical Research Institute (IDIBELL), Barcelona, Spain; CIBER Fisiopatología Obesidad y Nutrición (CIBERobn), Instituto de Salud Carlos III, Barcelona, Spain
| | - Andrew Harkin
- School of Pharmacy and Pharmaceutical Sciences, Trinity College Dublin, Ireland
| | - Simone Macrì
- Centre for Behavioural Sciences and Mental Health, Istituto Superiore di Sanità, 00161 Rome, Italy
| | - Bernat Mora-Maltas
- Department of Psychiatry, University Hospital of Bellvitge, Barcelona, Spain; Psychoneurobiology of Eating and Addictive Behaviors Group, Neurosciences Program, Bellvitge Biomedical Research Institute (IDIBELL), Barcelona, Spain
| | - Susana Jiménez-Murcia
- Department of Psychiatry, University Hospital of Bellvitge, Barcelona, Spain; Psychoneurobiology of Eating and Addictive Behaviors Group, Neurosciences Program, Bellvitge Biomedical Research Institute (IDIBELL), Barcelona, Spain; CIBER Fisiopatología Obesidad y Nutrición (CIBERobn), Instituto de Salud Carlos III, Barcelona, Spain; Department of Clinical Sciences, School of Medicine and Health Sciences, University of Barcelona, Barcelona, Spain
| | - Aet O'Leary
- University Hospital Frankfurt, Frankfurt, Germany
| | - Angela Maria Ottomana
- Centre for Behavioural Sciences and Mental Health, Istituto Superiore di Sanità, 00161 Rome, Italy; Neuroscience Unit, Department of Medicine, University of Parma, 43100 Parma, Italy
| | - Martina Presta
- Centre for Behavioural Sciences and Mental Health, Istituto Superiore di Sanità, 00161 Rome, Italy; Department of Physiology and Pharmacology, Sapienza University of Rome, 00185 Rome, Italy
| | | | | | - Jeffrey C Glennon
- Conway Institute of Biomedical and Biomolecular Research, School of Medicine, University College Dublin, Dublin, Ireland
| |
Collapse
|
15
|
Marguilho M, Figueiredo I, Castro-Rodrigues P. A unified model of ketamine's dissociative and psychedelic properties. J Psychopharmacol 2023; 37:14-32. [PMID: 36527355 PMCID: PMC9834329 DOI: 10.1177/02698811221140011] [Citation(s) in RCA: 10] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/23/2022]
Abstract
Ketamine is an N-methyl-d-aspartate antagonist which is increasingly being researched and used as a treatment for depression. In low doses, it can cause a transitory modification in consciousness which was classically labelled as 'dissociation'. However, ketamine is also commonly classified as an atypical psychedelic and it has been recently reported that ego dissolution experiences during ketamine administration are associated with greater antidepressant response. Neuroimaging studies have highlighted several similarities between the effects of ketamine and those of serotonergic psychedelics in the brain; however, no unified account has been proposed for ketamine's multi-level effects - from molecular to network and psychological levels. Here, we propose that the fast, albeit transient, antidepressant effects observed after ketamine infusions are mainly driven by its acute modulation of reward circuits and sub-acute increase in neuroplasticity, while its dissociative and psychedelic properties are driven by dose- and context-dependent disruption of large-scale functional networks. Computationally, as nodes of the salience network (SN) represent high-level priors about the body ('minimal' self) and nodes of the default-mode network (DMN) represent the highest-level priors about narrative self-experience ('biographical' self), we propose that transitory SN desegregation and disintegration accounts for ketamine's 'dissociative' state, while transitory DMN desegregation and disintegration accounts for ketamine's 'psychedelic' state. In psychedelic-assisted psychotherapy, a relaxation of the highest-level beliefs with psychotherapeutic support may allow a revision of pathological self-representation models, for which neuroplasticity plays a permissive role. Our account provides a multi-level rationale for using the psychedelic properties of ketamine to increase its long-term benefits.
Collapse
Affiliation(s)
| | | | - Pedro Castro-Rodrigues
- Centro Hospitalar Psiquiátrico de Lisboa, Lisbon, Portugal,NOVA Medical School, NMS, Universidade Nova de Lisboa, Lisbon, Portugal,Pedro Castro-Rodrigues, Centro Hospitalar Psiquiátrico de Lisboa, Avenida do Brasil, 53, Lisbon, 1749-002, Portugal.
| |
Collapse
|
16
|
Dourron HM, Strauss C, Hendricks PS. Self-Entropic Broadening Theory: Toward a New Understanding of Self and Behavior Change Informed by Psychedelics and Psychosis. Pharmacol Rev 2022; 74:982-1027. [PMID: 36113878 DOI: 10.1124/pharmrev.121.000514] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/12/2021] [Revised: 06/14/2022] [Accepted: 06/16/2022] [Indexed: 03/21/2025] Open
Abstract
The extremes of human experiences, such as those occasioned by classic psychedelics and psychosis, provide a rich contrast for understanding how components of these experiences impact well-being. In recent years, research has suggested that classic psychedelics display the potential to promote positive enduring psychologic and behavioral changes in clinical and nonclinical populations. Paradoxically, classic psychedelics have been described as psychotomimetics. This review offers a putative solution to this paradox by providing a theory of how classic psychedelics often facilitate persistent increases in well-being, whereas psychosis leads down a "darker" path. This will be done by providing an overview of the overlap between the states (i.e., entropic processing) and their core differences (i.e., self-focus). In brief, entropic processing can be defined as an enhanced overall attentional scope and decreased predictability in processing stimuli facilitating a hyperassociative style of thinking. However, the outcomes of entropic states vary depending on level of self-focus, or the degree to which the associations and information being processed are evaluated in a self-referential manner. We also describe potential points of overlap with less extreme experiences, such as creative thinking and positive emotion-induction. Self-entropic broadening theory offers a heuristically valuable perspective on classic psychedelics and their lasting effects and relation to other states by creating a novel synthesis of contemporary theories in psychology. SIGNIFICANCE STATEMENT: Self-entropic broadening theory provides a novel theory examining the psychedelic-psychotomimetic paradox, or how classic psychedelics can be therapeutic, yet mimic symptoms of psychosis. It also posits a framework for understanding the transdiagnostic applicability of classic psychedelics. We hope this model invigorates the field to provide more rigorous comparisons between classic psychedelic-induced states and psychosis and further examinations of how classic psychedelics facilitate long-term change. As a more psychedelic future of psychiatry appears imminent, a model that addresses these long-standing questions is crucial.
Collapse
Affiliation(s)
- Haley Maria Dourron
- Drug Use & Behavior Laboratory, School of Public Health, University of Alabama at Birmingham, Birmingham, Alabama (H.M.D., P.S.H.) and Princeton Neuroscience Institute, Princeton University, Princeton, New Jersey (C.S.)
| | - Camilla Strauss
- Drug Use & Behavior Laboratory, School of Public Health, University of Alabama at Birmingham, Birmingham, Alabama (H.M.D., P.S.H.) and Princeton Neuroscience Institute, Princeton University, Princeton, New Jersey (C.S.)
| | - Peter S Hendricks
- Drug Use & Behavior Laboratory, School of Public Health, University of Alabama at Birmingham, Birmingham, Alabama (H.M.D., P.S.H.) and Princeton Neuroscience Institute, Princeton University, Princeton, New Jersey (C.S.)
| |
Collapse
|
17
|
Presynaptic 5-HT2A-mGlu2/3 Receptor–Receptor Crosstalk in the Prefrontal Cortex: Metamodulation of Glutamate Exocytosis. Cells 2022; 11:cells11193035. [PMID: 36230998 PMCID: PMC9562019 DOI: 10.3390/cells11193035] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/19/2022] [Revised: 09/19/2022] [Accepted: 09/26/2022] [Indexed: 12/02/2022] Open
Abstract
The glutamatergic nerve endings of a rat prefrontal cortex (PFc) possess presynaptic 5-HT2A heteroreceptors and mGlu2/3 autoreceptors, whose activation inhibits glutamate exocytosis, and is measured as 15 mM KCl-evoked [3H]D-aspartate ([3H]D-asp) release (which mimics glutamate exocytosis). The concomitant activation of the two receptors nulls their inhibitory activities, whereas blockade of the 5-HT2A heteroreceptors with MDL11,939 (1 μM) strengthens the inhibitory effect elicited by the mGlu2/3 receptor agonist LY329268 (1 μM). 5-HT2A receptor antagonists (MDL11,939; ketanserin; trazodone) amplify the impact of low (3 nM) LY379268. Clozapine (0.1–10 μM) mimics the 5-HT2A agonist (±) DOI and inhibits the KCl-evoked [3H]D-asp overflow in a MDL11,939-dependent fashion, but does not modify the (±) DOI-induced effect. mGlu2 and 5-HT2A proteins do not co-immunoprecipitate from synaptosomal lysates, nor does the incubation of PFc synaptosomes with MDL11,939 (1 μM) or clozapine (10 µM) modify the insertion of mGlu2 subunits in synaptosomal plasma membranes. In conclusion, 5-HT2A and mGlu2/3 receptors colocalize, but do not physically associate, in PFc glutamatergic terminals, where they functionally interact in an antagonist-like fashion to control glutamate exocytosis. The mGlu2/3-5-HT2A metamodulation could be relevant to therapy for central neuropsychiatric disorders, including schizophrenia, but also unveil cellular events accounting for their development, which also influence the responsiveness to drugs regimens.
Collapse
|
18
|
Abstract
N,N-dimethyltryptamine (DMT) is a potent psychedelic naturally produced by many plants and animals, including humans. Whether or not DMT is significant to mammalian physiology, especially within the central nervous system, is a debate that started in the early 1960s and continues to this day. This review integrates historical and recent literature to clarify this issue, giving special attention to the most controversial subjects of DMT's biosynthesis, its storage in synaptic vesicles and the activation receptors like sigma-1. Less discussed topics, like DMT's metabolic regulation or the biased activation of serotonin receptors, are highlighted. We conclude that most of the arguments dismissing endogenous DMT's relevance are based on obsolete data or misleading assumptions. Data strongly suggest that DMT can be relevant as a neurotransmitter, neuromodulator, hormone and immunomodulator, as well as being important to pregnancy and development. Key experiments are addressed to definitely prove what specific roles DMT plays in mammalian physiology.
Collapse
Affiliation(s)
- Javier Hidalgo Jiménez
- ICEERS Foundation (International Center for Ethnobotanical Education, Research and Services), Barcelona, Spain
| | - José Carlos Bouso
- ICEERS Foundation (International Center for Ethnobotanical Education, Research and Services), Barcelona, Spain
| |
Collapse
|
19
|
Effect of Psilocybin and Ketamine on Brain Neurotransmitters, Glutamate Receptors, DNA and Rat Behavior. Int J Mol Sci 2022; 23:ijms23126713. [PMID: 35743159 PMCID: PMC9224489 DOI: 10.3390/ijms23126713] [Citation(s) in RCA: 34] [Impact Index Per Article: 11.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/28/2022] [Revised: 06/14/2022] [Accepted: 06/14/2022] [Indexed: 12/28/2022] Open
Abstract
Clinical studies provide evidence that ketamine and psilocybin could be used as fast-acting antidepressants, though their mechanisms and toxicity are still not fully understood. To address this issue, we have examined the effect of a single administration of ketamine and psilocybin on the extracellular levels of neurotransmitters in the rat frontal cortex and reticular nucleus of the thalamus using microdialysis. The genotoxic effect and density of glutamate receptor proteins was measured with comet assay and Western blot, respectively. An open field test, light–dark box test and forced swim test were conducted to examine rat behavior 24 h after drug administration. Ketamine (10 mg/kg) and psilocybin (2 and 10 mg/kg) increased dopamine, serotonin, glutamate and GABA extracellular levels in the frontal cortex, while psilocybin also increased GABA in the reticular nucleus of the thalamus. Oxidative DNA damage due to psilocybin was observed in the frontal cortex and from both drugs in the hippocampus. NR2A subunit levels were increased after psilocybin (10 mg/kg). Behavioral tests showed no antidepressant or anxiolytic effects, and only ketamine suppressed rat locomotor activity. The observed changes in neurotransmission might lead to genotoxicity and increased NR2A levels, while not markedly affecting animal behavior.
Collapse
|
20
|
Kojic M, Saelens J, Kadriu B, Zarate CA, Kraus C. Ketamine for Depression: Advances in Clinical Treatment, Rapid Antidepressant Mechanisms of Action, and a Contrast with Serotonergic Psychedelics. Curr Top Behav Neurosci 2022; 56:141-167. [PMID: 35312993 PMCID: PMC10500612 DOI: 10.1007/7854_2022_313] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 06/14/2023]
Abstract
The approval of ketamine for treatment-resistant depression has created a model for a novel class of rapid-acting glutamatergic antidepressants. Recent research into other novel rapid-acting antidepressants - most notably serotonergic psychedelics (SPs) - has also proven promising. Presently, the mechanisms of action of these substances are under investigation to improve these novel treatments, which also exhibit considerable side effects such as dissociation. This chapter lays out the historical development of ketamine as an antidepressant, outlines its efficacy and safety profile, reviews the evidence for ketamine's molecular mechanism of action, and compares it to the proposed mechanism of SPs. The evidence suggests that although ketamine and SPs act on distinct primary targets, both may lead to rapid restoration of synaptic deficits and downstream network reconfiguration. In both classes of drugs, a glutamate surge activates α-amino-3-hydroxy-5-methyl-4-isoxazolepropionic acid receptor (AMPAR) throughput and increases in brain-derived neurotrophic factor (BDNF) levels. Taken together, these novel antidepressant mechanisms may serve as a framework to explain the rapid and sustained antidepressant effects of ketamine and may be crucial for developing new rapid-acting antidepressants with an improved side effect profile.
Collapse
Affiliation(s)
- Marina Kojic
- Department of Pediatrics and Adolescent Medicine, Medical University of Vienna, Vienna, Austria
| | - Johan Saelens
- Department of Psychiatry and Psychotherapy, Medical University of Vienna, Vienna, Austria
| | - Bashkim Kadriu
- Section on the Neurobiology and Treatment of Mood Disorders, National Institute of Mental Health, National Institutes of Health, Bethesda, MD, USA
- Department of Neuroscience, Janssen Research & Development, LLC, San Diego, CA, USA
| | - Carlos A Zarate
- Section on the Neurobiology and Treatment of Mood Disorders, National Institute of Mental Health, National Institutes of Health, Bethesda, MD, USA
| | - Christoph Kraus
- Department of Psychiatry and Psychotherapy, Medical University of Vienna, Vienna, Austria.
- Section on the Neurobiology and Treatment of Mood Disorders, National Institute of Mental Health, National Institutes of Health, Bethesda, MD, USA.
| |
Collapse
|
21
|
Herian M, Skawski M, Wojtas A, Sobocińska MK, Noworyta K, Gołembiowska K. Tolerance to neurochemical and behavioral effects of the hallucinogen 25I-NBOMe. Psychopharmacology (Berl) 2021; 238:2349-2364. [PMID: 34032876 PMCID: PMC8292280 DOI: 10.1007/s00213-021-05860-5] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/02/2020] [Accepted: 04/22/2021] [Indexed: 12/25/2022]
Abstract
RATIONALE 4-Iodo-2,5-dimethoxy-N-(2-methoxybenzyl)phenethylamine (25I-NBOMe) is a potent serotonin 5-HT2A/2C receptor agonist with hallucinogenic activity. There is no data on the 25I-NBOMe effect on brain neurotransmission and animal performance after chronic administration. OBJECTIVES We examined the effect of a 7-day treatment with 25I-NBOMe (0.3 mg/kg/day) on neurotransmitters' release and rats' behavior in comparison to acute dose. METHODS Changes in dopamine (DA), serotonin (5-HT), acetylcholine (ACh), and glutamate release were studied using microdialysis in freely moving rats. The hallucinogenic activity was measured in the wet dog shake (WDS) test. The animal locomotion was examined in the open field (OF) test, short-term memory in the novel object recognition (NOR) test. The anxiogenic/anxiolytic properties of the drug were tested using the light/dark box (LDB) test. RESULTS Repeated administration of 25I-NBOMe decreased the response to a challenge dose of DA, 5-HT, and glutamatergic neurons in the frontal cortex as well as weakened the hallucinogenic activity in comparison to acute dose. In contrast, striatal and accumbal DA and 5-HT release and accumbal but not striatal glutamate release in response to the challenge dose of 25I-NBOMe was increased in comparison to acute treatment. The ACh release was increased in all brain regions. Behavioral tests showed a motor activity reduction and memory deficiency in comparison to a single dose and induction of anxiety after the drug's chronic and acute administration. CONCLUSIONS Our findings suggest that multiple injections of 25I-NBOMe induce tolerance to hallucinogenic activity and produce alterations in neurotransmission. 25I-NBOMe effect on short-term memory, locomotor function, and anxiety seems to be the result of complex interactions between neurotransmitter pathways.
Collapse
Affiliation(s)
- Monika Herian
- Department of Pharmacology, Polish Academy of Sciences, Maj Institute of Pharmacology, 12 Smętna, 31-343, Kraków, Poland
| | - Mateusz Skawski
- Department of Pharmacology, Polish Academy of Sciences, Maj Institute of Pharmacology, 12 Smętna, 31-343, Kraków, Poland
| | - Adam Wojtas
- Department of Pharmacology, Polish Academy of Sciences, Maj Institute of Pharmacology, 12 Smętna, 31-343, Kraków, Poland
| | - Małgorzata K Sobocińska
- Department of Pharmacology, Polish Academy of Sciences, Maj Institute of Pharmacology, 12 Smętna, 31-343, Kraków, Poland
| | - Karolina Noworyta
- Department of Pharmacology, Polish Academy of Sciences, Maj Institute of Pharmacology, 12 Smętna, 31-343, Kraków, Poland
| | - Krystyna Gołembiowska
- Department of Pharmacology, Polish Academy of Sciences, Maj Institute of Pharmacology, 12 Smętna, 31-343, Kraków, Poland.
| |
Collapse
|
22
|
Banks MI, Zahid Z, Jones NT, Sultan ZW, Wenthur CJ. Catalysts for change: the cellular neurobiology of psychedelics. Mol Biol Cell 2021; 32:1135-1144. [PMID: 34043427 PMCID: PMC8351556 DOI: 10.1091/mbc.e20-05-0340] [Citation(s) in RCA: 17] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/21/2021] [Revised: 03/23/2021] [Accepted: 03/31/2021] [Indexed: 01/18/2023] Open
Abstract
The resurgence of interest in the therapeutic potential of psychedelics for treating psychiatric disorders has rekindled efforts to elucidate their mechanism of action. In this Perspective, we focus on the ability of psychedelics to promote neural plasticity, postulated to be central to their therapeutic activity. We begin with a brief overview of the history and behavioral effects of the classical psychedelics. We then summarize our current understanding of the cellular and subcellular mechanisms underlying these drugs' behavioral effects, their effects on neural plasticity, and the roles of stress and inflammation in the acute and long-term effects of psychedelics. The signaling pathways activated by psychedelics couple to numerous potential mechanisms for producing long-term structural changes in the brain, a complexity that has barely begun to be disentangled. This complexity is mirrored by that of the neural mechanisms underlying psychiatric disorders and the transformations of consciousness, mood, and behavior that psychedelics promote in health and disease. Thus, beyond changes in the brain, psychedelics catalyze changes in our understanding of the neural basis of psychiatric disorders, as well as consciousness and human behavior.
Collapse
Affiliation(s)
- Matthew I. Banks
- Department of Anesthesiology, University of Wisconsin School of Medicine and Public Health, Madison, WI 53706
- Neuroscience Training Program, University of Wisconsin–Madison, Madison, WI 53706
| | - Zarmeen Zahid
- Neuroscience Training Program, University of Wisconsin–Madison, Madison, WI 53706
| | - Nathan T. Jones
- Molecular and Cellular Pharmacology Training Program, University of Wisconsin–Madison, Madison, WI 53706
| | - Ziyad W. Sultan
- Department of Anesthesiology, University of Wisconsin School of Medicine and Public Health, Madison, WI 53706
| | - Cody J. Wenthur
- Neuroscience Training Program, University of Wisconsin–Madison, Madison, WI 53706
- Molecular and Cellular Pharmacology Training Program, University of Wisconsin–Madison, Madison, WI 53706
- School of Pharmacy, University of Wisconsin–Madison, Madison, WI 53705
| |
Collapse
|
23
|
Wojtas A, Herian M, Skawski M, Sobocińska M, González-Marín A, Noworyta-Sokołowska K, Gołembiowska K. Neurochemical and Behavioral Effects of a New Hallucinogenic Compound 25B-NBOMe in Rats. Neurotox Res 2021; 39:305-326. [PMID: 33337517 PMCID: PMC7936972 DOI: 10.1007/s12640-020-00297-8] [Citation(s) in RCA: 16] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/29/2020] [Revised: 10/08/2020] [Accepted: 10/12/2020] [Indexed: 12/20/2022]
Abstract
4-Bromo-2,5-dimethoxy-N-(2-methoxybenzyl)phenethylamine (25B-NBOMe) is a hallucinogen exhibiting high binding affinity for 5-HT2A/C serotonin receptors. In the present work, we investigated its effect on dopamine (DA), serotonin (5-HT), acetylcholine (ACh), and glutamate release in the rat frontal cortex, striatum, and nucleus accumbens. Hallucinogenic activity, impact on cognitive and motor functions, and anxiogenic/anxiolytic properties of this compound were also tested. The release of DA, 5-HT, ACh, and glutamate was studied using microdialysis in freely moving animals. Hallucinogenic activity was investigated using head and body twitch response (WDS), cognitive functions were examined with the novel object recognition test (NOR), locomotor activity was studied in the open field (OF), while anxiogenic/anxiolytic effect was tested using the light/dark box (LDB). Neurotoxicity was evaluated with the comet assay. 25B-NBOMe increased DA, 5-HT, and glutamate release in all studied brain regions, induced hallucinogenic activity, and lowered the recognition index (Ri) vs. control in the NOR test. It also decreased locomotor activity of rats in the OF test. The effect of 25B-NBOMe in the NOR test was inhibited by scopolamine. In the LDB test, the time spent in the dark zone was longer in comparison to control and was dose-dependent. In contrast to MDMA, 25B-NBOMe showed subtle genotoxic effect observed in the comet assay.Our findings indicate that 25B-NBOMe shows hallucinogenic activity in the wide range of doses. The changes in neurotransmitter levels may be related to 25B-NBOMe affinity for 5-HT2A receptor. Alterations in the NOR, OF, and LDB indicate that 25B-NBOMe impacts short-term memory, locomotion, and may be anxiogenic.
Collapse
Affiliation(s)
- Adam Wojtas
- Maj Institute of Pharmacology, Polish Academy of Sciences, Department of Pharmacology, 12 Smętna, 31-343, Kraków, Poland
| | - Monika Herian
- Maj Institute of Pharmacology, Polish Academy of Sciences, Department of Pharmacology, 12 Smętna, 31-343, Kraków, Poland
| | - Mateusz Skawski
- Maj Institute of Pharmacology, Polish Academy of Sciences, Department of Pharmacology, 12 Smętna, 31-343, Kraków, Poland
| | - Małgorzata Sobocińska
- Maj Institute of Pharmacology, Polish Academy of Sciences, Department of Pharmacology, 12 Smętna, 31-343, Kraków, Poland
| | - Alejandro González-Marín
- Maj Institute of Pharmacology, Polish Academy of Sciences, Department of Pharmacology, 12 Smętna, 31-343, Kraków, Poland
| | - Karolina Noworyta-Sokołowska
- Maj Institute of Pharmacology, Polish Academy of Sciences, Department of Pharmacology, 12 Smętna, 31-343, Kraków, Poland
| | - Krystyna Gołembiowska
- Maj Institute of Pharmacology, Polish Academy of Sciences, Department of Pharmacology, 12 Smętna, 31-343, Kraków, Poland.
| |
Collapse
|
24
|
Contreras A, Khumnark M, Hines RM, Hines DJ. Behavioral arrest and a characteristic slow waveform are hallmark responses to selective 5-HT 2A receptor activation. Sci Rep 2021; 11:1925. [PMID: 33479368 PMCID: PMC7820508 DOI: 10.1038/s41598-021-81552-6] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/27/2020] [Accepted: 01/07/2021] [Indexed: 11/09/2022] Open
Abstract
Perception, emotion, and mood are powerfully modulated by serotonin receptor (5-HTR) agonists including hallucinogens. The 5-HT2AR subtype has been shown to be central to hallucinogen action, yet the precise mechanisms mediating the response to 5-HT2AR activation remain unclear. Hallucinogens induce the head twitch response (HTR) in rodents, which is the most commonly used behavioral readout of hallucinogen pharmacology. While the HTR provides a key behavioral signature, less is known about the meso level changes that are induced by 5-HT2AR activation. In response to administration of the potent and highly selective 5-HT2AR agonist 25I-NBOH in mice, we observe a disorganization of behavior which includes frequent episodes of behavioral arrest that consistently precede the HTR by a precise interval. By combining behavioral analysis with electroencephalogram (EEG) recordings we describe a characteristic pattern composed of two distinctive EEG waveforms, Phase 1 and Phase 2, that map onto behavioral arrest and the HTR respectively, with the same temporal separation. Phase 1, which underlies behavioral arrest, is a 3.5-4.5 Hz waveform, while Phase 2 is slower at 2.5-3.2 Hz. Nicotine pretreatment, considered an integral component of ritualistic hallucinogen practices, attenuates 25I-NBOH induced HTR and Phase 2 waveforms, yet increases behavioral arrest and Phase 1 waveforms. Our results suggest that in addition to the HTR, behavioral arrest and characteristic meso level slow waveforms are key hallmarks of the response to 5-HT2AR activation. Increased understanding of the response to serotonergic hallucinogens may provide mechanistic insights into perception and hallucinations, as well as regulation of mood.
Collapse
Affiliation(s)
- April Contreras
- University of Nevada, Las Vegas, 4505 Maryland Parkway, Las Vegas, NV, 89154, USA
| | - Matthew Khumnark
- University of Nevada, Las Vegas, 4505 Maryland Parkway, Las Vegas, NV, 89154, USA
| | - Rochelle M Hines
- University of Nevada, Las Vegas, 4505 Maryland Parkway, Las Vegas, NV, 89154, USA
| | - Dustin J Hines
- University of Nevada, Las Vegas, 4505 Maryland Parkway, Las Vegas, NV, 89154, USA.
| |
Collapse
|
25
|
Kadriu B, Greenwald M, Henter ID, Gilbert JR, Kraus C, Park LT, Zarate CA. Ketamine and Serotonergic Psychedelics: Common Mechanisms Underlying the Effects of Rapid-Acting Antidepressants. Int J Neuropsychopharmacol 2021; 24:8-21. [PMID: 33252694 PMCID: PMC7816692 DOI: 10.1093/ijnp/pyaa087] [Citation(s) in RCA: 67] [Impact Index Per Article: 16.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/30/2020] [Revised: 10/13/2020] [Accepted: 11/16/2020] [Indexed: 12/21/2022] Open
Abstract
BACKGROUND The glutamatergic modulator ketamine has created a blueprint for studying novel pharmaceuticals in the field. Recent studies suggest that "classic" serotonergic psychedelics (SPs) may also have antidepressant efficacy. Both ketamine and SPs appear to produce rapid, sustained antidepressant effects after a transient psychoactive period. METHODS This review summarizes areas of overlap between SP and ketamine research and considers the possibility of a common, downstream mechanism of action. The therapeutic relevance of the psychoactive state, overlapping cellular and molecular effects, and overlapping electrophysiological and neuroimaging observations are all reviewed. RESULTS Taken together, the evidence suggests a potentially shared mechanism wherein both ketamine and SPs may engender rapid neuroplastic effects in a glutamatergic activity-dependent manner. It is postulated that, though distinct, both ketamine and SPs appear to produce acute alterations in cortical network activity that may initially produce psychoactive effects and later produce milder, sustained changes in network efficiency associated with therapeutic response. However, despite some commonalities between the psychoactive component of these pharmacologically distinct therapies-such as engagement of the downstream glutamatergic pathway-the connection between psychoactive impact and antidepressant efficacy remains unclear and requires more rigorous research. CONCLUSIONS Rapid-acting antidepressants currently under investigation may share some downstream pharmacological effects, suggesting that their antidepressant effects may come about via related mechanisms. Given the prototypic nature of ketamine research and recent progress in this area, this platform could be used to investigate entirely new classes of antidepressants with rapid and robust actions.
Collapse
Affiliation(s)
- Bashkim Kadriu
- Section on the Neurobiology and Treatment of Mood Disorders, National Institute of Mental Health, National Institutes of Health, Bethesda, MD, USA
| | - Maximillian Greenwald
- Section on the Neurobiology and Treatment of Mood Disorders, National Institute of Mental Health, National Institutes of Health, Bethesda, MD, USA
| | - Ioline D Henter
- Section on the Neurobiology and Treatment of Mood Disorders, National Institute of Mental Health, National Institutes of Health, Bethesda, MD, USA
| | - Jessica R Gilbert
- Section on the Neurobiology and Treatment of Mood Disorders, National Institute of Mental Health, National Institutes of Health, Bethesda, MD, USA
| | - Christoph Kraus
- Section on the Neurobiology and Treatment of Mood Disorders, National Institute of Mental Health, National Institutes of Health, Bethesda, MD, USA
| | - Lawrence T Park
- Section on the Neurobiology and Treatment of Mood Disorders, National Institute of Mental Health, National Institutes of Health, Bethesda, MD, USA
| | - Carlos A Zarate
- Section on the Neurobiology and Treatment of Mood Disorders, National Institute of Mental Health, National Institutes of Health, Bethesda, MD, USA
| |
Collapse
|
26
|
Inserra A, De Gregorio D, Gobbi G. Psychedelics in Psychiatry: Neuroplastic, Immunomodulatory, and Neurotransmitter Mechanisms. Pharmacol Rev 2021; 73:202-277. [PMID: 33328244 DOI: 10.1124/pharmrev.120.000056] [Citation(s) in RCA: 118] [Impact Index Per Article: 29.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023] Open
Abstract
Mounting evidence suggests safety and efficacy of psychedelic compounds as potential novel therapeutics in psychiatry. Ketamine has been approved by the Food and Drug Administration in a new class of antidepressants, and 3,4-methylenedioxymethamphetamine (MDMA) is undergoing phase III clinical trials for post-traumatic stress disorder. Psilocybin and lysergic acid diethylamide (LSD) are being investigated in several phase II and phase I clinical trials. Hence, the concept of psychedelics as therapeutics may be incorporated into modern society. Here, we discuss the main known neurobiological therapeutic mechanisms of psychedelics, which are thought to be mediated by the effects of these compounds on the serotonergic (via 5-HT2A and 5-HT1A receptors) and glutamatergic [via N-methyl-d-aspartate (NMDA) and α-amino-3-hydroxy-5-methyl-4-isoxazolepropionic acid (AMPA) receptors] systems. We focus on 1) neuroplasticity mediated by the modulation of mammalian target of rapamycin-, brain-derived neurotrophic factor-, and early growth response-related pathways; 2) immunomodulation via effects on the hypothalamic-pituitary-adrenal axis, nuclear factor ĸB, and cytokines such as tumor necrosis factor-α and interleukin 1, 6, and 10 production and release; and 3) modulation of serotonergic, dopaminergic, glutamatergic, GABAergic, and norepinephrinergic receptors, transporters, and turnover systems. We discuss arising concerns and ways to assess potential neurobiological changes, dependence, and immunosuppression. Although larger cohorts are required to corroborate preliminary findings, the results obtained so far are promising and represent a critical opportunity for improvement of pharmacotherapies in psychiatry, an area that has seen limited therapeutic advancement in the last 20 years. Studies are underway that are trying to decouple the psychedelic effects from the therapeutic effects of these compounds. SIGNIFICANCE STATEMENT: Psychedelic compounds are emerging as potential novel therapeutics in psychiatry. However, understanding of molecular mechanisms mediating improvement remains limited. This paper reviews the available evidence concerning the effects of psychedelic compounds on pathways that modulate neuroplasticity, immunity, and neurotransmitter systems. This work aims to be a reference for psychiatrists who may soon be faced with the possibility of prescribing psychedelic compounds as medications, helping them assess which compound(s) and regimen could be most useful for decreasing specific psychiatric symptoms.
Collapse
Affiliation(s)
- Antonio Inserra
- Neurobiological Psychiatry Unit, Department of Psychiatry, McGill University, Montreal, Quebec, Canada
| | - Danilo De Gregorio
- Neurobiological Psychiatry Unit, Department of Psychiatry, McGill University, Montreal, Quebec, Canada
| | - Gabriella Gobbi
- Neurobiological Psychiatry Unit, Department of Psychiatry, McGill University, Montreal, Quebec, Canada
| |
Collapse
|
27
|
Herian M, Wojtas A, Sobocińska MK, Skawski M, González-Marín A, Gołembiowska K. Contribution of serotonin receptor subtypes to hallucinogenic activity of 25I-NBOMe and to its effect on neurotransmission. Pharmacol Rep 2020; 72:1593-1603. [PMID: 33174181 PMCID: PMC7704505 DOI: 10.1007/s43440-020-00181-4] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/31/2020] [Revised: 10/19/2020] [Accepted: 10/22/2020] [Indexed: 01/13/2023]
Abstract
BACKGROUND 4-Iodo-2,5-dimethoxy-N-(2-methoxybenzyl)phenethylamine (25I-NBOMe) is a potent serotonin (5-HT) receptor agonist with hallucinogenic properties. The aim of our research was to examine the role of the 5-HT2A, 5-HT2C and 5-HT1A serotonin receptor subtypes in 25I-NBOMe hallucinogenic activity and its effect on dopamine (DA), 5-HT and glutamate release in the rat frontal cortex. METHODS Hallucinogenic activity was investigated using the wet dog shake (WDS) test. The release of DA, 5-HT and glutamate in the rat frontal cortex was studied using a microdialysis in freely moving rats. Neurotransmitter levels were analyzed by HPLC with electrochemical detection. The selective antagonists of the 5-HT2A, 5-HT2C and 5-HT1A serotonin receptor subtypes: M100907, SB242084 and WAY100635, respectively were applied through a microdialysis probe. RESULTS The WDS response to 25I-NBOMe (1 and 3 mg/kg) was significantly reduced by local administration of M100907 and SB242084 (100 nM). The 25I-NBOMe-induced increase in glutamate, DA and 5-HT release was inhibited by M100907 and SB242084. WAY100635 had no effect on 25I-NBOMe-induced WDS and glutamate release, while it decreased DA and 5-HT release from cortical neuronal terminals. CONCLUSION The obtained results suggest that 5-HT2A and 5-HT2C receptors play a role in 25I-NBOMe-induced hallucinogenic activity and in glutamate, DA and 5-HT release in the rat frontal cortex as their respective antagonists attenuated the effect of this hallucinogen. The disinhibition of GABA cells by the 5-HT1A receptor antagonist seems to underlie the mechanism of decreased DA and 5-HT release from neuronal terminals in the frontal cortex.
Collapse
MESH Headings
- Animals
- Dimethoxyphenylethylamine/analogs & derivatives
- Dimethoxyphenylethylamine/pharmacology
- Dopamine/metabolism
- Frontal Lobe/drug effects
- Frontal Lobe/metabolism
- Glutamic Acid/metabolism
- Hallucinogens/pharmacology
- Male
- Microdialysis
- Rats
- Rats, Wistar
- Receptor, Serotonin, 5-HT1A/drug effects
- Receptor, Serotonin, 5-HT1A/metabolism
- Receptor, Serotonin, 5-HT2A/drug effects
- Receptor, Serotonin, 5-HT2A/metabolism
- Receptor, Serotonin, 5-HT2C/drug effects
- Receptor, Serotonin, 5-HT2C/metabolism
- Serotonin/metabolism
- Serotonin Receptor Agonists/pharmacology
- Synaptic Transmission/drug effects
Collapse
Affiliation(s)
- Monika Herian
- Department of Pharmacology, Maj Institute of Pharmacology, Polish Academy of Sciences, 12 Smętna, 31-343, Kraków, Poland
| | - Adam Wojtas
- Department of Pharmacology, Maj Institute of Pharmacology, Polish Academy of Sciences, 12 Smętna, 31-343, Kraków, Poland
| | | | - Mateusz Skawski
- Department of Pharmacology, Maj Institute of Pharmacology, Polish Academy of Sciences, 12 Smętna, 31-343, Kraków, Poland
| | - Alejandro González-Marín
- Department of Pharmacology, Maj Institute of Pharmacology, Polish Academy of Sciences, 12 Smętna, 31-343, Kraków, Poland
| | - Krystyna Gołembiowska
- Department of Pharmacology, Maj Institute of Pharmacology, Polish Academy of Sciences, 12 Smętna, 31-343, Kraków, Poland.
| |
Collapse
|
28
|
Mason NL, Kuypers KPC, Müller F, Reckweg J, Tse DHY, Toennes SW, Hutten NRPW, Jansen JFA, Stiers P, Feilding A, Ramaekers JG. Me, myself, bye: regional alterations in glutamate and the experience of ego dissolution with psilocybin. Neuropsychopharmacology 2020; 45:2003-2011. [PMID: 32446245 PMCID: PMC7547711 DOI: 10.1038/s41386-020-0718-8] [Citation(s) in RCA: 138] [Impact Index Per Article: 27.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/30/2020] [Accepted: 05/14/2020] [Indexed: 01/21/2023]
Abstract
There is growing interest in the therapeutic utility of psychedelic substances, like psilocybin, for disorders characterized by distortions of the self-experience, like depression. Accumulating preclinical evidence emphasizes the role of the glutamate system in the acute action of the drug on brain and behavior; however this has never been tested in humans. Following a double-blind, placebo-controlled, parallel group design, we utilized an ultra-high field multimodal brain imaging approach and demonstrated that psilocybin (0.17 mg/kg) induced region-dependent alterations in glutamate, which predicted distortions in the subjective experience of one's self (ego dissolution). Whereas higher levels of medial prefrontal cortical glutamate were associated with negatively experienced ego dissolution, lower levels in hippocampal glutamate were associated with positively experienced ego dissolution. Such findings provide further insights into the underlying neurobiological mechanisms of the psychedelic, as well as the baseline, state. Importantly, they may also provide a neurochemical basis for therapeutic effects as witnessed in ongoing clinical trials.
Collapse
Affiliation(s)
- N L Mason
- Department of Neuropsychology and Psychopharmacology, Faculty of Psychology and Neuroscience, Maastricht University, P.O. Box 616, 6200 MD, Maastricht, the Netherlands.
| | - K P C Kuypers
- Department of Neuropsychology and Psychopharmacology, Faculty of Psychology and Neuroscience, Maastricht University, P.O. Box 616, 6200 MD, Maastricht, the Netherlands
| | - F Müller
- Department of Neuropsychology and Psychopharmacology, Faculty of Psychology and Neuroscience, Maastricht University, P.O. Box 616, 6200 MD, Maastricht, the Netherlands
- Department of Psychiatry (UPK), University of Basel, Basel, Switzerland
| | - J Reckweg
- Department of Neuropsychology and Psychopharmacology, Faculty of Psychology and Neuroscience, Maastricht University, P.O. Box 616, 6200 MD, Maastricht, the Netherlands
| | - D H Y Tse
- Department of Neuropsychology and Psychopharmacology, Faculty of Psychology and Neuroscience, Maastricht University, P.O. Box 616, 6200 MD, Maastricht, the Netherlands
| | - S W Toennes
- Institute of Legal Medicine, University of Frankfurt, Kennedyallee 104, D-60596, Frankfurt/Main, Germany
| | - N R P W Hutten
- Department of Neuropsychology and Psychopharmacology, Faculty of Psychology and Neuroscience, Maastricht University, P.O. Box 616, 6200 MD, Maastricht, the Netherlands
| | - J F A Jansen
- Department of Radiology and Nuclear Medicine, Maastricht University Medical Center+ (MUMC+), Maastricht, the Netherlands
- School for Mental Health and Neuroscience, Maastricht University Medical Center, P. Debyelaan 25, Maastricht, the Netherlands
| | - P Stiers
- Department of Neuropsychology and Psychopharmacology, Faculty of Psychology and Neuroscience, Maastricht University, P.O. Box 616, 6200 MD, Maastricht, the Netherlands
| | - A Feilding
- The Beckley Foundation, Beckley Park, Oxford, OX3 9SY, UK
| | - J G Ramaekers
- Department of Neuropsychology and Psychopharmacology, Faculty of Psychology and Neuroscience, Maastricht University, P.O. Box 616, 6200 MD, Maastricht, the Netherlands.
| |
Collapse
|
29
|
Psychedelic drugs: neurobiology and potential for treatment of psychiatric disorders. Nat Rev Neurosci 2020; 21:611-624. [PMID: 32929261 DOI: 10.1038/s41583-020-0367-2] [Citation(s) in RCA: 245] [Impact Index Per Article: 49.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 08/07/2020] [Indexed: 12/13/2022]
Abstract
Renewed interest in the use of psychedelics in the treatment of psychiatric disorders warrants a better understanding of the neurobiological mechanisms underlying the effects of these substances. After a hiatus of about 50 years, state-of-the art studies have recently begun to close important knowledge gaps by elucidating the mechanisms of action of psychedelics with regard to their effects on receptor subsystems, systems-level brain activity and connectivity, and cognitive and emotional processing. In addition, functional studies have shown that changes in self-experience, emotional processing and social cognition may contribute to the potential therapeutic effects of psychedelics. These discoveries provide a scientific road map for the investigation and application of psychedelic substances in psychiatry.
Collapse
|
30
|
Barrett FS, Krimmel SR, Griffiths RR, Seminowicz DA, Mathur BN. Psilocybin acutely alters the functional connectivity of the claustrum with brain networks that support perception, memory, and attention. Neuroimage 2020; 218:116980. [PMID: 32454209 PMCID: PMC10792549 DOI: 10.1016/j.neuroimage.2020.116980] [Citation(s) in RCA: 100] [Impact Index Per Article: 20.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/29/2020] [Accepted: 05/18/2020] [Indexed: 02/06/2023] Open
Abstract
Psychedelic drugs, including the serotonin 2a (5-HT2A) receptor partial agonist psilocybin, are receiving renewed attention for their possible efficacy in treating a variety of neuropsychiatric disorders. Psilocybin induces widespread dysregulation of cortical activity, but circuit-level mechanisms underlying this effect are unclear. The claustrum is a subcortical nucleus that highly expresses 5-HT2A receptors and provides glutamatergic inputs to arguably all areas of the cerebral cortex. We therefore tested the hypothesis that psilocybin modulates claustrum function in humans. Fifteen healthy participants (10M, 5F) completed this within-subjects study in which whole-brain resting-state blood-oxygenation level-dependent (BOLD) signal was measured 100 min after blinded oral administration of placebo and 10 mg/70 kg psilocybin. Left and right claustrum signal was isolated using small region confound correction. Psilocybin significantly decreased both the amplitude of low frequency fluctuations as well as the variance of BOLD signal in the left and right claustrum. Psilocybin also significantly decreased functional connectivity of the right claustrum with auditory and default mode networks (DMN), increased right claustrum connectivity with the fronto-parietal task control network (FPTC), and decreased left claustrum connectivity with the FPTC. DMN integrity was associated with right-claustrum connectivity with the DMN, while FPTC integrity and modularity were associated with right claustrum and left claustrum connectivity with the FPTC, respectively. Subjective effects of psilocybin predicted changes in the amplitude of low frequency fluctuations and the variance of BOLD signal in the left and right claustrum. Observed effects were specific to claustrum, compared to flanking regions of interest (the left and right insula and putamen). This study used a pharmacological intervention to provide the first empirical evidence in any species for a significant role of 5-HT2A receptor signaling in claustrum functioning, and supports a possible role of the claustrum in the subjective and therapeutic effects of psilocybin.
Collapse
Affiliation(s)
- Frederick S Barrett
- Department of Psychiatry and Behavioral Sciences, Johns Hopkins University School of Medicine, Baltimore, MD, 21224, USA; Center for Psychedelic and Consciousness Research, Johns Hopkins University School of Medicine, Baltimore, MD, 21224, USA.
| | - Samuel R Krimmel
- Department of Neural and Pain Sciences, School of Dentistry, and Center to Advance Chronic Pain Research, University of Maryland, Baltimore, MD, 21201, USA
| | - Roland R Griffiths
- Department of Psychiatry and Behavioral Sciences, Johns Hopkins University School of Medicine, Baltimore, MD, 21224, USA; Center for Psychedelic and Consciousness Research, Johns Hopkins University School of Medicine, Baltimore, MD, 21224, USA; Department of Neuroscience, Johns Hopkins University School of Medicine, Baltimore, MD, 21224, USA
| | - David A Seminowicz
- Department of Neural and Pain Sciences, School of Dentistry, and Center to Advance Chronic Pain Research, University of Maryland, Baltimore, MD, 21201, USA
| | - Brian N Mathur
- Department of Pharmacology, School of Medicine, University of Maryland, Baltimore, MD, 21201, USA
| |
Collapse
|
31
|
Rantamäki T, Kohtala S. Encoding, Consolidation, and Renormalization in Depression: Synaptic Homeostasis, Plasticity, and Sleep Integrate Rapid Antidepressant Effects. Pharmacol Rev 2020; 72:439-465. [PMID: 32139613 DOI: 10.1124/pr.119.018697] [Citation(s) in RCA: 26] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 03/21/2025] Open
Abstract
Recent studies have strived to find an association between rapid antidepressant effects and a specific subset of pharmacological targets and molecular pathways. Here, we propose a broader hypothesis of encoding, consolidation, and renormalization in depression (ENCORE-D), which suggests that, fundamentally, rapid and sustained antidepressant effects rely on intrinsic homeostatic mechanisms evoked as a response to the acute pharmacological or physiologic effects triggered by the treatment. We review evidence that supports the notion that various treatments with a rapid onset of action, such as ketamine, electroconvulsive therapy, and sleep deprivation, share the ability to acutely excite cortical networks, which increases synaptic potentiation, alters patterns of functional connectivity, and ameliorates depressive symptoms. We proceed to examine how the initial effects are short-lived and, as such, require both consolidation during wake and maintenance throughout sleep to remain sustained. Here, we incorporate elements from the synaptic homeostasis hypothesis and theorize that the fundamental mechanisms of synaptic plasticity and sleep, particularly the homeostatic emergence of slow-wave electroencephalogram activity and the renormalization of synaptic strength, are at the center of sustained antidepressant effects. We conclude by discussing the various implications of the ENCORE-D hypothesis and offer several considerations for future experimental and clinical research. SIGNIFICANCE STATEMENT: Proposed molecular perspectives of rapid antidepressant effects fail to appreciate the temporal distribution of the effects of ketamine on cortical excitation and plasticity as well as the prolonged influence on depressive symptoms. The encoding, consolidation, and renormalization in depression hypothesis proposes that the lasting clinical effects can be best explained by adaptive functional and structural alterations in neural circuitries set in motion in response to the acute pharmacological effects of ketamine (i.e., changes evoked during the engagement of receptor targets such as N-methyl-D-aspartate receptors) or other putative rapid-acting antidepressants. The present hypothesis opens a completely new avenue for conceptualizing and targeting brain mechanisms that are important for antidepressant effects wherein sleep and synaptic homeostasis are at the center stage.
Collapse
Affiliation(s)
- Tomi Rantamäki
- Laboratory of Neurotherapeutics, Drug Research Program, Division of Pharmacology and Pharmacotherapy, Faculty of Pharmacy (T.R., S.K.) and SleepWell Research Program, Faculty of Medicine (T.R., S.K.), University of Helsinki, Helsinki, Finland
| | - Samuel Kohtala
- Laboratory of Neurotherapeutics, Drug Research Program, Division of Pharmacology and Pharmacotherapy, Faculty of Pharmacy (T.R., S.K.) and SleepWell Research Program, Faculty of Medicine (T.R., S.K.), University of Helsinki, Helsinki, Finland
| |
Collapse
|
32
|
Zawilska JB, Kacela M, Adamowicz P. NBOMes-Highly Potent and Toxic Alternatives of LSD. Front Neurosci 2020; 14:78. [PMID: 32174803 PMCID: PMC7054380 DOI: 10.3389/fnins.2020.00078] [Citation(s) in RCA: 43] [Impact Index Per Article: 8.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/26/2019] [Accepted: 01/20/2020] [Indexed: 12/29/2022] Open
Abstract
Recently, a new class of psychedelic compounds named NBOMe (or 25X-NBOMe) has appeared on the illegal drug market. NBOMes are analogs of the 2C family of phenethylamine drugs, originally synthesized by Alexander Shulgin, that contain a N-(2-methoxy)benzyl substituent. The most frequently reported drugs from this group are 25I-NBOMe, 25B-NBOMe, and 25C-NBOMe. NBOMe compounds are ultrapotent and highly efficacious agonists of serotonin 5-HT2A and 5-HT2C receptors (Ki values in low nanomolar range) with more than 1000-fold selectivity for 5-HT2A compared with 5-HT1A. They display higher affinity for 5-HT2A receptors than their 2C counterparts and have markedly lower affinity, potency, and efficacy at the 5-HT2B receptor compared to 5-HT2A or 5-HT2C. The drugs are sold as blotter papers, or in powder, liquid, or tablet form, and they are administered sublingually/buccally, intravenously, via nasal insufflations, or by smoking. Since their introduction in the early 2010s, numerous reports have been published on clinical intoxications and fatalities resulting from the consumption of NBOMe compounds. Commonly observed adverse effects include visual and auditory hallucinations, confusion, anxiety, panic and fear, agitation, uncontrollable violent behavior, seizures, excited delirium, and sympathomimetic signs such mydriasis, tachycardia, hypertension, hyperthermia, and diaphoresis. Rhabdomyolysis, disseminated intravascular coagulation, hypoglycemia, metabolic acidosis, and multiorgan failure were also reported. This survey provides an updated overview of the pharmacological properties, pattern of use, metabolism, and desired effects associated with NBOMe use. Special emphasis is given to cases of non-fatal and lethal intoxication involving these compounds. As the analysis of NBOMes in biological materials can be challenging even for laboratories applying modern sensitive techniques, this paper also presents the analytical methods most commonly used for detection and identification of NBOMes and their metabolites.
Collapse
Affiliation(s)
- Jolanta B Zawilska
- Department of Pharmacodynamics, Medical University of Łódź, Łódź, Poland
| | - Monika Kacela
- Department of Pharmacodynamics, Medical University of Łódź, Łódź, Poland
| | - Piotr Adamowicz
- Department of Forensic Toxicology, Institute of Forensic Research, Kraków, Poland
| |
Collapse
|
33
|
Serotonin 2A receptors are a stress response system: implications for post-traumatic stress disorder. Behav Pharmacol 2020; 30:151-162. [PMID: 30632995 DOI: 10.1097/fbp.0000000000000459] [Citation(s) in RCA: 41] [Impact Index Per Article: 8.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/31/2022]
Abstract
Serotonin, one of the first neurotransmitters to be identified, is an evolutionarily old molecule that is highly conserved across the animal kingdom, and widely used throughout the brain. Despite this, ascribing a specific set of functions to brain serotonin and its receptors has been difficult and controversial. The 2A subtype of serotonin receptors (5-HT2A receptor) is the major excitatory serotonin receptor in the brain and has been linked to the effects of drugs that produce profound sensory and cognitive changes. Numerous studies have shown that this receptor is upregulated by a broad variety of stressors, and have related 5-HT2A receptor function to associative learning. This review proposes that stress, particularly stress related to danger and existential threats, increases the expression and function of 5-HT2A receptors. It is argued that this is a neurobiological adaptation to promote learning and avoidance of danger in the future. Upregulation of 5-HT2A receptors during stressful events forms associations that tune the brain to environmental cues that signal danger. It is speculated that life-threatening situations may activate this system and contribute to the symptoms associated with post-traumatic stress disorder (PTSD). 3,4-Methylenedioxymethamphetamine, which activates 5-HT2A receptors, has been successful in the treatment of PTSD and has recently achieved status as a breakthrough therapy. An argument is presented that 3,4-methylenedioxymethamphetamine may paradoxically act through these same 5-HT2A receptors to ameliorate the symptoms of PTSD. The central thematic contention is that a key role of serotonin may be to function as a stress detection and response system.
Collapse
|
34
|
Effects of the synthetic psychedelic 2,5-dimethoxy-4-iodoamphetamine (DOI) on ethanol consumption and place conditioning in male mice. Psychopharmacology (Berl) 2019; 236:3567-3578. [PMID: 31309240 PMCID: PMC6895420 DOI: 10.1007/s00213-019-05328-7] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/07/2018] [Accepted: 07/08/2019] [Indexed: 12/27/2022]
Abstract
RATIONALE Approximately 20 million adults in the USA have an alcohol use disorder (AUD). There are clinical and preclinical data suggesting that psychedelics may have benefits for AUD. OBJECTIVE To investigate the effects of the synthetic psychedelic 2,5-dimethoxy-4-iodoamphetamine (DOI) on the behavioral effects of ethanol. METHODS The effects of DOI were examined using ethanol-induced place conditioning (1.8 g/kg ethanol) and 2-bottle choice ethanol drinking (20% v/v), using a dose of DOI (3 mg/kg) that produced the maximal response in the serotonin 2A (5-HT2A) receptor-dependent head-twitch assay. Interactions between DOI and ethanol (3 g/kg) were examined using the ethanol-induced loss of righting reflex procedure and blood-ethanol analysis. To examine additional mechanisms by which psychedelics may interact with ethanol, we determined whether DOI reverses ethanol-induced nitric oxide release in macrophages, a marker of inflammation. RESULTS DOI significantly attenuated ethanol-induced place conditioning and ethanol drinking. DOI-induced suppression of alcohol drinking depended upon 5-HT2A receptors, was selective for alcohol over water, and was selective for high alcohol-preferring subjects. DOI had no apparent pharmacokinetic interactions with ethanol, and DOI reduced ethanol-induced nitric oxide release. CONCLUSIONS Our findings demonstrate that DOI blocks ethanol place conditioning and selectively reduces voluntary ethanol consumption. This may be related to modulation of the effects of ethanol in the reward circuitry of the brain, ethanol-induced neuroinflammation, or a combination of both. Additional studies to elucidate the mechanisms through which psychedelics attenuate the effects of ethanol would inform the pathophysiology of AUD and potentially provide new treatment options.
Collapse
|
35
|
Canal CE. Serotonergic Psychedelics: Experimental Approaches for Assessing Mechanisms of Action. Handb Exp Pharmacol 2019; 252:227-260. [PMID: 29532180 PMCID: PMC6136989 DOI: 10.1007/164_2018_107] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/23/2023]
Abstract
Recent, well-controlled - albeit small-scale - clinical trials show that serotonergic psychedelics, including psilocybin and lysergic acid diethylamide, possess great promise for treating psychiatric disorders, including treatment-resistant depression. Additionally, fresh results from a deluge of clinical neuroimaging studies are unveiling the dynamic effects of serotonergic psychedelics on functional activity within, and connectivity across, discrete neural systems. These observations have led to testable hypotheses regarding neural processing mechanisms that contribute to psychedelic effects and therapeutic benefits. Despite these advances and a plethora of preclinical and clinical observations supporting a central role for brain serotonin 5-HT2A receptors in producing serotonergic psychedelic effects, lingering and new questions about mechanisms abound. These chiefly pertain to molecular neuropharmacology. This chapter is devoted to illuminating and discussing such questions in the context of preclinical experimental approaches for studying mechanisms of action of serotonergic psychedelics, classic and new.
Collapse
Affiliation(s)
- Clinton E Canal
- Department of Pharmaceutical Sciences, College of Pharmacy, Mercer University, Atlanta, GA, USA.
| |
Collapse
|
36
|
Herian M, Wojtas A, Kamińska K, Świt P, Wach A, Gołembiowska K. Hallucinogen-Like Action of the Novel Designer Drug 25I-NBOMe and Its Effect on Cortical Neurotransmitters in Rats. Neurotox Res 2019; 36:91-100. [PMID: 30989482 PMCID: PMC6570696 DOI: 10.1007/s12640-019-00033-x] [Citation(s) in RCA: 28] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/15/2018] [Revised: 03/20/2019] [Accepted: 03/26/2019] [Indexed: 12/22/2022]
Abstract
NBOMes are N-benzylmethoxy derivatives of the 2C family hallucinogens. 4-Iodo-2,5-dimethoxy-N-(2-methoxybenzyl)phenethylamine (25I-NBOMe) is one of the commonly used illicit drugs. It exhibits high binding affinity for 5-HT2A/C and 5-HT1A serotonin receptors. Activation of 5-HT2A receptor induces head-twitch response (HTR) in rodents, a behavioral marker of hallucinogen effect in humans. There is not much data on neurochemical properties of NBOMes. Therefore, we aimed to investigate the effect of 25I-NBOMe on extracellular level of dopamine (DA), serotonin (5-HT), and glutamate (GLU) in the rat frontal cortex, tissue contents of monoamines, and hallucinogenic activity in rats. The extracellular levels of DA, 5-HT, and GLU were studied using microdialysis in freely moving animals. The tissue contents of DA, 5-HT and their metabolites 3,4-dihydroxyphenylacetic acid (DOPAC), homovanillic acid (HVA), and 5-hydroxyindoleacetic acid (5-HIAA) were determined in the rat frontal cortex. We also tested a drug-elicited HTR. 25I-NBOMe at doses 1, 3, and 10 mg/kg (sc) increased extracellular DA, 5-HT, and GLU levels, enhanced tissue content of 5-HT and 5-HIAA, but did not affect tissue level of DA and its metabolites. The compound exhibited an inverted U-shaped dose-response curve with respect to the effect on extracellular DA and 5-HT levels, but a U-shaped dose-response curve was observed for its effect on GLU release and HTR. The data from our study suggest that hallucinogenic activity of 25I-NBOMe seems to be related with the increase in extracellular GLU level-mediated via cortical 5-HT2A receptors. The influence of 25I-NBOMe on 5-HT2C and 5-HT1A receptors may modulate its effect on neurotransmitters and HTR.
Collapse
Affiliation(s)
- Monika Herian
- Department of Pharmacology, Institute of Pharmacology, Polish Academy of Sciences, 12 Smętna, 31-343, Kraków, Poland
| | - Adam Wojtas
- Department of Pharmacology, Institute of Pharmacology, Polish Academy of Sciences, 12 Smętna, 31-343, Kraków, Poland
| | - Katarzyna Kamińska
- Department of Pharmacology, Institute of Pharmacology, Polish Academy of Sciences, 12 Smętna, 31-343, Kraków, Poland
| | - Paweł Świt
- Department of Pharmacology, Institute of Pharmacology, Polish Academy of Sciences, 12 Smętna, 31-343, Kraków, Poland
| | - Anna Wach
- Department of Pharmacology, Institute of Pharmacology, Polish Academy of Sciences, 12 Smętna, 31-343, Kraków, Poland
| | - Krystyna Gołembiowska
- Department of Pharmacology, Institute of Pharmacology, Polish Academy of Sciences, 12 Smętna, 31-343, Kraków, Poland.
| |
Collapse
|
37
|
Taskiran M, Tasdemir A, Ayyildiz N, Ayyildiz M, Agar E. The effect of serotonin on penicillin-induced epileptiform activity. Int J Neurosci 2019; 129:687-697. [DOI: 10.1080/00207454.2018.1557166] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/16/2023]
Affiliation(s)
- Mehmet Taskiran
- Department of Biology, Faculty of Science, Erciyes University, Kayseri, Turkey
| | - Abdulkadir Tasdemir
- Graduate School of Natural and Applied Sciences, Erciyes University, Kayseri, Turkey
| | - Nusret Ayyildiz
- Department of Biology, Faculty of Science, Erciyes University, Kayseri, Turkey
| | - Mustafa Ayyildiz
- Department of Physiology, Faculty of Medicine, Ondokuz Mayıs University, Samsun, Turkey
| | - Erdal Agar
- Department of Physiology, Faculty of Medicine, Ondokuz Mayıs University, Samsun, Turkey
| |
Collapse
|
38
|
Benvenga MJ, Chaney SF, Baez M, Britton TC, Hornback WJ, Monn JA, Marek GJ. Metabotropic Glutamate 2 Receptors Play a Key Role in Modulating Head Twitches Induced by a Serotonergic Hallucinogen in Mice. Front Pharmacol 2018; 9:208. [PMID: 29599719 PMCID: PMC5862811 DOI: 10.3389/fphar.2018.00208] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/31/2017] [Accepted: 02/23/2018] [Indexed: 11/29/2022] Open
Abstract
There is substantial evidence that glutamate can modulate the effects of 5-hydroxytryptamine2A (5-HT2A) receptor activation through stimulation of metabotropic glutamate2/3 (mGlu2/3) receptors in the prefrontal cortex. Here we show that constitutive deletion of the mGlu2 gene profoundly attenuates an effect of 5-HT2A receptor activation using the mouse head twitch response (HTR). MGlu2 and mGlu3 receptor knockout (KO) as well as age-matched ICR (CD-1) wild type (WT) mice were administered (±)1-(2,5-dimethoxy-4-iodophenyl)-2-aminopropane (DOI) and observed for head twitch activity. DOI failed to produce significant head twitches in mGlu2 receptor KO mice at a dose 10-fold higher than the peak effective dose in WT or mGlu3 receptor KO mice. In addition, the mGlu2/3 receptor agonist LY379268, and the mGlu2 receptor positive allosteric modulator (PAM) CBiPES, potently blocked the HTR to DOI in WT and mGlu3 receptor KO mice. Conversely, the mGlu2/3 receptor antagonist LY341495 (10 mg/kg) increased the HTR produced by DOI (3 mg/kg) in mGlu3 receptor KO mice. Finally, the mGlu2 receptor potentiator CBiPES was able to attenuate the increase in the HTR produced by LY341495 in mGlu3 receptor KO mice. Taken together, all of these results are consistent with the hypothesis that that DOI-induced head twitches are modulated by mGlu2 receptor activation. These results also are in keeping with a critical autoreceptor function for mGlu2 receptors in the prefrontal cortex with differential effects of acute vs. chronic perturbation (e.g., constitutive mGlu2 receptor KO mice). The robust attenuation of DOI-induced head twitches in the mGlu2 receptor KO mice appears to reflect the critical role of glutamate in ongoing regulation of 5-HT2A receptors in the prefrontal cortex. Future experiments with inducible knockouts for the mGlu2 receptor and/or selective mGlu3 receptor agonists/PAMs/antagonists could provide an important tools in understanding glutamatergic modulation of prefrontal cortical 5-HT2A receptor function.
Collapse
Affiliation(s)
- Mark J Benvenga
- Neuroscience Discovery Research, Lilly Research Laboratories, Eli Lilly and Company, Indianapolis, IN, United States.,Lilly Corporate Center, Eli Lilly and Company, Indianapolis, IN, United States
| | - Stephen F Chaney
- Neuroscience Discovery Research, Lilly Research Laboratories, Eli Lilly and Company, Indianapolis, IN, United States
| | - Melvyn Baez
- Neuroscience Discovery Research, Lilly Research Laboratories, Eli Lilly and Company, Indianapolis, IN, United States
| | - Thomas C Britton
- Discovery Chemistry Research, Lilly Research Laboratories, Eli Lilly and Company, Indianapolis, IN, United States
| | - William J Hornback
- Discovery Chemistry Research, Lilly Research Laboratories, Eli Lilly and Company, Indianapolis, IN, United States
| | - James A Monn
- Discovery Chemistry Research, Lilly Research Laboratories, Eli Lilly and Company, Indianapolis, IN, United States
| | - Gerard J Marek
- Neuroscience Discovery Research, Lilly Research Laboratories, Eli Lilly and Company, Indianapolis, IN, United States
| |
Collapse
|
39
|
Buchborn T, Lyons T, Knöpfel T. Tolerance and Tachyphylaxis to Head Twitches Induced by the 5-HT2A Agonist 25CN-NBOH in Mice. Front Pharmacol 2018; 9:17. [PMID: 29467649 PMCID: PMC5808243 DOI: 10.3389/fphar.2018.00017] [Citation(s) in RCA: 32] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/19/2017] [Accepted: 01/08/2018] [Indexed: 12/20/2022] Open
Abstract
The serotonin (5-HT) 2A receptor is the primary molecular target of serotonergic hallucinogens, which trigger large-scale perturbations of the cortex. Our understanding of how 5-HT2A activation may cause the effects of hallucinogens has been hampered by the receptor unselectivity of most of the drugs of this class. Here we used 25CN-NBOH (N-(2-hydroxybenzyl)-2,5-dimethoxy-4-cyanophenylethylamine), a newly developed selective 5-HT2A agonist, and tested it with regard to the head-twitch-response (HTR) model of 5-HT2A activity and effects on locomotion. 25CN-NBOH evoked HTRs with an inverted u-shape-like dose-response curve and highest efficacy at 1.5 mg/kg, i.p. HTR occurrence peaked within 5 min after agonist injection, and exponentially decreased to half-maximal frequency at ~11 min. Thorough habituation to the experimental procedures (including handling, saline injection, and exposure to the observational boxes 1 day before the experiment) facilitated the animals' response to 25CN-NBOH. 25CN-NBOH (1.5 mg/kg, i.p.) induced HTRs were blocked by the 5-HT2A antagonist ketanserin (0.75 mg/kg, 30 min pre), but not by the 5-HT2C antagonist SB-242084 (0.5 mg/kg, i.p., 30 min pre). SB-242084 instead slightly increased the number of HTRs occurring at a 3.0-mg/kg dose of the agonist. Apart from HTR induction, 25CN-NBOH also modestly increased locomotor activity of the mice. Repeated once-per-day injections (1.5 mg/kg, i.p.) led to reduced occurrence of 25CN-NBOH induced HTRs. This intermediate tolerance was augmented when a second (higher) dose of the drug (3.0 mg/kg) was interspersed. Short-interval tolerance (i.e., tachyphylaxis) was observed when the drug was injected twice at intervals of 1.0 and 1.5 h at either dose tested (1.5 mg/kg and 0.75 mg/kg, respectively). Inducing ketanserin-sensitive HTRs, which are dependent on environmental valences and which show signs of tachyphylaxis and tolerance, 25CN-NBOH shares striking features common to serotonergic hallucinogens. Given its distinct in vitro selectivity for 5-HT2A over non5-HT2 receptors and its behavioral dynamics, 25CN-NBOH appears to be a powerful tool for dissection of receptor-specific cortical circuit dynamics, including 5-HT2A related psychoactivity.
Collapse
Affiliation(s)
- Tobias Buchborn
- Laboratory for Neuronal Circuit Dynamics, Department of Medicine, Imperial College London, London, United Kingdom
| | - Taylor Lyons
- Laboratory for Neuronal Circuit Dynamics, Department of Medicine, Imperial College London, London, United Kingdom
| | - Thomas Knöpfel
- Laboratory for Neuronal Circuit Dynamics, Department of Medicine, Imperial College London, London, United Kingdom.,Centre for Neurotechnology, Institute of Biomedical Engineering, Imperial College London, London, United Kingdom
| |
Collapse
|
40
|
Abstract
Because of the ethical and regulatory hurdles associated with human studies, much of what is known about the psychopharmacology of hallucinogens has been derived from animal models. However, developing reliable animal models has proven to be a challenging task due to the complexity and variability of hallucinogen effects in humans. This chapter focuses on three animal models that are frequently used to test the effects of hallucinogens on unconditioned behavior: head twitch response (HTR), prepulse inhibition of startle (PPI), and exploratory behavior. The HTR has demonstrated considerable utility in the neurochemical actions of hallucinogens. However, the latter two models have clearer conceptual bridges to human phenomenology. Consistent with the known mechanism of action of hallucinogens in humans, the behavioral effects of hallucinogens in rodents are mediated primarily by activation of 5-HT2A receptors. There is evidence, however, that other receptors may play secondary roles. The structure-activity relationships (SAR) of hallucinogens are reviewed in relation to each model, with a focus on the HTR in rats and mice.
Collapse
Affiliation(s)
- Adam L Halberstadt
- Department of Psychiatry, University of California San Diego, La Jolla, CA, 92093-0804, USA.
- Research Service, VA San Diego Healthcare System, San Diego, CA, USA.
| | - Mark A Geyer
- Department of Psychiatry, University of California San Diego, La Jolla, CA, 92093-0804, USA
- Research Service, VA San Diego Healthcare System, San Diego, CA, USA
| |
Collapse
|
41
|
Müller F, Lenz C, Dolder P, Lang U, Schmidt A, Liechti M, Borgwardt S. Increased thalamic resting-state connectivity as a core driver of LSD-induced hallucinations. Acta Psychiatr Scand 2017; 136:648-657. [PMID: 28940312 PMCID: PMC5698745 DOI: 10.1111/acps.12818] [Citation(s) in RCA: 97] [Impact Index Per Article: 12.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Accepted: 09/05/2017] [Indexed: 12/12/2022]
Abstract
OBJECTIVE It has been proposed that the thalamocortical system is an important site of action of hallucinogenic drugs and an essential component of the neural correlates of consciousness. Hallucinogenic drugs such as LSD can be used to induce profoundly altered states of consciousness, and it is thus of interest to test the effects of these drugs on this system. METHOD 100 μg LSD was administrated orally to 20 healthy participants prior to fMRI assessment. Whole brain thalamic functional connectivity was measured using ROI-to-ROI and ROI-to-voxel approaches. Correlation analyses were used to explore relationships between thalamic connectivity to regions involved in auditory and visual hallucinations and subjective ratings on auditory and visual drug effects. RESULTS LSD caused significant alterations in all dimensions of the 5D-ASC scale and significantly increased thalamic functional connectivity to various cortical regions. Furthermore, LSD-induced functional connectivity measures between the thalamus and the right fusiform gyrus and insula correlated significantly with subjective auditory and visual drug effects. CONCLUSION Hallucinogenic drug effects might be provoked by facilitations of cortical excitability via thalamocortical interactions. Our findings have implications for the understanding of the mechanism of action of hallucinogenic drugs and provide further insight into the role of the 5-HT2A -receptor in altered states of consciousness.
Collapse
Affiliation(s)
- F. Müller
- Department of Psychiatry (UPK)University of BaselBaselSwitzerland
| | - C. Lenz
- Department of Psychiatry (UPK)University of BaselBaselSwitzerland
| | - P. Dolder
- Division of Clinical Pharmacology and ToxicologyDepartment of Biomedicine and Department of Clinical ResearchUniversity Hospital BaselUniversity of BaselBaselSwitzerland
| | - U. Lang
- Department of Psychiatry (UPK)University of BaselBaselSwitzerland
| | - A. Schmidt
- Department of Psychiatry (UPK)University of BaselBaselSwitzerland
| | - M. Liechti
- Division of Clinical Pharmacology and ToxicologyDepartment of Biomedicine and Department of Clinical ResearchUniversity Hospital BaselUniversity of BaselBaselSwitzerland
| | - S. Borgwardt
- Department of Psychiatry (UPK)University of BaselBaselSwitzerland
| |
Collapse
|
42
|
Sampedro F, de la Fuente Revenga M, Valle M, Roberto N, Domínguez-Clavé E, Elices M, Luna LE, Crippa JAS, Hallak JEC, de Araujo DB, Friedlander P, Barker SA, Álvarez E, Soler J, Pascual JC, Feilding A, Riba J. Assessing the Psychedelic "After-Glow" in Ayahuasca Users: Post-Acute Neurometabolic and Functional Connectivity Changes Are Associated with Enhanced Mindfulness Capacities. Int J Neuropsychopharmacol 2017; 20:698-711. [PMID: 28525587 PMCID: PMC5581489 DOI: 10.1093/ijnp/pyx036] [Citation(s) in RCA: 100] [Impact Index Per Article: 12.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/04/2016] [Accepted: 05/17/2017] [Indexed: 12/12/2022] Open
Abstract
Background Ayahuasca is a plant tea containing the psychedelic 5-HT2A agonist N,N-dimethyltryptamine and harmala monoamine-oxidase inhibitors. Acute administration leads to neurophysiological modifications in brain regions of the default mode network, purportedly through a glutamatergic mechanism. Post-acutely, ayahuasca potentiates mindfulness capacities in volunteers and induces rapid and sustained antidepressant effects in treatment-resistant patients. However, the mechanisms underlying these fast and maintained effects are poorly understood. Here, we investigated in an open-label uncontrolled study in 16 healthy volunteers ayahuasca-induced post-acute neurometabolic and connectivity modifications and their association with mindfulness measures. Methods Using 1H-magnetic resonance spectroscopy and functional connectivity, we compared baseline and post-acute neurometabolites and seed-to-voxel connectivity in the posterior and anterior cingulate cortex after a single ayahuasca dose. Results Magnetic resonance spectroscopy showed post-acute reductions in glutamate+glutamine, creatine, and N-acetylaspartate+N-acetylaspartylglutamate in the posterior cingulate cortex. Connectivity was increased between the posterior cingulate cortex and the anterior cingulate cortex, and between the anterior cingulate cortex and limbic structures in the right medial temporal lobe. Glutamate+glutamine reductions correlated with increases in the "nonjudging" subscale of the Five Facets Mindfulness Questionnaire. Increased anterior cingulate cortex-medial temporal lobe connectivity correlated with increased scores on the self-compassion questionnaire. Post-acute neural changes predicted sustained elevations in nonjudging 2 months later. Conclusions These results support the involvement of glutamate neurotransmission in the effects of psychedelics in humans. They further suggest that neurometabolic changes in the posterior cingulate cortex, a key region within the default mode network, and increased connectivity between the anterior cingulate cortex and medial temporal lobe structures involved in emotion and memory potentially underlie the post-acute psychological effects of ayahuasca.
Collapse
Affiliation(s)
- Frederic Sampedro
- School of Medicine, Autonomous University of Barcelona, Barcelona, Spain (Mr Sampedro); Human Neuropsychopharmacology Research Group, Sant Pau Institute of Biomedical Research, Barcelona, Spain (Dr de la Fuente Revenga, Ms Roberto, and Dr Riba); Pharmacokinetic and Pharmacodynamic Modelling and Simulation, Sant Pau Institute of Biomedical Research, Barcelona, Spain (Dr Valle); Centre d’Investigació de Medicaments, Servei de Farmacologia Clínica, Hospital de la Santa Creu i Sant Pau, Barcelona, Spain (Drs Valle and Riba); Centro de Investigación Biomédica en Red de Salud Mental, CIBERSAM, Spain (Drs Valle, Elices, Álvarez, Soler, Pascual, and Riba); Department of Pharmacology and Therapeutics, Autonomous University of Barcelona, Barcelona, Spain (Dr Valle); Department of Psychiatry, Hospital de la Santa Creu i Sant Pau, Barcelona, Spain (Ms Domínguez-Clavé and Drs Elices, Álvarez, Soler, and Pascual); Department of Psychiatry and Forensic Medicine, School of Medicine, Universitat Autònoma de Barcelona, Barcelona, Spain (Ms Domínguez-Clavé and Drs Elices, Álvarez, and Pascual); Research Center for the Study of Psychointegrator Plants, Visionary Art and Consciousness, Florianópolis, Santa Catarina, Brazil (Dr Luna); Department of Neuroscience and Behavior, Medical School of Ribeirão Preto, University of São Paulo, São Paulo, Brazil and National Institute for Translational Medicine, Ribeirão Preto, Brazil (Drs Crippa and Hallak); Brain Institute/Hospital Universitario Onofre Lopes, Natal, Brazil (Dr de Araujo); The Beckley Foundation, Beckley Park, Oxford, United Kingdom (Mr Friedlander and Mrs Feilding); Department of Comparative Biomedical Sciences, School of Veterinary Medicine, Louisiana State University, Skip Bertman Drive at River Road, Baton Rouge, Louisiana (Dr Barker); Department of Clinical and Health Psychology, School of Psychology, Autonomous University of Barcelona, Barcelona, Spain (Dr Soler)
| | - Mario de la Fuente Revenga
- School of Medicine, Autonomous University of Barcelona, Barcelona, Spain (Mr Sampedro); Human Neuropsychopharmacology Research Group, Sant Pau Institute of Biomedical Research, Barcelona, Spain (Dr de la Fuente Revenga, Ms Roberto, and Dr Riba); Pharmacokinetic and Pharmacodynamic Modelling and Simulation, Sant Pau Institute of Biomedical Research, Barcelona, Spain (Dr Valle); Centre d’Investigació de Medicaments, Servei de Farmacologia Clínica, Hospital de la Santa Creu i Sant Pau, Barcelona, Spain (Drs Valle and Riba); Centro de Investigación Biomédica en Red de Salud Mental, CIBERSAM, Spain (Drs Valle, Elices, Álvarez, Soler, Pascual, and Riba); Department of Pharmacology and Therapeutics, Autonomous University of Barcelona, Barcelona, Spain (Dr Valle); Department of Psychiatry, Hospital de la Santa Creu i Sant Pau, Barcelona, Spain (Ms Domínguez-Clavé and Drs Elices, Álvarez, Soler, and Pascual); Department of Psychiatry and Forensic Medicine, School of Medicine, Universitat Autònoma de Barcelona, Barcelona, Spain (Ms Domínguez-Clavé and Drs Elices, Álvarez, and Pascual); Research Center for the Study of Psychointegrator Plants, Visionary Art and Consciousness, Florianópolis, Santa Catarina, Brazil (Dr Luna); Department of Neuroscience and Behavior, Medical School of Ribeirão Preto, University of São Paulo, São Paulo, Brazil and National Institute for Translational Medicine, Ribeirão Preto, Brazil (Drs Crippa and Hallak); Brain Institute/Hospital Universitario Onofre Lopes, Natal, Brazil (Dr de Araujo); The Beckley Foundation, Beckley Park, Oxford, United Kingdom (Mr Friedlander and Mrs Feilding); Department of Comparative Biomedical Sciences, School of Veterinary Medicine, Louisiana State University, Skip Bertman Drive at River Road, Baton Rouge, Louisiana (Dr Barker); Department of Clinical and Health Psychology, School of Psychology, Autonomous University of Barcelona, Barcelona, Spain (Dr Soler)
| | - Marta Valle
- School of Medicine, Autonomous University of Barcelona, Barcelona, Spain (Mr Sampedro); Human Neuropsychopharmacology Research Group, Sant Pau Institute of Biomedical Research, Barcelona, Spain (Dr de la Fuente Revenga, Ms Roberto, and Dr Riba); Pharmacokinetic and Pharmacodynamic Modelling and Simulation, Sant Pau Institute of Biomedical Research, Barcelona, Spain (Dr Valle); Centre d’Investigació de Medicaments, Servei de Farmacologia Clínica, Hospital de la Santa Creu i Sant Pau, Barcelona, Spain (Drs Valle and Riba); Centro de Investigación Biomédica en Red de Salud Mental, CIBERSAM, Spain (Drs Valle, Elices, Álvarez, Soler, Pascual, and Riba); Department of Pharmacology and Therapeutics, Autonomous University of Barcelona, Barcelona, Spain (Dr Valle); Department of Psychiatry, Hospital de la Santa Creu i Sant Pau, Barcelona, Spain (Ms Domínguez-Clavé and Drs Elices, Álvarez, Soler, and Pascual); Department of Psychiatry and Forensic Medicine, School of Medicine, Universitat Autònoma de Barcelona, Barcelona, Spain (Ms Domínguez-Clavé and Drs Elices, Álvarez, and Pascual); Research Center for the Study of Psychointegrator Plants, Visionary Art and Consciousness, Florianópolis, Santa Catarina, Brazil (Dr Luna); Department of Neuroscience and Behavior, Medical School of Ribeirão Preto, University of São Paulo, São Paulo, Brazil and National Institute for Translational Medicine, Ribeirão Preto, Brazil (Drs Crippa and Hallak); Brain Institute/Hospital Universitario Onofre Lopes, Natal, Brazil (Dr de Araujo); The Beckley Foundation, Beckley Park, Oxford, United Kingdom (Mr Friedlander and Mrs Feilding); Department of Comparative Biomedical Sciences, School of Veterinary Medicine, Louisiana State University, Skip Bertman Drive at River Road, Baton Rouge, Louisiana (Dr Barker); Department of Clinical and Health Psychology, School of Psychology, Autonomous University of Barcelona, Barcelona, Spain (Dr Soler)
| | - Natalia Roberto
- School of Medicine, Autonomous University of Barcelona, Barcelona, Spain (Mr Sampedro); Human Neuropsychopharmacology Research Group, Sant Pau Institute of Biomedical Research, Barcelona, Spain (Dr de la Fuente Revenga, Ms Roberto, and Dr Riba); Pharmacokinetic and Pharmacodynamic Modelling and Simulation, Sant Pau Institute of Biomedical Research, Barcelona, Spain (Dr Valle); Centre d’Investigació de Medicaments, Servei de Farmacologia Clínica, Hospital de la Santa Creu i Sant Pau, Barcelona, Spain (Drs Valle and Riba); Centro de Investigación Biomédica en Red de Salud Mental, CIBERSAM, Spain (Drs Valle, Elices, Álvarez, Soler, Pascual, and Riba); Department of Pharmacology and Therapeutics, Autonomous University of Barcelona, Barcelona, Spain (Dr Valle); Department of Psychiatry, Hospital de la Santa Creu i Sant Pau, Barcelona, Spain (Ms Domínguez-Clavé and Drs Elices, Álvarez, Soler, and Pascual); Department of Psychiatry and Forensic Medicine, School of Medicine, Universitat Autònoma de Barcelona, Barcelona, Spain (Ms Domínguez-Clavé and Drs Elices, Álvarez, and Pascual); Research Center for the Study of Psychointegrator Plants, Visionary Art and Consciousness, Florianópolis, Santa Catarina, Brazil (Dr Luna); Department of Neuroscience and Behavior, Medical School of Ribeirão Preto, University of São Paulo, São Paulo, Brazil and National Institute for Translational Medicine, Ribeirão Preto, Brazil (Drs Crippa and Hallak); Brain Institute/Hospital Universitario Onofre Lopes, Natal, Brazil (Dr de Araujo); The Beckley Foundation, Beckley Park, Oxford, United Kingdom (Mr Friedlander and Mrs Feilding); Department of Comparative Biomedical Sciences, School of Veterinary Medicine, Louisiana State University, Skip Bertman Drive at River Road, Baton Rouge, Louisiana (Dr Barker); Department of Clinical and Health Psychology, School of Psychology, Autonomous University of Barcelona, Barcelona, Spain (Dr Soler)
| | - Elisabet Domínguez-Clavé
- School of Medicine, Autonomous University of Barcelona, Barcelona, Spain (Mr Sampedro); Human Neuropsychopharmacology Research Group, Sant Pau Institute of Biomedical Research, Barcelona, Spain (Dr de la Fuente Revenga, Ms Roberto, and Dr Riba); Pharmacokinetic and Pharmacodynamic Modelling and Simulation, Sant Pau Institute of Biomedical Research, Barcelona, Spain (Dr Valle); Centre d’Investigació de Medicaments, Servei de Farmacologia Clínica, Hospital de la Santa Creu i Sant Pau, Barcelona, Spain (Drs Valle and Riba); Centro de Investigación Biomédica en Red de Salud Mental, CIBERSAM, Spain (Drs Valle, Elices, Álvarez, Soler, Pascual, and Riba); Department of Pharmacology and Therapeutics, Autonomous University of Barcelona, Barcelona, Spain (Dr Valle); Department of Psychiatry, Hospital de la Santa Creu i Sant Pau, Barcelona, Spain (Ms Domínguez-Clavé and Drs Elices, Álvarez, Soler, and Pascual); Department of Psychiatry and Forensic Medicine, School of Medicine, Universitat Autònoma de Barcelona, Barcelona, Spain (Ms Domínguez-Clavé and Drs Elices, Álvarez, and Pascual); Research Center for the Study of Psychointegrator Plants, Visionary Art and Consciousness, Florianópolis, Santa Catarina, Brazil (Dr Luna); Department of Neuroscience and Behavior, Medical School of Ribeirão Preto, University of São Paulo, São Paulo, Brazil and National Institute for Translational Medicine, Ribeirão Preto, Brazil (Drs Crippa and Hallak); Brain Institute/Hospital Universitario Onofre Lopes, Natal, Brazil (Dr de Araujo); The Beckley Foundation, Beckley Park, Oxford, United Kingdom (Mr Friedlander and Mrs Feilding); Department of Comparative Biomedical Sciences, School of Veterinary Medicine, Louisiana State University, Skip Bertman Drive at River Road, Baton Rouge, Louisiana (Dr Barker); Department of Clinical and Health Psychology, School of Psychology, Autonomous University of Barcelona, Barcelona, Spain (Dr Soler)
| | - Matilde Elices
- School of Medicine, Autonomous University of Barcelona, Barcelona, Spain (Mr Sampedro); Human Neuropsychopharmacology Research Group, Sant Pau Institute of Biomedical Research, Barcelona, Spain (Dr de la Fuente Revenga, Ms Roberto, and Dr Riba); Pharmacokinetic and Pharmacodynamic Modelling and Simulation, Sant Pau Institute of Biomedical Research, Barcelona, Spain (Dr Valle); Centre d’Investigació de Medicaments, Servei de Farmacologia Clínica, Hospital de la Santa Creu i Sant Pau, Barcelona, Spain (Drs Valle and Riba); Centro de Investigación Biomédica en Red de Salud Mental, CIBERSAM, Spain (Drs Valle, Elices, Álvarez, Soler, Pascual, and Riba); Department of Pharmacology and Therapeutics, Autonomous University of Barcelona, Barcelona, Spain (Dr Valle); Department of Psychiatry, Hospital de la Santa Creu i Sant Pau, Barcelona, Spain (Ms Domínguez-Clavé and Drs Elices, Álvarez, Soler, and Pascual); Department of Psychiatry and Forensic Medicine, School of Medicine, Universitat Autònoma de Barcelona, Barcelona, Spain (Ms Domínguez-Clavé and Drs Elices, Álvarez, and Pascual); Research Center for the Study of Psychointegrator Plants, Visionary Art and Consciousness, Florianópolis, Santa Catarina, Brazil (Dr Luna); Department of Neuroscience and Behavior, Medical School of Ribeirão Preto, University of São Paulo, São Paulo, Brazil and National Institute for Translational Medicine, Ribeirão Preto, Brazil (Drs Crippa and Hallak); Brain Institute/Hospital Universitario Onofre Lopes, Natal, Brazil (Dr de Araujo); The Beckley Foundation, Beckley Park, Oxford, United Kingdom (Mr Friedlander and Mrs Feilding); Department of Comparative Biomedical Sciences, School of Veterinary Medicine, Louisiana State University, Skip Bertman Drive at River Road, Baton Rouge, Louisiana (Dr Barker); Department of Clinical and Health Psychology, School of Psychology, Autonomous University of Barcelona, Barcelona, Spain (Dr Soler)
| | - Luís Eduardo Luna
- School of Medicine, Autonomous University of Barcelona, Barcelona, Spain (Mr Sampedro); Human Neuropsychopharmacology Research Group, Sant Pau Institute of Biomedical Research, Barcelona, Spain (Dr de la Fuente Revenga, Ms Roberto, and Dr Riba); Pharmacokinetic and Pharmacodynamic Modelling and Simulation, Sant Pau Institute of Biomedical Research, Barcelona, Spain (Dr Valle); Centre d’Investigació de Medicaments, Servei de Farmacologia Clínica, Hospital de la Santa Creu i Sant Pau, Barcelona, Spain (Drs Valle and Riba); Centro de Investigación Biomédica en Red de Salud Mental, CIBERSAM, Spain (Drs Valle, Elices, Álvarez, Soler, Pascual, and Riba); Department of Pharmacology and Therapeutics, Autonomous University of Barcelona, Barcelona, Spain (Dr Valle); Department of Psychiatry, Hospital de la Santa Creu i Sant Pau, Barcelona, Spain (Ms Domínguez-Clavé and Drs Elices, Álvarez, Soler, and Pascual); Department of Psychiatry and Forensic Medicine, School of Medicine, Universitat Autònoma de Barcelona, Barcelona, Spain (Ms Domínguez-Clavé and Drs Elices, Álvarez, and Pascual); Research Center for the Study of Psychointegrator Plants, Visionary Art and Consciousness, Florianópolis, Santa Catarina, Brazil (Dr Luna); Department of Neuroscience and Behavior, Medical School of Ribeirão Preto, University of São Paulo, São Paulo, Brazil and National Institute for Translational Medicine, Ribeirão Preto, Brazil (Drs Crippa and Hallak); Brain Institute/Hospital Universitario Onofre Lopes, Natal, Brazil (Dr de Araujo); The Beckley Foundation, Beckley Park, Oxford, United Kingdom (Mr Friedlander and Mrs Feilding); Department of Comparative Biomedical Sciences, School of Veterinary Medicine, Louisiana State University, Skip Bertman Drive at River Road, Baton Rouge, Louisiana (Dr Barker); Department of Clinical and Health Psychology, School of Psychology, Autonomous University of Barcelona, Barcelona, Spain (Dr Soler)
| | - José Alexandre S Crippa
- School of Medicine, Autonomous University of Barcelona, Barcelona, Spain (Mr Sampedro); Human Neuropsychopharmacology Research Group, Sant Pau Institute of Biomedical Research, Barcelona, Spain (Dr de la Fuente Revenga, Ms Roberto, and Dr Riba); Pharmacokinetic and Pharmacodynamic Modelling and Simulation, Sant Pau Institute of Biomedical Research, Barcelona, Spain (Dr Valle); Centre d’Investigació de Medicaments, Servei de Farmacologia Clínica, Hospital de la Santa Creu i Sant Pau, Barcelona, Spain (Drs Valle and Riba); Centro de Investigación Biomédica en Red de Salud Mental, CIBERSAM, Spain (Drs Valle, Elices, Álvarez, Soler, Pascual, and Riba); Department of Pharmacology and Therapeutics, Autonomous University of Barcelona, Barcelona, Spain (Dr Valle); Department of Psychiatry, Hospital de la Santa Creu i Sant Pau, Barcelona, Spain (Ms Domínguez-Clavé and Drs Elices, Álvarez, Soler, and Pascual); Department of Psychiatry and Forensic Medicine, School of Medicine, Universitat Autònoma de Barcelona, Barcelona, Spain (Ms Domínguez-Clavé and Drs Elices, Álvarez, and Pascual); Research Center for the Study of Psychointegrator Plants, Visionary Art and Consciousness, Florianópolis, Santa Catarina, Brazil (Dr Luna); Department of Neuroscience and Behavior, Medical School of Ribeirão Preto, University of São Paulo, São Paulo, Brazil and National Institute for Translational Medicine, Ribeirão Preto, Brazil (Drs Crippa and Hallak); Brain Institute/Hospital Universitario Onofre Lopes, Natal, Brazil (Dr de Araujo); The Beckley Foundation, Beckley Park, Oxford, United Kingdom (Mr Friedlander and Mrs Feilding); Department of Comparative Biomedical Sciences, School of Veterinary Medicine, Louisiana State University, Skip Bertman Drive at River Road, Baton Rouge, Louisiana (Dr Barker); Department of Clinical and Health Psychology, School of Psychology, Autonomous University of Barcelona, Barcelona, Spain (Dr Soler)
| | - Jaime E C Hallak
- School of Medicine, Autonomous University of Barcelona, Barcelona, Spain (Mr Sampedro); Human Neuropsychopharmacology Research Group, Sant Pau Institute of Biomedical Research, Barcelona, Spain (Dr de la Fuente Revenga, Ms Roberto, and Dr Riba); Pharmacokinetic and Pharmacodynamic Modelling and Simulation, Sant Pau Institute of Biomedical Research, Barcelona, Spain (Dr Valle); Centre d’Investigació de Medicaments, Servei de Farmacologia Clínica, Hospital de la Santa Creu i Sant Pau, Barcelona, Spain (Drs Valle and Riba); Centro de Investigación Biomédica en Red de Salud Mental, CIBERSAM, Spain (Drs Valle, Elices, Álvarez, Soler, Pascual, and Riba); Department of Pharmacology and Therapeutics, Autonomous University of Barcelona, Barcelona, Spain (Dr Valle); Department of Psychiatry, Hospital de la Santa Creu i Sant Pau, Barcelona, Spain (Ms Domínguez-Clavé and Drs Elices, Álvarez, Soler, and Pascual); Department of Psychiatry and Forensic Medicine, School of Medicine, Universitat Autònoma de Barcelona, Barcelona, Spain (Ms Domínguez-Clavé and Drs Elices, Álvarez, and Pascual); Research Center for the Study of Psychointegrator Plants, Visionary Art and Consciousness, Florianópolis, Santa Catarina, Brazil (Dr Luna); Department of Neuroscience and Behavior, Medical School of Ribeirão Preto, University of São Paulo, São Paulo, Brazil and National Institute for Translational Medicine, Ribeirão Preto, Brazil (Drs Crippa and Hallak); Brain Institute/Hospital Universitario Onofre Lopes, Natal, Brazil (Dr de Araujo); The Beckley Foundation, Beckley Park, Oxford, United Kingdom (Mr Friedlander and Mrs Feilding); Department of Comparative Biomedical Sciences, School of Veterinary Medicine, Louisiana State University, Skip Bertman Drive at River Road, Baton Rouge, Louisiana (Dr Barker); Department of Clinical and Health Psychology, School of Psychology, Autonomous University of Barcelona, Barcelona, Spain (Dr Soler)
| | - Draulio B de Araujo
- School of Medicine, Autonomous University of Barcelona, Barcelona, Spain (Mr Sampedro); Human Neuropsychopharmacology Research Group, Sant Pau Institute of Biomedical Research, Barcelona, Spain (Dr de la Fuente Revenga, Ms Roberto, and Dr Riba); Pharmacokinetic and Pharmacodynamic Modelling and Simulation, Sant Pau Institute of Biomedical Research, Barcelona, Spain (Dr Valle); Centre d’Investigació de Medicaments, Servei de Farmacologia Clínica, Hospital de la Santa Creu i Sant Pau, Barcelona, Spain (Drs Valle and Riba); Centro de Investigación Biomédica en Red de Salud Mental, CIBERSAM, Spain (Drs Valle, Elices, Álvarez, Soler, Pascual, and Riba); Department of Pharmacology and Therapeutics, Autonomous University of Barcelona, Barcelona, Spain (Dr Valle); Department of Psychiatry, Hospital de la Santa Creu i Sant Pau, Barcelona, Spain (Ms Domínguez-Clavé and Drs Elices, Álvarez, Soler, and Pascual); Department of Psychiatry and Forensic Medicine, School of Medicine, Universitat Autònoma de Barcelona, Barcelona, Spain (Ms Domínguez-Clavé and Drs Elices, Álvarez, and Pascual); Research Center for the Study of Psychointegrator Plants, Visionary Art and Consciousness, Florianópolis, Santa Catarina, Brazil (Dr Luna); Department of Neuroscience and Behavior, Medical School of Ribeirão Preto, University of São Paulo, São Paulo, Brazil and National Institute for Translational Medicine, Ribeirão Preto, Brazil (Drs Crippa and Hallak); Brain Institute/Hospital Universitario Onofre Lopes, Natal, Brazil (Dr de Araujo); The Beckley Foundation, Beckley Park, Oxford, United Kingdom (Mr Friedlander and Mrs Feilding); Department of Comparative Biomedical Sciences, School of Veterinary Medicine, Louisiana State University, Skip Bertman Drive at River Road, Baton Rouge, Louisiana (Dr Barker); Department of Clinical and Health Psychology, School of Psychology, Autonomous University of Barcelona, Barcelona, Spain (Dr Soler)
| | - Pablo Friedlander
- School of Medicine, Autonomous University of Barcelona, Barcelona, Spain (Mr Sampedro); Human Neuropsychopharmacology Research Group, Sant Pau Institute of Biomedical Research, Barcelona, Spain (Dr de la Fuente Revenga, Ms Roberto, and Dr Riba); Pharmacokinetic and Pharmacodynamic Modelling and Simulation, Sant Pau Institute of Biomedical Research, Barcelona, Spain (Dr Valle); Centre d’Investigació de Medicaments, Servei de Farmacologia Clínica, Hospital de la Santa Creu i Sant Pau, Barcelona, Spain (Drs Valle and Riba); Centro de Investigación Biomédica en Red de Salud Mental, CIBERSAM, Spain (Drs Valle, Elices, Álvarez, Soler, Pascual, and Riba); Department of Pharmacology and Therapeutics, Autonomous University of Barcelona, Barcelona, Spain (Dr Valle); Department of Psychiatry, Hospital de la Santa Creu i Sant Pau, Barcelona, Spain (Ms Domínguez-Clavé and Drs Elices, Álvarez, Soler, and Pascual); Department of Psychiatry and Forensic Medicine, School of Medicine, Universitat Autònoma de Barcelona, Barcelona, Spain (Ms Domínguez-Clavé and Drs Elices, Álvarez, and Pascual); Research Center for the Study of Psychointegrator Plants, Visionary Art and Consciousness, Florianópolis, Santa Catarina, Brazil (Dr Luna); Department of Neuroscience and Behavior, Medical School of Ribeirão Preto, University of São Paulo, São Paulo, Brazil and National Institute for Translational Medicine, Ribeirão Preto, Brazil (Drs Crippa and Hallak); Brain Institute/Hospital Universitario Onofre Lopes, Natal, Brazil (Dr de Araujo); The Beckley Foundation, Beckley Park, Oxford, United Kingdom (Mr Friedlander and Mrs Feilding); Department of Comparative Biomedical Sciences, School of Veterinary Medicine, Louisiana State University, Skip Bertman Drive at River Road, Baton Rouge, Louisiana (Dr Barker); Department of Clinical and Health Psychology, School of Psychology, Autonomous University of Barcelona, Barcelona, Spain (Dr Soler)
| | - Steven A Barker
- School of Medicine, Autonomous University of Barcelona, Barcelona, Spain (Mr Sampedro); Human Neuropsychopharmacology Research Group, Sant Pau Institute of Biomedical Research, Barcelona, Spain (Dr de la Fuente Revenga, Ms Roberto, and Dr Riba); Pharmacokinetic and Pharmacodynamic Modelling and Simulation, Sant Pau Institute of Biomedical Research, Barcelona, Spain (Dr Valle); Centre d’Investigació de Medicaments, Servei de Farmacologia Clínica, Hospital de la Santa Creu i Sant Pau, Barcelona, Spain (Drs Valle and Riba); Centro de Investigación Biomédica en Red de Salud Mental, CIBERSAM, Spain (Drs Valle, Elices, Álvarez, Soler, Pascual, and Riba); Department of Pharmacology and Therapeutics, Autonomous University of Barcelona, Barcelona, Spain (Dr Valle); Department of Psychiatry, Hospital de la Santa Creu i Sant Pau, Barcelona, Spain (Ms Domínguez-Clavé and Drs Elices, Álvarez, Soler, and Pascual); Department of Psychiatry and Forensic Medicine, School of Medicine, Universitat Autònoma de Barcelona, Barcelona, Spain (Ms Domínguez-Clavé and Drs Elices, Álvarez, and Pascual); Research Center for the Study of Psychointegrator Plants, Visionary Art and Consciousness, Florianópolis, Santa Catarina, Brazil (Dr Luna); Department of Neuroscience and Behavior, Medical School of Ribeirão Preto, University of São Paulo, São Paulo, Brazil and National Institute for Translational Medicine, Ribeirão Preto, Brazil (Drs Crippa and Hallak); Brain Institute/Hospital Universitario Onofre Lopes, Natal, Brazil (Dr de Araujo); The Beckley Foundation, Beckley Park, Oxford, United Kingdom (Mr Friedlander and Mrs Feilding); Department of Comparative Biomedical Sciences, School of Veterinary Medicine, Louisiana State University, Skip Bertman Drive at River Road, Baton Rouge, Louisiana (Dr Barker); Department of Clinical and Health Psychology, School of Psychology, Autonomous University of Barcelona, Barcelona, Spain (Dr Soler)
| | - Enrique Álvarez
- School of Medicine, Autonomous University of Barcelona, Barcelona, Spain (Mr Sampedro); Human Neuropsychopharmacology Research Group, Sant Pau Institute of Biomedical Research, Barcelona, Spain (Dr de la Fuente Revenga, Ms Roberto, and Dr Riba); Pharmacokinetic and Pharmacodynamic Modelling and Simulation, Sant Pau Institute of Biomedical Research, Barcelona, Spain (Dr Valle); Centre d’Investigació de Medicaments, Servei de Farmacologia Clínica, Hospital de la Santa Creu i Sant Pau, Barcelona, Spain (Drs Valle and Riba); Centro de Investigación Biomédica en Red de Salud Mental, CIBERSAM, Spain (Drs Valle, Elices, Álvarez, Soler, Pascual, and Riba); Department of Pharmacology and Therapeutics, Autonomous University of Barcelona, Barcelona, Spain (Dr Valle); Department of Psychiatry, Hospital de la Santa Creu i Sant Pau, Barcelona, Spain (Ms Domínguez-Clavé and Drs Elices, Álvarez, Soler, and Pascual); Department of Psychiatry and Forensic Medicine, School of Medicine, Universitat Autònoma de Barcelona, Barcelona, Spain (Ms Domínguez-Clavé and Drs Elices, Álvarez, and Pascual); Research Center for the Study of Psychointegrator Plants, Visionary Art and Consciousness, Florianópolis, Santa Catarina, Brazil (Dr Luna); Department of Neuroscience and Behavior, Medical School of Ribeirão Preto, University of São Paulo, São Paulo, Brazil and National Institute for Translational Medicine, Ribeirão Preto, Brazil (Drs Crippa and Hallak); Brain Institute/Hospital Universitario Onofre Lopes, Natal, Brazil (Dr de Araujo); The Beckley Foundation, Beckley Park, Oxford, United Kingdom (Mr Friedlander and Mrs Feilding); Department of Comparative Biomedical Sciences, School of Veterinary Medicine, Louisiana State University, Skip Bertman Drive at River Road, Baton Rouge, Louisiana (Dr Barker); Department of Clinical and Health Psychology, School of Psychology, Autonomous University of Barcelona, Barcelona, Spain (Dr Soler)
| | - Joaquim Soler
- School of Medicine, Autonomous University of Barcelona, Barcelona, Spain (Mr Sampedro); Human Neuropsychopharmacology Research Group, Sant Pau Institute of Biomedical Research, Barcelona, Spain (Dr de la Fuente Revenga, Ms Roberto, and Dr Riba); Pharmacokinetic and Pharmacodynamic Modelling and Simulation, Sant Pau Institute of Biomedical Research, Barcelona, Spain (Dr Valle); Centre d’Investigació de Medicaments, Servei de Farmacologia Clínica, Hospital de la Santa Creu i Sant Pau, Barcelona, Spain (Drs Valle and Riba); Centro de Investigación Biomédica en Red de Salud Mental, CIBERSAM, Spain (Drs Valle, Elices, Álvarez, Soler, Pascual, and Riba); Department of Pharmacology and Therapeutics, Autonomous University of Barcelona, Barcelona, Spain (Dr Valle); Department of Psychiatry, Hospital de la Santa Creu i Sant Pau, Barcelona, Spain (Ms Domínguez-Clavé and Drs Elices, Álvarez, Soler, and Pascual); Department of Psychiatry and Forensic Medicine, School of Medicine, Universitat Autònoma de Barcelona, Barcelona, Spain (Ms Domínguez-Clavé and Drs Elices, Álvarez, and Pascual); Research Center for the Study of Psychointegrator Plants, Visionary Art and Consciousness, Florianópolis, Santa Catarina, Brazil (Dr Luna); Department of Neuroscience and Behavior, Medical School of Ribeirão Preto, University of São Paulo, São Paulo, Brazil and National Institute for Translational Medicine, Ribeirão Preto, Brazil (Drs Crippa and Hallak); Brain Institute/Hospital Universitario Onofre Lopes, Natal, Brazil (Dr de Araujo); The Beckley Foundation, Beckley Park, Oxford, United Kingdom (Mr Friedlander and Mrs Feilding); Department of Comparative Biomedical Sciences, School of Veterinary Medicine, Louisiana State University, Skip Bertman Drive at River Road, Baton Rouge, Louisiana (Dr Barker); Department of Clinical and Health Psychology, School of Psychology, Autonomous University of Barcelona, Barcelona, Spain (Dr Soler)
| | - Juan C Pascual
- School of Medicine, Autonomous University of Barcelona, Barcelona, Spain (Mr Sampedro); Human Neuropsychopharmacology Research Group, Sant Pau Institute of Biomedical Research, Barcelona, Spain (Dr de la Fuente Revenga, Ms Roberto, and Dr Riba); Pharmacokinetic and Pharmacodynamic Modelling and Simulation, Sant Pau Institute of Biomedical Research, Barcelona, Spain (Dr Valle); Centre d’Investigació de Medicaments, Servei de Farmacologia Clínica, Hospital de la Santa Creu i Sant Pau, Barcelona, Spain (Drs Valle and Riba); Centro de Investigación Biomédica en Red de Salud Mental, CIBERSAM, Spain (Drs Valle, Elices, Álvarez, Soler, Pascual, and Riba); Department of Pharmacology and Therapeutics, Autonomous University of Barcelona, Barcelona, Spain (Dr Valle); Department of Psychiatry, Hospital de la Santa Creu i Sant Pau, Barcelona, Spain (Ms Domínguez-Clavé and Drs Elices, Álvarez, Soler, and Pascual); Department of Psychiatry and Forensic Medicine, School of Medicine, Universitat Autònoma de Barcelona, Barcelona, Spain (Ms Domínguez-Clavé and Drs Elices, Álvarez, and Pascual); Research Center for the Study of Psychointegrator Plants, Visionary Art and Consciousness, Florianópolis, Santa Catarina, Brazil (Dr Luna); Department of Neuroscience and Behavior, Medical School of Ribeirão Preto, University of São Paulo, São Paulo, Brazil and National Institute for Translational Medicine, Ribeirão Preto, Brazil (Drs Crippa and Hallak); Brain Institute/Hospital Universitario Onofre Lopes, Natal, Brazil (Dr de Araujo); The Beckley Foundation, Beckley Park, Oxford, United Kingdom (Mr Friedlander and Mrs Feilding); Department of Comparative Biomedical Sciences, School of Veterinary Medicine, Louisiana State University, Skip Bertman Drive at River Road, Baton Rouge, Louisiana (Dr Barker); Department of Clinical and Health Psychology, School of Psychology, Autonomous University of Barcelona, Barcelona, Spain (Dr Soler)
| | - Amanda Feilding
- School of Medicine, Autonomous University of Barcelona, Barcelona, Spain (Mr Sampedro); Human Neuropsychopharmacology Research Group, Sant Pau Institute of Biomedical Research, Barcelona, Spain (Dr de la Fuente Revenga, Ms Roberto, and Dr Riba); Pharmacokinetic and Pharmacodynamic Modelling and Simulation, Sant Pau Institute of Biomedical Research, Barcelona, Spain (Dr Valle); Centre d’Investigació de Medicaments, Servei de Farmacologia Clínica, Hospital de la Santa Creu i Sant Pau, Barcelona, Spain (Drs Valle and Riba); Centro de Investigación Biomédica en Red de Salud Mental, CIBERSAM, Spain (Drs Valle, Elices, Álvarez, Soler, Pascual, and Riba); Department of Pharmacology and Therapeutics, Autonomous University of Barcelona, Barcelona, Spain (Dr Valle); Department of Psychiatry, Hospital de la Santa Creu i Sant Pau, Barcelona, Spain (Ms Domínguez-Clavé and Drs Elices, Álvarez, Soler, and Pascual); Department of Psychiatry and Forensic Medicine, School of Medicine, Universitat Autònoma de Barcelona, Barcelona, Spain (Ms Domínguez-Clavé and Drs Elices, Álvarez, and Pascual); Research Center for the Study of Psychointegrator Plants, Visionary Art and Consciousness, Florianópolis, Santa Catarina, Brazil (Dr Luna); Department of Neuroscience and Behavior, Medical School of Ribeirão Preto, University of São Paulo, São Paulo, Brazil and National Institute for Translational Medicine, Ribeirão Preto, Brazil (Drs Crippa and Hallak); Brain Institute/Hospital Universitario Onofre Lopes, Natal, Brazil (Dr de Araujo); The Beckley Foundation, Beckley Park, Oxford, United Kingdom (Mr Friedlander and Mrs Feilding); Department of Comparative Biomedical Sciences, School of Veterinary Medicine, Louisiana State University, Skip Bertman Drive at River Road, Baton Rouge, Louisiana (Dr Barker); Department of Clinical and Health Psychology, School of Psychology, Autonomous University of Barcelona, Barcelona, Spain (Dr Soler)
| | - Jordi Riba
- School of Medicine, Autonomous University of Barcelona, Barcelona, Spain (Mr Sampedro); Human Neuropsychopharmacology Research Group, Sant Pau Institute of Biomedical Research, Barcelona, Spain (Dr de la Fuente Revenga, Ms Roberto, and Dr Riba); Pharmacokinetic and Pharmacodynamic Modelling and Simulation, Sant Pau Institute of Biomedical Research, Barcelona, Spain (Dr Valle); Centre d’Investigació de Medicaments, Servei de Farmacologia Clínica, Hospital de la Santa Creu i Sant Pau, Barcelona, Spain (Drs Valle and Riba); Centro de Investigación Biomédica en Red de Salud Mental, CIBERSAM, Spain (Drs Valle, Elices, Álvarez, Soler, Pascual, and Riba); Department of Pharmacology and Therapeutics, Autonomous University of Barcelona, Barcelona, Spain (Dr Valle); Department of Psychiatry, Hospital de la Santa Creu i Sant Pau, Barcelona, Spain (Ms Domínguez-Clavé and Drs Elices, Álvarez, Soler, and Pascual); Department of Psychiatry and Forensic Medicine, School of Medicine, Universitat Autònoma de Barcelona, Barcelona, Spain (Ms Domínguez-Clavé and Drs Elices, Álvarez, and Pascual); Research Center for the Study of Psychointegrator Plants, Visionary Art and Consciousness, Florianópolis, Santa Catarina, Brazil (Dr Luna); Department of Neuroscience and Behavior, Medical School of Ribeirão Preto, University of São Paulo, São Paulo, Brazil and National Institute for Translational Medicine, Ribeirão Preto, Brazil (Drs Crippa and Hallak); Brain Institute/Hospital Universitario Onofre Lopes, Natal, Brazil (Dr de Araujo); The Beckley Foundation, Beckley Park, Oxford, United Kingdom (Mr Friedlander and Mrs Feilding); Department of Comparative Biomedical Sciences, School of Veterinary Medicine, Louisiana State University, Skip Bertman Drive at River Road, Baton Rouge, Louisiana (Dr Barker); Department of Clinical and Health Psychology, School of Psychology, Autonomous University of Barcelona, Barcelona, Spain (Dr Soler)
| |
Collapse
|
43
|
Involvement of serotonin 2A receptor activation in modulating medial prefrontal cortex and amygdala neuronal activation during novelty-exposure. Behav Brain Res 2017; 326:1-12. [PMID: 28263831 DOI: 10.1016/j.bbr.2017.02.050] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/11/2016] [Revised: 02/23/2017] [Accepted: 02/28/2017] [Indexed: 12/11/2022]
Abstract
The medial prefrontal cortex (PFC) plays a major role in executive function by exerting a top-down control onto subcortical areas. Novelty-induced frontal cortex activation is 5-HT2A receptor (5-HT2AR) dependent. Here, we further investigated how blockade of 5-HT2ARs in mice exposed to a novel open-field arena affects medial PFC activation and basolateral amygdala (BLA) reactivity. We used c-Fos immunoreactivity (IR) as a marker of neuronal activation and stereological quantification for obtaining the total number of c-Fos-IR neurons as a measure of regional activation. We further examined the impact of 5-HT2AR blockade on the striatal-projecting BLA neurons. Systemic administration of ketanserin (0.5mg/kg) prior to novel open-field exposure resulted in reduced total numbers of c-Fos-IR cells in dorsomedial PFC areas and the BLA. Moreover, there was a positive correlation between the relative time spent in the centre of the open-field and BLA c-Fos-IR in the ketanserin-treated animals. Unilateral medial PFC lesions blocked this effect, ascertaining an involvement of this frontal cortex area. On the other hand, medial PFC lesioning exacerbated the more anxiogenic-like behaviour of the ketanserin-treated animals, upholding its involvement in modulating averseness. Ketanserin did not affect the number of activated striatal-projecting BLA neurons (measured by number of Cholera Toxin b (CTb) retrograde labelled neurons also being c-Fos-IR) following CTb injection in the ventral striatum. These results support a role of 5-HT2AR activation in modulating mPFC and BLA activation during exposure to a novel environment, which may be interrelated. Conversely, 5-HT2AR blockade does not seem to affect the amygdala-striatal projection.
Collapse
|
44
|
Abstract
The classic serotonergic hallucinogens, or psychedelics, have the ability to profoundly alter perception and behavior. These can include visual distortions, hallucinations, detachment from reality, and mystical experiences. Some psychedelics, like LSD, are able to produce these effects with remarkably low doses of drug. Others, like psilocybin, have recently been demonstrated to have significant clinical efficacy in the treatment of depression, anxiety, and addiction that persist for at least several months after only a single therapeutic session. How does this occur? Much work has recently been published from imaging studies showing that psychedelics alter brain network connectivity. They facilitate a disintegration of the default mode network, producing a hyperconnectivity between brain regions that allow centers that do not normally communicate with each other to do so. The immediate and acute effects on both behaviors and network connectivity are likely mediated by effector pathways downstream of serotonin 5-HT2A receptor activation. These acute molecular processes also influence gene expression changes, which likely influence synaptic plasticity and facilitate more long-term changes in brain neurochemistry ultimately underlying the therapeutic efficacy of a single administration to achieve long-lasting effects. In this review, we summarize what is currently known about the molecular genetic responses to psychedelics within the brain and discuss how gene expression changes may contribute to altered cellular physiology and behaviors.
Collapse
|
45
|
Kapócs G, Scholkmann F, Salari V, Császár N, Szőke H, Bókkon I. Possible role of biochemiluminescent photons for lysergic acid diethylamide (LSD)-induced phosphenes and visual hallucinations. Rev Neurosci 2017; 28:77-86. [PMID: 27732562 DOI: 10.1515/revneuro-2016-0047] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/03/2016] [Accepted: 09/03/2016] [Indexed: 11/15/2022]
Abstract
AbstractToday, there is an increased interest in research on lysergic acid diethylamide (LSD) because it may offer new opportunities in psychotherapy under controlled settings. The more we know about how a drug works in the brain, the more opportunities there will be to exploit it in medicine. Here, based on our previously published papers and investigations, we suggest that LSD-induced visual hallucinations/phosphenes may be due to the transient enhancement of bioluminescent photons in the early retinotopic visual system in blind as well as healthy people.
Collapse
Affiliation(s)
- Gábor Kapócs
- 1Social Home for Psychiatric Patients, H-9970, Szentgotthard, Hungary
- 2Institute of Behavioral Sciences, Semmelweis University, H-1089, Budapest, Hungary
| | - Felix Scholkmann
- 3Biomedical Optics Research Laboratory, Department of Neonatology, University Hospital Zurich, University of Zurich, CH-8091 Zurich, Switzerland
- 4Research Office for Complex Physical and Biological Systems (ROCoS), CH-8038 Zurich, Switzerland
| | - Vahid Salari
- 5Department of Physics, Isfahan University of Technology, Isfahan 84156-83111, Iran (Islamic Republic of)
- 6School of Physics, Institute for Research in Fundamental Sciences (IPM), Tehran 19395-5531, Iran (Islamic Republic of)
| | - Noémi Császár
- 7Psychoszomatic OutPatient Department, H-1037, Budapest, Hungary
- 8Gaspar Karoly University Psychological Institute, H-1091 Budapest, Hungary
| | - Henrik Szőke
- 9Doctors School of Health Sciences, University of Pécs, H-7621 Pécs, Hungary
| | - István Bókkon
- 7Psychoszomatic OutPatient Department, H-1037, Budapest, Hungary
- 10Vision Research Institute, Neuroscience and Consciousness Research Department, Lowell, MA 01854, United States of America
| |
Collapse
|
46
|
Noworyta-Sokołowska K, Kamińska K, Kreiner G, Rogóż Z, Gołembiowska K. Neurotoxic Effects of 5-MeO-DIPT: A Psychoactive Tryptamine Derivative in Rats. Neurotox Res 2016; 30:606-619. [PMID: 27461536 PMCID: PMC5047954 DOI: 10.1007/s12640-016-9654-0] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/21/2016] [Revised: 07/05/2016] [Accepted: 07/13/2016] [Indexed: 11/01/2022]
Abstract
5-Methoxy-N,N-diisopropyltryptamine (5-MeO-DIPT, 'foxy') is one of the most popular tryptamine hallucinogens in the illicit drug market. It produces serious adverse effects, but its pharmacological profile is not well recognized. In vitro data have shown that 5-MeO-DIPT acts as a potent serotonin transporter (SERT) inhibitor and displays high affinity at serotonin 5-HT1A, 5-HT2A, and 5-HT2C receptors. In this study, using microdialysis in freely moving rats, we examined the effect of 5-MeO-DIPT on dopamine (DA), serotonin (5-HT), and glutamate release in the rat striatum, nucleus accumbens, and frontal cortex. In search of a possible neurotoxic effect of 5-MeO-DIPT, we measured DA and 5-HT tissue content in the above rat brain regions and also determined the oxidative DNA damage with the comet assay. Moreover, we tested drug-elicited head-twitch response and a forepaw treading induced by 8-OH-DPAT. 5-MeO-DIPT at doses of 5, 10, and 20 mg/kg increased extracellular DA, 5-HT, and glutamate level but the differences in the potency were found between brain regions. 5-MeO-DIPT increased 5-HT and decreased 5-HIAA tissue content which seems to result from SERT inhibition. On the other hand, a decrease in DA, DOPAC, and HVA tissue contents suggests possible adaptive changes in DA turnover or damage of DA terminals by 5-MeO-DIPT. DNA single and double-strand breaks persisted up to 60 days after the treatment, indicating marked neurotoxicity of 5-MeO-DIPT. The induction of head-twitch response and potentiation of forepaw treading induced by 8-OH-DPAT indicate that hallucinogenic activity seems to be mediated through the stimulation of 5-HT2A and 5-HT1A receptors by 5-MeO-DIPT.
Collapse
Affiliation(s)
- Karolina Noworyta-Sokołowska
- Department of Pharmacology, Institute of Pharmacology, Polish Academy of Sciences, 12 Smętna, 31-343, Kraków, Poland
| | - Katarzyna Kamińska
- Department of Pharmacology, Institute of Pharmacology, Polish Academy of Sciences, 12 Smętna, 31-343, Kraków, Poland
| | - Grzegorz Kreiner
- Department of Biochemistry, Polish Academy of Sciences, 12 Smętna, 31-343, Kraków, Poland
| | - Zofia Rogóż
- Department of Pharmacology, Institute of Pharmacology, Polish Academy of Sciences, 12 Smętna, 31-343, Kraków, Poland
| | - Krystyna Gołembiowska
- Department of Pharmacology, Institute of Pharmacology, Polish Academy of Sciences, 12 Smętna, 31-343, Kraków, Poland.
| |
Collapse
|
47
|
5-HT2A and mGlu2/3 receptor interactions: on their relevance to cognitive function and psychosis. Behav Pharmacol 2016; 27:1-11. [PMID: 26292187 DOI: 10.1097/fbp.0000000000000183] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/26/2022]
Abstract
Serotonin [5-hydroxytryptamine (5-HT)] and glutamate have both been implicated in the pathophysiology of neuropsychiatric disorders but also in the mechanism of antipsychotic and hallucinogenic drug actions. Furthermore, close antagonistic interactions between 5-HT2A and metabotropic glutamate (mGlu)2/3 receptors have been established over the last decades on the basis of numerous electrophysiological, biochemical, and behavioral studies. Besides synaptic mechanisms, more recent findings suggested that heterodimeric 5-HT2A-mGlu2 receptor complexes in the prefrontal cortex may account for the functional crosstalk between these two receptor subtypes. In this review, we focus on in-vitro and in-vivo studies documenting the important relationship between 5-HT2A and mGlu2/3 receptors, with relevance to both normal behavioral function and psychosis.
Collapse
|
48
|
Abstract
Psychedelics (serotonergic hallucinogens) are powerful psychoactive substances that alter perception and mood and affect numerous cognitive processes. They are generally considered physiologically safe and do not lead to dependence or addiction. Their origin predates written history, and they were employed by early cultures in many sociocultural and ritual contexts. After the virtually contemporaneous discovery of (5R,8R)-(+)-lysergic acid-N,N-diethylamide (LSD)-25 and the identification of serotonin in the brain, early research focused intensively on the possibility that LSD and other psychedelics had a serotonergic basis for their action. Today there is a consensus that psychedelics are agonists or partial agonists at brain serotonin 5-hydroxytryptamine 2A receptors, with particular importance on those expressed on apical dendrites of neocortical pyramidal cells in layer V. Several useful rodent models have been developed over the years to help unravel the neurochemical correlates of serotonin 5-hydroxytryptamine 2A receptor activation in the brain, and a variety of imaging techniques have been employed to identify key brain areas that are directly affected by psychedelics. Recent and exciting developments in the field have occurred in clinical research, where several double-blind placebo-controlled phase 2 studies of psilocybin-assisted psychotherapy in patients with cancer-related psychosocial distress have demonstrated unprecedented positive relief of anxiety and depression. Two small pilot studies of psilocybin-assisted psychotherapy also have shown positive benefit in treating both alcohol and nicotine addiction. Recently, blood oxygen level-dependent functional magnetic resonance imaging and magnetoencephalography have been employed for in vivo brain imaging in humans after administration of a psychedelic, and results indicate that intravenously administered psilocybin and LSD produce decreases in oscillatory power in areas of the brain's default mode network.
Collapse
Affiliation(s)
- David E Nichols
- Eschelman School of Pharmacy, University of North Carolina, Chapel Hill, North Carolina
| |
Collapse
|
49
|
Maternal lipopolysaccharide treatment differentially affects 5-HT2A and mGlu2/3 receptor function in the adult male and female rat offspring. Neuropharmacology 2015; 97:275-88. [DOI: 10.1016/j.neuropharm.2015.05.029] [Citation(s) in RCA: 32] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/08/2015] [Revised: 05/08/2015] [Accepted: 05/22/2015] [Indexed: 12/17/2022]
|
50
|
mGlu5-GABAB interplay in animal models of positive, negative and cognitive symptoms of schizophrenia. Neurochem Int 2015; 88:97-109. [DOI: 10.1016/j.neuint.2015.03.010] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/17/2014] [Revised: 03/17/2015] [Accepted: 03/23/2015] [Indexed: 11/19/2022]
|