1
|
Halman A, Conyers R, Moore C, Khatri D, Sarris J, Perkins D. Harnessing Pharmacogenomics in Clinical Research on Psychedelic-Assisted Therapy. Clin Pharmacol Ther 2025; 117:106-115. [PMID: 39345195 DOI: 10.1002/cpt.3459] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/08/2024] [Accepted: 09/18/2024] [Indexed: 10/01/2024]
Abstract
Psychedelics have recently re-emerged as potential treatments for various psychiatric conditions that impose major public health costs and for which current treatment options have limited efficacy. At the same time, personalized medicine is increasingly being implemented in psychiatry to provide individualized drug dosing recommendations based on genetics. This review brings together these topics to explore the utility of pharmacogenomics (a key component of personalized medicine) in psychedelic-assisted therapies. We summarized the literature and explored the potential implications of genetic variability on the pharmacodynamics and pharmacokinetics of psychedelic drugs including lysergic acid diethylamide (LSD), psilocybin, N,N-dimethyltryptamine (DMT), 5-methoxy-N,N-dimethyltryptamine (5-MeO-DMT), ibogaine and 3,4-methylenedioxymethamphetamine (MDMA). Although existing evidence is limited, particularly concerning pharmacodynamics, studies investigating pharmacokinetics indicate that genetic variants in drug-metabolizing enzymes, such as cytochrome P450, impact the intensity of acute psychedelic effects for LSD and ibogaine, and that a dose reduction for CYP2D6 poor metabolizers may be appropriate. Furthermore, based on the preclinical evidence, it can be hypothesized that CYP2D6 metabolizer status might contribute to altered acute psychedelic experiences with 5-MeO-DMT and psilocybin when combined with monoamine oxidase inhibitors. In conclusion, considering early evidence that genetic factors can influence the effects of certain psychedelics, we suggest that pharmacogenomic testing should be further investigated in clinical research. This is necessary to evaluate its utility in improving the safety and therapeutic profile of psychedelic therapies and a potential future role in personalizing psychedelic-assisted therapies, should these treatments become available.
Collapse
Affiliation(s)
- Andreas Halman
- Psychae Therapeutics, Melbourne, Victoria, Australia
- Cancer Therapies, Stem Cell Medicine, Murdoch Children's Research Institute, Parkville, Victoria, Australia
- School of Population and Global Health, The University of Melbourne, Melbourne, Victoria, Australia
| | - Rachel Conyers
- Cancer Therapies, Stem Cell Medicine, Murdoch Children's Research Institute, Parkville, Victoria, Australia
- Department of Paediatrics, The University of Melbourne, Melbourne, Victoria, Australia
| | - Claire Moore
- Cancer Therapies, Stem Cell Medicine, Murdoch Children's Research Institute, Parkville, Victoria, Australia
- Department of Paediatrics, The University of Melbourne, Melbourne, Victoria, Australia
| | - Dhrita Khatri
- Cancer Therapies, Stem Cell Medicine, Murdoch Children's Research Institute, Parkville, Victoria, Australia
| | - Jerome Sarris
- Psychae Therapeutics, Melbourne, Victoria, Australia
- Centre for Mental Health, Swinburne University, Melbourne, Victoria, Australia
- NICM Health Research Institute, Western Sydney University, Westmead, New South Wales, Australia
- The Florey Institute of Neuroscience and Mental Health & The Department of Psychiatry, Melbourne University, Melbourne, Victoria, Australia
| | - Daniel Perkins
- Psychae Therapeutics, Melbourne, Victoria, Australia
- School of Population and Global Health, The University of Melbourne, Melbourne, Victoria, Australia
- Centre for Mental Health, Swinburne University, Melbourne, Victoria, Australia
| |
Collapse
|
2
|
Khakpai F, Golshani SP, Alijanpour S, Ebrahimi-Ghiri M, Zarrindast MR. Anxiolytic- and antidepressive-like effects of harmaline in mice are mediated via histamine H3 receptor blockade. Biochem Biophys Res Commun 2024; 736:150879. [PMID: 39467356 DOI: 10.1016/j.bbrc.2024.150879] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/09/2024] [Accepted: 10/22/2024] [Indexed: 10/30/2024]
Abstract
Many neuropsychiatric disorders can be caused by neurotransmitter dysfunction. Experimental studies have demonstrated that histamine and the harmaline affect physiological processes through interaction with other neurotransmitter systems. The objective of these experiments was to investigate the involvement of the histaminergic system in the effects of harmaline on anxiety- and depressive-related effects in male NMRI mice. Behavioral tests were employed to evaluate anxiety-related symptoms (elevated plus maze; EPM), depressive-like symptoms (forced swim test; FST), and cognitive decline (step-down test). The histamine H3 receptor (H3R) agonist α-methylhistamine dihydrobromide (α-MH; 5 mg/kg, i.p.) had anxiolytic- and depressive-like effects, while the H3R antagonist thioperamide (10 mg/kg, i.p.) showed an antidepressive-like property. The subthreshold dose of α-MH resulted in an increase in the tendency of mice treated with the harmaline (2.5 mg/kg) to remain in the EPM open-arms. A subthreshold dose of thioperamide (5 mg/kg) increased the time spent in the open-arms in mice treated with harmaline (2.5 and 5 mg/kg) while a high dose of harmaline decreased the immobility time. Furthermore, two higher doses of harmaline resulted in a reduction in the number of open-arm entries. Similarly, mice administered with thioperamide and a low dose of harmaline decreased locomotor activity in the EPM. Ultimately, the combined thioperamide and harmaline did not impair memory retrieval of mice. These experiments demonstrate that the histaminergic system is implicated in the anxiety- and depressive-related effects of harmaline. The combination of thioperamide and harmaline is effective in treating anxiety and depression without having an adverse effect on memory formation.
Collapse
Affiliation(s)
- Fatemeh Khakpai
- Department of Physiology, Faculty of Medicine, Tehran Medical Sciences, Islamic Azad University, Iran
| | - Seyed Parsa Golshani
- Department of Pharmacology, School of Medicine, Tehran University of Medical Sciences, Tehran, Iran
| | - Sakineh Alijanpour
- Department of Biology, Faculty of Science, Gonbad Kavous University, Gonbad Kavous, Iran
| | | | - Mohammad-Reza Zarrindast
- Department of Pharmacology, School of Medicine, Tehran University of Medical Sciences, Tehran, Iran; Iranian National Center for Addiction Studies, Tehran University of Medical Sciences, Tehran, Iran
| |
Collapse
|
3
|
de Camargo RW, Joaquim L, Machado RS, de Souza Ramos S, da Rosa LR, de Novais Junior LR, Mathias K, Maximiano L, Strickert YR, Nord R, Gava ML, Scarpari E, Martins HM, Lins EMF, Chaves JS, da Silva LE, de Oliveira MP, da Silva MR, Fernandes BB, Tiscoski ADB, Piacentini N, Santos FP, Inserra A, Bobinski F, Rezin GT, Yonamine M, Petronilho F, de Bitencourt RM. Ayahuasca Pretreatment Prevents Sepsis-Induced Anxiety-Like Behavior, Neuroinflammation, and Oxidative Stress, and Increases Brain-Derived Neurotrophic Factor. Mol Neurobiol 2024:10.1007/s12035-024-04597-4. [PMID: 39613951 DOI: 10.1007/s12035-024-04597-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/28/2024] [Accepted: 10/26/2024] [Indexed: 12/01/2024]
Abstract
The psychoactive decoction Ayahuasca (AYA) used for therapeutic and religious purposes by indigenous groups and peoples from Amazonian regions produces anti-inflammatory and neuroprotective effects. Thus, it may be useful to attenuate the neuroinflammation and related anxiety- and depressive-like symptoms elicited by inflammatory insults such as sepsis. Rats were pretreated for 3 days with different doses of AYA. Twenty-four hours after, cecal ligation and puncture (CLP) was performed. On days 1-4, post-CLP behavioral tests to assess anxiety-like behavior were performed. After 24-h, neuroinflammation, oxidative stress, myeloperoxidase activity, and mitochondrial metabolism were assessed in the prefrontal cortex (PFC), hippocampus (HP), and cortex. AYA pretreatment increased the time spent in the open arms of the elevated plus maze and prevented the sepsis-induced hyper-grooming and -rearing behavior, suggesting an anxiolytic effect. AYA pretreatment increased the levels of the anti-inflammatory interleukin 4, in the PFC and the cortex, and brain-derived neurotrophic factor in the cortex. Moreover, AYA pretreatment increased myeloperoxidase activity in the PFC and the HP and decreased nitrite/nitrate concentration in the PFC, HP, and cortex of septic rats, suggesting enhanced neutrophil activation and decreased nitric oxide signaling. Furthermore, AYA pretreatment prevented lipid peroxidation in the PFC, HP, and cortex of septic rats as measured by decreased levels of thiobarbituric acid reactive substances. Levels of protein carbonyls and activity of superoxide dismutase, citrate synthase, succinate dehydrogenase, and mitochondrial respiratory chain were not affected. Together, AYA represents a promising approach to prevent sepsis-induced neuroinflammatory and oxidative stress and associated anxiety-like symptoms.
Collapse
Affiliation(s)
- Rick Wilhiam de Camargo
- Behavioral Neuroscience Laboratory, Postgraduate Program in Health Sciences, University of South Santa Catarina (UNISUL), Tubarão, Santa Catarina, Brazil
| | - Larissa Joaquim
- Behavioral Neuroscience Laboratory, Postgraduate Program in Health Sciences, University of South Santa Catarina (UNISUL), Tubarão, Santa Catarina, Brazil
| | - Richard Simon Machado
- Laboratory of Neurobiology of Inflammatory and Metabolic Processes, Postgraduate Program in Health Sciences, University of South Santa Catarina (UNISUL), Tubarão, Santa Catarina, Brazil
| | - Suelen de Souza Ramos
- Behavioral Neuroscience Laboratory, Postgraduate Program in Health Sciences, University of South Santa Catarina (UNISUL), Tubarão, Santa Catarina, Brazil
| | - Lara Rodrigues da Rosa
- Behavioral Neuroscience Laboratory, Postgraduate Program in Health Sciences, University of South Santa Catarina (UNISUL), Tubarão, Santa Catarina, Brazil
| | - Linério Ribeiro de Novais Junior
- Behavioral Neuroscience Laboratory, Postgraduate Program in Health Sciences, University of South Santa Catarina (UNISUL), Tubarão, Santa Catarina, Brazil
| | - Khiany Mathias
- Laboratory of Experimental Neurology, Graduate Program in Health Sciences, University of Southern Santa Catarina (UNESC), Criciúma, Santa Catarina, Brazil
| | - Lara Maximiano
- Behavioral Neuroscience Laboratory, Postgraduate Program in Health Sciences, University of South Santa Catarina (UNISUL), Tubarão, Santa Catarina, Brazil
| | - Yasmin Ribeiro Strickert
- Behavioral Neuroscience Laboratory, Postgraduate Program in Health Sciences, University of South Santa Catarina (UNISUL), Tubarão, Santa Catarina, Brazil
| | - Rafael Nord
- Behavioral Neuroscience Laboratory, Postgraduate Program in Health Sciences, University of South Santa Catarina (UNISUL), Tubarão, Santa Catarina, Brazil
| | - Maria Laura Gava
- Behavioral Neuroscience Laboratory, Postgraduate Program in Health Sciences, University of South Santa Catarina (UNISUL), Tubarão, Santa Catarina, Brazil
| | - Eduarda Scarpari
- Behavioral Neuroscience Laboratory, Postgraduate Program in Health Sciences, University of South Santa Catarina (UNISUL), Tubarão, Santa Catarina, Brazil
| | - Helena Mafra Martins
- Experimental Neuroscience Laboratory (LaNEx), Postgraduate Program in Health Sciences, University of South Santa Catarina (UNISUL), Palhoça, Santa Catarina, Brazil
| | - Elisa Mitkus Flores Lins
- Experimental Neuroscience Laboratory (LaNEx), Postgraduate Program in Health Sciences, University of South Santa Catarina (UNISUL), Palhoça, Santa Catarina, Brazil
| | - Jéssica Schaefer Chaves
- Experimental Neuroscience Laboratory (LaNEx), Postgraduate Program in Health Sciences, University of South Santa Catarina (UNISUL), Palhoça, Santa Catarina, Brazil
| | - Larissa Espindola da Silva
- Laboratory of Neurobiology of Inflammatory and Metabolic Processes, Postgraduate Program in Health Sciences, University of South Santa Catarina (UNISUL), Tubarão, Santa Catarina, Brazil
| | - Mariana Pacheco de Oliveira
- Laboratory of Neurobiology of Inflammatory and Metabolic Processes, Postgraduate Program in Health Sciences, University of South Santa Catarina (UNISUL), Tubarão, Santa Catarina, Brazil
| | - Mariella Reinol da Silva
- Laboratory of Neurobiology of Inflammatory and Metabolic Processes, Postgraduate Program in Health Sciences, University of South Santa Catarina (UNISUL), Tubarão, Santa Catarina, Brazil
| | - Bruna Barros Fernandes
- Laboratory of Neurobiology of Inflammatory and Metabolic Processes, Postgraduate Program in Health Sciences, University of South Santa Catarina (UNISUL), Tubarão, Santa Catarina, Brazil
| | - Anita Dal Bó Tiscoski
- Laboratory of Experimental Neurology, Graduate Program in Health Sciences, University of Southern Santa Catarina (UNESC), Criciúma, Santa Catarina, Brazil
| | - Natália Piacentini
- Laboratory of Experimental Neurology, Graduate Program in Health Sciences, University of Southern Santa Catarina (UNESC), Criciúma, Santa Catarina, Brazil
| | - Fabiana Pereira Santos
- Department of Clinical and Toxicological Analyses, School of Pharmaceutical Sciences, University of São Paulo (USP), São Paulo, Brazil
| | - Antonio Inserra
- Behavioral Neuroscience Laboratory, Postgraduate Program in Health Sciences, University of South Santa Catarina (UNISUL), Tubarão, Santa Catarina, Brazil
- Previous Affiliation: Department of Psychiatry, McGill University, Montreal, Canada
| | - Franciane Bobinski
- Experimental Neuroscience Laboratory (LaNEx), Postgraduate Program in Health Sciences, University of South Santa Catarina (UNISUL), Palhoça, Santa Catarina, Brazil
| | - Gislaine Tezza Rezin
- Laboratory of Neurobiology of Inflammatory and Metabolic Processes, Postgraduate Program in Health Sciences, University of South Santa Catarina (UNISUL), Tubarão, Santa Catarina, Brazil
| | - Mauricio Yonamine
- Department of Clinical and Toxicological Analyses, School of Pharmaceutical Sciences, University of São Paulo (USP), São Paulo, Brazil
| | - Fabrícia Petronilho
- Laboratory of Experimental Neurology, Graduate Program in Health Sciences, University of Southern Santa Catarina (UNESC), Criciúma, Santa Catarina, Brazil
| | - Rafael Mariano de Bitencourt
- Behavioral Neuroscience Laboratory, Postgraduate Program in Health Sciences, University of South Santa Catarina (UNISUL), Tubarão, Santa Catarina, Brazil.
| |
Collapse
|
4
|
Dos Santos RG, Hallak JEC. Ayahuasca: pharmacology, safety, and therapeutic effects. CNS Spectr 2024:1-9. [PMID: 39564645 DOI: 10.1017/s109285292400213x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/21/2024]
Abstract
Ayahuasca is a botanical hallucinogen traditionally used for therapeutic and ritual purposes by indigenous groups from Northwestern Amazonian countries such as Brazil, Peru, Colombia, and Ecuador. Ayahuasca is made by the decoction of two plants, which are rich in the 5-HT1A/2A partial agonist dimethyltryptamine or DMT (from the leaves of the Psychotria viridis bush) and β-carbolines such as harmine, from the stalks of the Banisteriopsis caapi vine. There is an increasing interest in the possible therapeutic effects of ayahuasca, especially for psychiatric disorders (major depression, posttraumatic stress disorder, and substance use disorder). This review summarizes information on the pharmacology, safety, and therapeutic potentials of ayahuasca. Although human experimental and naturalist studies published until now suggest a good safety and tolerability profile, often associated with improvements in depressive and anxious symptoms, there are few controlled studies, with small sample sizes, using only single doses, and with short follow-ups. Potential benefits of ayahuasca should be evaluated in larger samples in both experimental and observational studies and using different doses in controlled trials.
Collapse
Affiliation(s)
- Rafael Guimarães Dos Santos
- Department of Neuroscience and Behavior, Ribeirão Preto Medical School, University of São Paulo, Ribeirão Preto, Brazil
- National Institute of Science and Technology Translational Medicine (INCT-TM), Brazil
| | - Jaime Eduardo Cecilio Hallak
- Department of Neuroscience and Behavior, Ribeirão Preto Medical School, University of São Paulo, Ribeirão Preto, Brazil
- National Institute of Science and Technology Translational Medicine (INCT-TM), Brazil
| |
Collapse
|
5
|
Wang W, Zhai GQ, Xin M, Li J, Liao JJ, Liang J, Li CB. Integrated network pharmacology and transcriptomics to reveal the mechanism of Passiflora against depressive disorder: An observational study. Medicine (Baltimore) 2024; 103:e39309. [PMID: 39465851 PMCID: PMC11479432 DOI: 10.1097/md.0000000000039309] [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: 10/02/2023] [Accepted: 07/25/2024] [Indexed: 10/29/2024] Open
Abstract
Relevant studies have pointed out that Passiflora could relieve depressive disorder (DD) related symptoms, such as anxiety and insomnia, but its mechanism in DD has not been reported. In this study, DD-related transcriptome data was extracted from the Gene Expression Omnibus (GEO) database. Subsequently, 50 differentially expressed genes (DEGs) were screened by "limma," and the enrichment analysis of these DEGs revealed that they were associated with neuro-inflammatory-related signaling pathways, including IL-17, TNF, NF-kappa B, etc signaling pathways. Then, CCDC58, CXCL5, EGR1, LOC101929855, SCML1, and THBS1 were screened as biomarkers of DD by the least absolute shrinkage and selection operator (LASSO) analysis. Moreover, Harmaline, Harmine, Quercetin, and Kaempferol were the key chemically active ingredients of Passiflora. Noticeable, THBS1 and Quercetin were connected closely. In addition, the quantitative real-time polymerase chain reaction (qRT-PCR) confirmed that the key biomarkers (EGR1 and THBS1) were significantly lowly expressed in DD samples. In summary, we identified 2 key biomarkers of DD and 4 key chemically active ingredients of Passiflora. The potential mechanism of antidepressant effect of DD associated with neuro-inflammatory responses and neurotransmitter function. These might related to the synergistic activity of its key active ingredients with TNF-α, IL-1β, IL-6, etc, which work with EGR1 and THBS1 to regulate IL-17, NF-kappa B, TNF, etc signaling pathways. These findings might help to deepen the understanding of the mechanism of Passiflora in clinical treatment of DD.
Collapse
Affiliation(s)
- Wei Wang
- Department of Urology, Guangxi Hospital Division of The First Affiliated Hospital, Sun Yat-sen University, Nanning, Guangxi Zhuang Autonomous Region, PR China
| | - Gao-Qiang Zhai
- Department of Urology, Guangxi Hospital Division of The First Affiliated Hospital, Sun Yat-sen University, Nanning, Guangxi Zhuang Autonomous Region, PR China
| | - Ming Xin
- Agro-food Science and Technology Research Institute, Guangxi Academy of Agricultural Sciences, Nanning, Guangxi Zhuang Autonomous Region, PR China
| | - Jun Li
- Department of Urology, Guangxi Hospital Division of The First Affiliated Hospital, Sun Yat-sen University, Nanning, Guangxi Zhuang Autonomous Region, PR China
| | - Jun-Juan Liao
- Department of Urology, Guangxi Hospital Division of The First Affiliated Hospital, Sun Yat-sen University, Nanning, Guangxi Zhuang Autonomous Region, PR China
| | - Jia Liang
- Psychopsychology Department, People’s Hospital of Guangxi Zhuang Autonomous Region, Nanning, Guangxi Zhuang Autonomous Region, PR China
| | - Chang-Bao Li
- Agro-food Science and Technology Research Institute, Guangxi Academy of Agricultural Sciences, Nanning, Guangxi Zhuang Autonomous Region, PR China
| |
Collapse
|
6
|
Arruda Sanchez T, Ramos LR, Araujo F, Schenberg EE, Yonamine M, Lobo I, de Araujo DB, Luna LE. Emotion regulation effects of Ayahuasca in experienced subjects during implicit aversive stimulation: An fMRI study. JOURNAL OF ETHNOPHARMACOLOGY 2024; 320:117430. [PMID: 37979818 DOI: 10.1016/j.jep.2023.117430] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/21/2023] [Revised: 08/15/2023] [Accepted: 11/12/2023] [Indexed: 11/20/2023]
Abstract
ETHNOPHARMACOLOGICAL RELEVANCE Ayahuasca is a beverage used in Amazonian traditional medicine and it has been part of the human experience for millennia as well as other different psychoactive plants. Although Ayahuasca has been proposed as potentially therapeutic as an anxiolytic and antidepressant, whilst no studies have been carried out so far investigating their direct effect on brain emotional processing. AIM OF THE STUDY This study aimed to measure the emotional acute effect of Ayahuasca on brain response to implicit aversive stimulation using a face recognition task in functional magnetic resonance imaging (fMRI). MATERIALS AND METHODS Nineteen male experienced Ayahuasca users participated in this study in two fMRI sessions before and after 50 min of the Ayahuasca ingestion. Subjects were presented with pictures of neutral (A) and aversive (B) (fearful or disgusted) faces from the Pictures of Facial Affect Series. Subjects were instructed to identify the gender of the faces (gender discrimination task) while the emotional content was implicit. Subjective mood states were also evaluated before Ayahuasca intake and after the second fMRI session, using a visual analogue mood scale (VAMS). RESULTS During the aversive stimuli, the activity in the bilateral amygdala was attenuated by Ayahuasca (qFDR<0.05). Furthermore, in an exploratory analysis of the effects after intake, Ayahuasca enhances the activation in the insular cortex bilaterally, as well as in the right dorsolateral prefrontal cortex (qFDR<0.05). In the psychometric VAMS scale, subjects reported attenuation of both anxiety and mental sedation (p < 0.01) during acute effects. CONCLUSIONS Together, all reported results including neuroimaging, behavioral data and psychometric self-report suggest that Ayahuasca can promote an emotion regulation mechanism in response to aversive stimuli with corresponding improved cognition including reduced anxiety and mental sedation.
Collapse
Affiliation(s)
- Tiago Arruda Sanchez
- Laboratory of Neuroimaging and Psychophysiology, Faculdade de Medicina, Universidade Federal do Rio de Janeiro (UFRJ), Rio de Janeiro, RJ, Brazil.
| | - Lucas Rego Ramos
- Laboratory of Neuroimaging and Psychophysiology, Faculdade de Medicina, Universidade Federal do Rio de Janeiro (UFRJ), Rio de Janeiro, RJ, Brazil
| | - Felipe Araujo
- Laboratory of Neuroimaging and Psychophysiology, Faculdade de Medicina, Universidade Federal do Rio de Janeiro (UFRJ), Rio de Janeiro, RJ, Brazil
| | | | - Mauricio Yonamine
- Faculdade de Ciências Farmacêuticas, Universidade de São Paulo (USP), São Paulo, SP, Brazil
| | - Isabela Lobo
- Instituto de Biodiversidade e Sustentabilidade (NUPEM), UFRJ, Macaé, RJ, Brazil
| | - Draulio Barros de Araujo
- Brain Institute / Hospital Universitário Onofre Lopes, Universidade Federal do Rio Grande do Norte (UFRN), Natal, RN, Brazil
| | - Luis Eduardo Luna
- Research Centre for the study of psychointegrator plants, Visionary Art and Consciousness - Wasiwaska, Florianópolis, SC, Brazil
| |
Collapse
|
7
|
Daldegan-Bueno D, Simionato NM, Favaro VM, Maia LO. The current state of ayahuasca research in animal models: A systematic review. Prog Neuropsychopharmacol Biol Psychiatry 2023; 125:110738. [PMID: 36863501 DOI: 10.1016/j.pnpbp.2023.110738] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/14/2022] [Revised: 02/22/2023] [Accepted: 02/25/2023] [Indexed: 03/04/2023]
Abstract
RATIONALE The psychedelic brew ayahuasca is increasingly being investigated for its therapeutic potential. Animal models are essential to investigate the pharmacological effects of ayahuasca since they can control important factors influencing it, such as the set and setting. OBJECTIVE Review and summarise data available on ayahuasca research using animal models. METHODS We systematically searched five databases (PubMed, Web of Science, EMBASE, LILACS and PsycInfo) for peer-reviewed studies in English, Portuguese or Spanish published up to July 2022. The search strategy included ayahuasca- and animal model-related terms adapted from the SYRCLE search syntax. RESULTS We identified 32 studies investigating ayahuasca effects on toxicological, behavioural and (neuro)biological parameters in rodents, primates and zebrafish. Toxicological results show that ayahuasca is safe at ceremonial-based doses but toxic at high doses. Behavioural results indicate an antidepressant effect and a potential to reduce the reward effects of ethanol and amphetamines, while the anxiety-related outcomes are yet inconclusive; also, ayahuasca can influence locomotor activity, highlighting the importance of controlling the analysis for locomotion when using tasks depending on it. Neurobiological results show that ayahuasca affects brain structures involved in memory, emotion and learning and that other neuropathways, besides the serotonergic action, are important in modulating its effects. CONCLUSIONS Studies using animal models indicate that ayahuasca is toxicologically safe in ceremonial-comparable doses and indicates a therapeutic potential for depression and substance use disorder while not supporting an anxiolytic effect. Essential gaps in the ayahuasca field can still be sufficed using animal models.
Collapse
Affiliation(s)
- Dimitri Daldegan-Bueno
- Interdisciplinary Cooperation for Ayahuasca Research and Outreach (ICARO), School of Medical Sciences, University of Campinas (UNICAMP), Campinas, Brazil; Centre for Applied Research in Mental Health and Addiction, Faculty of Health Sciences, Simon Fraser University, Vancouver, British Columbia, Canada
| | | | - Vanessa Manchim Favaro
- Division of Neuromuscular Diseases, Department of Neurology and Neurosurgery, Federal University of São Paulo (UNIFESP), São Paulo, Brazil
| | - Lucas Oliveira Maia
- Interdisciplinary Cooperation for Ayahuasca Research and Outreach (ICARO), School of Medical Sciences, University of Campinas (UNICAMP), Campinas, Brazil; Brain Institute, Federal University of Rio Grande do Norte (UFRN), Natal, Brazil; Interdisciplinary Center for Studies in Palliative Care (CIECP), School of Nursing, Federal University of Alfenas (UNIFAL-MG), Alfenas, Brazil
| |
Collapse
|
8
|
Stocco MR, Tyndale RF. Cytochrome P450 enzymes and metabolism of drugs and neurotoxins within the mammalian brain. ADVANCES IN PHARMACOLOGY (SAN DIEGO, CALIF.) 2022; 95:73-106. [PMID: 35953164 DOI: 10.1016/bs.apha.2022.04.003] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/15/2023]
Abstract
Cytochrome P450 enzymes (CYPs) that metabolize xenobiotics are expressed and active in the brain. These CYPs contribute to the metabolism of many centrally acting compounds, including clinically used drugs, drugs of abuse, and neurotoxins. Although CYP levels are lower in the brain than in the liver, they may influence central substrate and metabolite concentrations, which could alter resulting centrally-mediated responses to these compounds. Additionally, xenobiotic metabolizing CYPs are highly variable due to genetic polymorphisms and regulation by endogenous and xenobiotic molecules. In the brain, these CYPs are sensitive to xenobiotic induction. As a result, CYPs in the brain vary widely, including among humans, and this CYP variation may influence central metabolism and resulting response to centrally acting compounds. It has been demonstrated, using experimental manipulation of CYP activity in vivo selectively within the brain, that CYP metabolism in the brain alters central substrate and metabolite concentrations, as well as drug response and neurotoxic effects. This suggests that variability in xenobiotic metabolizing CYPs in the human brain may meaningfully contribute to individual differences in response to, and effects of, centrally acting drugs and neurotoxins. This chapter will provide an overview of CYP expression in the brain, endogenous- and xenobiotic-mediated CYP regulation, and the functional impact of CYP-mediated metabolism of drugs and neurotoxins in the brain, with a focus on experimental approaches in mice, rats, and non-human primates, and a discussion regarding the potential role of xenobiotic metabolizing CYPs in the human brain.
Collapse
Affiliation(s)
- Marlaina R Stocco
- Department of Psychological and Brain Sciences, University of California, Santa Barbara, Santa Barbara, CA, United States
| | - Rachel F Tyndale
- Department of Pharmacology and Toxicology, University of Toronto, Toronto, ON, Canada; Campbell Family Mental Health Research Institute, CAMH, Toronto, ON, Canada; Department of Psychiatry, University of Toronto, Toronto, ON, Canada.
| |
Collapse
|
9
|
Mosaffa S, Ahmadi H, Khakpai F, Ebrahimi-Ghiri M, Zarrindast MR. Synergistic antidepressant- and anxiolytic-like effects of harmaline along with cinanserin in acute restraint stress-treated mice. Psychopharmacology (Berl) 2021; 238:259-269. [PMID: 33190164 PMCID: PMC7666640 DOI: 10.1007/s00213-020-05679-6] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/06/2020] [Accepted: 10/07/2020] [Indexed: 02/06/2023]
Abstract
RATIONALE Acute restraint stress (ARS) is an experimental paradigm used for the induction of rodent models of stress-produced neuropsychiatric disorders, such as depression and anxiety. β-carbolines and serotonin (5-HT) systems are involved in the modulation of depression and anxiety behaviors. OBJECTIVE This study was designed to examine the effects of intracerebroventricular (i.c.v.) injection of cinanserin (5-HT2 receptor antagonist) on harmaline-induced responses on depression- and anxiety-like behaviors in the ARS mice. METHODS For i.c.v. infusion, guide cannula was surgically implanted in the left lateral ventricle of mice. The ARS model was conducted via movement restraint at a period of 4 h. Depression- and anxiety-related behaviors were evaluated by forced swim test (FST) and elevated plus maze (EPM), respectively. RESULTS The results displayed that the ARS mice showed depressive- and anxiety-like responses. I.p. administration of different doses of harmaline (0.31, 0.625 and 1.25 mg/kg) or i.c.v. microinjection of cinanserin (1, 2.5, and 5 μg/mouse) blocked depression- and anxiogenic-like behaviors in the ARS mice. Furthermore, co-administration of harmaline (1.25 mg/kg; i.p.) and cinanserin (5 μg/mouse; i.c.v.) prevented the depression- and anxiogenic-like effects in the ARS mice. We found a synergistic antidepressant- and anxiolytic-like effects of harmaline and cinanserin in the ARS mice. CONCLUSIONS These results propose an interaction between harmaline and cinanserin to prevent depressive- and anxiogenic-like behaviors in the ARS mice.
Collapse
Affiliation(s)
- Sajedeh Mosaffa
- Department of Pharmacology, School of Medicine, Tehran University of Medical Sciences, P.O.Box 13145-784, Tehran, Iran
| | - Hanieh Ahmadi
- Department of Pharmacology, School of Medicine, Tehran University of Medical Sciences, P.O.Box 13145-784, Tehran, Iran
| | - Fatemeh Khakpai
- Cognitive and Neuroscience Research Center (CNRC), Tehran Medical Sciences, Islamic Azad University, Tehran, Iran
| | | | - Mohammad-Reza Zarrindast
- Department of Pharmacology, School of Medicine, Tehran University of Medical Sciences, P.O.Box 13145-784, Tehran, Iran. .,Iranian National Center for Addiction Studies, Tehran University of Medical Sciences, Tehran, Iran. .,Department of Neuroendocrinology, Endocrinology and Metabolism Clinical Sciences Institute, Tehran University of Medical Sciences, Tehran, Iran.
| |
Collapse
|
10
|
Ruffell S, Netzband N, Bird C, Young AH, Juruena MF. The pharmacological interaction of compounds in ayahuasca: a systematic review. REVISTA BRASILEIRA DE PSIQUIATRIA (SAO PAULO, BRAZIL : 1999) 2020; 42:646-656. [PMID: 32638916 PMCID: PMC7678905 DOI: 10.1590/1516-4446-2020-0884] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Received: 01/31/2020] [Accepted: 05/06/2020] [Indexed: 02/08/2023]
Abstract
Ayahuasca is a South American psychoactive plant brew used as traditional medicine in spiritual and in cultural rituals. This is a review of the current understanding about the pharmacological mechanisms that may be interacting in ayahuasca. Searches were performed using PubMed, PsycINFO, and Web of Science databases and 16 papers were selected. As hypothesized, the primary narrative in existing research revolved around prevention of deamination of N,N-dimethyltryptamine (N,N-DMT, also referred to as DMT) by monoamine oxidase inhibitors (MAOIs) in ayahuasca. Two of the constituents, DMT and harmine, have been studied more than the secondary harmala alkaloids. At present, it is unclear whether the pharmacological interactions in ayahuasca act synergistically or additively to produce psychoactive drug effects. The included studies suggest that our current understanding of the preparation's synergistic mechanisms is limited and that more complex processes may be involved; there is not yet enough data to determine any potential synergistic interaction between the known compounds in ayahuasca. Our pharmacological understanding of its compounds must be increased to avoid the potential risks of ayahuasca use.
Collapse
Affiliation(s)
- Simon Ruffell
- Institute of Psychiatry, Psychology and Neuroscience, King’s College London, London, UK
- Correspondence: Mario F. Juruena, Institute of Psychiatry, Psychology and Neuroscience, King’s College London, Room M3.24, PO72 De Crespigny Park, Denmark Hill, London SE5 8AF, UK. E-mail:
| | - Nige Netzband
- University of the West of England, Bristol, UK
- Correspondence: Mario F. Juruena, Institute of Psychiatry, Psychology and Neuroscience, King’s College London, Room M3.24, PO72 De Crespigny Park, Denmark Hill, London SE5 8AF, UK. E-mail:
| | - Catherine Bird
- Institute of Psychiatry, Psychology and Neuroscience, King’s College London, London, UK
| | - Allan H. Young
- Institute of Psychiatry, Psychology and Neuroscience, King’s College London, London, UK
| | - Mario F. Juruena
- Institute of Psychiatry, Psychology and Neuroscience, King’s College London, London, UK
- Correspondence: Mario F. Juruena, Institute of Psychiatry, Psychology and Neuroscience, King’s College London, Room M3.24, PO72 De Crespigny Park, Denmark Hill, London SE5 8AF, UK. E-mail:
| |
Collapse
|
11
|
Stocco MR, Tolledo C, Wadji FB, Gonzalez FJ, Miksys S, Tyndale RF. Human CYP2D6 in the Brain Is Protective Against Harmine-Induced Neurotoxicity: Evidence from Humanized CYP2D6 Transgenic Mice. Mol Neurobiol 2020; 57:4608-4621. [PMID: 32761352 PMCID: PMC8865091 DOI: 10.1007/s12035-020-02050-w] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/26/2020] [Accepted: 07/28/2020] [Indexed: 10/25/2022]
Abstract
CYP2D6 metabolically inactivates several neurotoxins, including beta-carbolines, which are implicated in neurodegenerative diseases. Variation in CYP2D6 within the brain may alter local inactivation of neurotoxic beta-carbolines, thereby influencing neurotoxicity. The beta-carboline harmine, which induces hypothermia and tremor, is metabolized by CYP2D6 to the non-hypothermic/non-tremorgenic harmol. Transgenic mice (TG), expressing human CYP2D6 in addition to their endogenous mouse CYP2D, experience less harmine-induced hypothermia and tremor compared with wild-type mice (WT). We first sought to elucidate the role of CYP2D in general within the brain in harmine-induced hypothermia and tremor severity. A 4-h intracerebroventricular (ICV) pretreatment with the CYP2D inhibitor propranolol increased harmine-induced hypothermia and tremor in TG and increased harmine-induced hypothermia in WT. We next sought to specifically demonstrate that human CYP2D6 expressed in TG brain altered harmine response severity. A 24-h ICV propranolol pretreatment, which selectively and irreversibly inhibits human CYP2D6 in TG brain, increased harmine-induced hypothermia. This 24-h pretreatment had no impact on harmine response in WT, as propranolol is not an irreversible inhibitor of mouse CYP2D in the brain, thus confirming no off-target effects of ICV propranolol pretreatment. Human CYP2D6 activity in TG brain was sufficient in vivo to mitigate harmine-induced neurotoxicity. These findings suggest that human CYP2D6 in the brain is protective against beta-carboline-induced neurotoxicity and that the extensive interindividual variability in CYP2D6 expression in human brain may contribute to variation in susceptibility to certain neurotoxin-associated neurodegenerative disorders.
Collapse
Affiliation(s)
- Marlaina R Stocco
- Campbell Family Mental Health Research Institute, Centre for Addiction and Mental Health (CAMH), Toronto, Ontario, Canada
- Department of Pharmacology and Toxicology, University of Toronto, Toronto, Ontario, Canada
| | - Cole Tolledo
- Campbell Family Mental Health Research Institute, Centre for Addiction and Mental Health (CAMH), Toronto, Ontario, Canada
- Department of Pharmacology and Toxicology, University of Toronto, Toronto, Ontario, Canada
| | - Fariba Baghai Wadji
- Department of Pharmacology and Toxicology, University of Toronto, Toronto, Ontario, Canada
| | - Frank J Gonzalez
- Laboratory of Metabolism, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, MD, USA
| | - Sharon Miksys
- Campbell Family Mental Health Research Institute, Centre for Addiction and Mental Health (CAMH), Toronto, Ontario, Canada
- Department of Pharmacology and Toxicology, University of Toronto, Toronto, Ontario, Canada
| | - Rachel F Tyndale
- Campbell Family Mental Health Research Institute, Centre for Addiction and Mental Health (CAMH), Toronto, Ontario, Canada.
- Department of Pharmacology and Toxicology, University of Toronto, Toronto, Ontario, Canada.
- Department of Psychiatry, University of Toronto, 1 King's College Circle, Toronto, Ontario, M5S 1A8, Canada.
| |
Collapse
|
12
|
Rao T, Tan Z, Peng J, Guo Y, Chen Y, Zhou H, Ouyang D. The pharmacogenetics of natural products: A pharmacokinetic and pharmacodynamic perspective. Pharmacol Res 2019; 146:104283. [PMID: 31129178 DOI: 10.1016/j.phrs.2019.104283] [Citation(s) in RCA: 56] [Impact Index Per Article: 9.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/04/2019] [Revised: 05/17/2019] [Accepted: 05/21/2019] [Indexed: 11/19/2022]
Abstract
Natural products have represented attractive alternatives for disease prevention and treatment over the course of human history and have contributed to the development of modern drugs. These natural products possess beneficial efficacies as well as adverse efffects, which vary largely among individuals because of genetic variations in their pharmacokinetics and pharmacodynamics. As with other synthetic chemical drugs, the dosing of natural products can be optimized to improve efficacy and reduce toxicity according to the pharmacogenetic properties. With the emergence and development of pharmacogenomics, it is possible to discover and identify the targets/mechanisms of pharmacological effects and therapeutic responses of natural products effectively and efficiently on the whole genome level. This review covers the effects of genetic variations in drug metabolizing enzymes, drug transporters, and direct and indirect interactions with the pharmacological targets/pathways on the individual response to natural products, and provides suggestions on dosing regimen adjustments of natural products based on their pharmacokinetic and pharmacogenetic paratmeters. Finally, we provide our viewpoints on the importance and necessity of pharmacogenetic and pharmacogenomic research of natural products in natural medicine's rational development and clinical application of precision medicine.
Collapse
Affiliation(s)
- Tai Rao
- Department of Clinical Pharmacology, Xiangya Hospital, Central South University, Changsha, PR China; Institute of Clinical Pharmacology, Central South University, Changsha, PR China; Engineering Research Center of Applied Technology of Pharmacogenomics, Ministry of Education, Changsha, PR China; National Clinical Research Center for Geriatric Disorders, Changsha, Hunan, PR China
| | - Zhirong Tan
- Department of Clinical Pharmacology, Xiangya Hospital, Central South University, Changsha, PR China; Institute of Clinical Pharmacology, Central South University, Changsha, PR China; Engineering Research Center of Applied Technology of Pharmacogenomics, Ministry of Education, Changsha, PR China; National Clinical Research Center for Geriatric Disorders, Changsha, Hunan, PR China
| | - Jingbo Peng
- Department of Clinical Pharmacology, Xiangya Hospital, Central South University, Changsha, PR China; Institute of Clinical Pharmacology, Central South University, Changsha, PR China; Engineering Research Center of Applied Technology of Pharmacogenomics, Ministry of Education, Changsha, PR China; National Clinical Research Center for Geriatric Disorders, Changsha, Hunan, PR China
| | - Ying Guo
- Department of Clinical Pharmacology, Xiangya Hospital, Central South University, Changsha, PR China; Institute of Clinical Pharmacology, Central South University, Changsha, PR China; Engineering Research Center of Applied Technology of Pharmacogenomics, Ministry of Education, Changsha, PR China; National Clinical Research Center for Geriatric Disorders, Changsha, Hunan, PR China
| | - Yao Chen
- Department of Clinical Pharmacology, Xiangya Hospital, Central South University, Changsha, PR China; Institute of Clinical Pharmacology, Central South University, Changsha, PR China; Engineering Research Center of Applied Technology of Pharmacogenomics, Ministry of Education, Changsha, PR China; National Clinical Research Center for Geriatric Disorders, Changsha, Hunan, PR China
| | - Honghao Zhou
- Department of Clinical Pharmacology, Xiangya Hospital, Central South University, Changsha, PR China; Institute of Clinical Pharmacology, Central South University, Changsha, PR China; Engineering Research Center of Applied Technology of Pharmacogenomics, Ministry of Education, Changsha, PR China; National Clinical Research Center for Geriatric Disorders, Changsha, Hunan, PR China
| | - Dongsheng Ouyang
- Department of Clinical Pharmacology, Xiangya Hospital, Central South University, Changsha, PR China; Institute of Clinical Pharmacology, Central South University, Changsha, PR China; Engineering Research Center of Applied Technology of Pharmacogenomics, Ministry of Education, Changsha, PR China; National Clinical Research Center for Geriatric Disorders, Changsha, Hunan, PR China.
| |
Collapse
|
13
|
Ebrahimi-Ghiri M, Nasehi M, Zarrindast MR. Anxiolytic and antidepressant effects of ACPA and harmaline co-treatment. Behav Brain Res 2019; 364:296-302. [DOI: 10.1016/j.bbr.2019.02.034] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/29/2018] [Revised: 02/12/2019] [Accepted: 02/20/2019] [Indexed: 01/19/2023]
|
14
|
Bissig KD, Han W, Barzi M, Kovalchuk N, Ding L, Fan X, Pankowicz FP, Zhang QY, Ding X. P450-Humanized and Human Liver Chimeric Mouse Models for Studying Xenobiotic Metabolism and Toxicity. Drug Metab Dispos 2018; 46:1734-1744. [PMID: 30093418 DOI: 10.1124/dmd.118.083303] [Citation(s) in RCA: 39] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/25/2018] [Accepted: 08/03/2018] [Indexed: 01/01/2023] Open
Abstract
Preclinical evaluation of drug candidates in experimental animal models is an essential step in drug development. Humanized mouse models have emerged as a promising alternative to traditional animal models. The purpose of this mini-review is to provide a brief survey of currently available mouse models for studying human xenobiotic metabolism. Here, we describe both genetic humanization and human liver chimeric mouse models, focusing on the advantages and limitations while outlining their key features and applications. Although this field of biomedical science is relatively young, these humanized mouse models have the potential to transform preclinical drug testing and eventually lead to a more cost-effective and rapid development of new therapies.
Collapse
Affiliation(s)
- Karl-Dimiter Bissig
- Baylor College of Medicine, Houston, Texas (K.-D.B., M.B., F.P.P.); and Department of Pharmacology and Toxicology, College of Pharmacy, University of Arizona, Tucson, Arizona (W.H., N.K., L.D., X.F., Q.-Y.Z., X.D.)
| | - Weiguo Han
- Baylor College of Medicine, Houston, Texas (K.-D.B., M.B., F.P.P.); and Department of Pharmacology and Toxicology, College of Pharmacy, University of Arizona, Tucson, Arizona (W.H., N.K., L.D., X.F., Q.-Y.Z., X.D.)
| | - Mercedes Barzi
- Baylor College of Medicine, Houston, Texas (K.-D.B., M.B., F.P.P.); and Department of Pharmacology and Toxicology, College of Pharmacy, University of Arizona, Tucson, Arizona (W.H., N.K., L.D., X.F., Q.-Y.Z., X.D.)
| | - Nataliia Kovalchuk
- Baylor College of Medicine, Houston, Texas (K.-D.B., M.B., F.P.P.); and Department of Pharmacology and Toxicology, College of Pharmacy, University of Arizona, Tucson, Arizona (W.H., N.K., L.D., X.F., Q.-Y.Z., X.D.)
| | - Liang Ding
- Baylor College of Medicine, Houston, Texas (K.-D.B., M.B., F.P.P.); and Department of Pharmacology and Toxicology, College of Pharmacy, University of Arizona, Tucson, Arizona (W.H., N.K., L.D., X.F., Q.-Y.Z., X.D.)
| | - Xiaoyu Fan
- Baylor College of Medicine, Houston, Texas (K.-D.B., M.B., F.P.P.); and Department of Pharmacology and Toxicology, College of Pharmacy, University of Arizona, Tucson, Arizona (W.H., N.K., L.D., X.F., Q.-Y.Z., X.D.)
| | - Francis P Pankowicz
- Baylor College of Medicine, Houston, Texas (K.-D.B., M.B., F.P.P.); and Department of Pharmacology and Toxicology, College of Pharmacy, University of Arizona, Tucson, Arizona (W.H., N.K., L.D., X.F., Q.-Y.Z., X.D.)
| | - Qing-Yu Zhang
- Baylor College of Medicine, Houston, Texas (K.-D.B., M.B., F.P.P.); and Department of Pharmacology and Toxicology, College of Pharmacy, University of Arizona, Tucson, Arizona (W.H., N.K., L.D., X.F., Q.-Y.Z., X.D.)
| | - Xinxin Ding
- Baylor College of Medicine, Houston, Texas (K.-D.B., M.B., F.P.P.); and Department of Pharmacology and Toxicology, College of Pharmacy, University of Arizona, Tucson, Arizona (W.H., N.K., L.D., X.F., Q.-Y.Z., X.D.)
| |
Collapse
|
15
|
Harmine enhances GABAergic transmission onto basoamygdala projection neurons in mice. Brain Res Bull 2018; 137:294-300. [DOI: 10.1016/j.brainresbull.2018.01.004] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/03/2017] [Revised: 12/12/2017] [Accepted: 01/05/2018] [Indexed: 12/22/2022]
|
16
|
Wagner DJ, Duan H, Chapron A, Lee RW, Wang J. Potent inhibition of human organic cation transporter 2 (hOCT2) by β-carboline alkaloids. Xenobiotica 2017; 47:1112-1120. [PMID: 27977936 PMCID: PMC5648609 DOI: 10.1080/00498254.2016.1271160] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/08/2016] [Accepted: 12/07/2016] [Indexed: 02/08/2023]
Abstract
1. Beta-carbolines are indole alkaloids with a wide range of pharmacological and toxicological activities. Beta-carbolines are structurally related to the neurotoxin 1-methyl-4-phenylpyridinium (MPP+), a known substrate of organic cation transporters (OCTs). The goal of this study is to determine the interaction of β-carbolines with human OCT1, 2, and 3 (SLC22A1-3). 2. Dose-dependent inhibition studies were performed for five commercially available β-carbolines using a fluorescent substrate assay in HEK293 cells stably expressing hOCT1-3. The substrate potential was evaluated by uptake assays and the impact of active transport on cellular toxicity examined. 3. All tested β-carbolines potently inhibited hOCT2 with IC50 values in the sub- or low micromolar range. Harmaline is the most potent hOCT2 inhibitor (IC50 = 0.50 ± 0.08 μM). hOCT1 and hOCT3 are less sensitive to β-carboline inhibition. Harmaline, norharmanium, and 2,9-dimethyl-4,9-dihydro-3H-β-carbolinium accumulated 2- to 7-fold higher in cells expressing hOCT1-3. HEK293 cells expressing hOCT1-3 were 6.5- to 13-fold more sensitive to harmane and norharmanium toxicity. 4. Our data support a significant role of hOCT1-3 in tissue uptake and disposition of β-carbolines. Importantly, the potent inhibition of hOCT2 by β-carbolines also raises the concern of potential drug interactions between naturally occurring bioactive alkaloids and drugs eliminated by hOCT2.
Collapse
Affiliation(s)
- David J. Wagner
- Department of Pharmaceutics, University of Washington, Seattle, WA, USA
| | - Haichuan Duan
- Department of Pharmaceutics, University of Washington, Seattle, WA, USA
| | - Alenka Chapron
- Department of Pharmaceutics, University of Washington, Seattle, WA, USA
| | - Richard W. Lee
- School of Pharmacy, University of Washington, Seattle, WA, USA
| | - Joanne Wang
- Department of Pharmaceutics, University of Washington, Seattle, WA, USA
| |
Collapse
|
17
|
Li S, Zhang Y, Deng G, Wang Y, Qi S, Cheng X, Ma Y, Xie Y, Wang C. Exposure Characteristics of the Analogous β-Carboline Alkaloids Harmaline and Harmine Based on the Efflux Transporter of Multidrug Resistance Protein 2. Front Pharmacol 2017; 8:541. [PMID: 28871225 PMCID: PMC5566973 DOI: 10.3389/fphar.2017.00541] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/15/2017] [Accepted: 08/03/2017] [Indexed: 12/17/2022] Open
Abstract
Harmaline and harmine occur naturally in plants and are distributed endogenously in human and animal tissues. The two β-carboline alkaloids possess potential for treating Alzheimer's disease, Parkinson's disease, depression and other central nervous system diseases. However, studies have showed that the two compounds have similar structures but with quite different bioavailability. The aim of this study was to elucidate the exposure difference and characterize the in vitro transport, metabolism, and pharmacokinetic properties of harmaline and harmine. The results showed that the harmaline and harmine transport across the Caco-2 and MDCK cell monolayers was varied as the time, concentration, pH and temperature changed. The absorption of harmaline and harmine was significantly decreased when ES (OATPs inhibitor), TEA (OCTs/OCTNs substrate), NaN3 (adenosine triphosphate inhibitor), or sodium vanadate (ATPase Na+/K+-dependent inhibitor) was added. However, when given MK571 and probenecid (the typical MRP2 inhibitor), the PappAB of harmine was increased (1.62- and 1.27-folds), and the efflux ratio was decreased from 1.59 to 0.98 and from 1.59 to 1.19, respectively. In addition, the uptake ratio of harmine at 1 μM was >2.65 in the membrane vesicles expressing human MRP2. Furthermore, harmine could slightly up-regulate the expression of MRP2, which implying harmine might be the substrate of MRP2. Particularly, the CLint-value for harmine was ~1.49-folds greater than that of harmaline in human liver microsomes. It was worth noting that the F-value of harmine was increased 1.96-folds after harmine co-administration with probenecid. To summarize, comprehensive analysis indicated that harmaline and harmine were absorbed by transcellular passive diffusion and a pH- and Na+-dependent mechanism might be mediated by OATPs and OCTs/OCTNs. MRP2 but MDR1 or BCRP might be involved in the transport of harmine. Furthermore, harmine was more unstable and easily metabolized than harmaline. All these findings suggested that harmine not only appears be an MRP2 substrate, but also possesses weak metabolic stability, and eventually leads to a low oral bioavailability. Taken together, the elucidated absorption, transport, metabolism as well as pharmacokinetic characteristics of harmaline and harmine provide useful information for designing delivery systems, pharmacological applications and avoiding drug-drug interactions.
Collapse
Affiliation(s)
- Shuping Li
- The MOE Key Laboratory for Standardization of Chinese Medicines, The SATCM Key Laboratory for New Resources and Quality Evaluation of Chinese, Institute of Chinese Materia Medica, Shanghai University of Traditional Chinese MedicineShanghai, China
| | - Yunpeng Zhang
- The MOE Key Laboratory for Standardization of Chinese Medicines, The SATCM Key Laboratory for New Resources and Quality Evaluation of Chinese, Institute of Chinese Materia Medica, Shanghai University of Traditional Chinese MedicineShanghai, China
| | - Gang Deng
- The MOE Key Laboratory for Standardization of Chinese Medicines, The SATCM Key Laboratory for New Resources and Quality Evaluation of Chinese, Institute of Chinese Materia Medica, Shanghai University of Traditional Chinese MedicineShanghai, China
| | - Yuwen Wang
- The MOE Key Laboratory for Standardization of Chinese Medicines, The SATCM Key Laboratory for New Resources and Quality Evaluation of Chinese, Institute of Chinese Materia Medica, Shanghai University of Traditional Chinese MedicineShanghai, China
| | - Shenglan Qi
- The MOE Key Laboratory for Standardization of Chinese Medicines, The SATCM Key Laboratory for New Resources and Quality Evaluation of Chinese, Institute of Chinese Materia Medica, Shanghai University of Traditional Chinese MedicineShanghai, China
| | - Xuemei Cheng
- The MOE Key Laboratory for Standardization of Chinese Medicines, The SATCM Key Laboratory for New Resources and Quality Evaluation of Chinese, Institute of Chinese Materia Medica, Shanghai University of Traditional Chinese MedicineShanghai, China.,Shanghai R&D Centre for Standardization of Chinese MedicinesShanghai, China
| | - Yueming Ma
- Laboratory of Pharmacokinetics, Shanghai University of Traditional Chinese MedicineShanghai, China
| | - Yan Xie
- Research Center for Health and Nutrition, Shanghai University of Traditional Chinese MedicineShanghai, China
| | - Changhong Wang
- The MOE Key Laboratory for Standardization of Chinese Medicines, The SATCM Key Laboratory for New Resources and Quality Evaluation of Chinese, Institute of Chinese Materia Medica, Shanghai University of Traditional Chinese MedicineShanghai, China.,Shanghai R&D Centre for Standardization of Chinese MedicinesShanghai, China
| |
Collapse
|
18
|
Dahmardeh N, Asadi-Shekaari M, Arjmand S, Kalantaripour T, Basiri M, Shabani M. Modulation of sphingosine-1-phosphate receptor ameliorates harmaline-induced essential tremor in rat. Neurosci Lett 2017. [PMID: 28627375 DOI: 10.1016/j.neulet.2017.06.015] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
Essential tremor (ET) is one of the most common movement disorders with unknown etiology. Despite lack of effective clinical treatments, some potential therapeutic factors and modulation of some neurotransmitters have been utilized to ameliorate motor symptoms in the animal models of tremor. In the current study, male Wistar rats (n=10 in each group) weighing 40-60g were divided into vehicle control groups (saline or DMSO), saline/DMSO+harmaline (30mg/kg, i.p.)+fingolimod (FTY720) (1mg/kg, i.p, 1h before harmaline injection) groups. Open field, rotarod, wire grip and foot print tests were used to evaluate motor function. The results demonstrated that administration of FTY720 can improve harmaline-induced tremor in rats. Moreover, FTY720 ameliorated gait disturbance. The results showed that FTY720 can recover step width, left and right step length; however, FTY720 failed to recover mobility duration. FTY720 also improved falling time and time spent in wire grip and rotarod, respectively. The current study provides the first evidence for the effectiveness of FTY720 on motor function in the harmaline model of ET. Furthermore, neuroprotective effects of FTY720 demonstrated in this study offer sphingosine-1-phosphate receptor (S1PR) modulators as a potential neuroprotective candidate against substance-induced tremor and a possible strategy for the treatment of patients with tremor.
Collapse
Affiliation(s)
- Narjes Dahmardeh
- Intracellular Recording Lab, Kerman Neuroscience Research Center, Neuropharmacology Institute, Kerman University of Medical Sciences, Kerman, Iran; Department of Anatomical Sciences, Afzalipour Medical Faculty, Kerman University of Medical Sciences, Kerman, Iran
| | - Majid Asadi-Shekaari
- Intracellular Recording Lab, Kerman Neuroscience Research Center, Neuropharmacology Institute, Kerman University of Medical Sciences, Kerman, Iran
| | - Shokouh Arjmand
- Intracellular Recording Lab, Kerman Neuroscience Research Center, Neuropharmacology Institute, Kerman University of Medical Sciences, Kerman, Iran
| | - Tajpari Kalantaripour
- Department of Physiology, School of Medicine, Islamic Azad University, Branch of Kerman, Kerman, Iran
| | - Mohsen Basiri
- Department of Anatomical Sciences, Afzalipour Medical Faculty, Kerman University of Medical Sciences, Kerman, Iran
| | - Mohammad Shabani
- Intracellular Recording Lab, Kerman Neuroscience Research Center, Neuropharmacology Institute, Kerman University of Medical Sciences, Kerman, Iran.
| |
Collapse
|
19
|
Dos Santos RG, Osório FL, Crippa JAS, Hallak JEC. Antidepressive and anxiolytic effects of ayahuasca: a systematic literature review of animal and human studies. BRAZILIAN JOURNAL OF PSYCHIATRY 2017; 38:65-72. [PMID: 27111702 PMCID: PMC7115465 DOI: 10.1590/1516-4446-2015-1701] [Citation(s) in RCA: 82] [Impact Index Per Article: 10.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 03/02/2015] [Accepted: 05/05/2015] [Indexed: 01/24/2023]
Abstract
OBJECTIVE To conduct a systematic literature review of animal and human studies reporting anxiolytic or antidepressive effects of ayahuasca or some of its isolated alkaloids (dimethyltryptamine, harmine, tetrahydroharmine, and harmaline). METHODS Papers published until 3 April 2015 were retrieved from the PubMed, LILACS and SciELO databases following a comprehensive search strategy and using a predetermined set of criteria for article selection. RESULTS Five hundred and fourteen studies were identified, of which 21 met the established criteria. Studies in animals have shown anxiolytic and antidepressive effects of ayahuasca, harmine, and harmaline, and experimental studies in humans and mental health assessments of experienced ayahuasca consumers also suggest that ayahuasca is associated with reductions in anxiety and depressive symptoms. A pilot study reported rapid antidepressive effects of a single ayahuasca dose in six patients with recurrent depression. CONCLUSION Considering the need for new drugs that produce fewer adverse effects and are more effective in reducing anxiety and depression symptomatology, the described effects of ayahuasca and its alkaloids should be further investigated.
Collapse
Affiliation(s)
- Rafael G Dos Santos
- Departamento de Neurociências e Ciências do Comportamento, Faculdade de Medicina de Ribeirão Preto, Universidade de São Paulo (USP), Ribeirão Preto, SP, Brazil
| | - Flávia L Osório
- Departamento de Neurociências e Ciências do Comportamento, Faculdade de Medicina de Ribeirão Preto, Universidade de São Paulo (USP), Ribeirão Preto, SP, Brazil
| | - José Alexandre S Crippa
- Departamento de Neurociências e Ciências do Comportamento, Faculdade de Medicina de Ribeirão Preto, Universidade de São Paulo (USP), Ribeirão Preto, SP, Brazil
| | - Jaime E C Hallak
- Departamento de Neurociências e Ciências do Comportamento, Faculdade de Medicina de Ribeirão Preto, Universidade de São Paulo (USP), Ribeirão Preto, SP, Brazil
| |
Collapse
|
20
|
Development of a mechanism-based pharmacokinetic/pharmacodynamic model to characterize the thermoregulatory effects of serotonergic drugs in mice. Acta Pharm Sin B 2016; 6:492-503. [PMID: 27709018 PMCID: PMC5045556 DOI: 10.1016/j.apsb.2016.07.007] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/06/2016] [Revised: 04/25/2016] [Accepted: 04/27/2016] [Indexed: 01/01/2023] Open
Abstract
We have shown recently that concurrent harmaline, a monoamine oxidase-A inhibitor (MAOI), potentiates serotonin (5-HT) receptor agonist 5-methoxy-N,N-dimethyltryptamine (5-MeO-DMT)-induced hyperthermia. The objective of this study was to develop an integrated pharmacokinetic/pharmacodynamic (PK/PD) model to characterize and predict the thermoregulatory effects of such serotonergic drugs in mice. Physiological thermoregulation was described by a mechanism-based indirect-response model with adaptive feedback control. Harmaline-induced hypothermia and 5-MeO-DMT–elicited hyperthermia were attributable to the loss of heat through the activation of 5-HT1A receptor and thermogenesis via the stimulation of 5-HT2A receptor, respectively. Thus serotonergic 5-MeO-DMT–induced hyperthermia was readily distinguished from handling/injection stress-provoked hyperthermic effects. This PK/PD model was able to simultaneously describe all experimental data including the impact of drug-metabolizing enzyme status on 5-MeO-DMT and harmaline PK properties, and drug- and stress-induced simple hypo/hyperthermic and complex biphasic effects. Furthermore, the modeling results revealed a 4-fold decrease of apparent SC50 value (1.88–0.496 µmol/L) for 5-MeO-DMT when harmaline was co-administered, providing a quantitative assessment for the impact of concurrent MAOI harmaline on 5-MeO-DMT–induced hyperthermia. In addition, the hyperpyrexia caused by toxic dose combinations of harmaline and 5-MeO-DMT were linked to the increased systemic exposure to harmaline rather than 5-MeO-DMT, although the body temperature profiles were mispredicted by the model. The results indicate that current PK/PD model may be used as a new conceptual framework to define the impact of serotonergic agents and stress factors on thermoregulation.
Collapse
|
21
|
Li S, Teng L, Liu W, Cheng X, Jiang B, Wang Z, Wang C. Interspecies metabolic diversity of harmaline and harmine in in vitro
11 mammalian liver microsomes. Drug Test Anal 2016; 9:754-768. [DOI: 10.1002/dta.2028] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/22/2016] [Revised: 05/31/2016] [Accepted: 06/05/2016] [Indexed: 12/18/2022]
Affiliation(s)
- Shuping Li
- Institute of Chinese Materia Medica; Shanghai University of Traditional Chinese Medicine; The MOE Key Laboratory for Standardization of Chinese Medicines and The SATCM Key Laboratory for New Resources and Quality Evaluation of Chinese Medicine; Shanghai China
| | - Liang Teng
- Institute of Chinese Materia Medica; Shanghai University of Traditional Chinese Medicine; The MOE Key Laboratory for Standardization of Chinese Medicines and The SATCM Key Laboratory for New Resources and Quality Evaluation of Chinese Medicine; Shanghai China
- Pharmacy Department; the First Affiliated Hospital of Xinjiang Medical University; Urumqi China
| | - Wei Liu
- Institute of Chinese Materia Medica; Shanghai University of Traditional Chinese Medicine; The MOE Key Laboratory for Standardization of Chinese Medicines and The SATCM Key Laboratory for New Resources and Quality Evaluation of Chinese Medicine; Shanghai China
| | - Xuemei Cheng
- Institute of Chinese Materia Medica; Shanghai University of Traditional Chinese Medicine; The MOE Key Laboratory for Standardization of Chinese Medicines and The SATCM Key Laboratory for New Resources and Quality Evaluation of Chinese Medicine; Shanghai China
- Shanghai R&D Centre for Standardization of Chinese Medicines; Shanghai China
| | - Bo Jiang
- Institute of Chinese Materia Medica; Shanghai University of Traditional Chinese Medicine; The MOE Key Laboratory for Standardization of Chinese Medicines and The SATCM Key Laboratory for New Resources and Quality Evaluation of Chinese Medicine; Shanghai China
| | - Zhengtao Wang
- Institute of Chinese Materia Medica; Shanghai University of Traditional Chinese Medicine; The MOE Key Laboratory for Standardization of Chinese Medicines and The SATCM Key Laboratory for New Resources and Quality Evaluation of Chinese Medicine; Shanghai China
- Shanghai R&D Centre for Standardization of Chinese Medicines; Shanghai China
| | - Changhong Wang
- Institute of Chinese Materia Medica; Shanghai University of Traditional Chinese Medicine; The MOE Key Laboratory for Standardization of Chinese Medicines and The SATCM Key Laboratory for New Resources and Quality Evaluation of Chinese Medicine; Shanghai China
- Shanghai R&D Centre for Standardization of Chinese Medicines; Shanghai China
| |
Collapse
|
22
|
dos Santos RG, Osório FL, Crippa JAS, Riba J, Zuardi AW, Hallak JEC. Antidepressive, anxiolytic, and antiaddictive effects of ayahuasca, psilocybin and lysergic acid diethylamide (LSD): a systematic review of clinical trials published in the last 25 years. Ther Adv Psychopharmacol 2016; 6:193-213. [PMID: 27354908 PMCID: PMC4910400 DOI: 10.1177/2045125316638008] [Citation(s) in RCA: 155] [Impact Index Per Article: 17.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/18/2022] Open
Abstract
To date, pharmacological treatments for mood and anxiety disorders and for drug dependence show limited efficacy, leaving a large number of patients suffering severe and persistent symptoms. Preliminary studies in animals and humans suggest that ayahuasca, psilocybin and lysergic acid diethylamide (LSD) may have antidepressive, anxiolytic, and antiaddictive properties. Thus, we conducted a systematic review of clinical trials published from 1990 until 2015, assessing these therapeutic properties. Electronic searches were performed using the PubMed, LILACS, and SciELO databases. Only clinical trials published in peer-reviewed journals were included. Of these, 151 studies were identified, of which six met the established criteria. Reviewed studies suggest beneficial effects for treatment-resistant depression, anxiety and depression associated with life-threatening diseases, and tobacco and alcohol dependence. All drugs were well tolerated. In conclusion, ayahuasca, psilocybin and LSD may be useful pharmacological tools for the treatment of drug dependence, and anxiety and mood disorders, especially in treatment-resistant patients. These drugs may also be useful pharmacological tools to understand psychiatric disorders and to develop new therapeutic agents. However, all studies reviewed had small sample sizes, and half of them were open-label, proof-of-concept studies. Randomized, double-blind, placebo-controlled studies with more patients are needed to replicate these preliminary findings.
Collapse
Affiliation(s)
- Rafael G. dos Santos
- Departamento de Neurociências e Ciências do Comportamento, Faculdade de Medicina de Ribeirão Preto, Universidade de São Paulo, Hospital das Clínicas, Terceiro Andar, Av. Bandeirantes, 3900, Ribeirão Preto, São Paulo, Brazil
| | - Flávia L. Osório
- Department of Neuroscience and Behavior, Ribeirão Preto Medical School, University of São Paulo, SP, Brazil National Institute for Translational Medicine (INCT-TM), CNPq, Brazil
| | - José Alexandre S. Crippa
- Department of Neuroscience and Behavior, Ribeirão Preto Medical School, University of São Paulo, SP, Brazil National Institute for Translational Medicine (INCT-TM), CNPq, Brazil
| | - Jordi Riba
- Centre d’Investigació de Medicaments, Servei de Farmacologia Clínica, Hospital de la Santa Creu i Sant Pau, Barcelona, Spain
- Human Experimental Neuropsy-chopharmacology, Institut de Recerca, Hospital de la Santa Creu i Sant Pau, Barcelona, Spain
- Departament de Farmacologia i Terapèutica, Universitat Autònoma de Barcelona, Spain
- Centro de Investigación Biomédica en Red de Salud Mental, CIBERSAM, Barcelona, Spain
| | - Antônio W. Zuardi
- Department of Neuroscience and Behavior, Ribeirão Preto Medical School, University of São Paulo, SP, Brazil National Institute for Translational Medicine (INCT-TM), CNPq, Brazil
| | - Jaime E. C. Hallak
- Department of Neuroscience and Behavior, Ribeirão Preto Medical School, University of São Paulo, SP, Brazil National Institute for Translational Medicine (INCT-TM), CNPq, Brazil
| |
Collapse
|
23
|
Jiang XL, Shen HW, Yu AM. Modification of 5-methoxy-N,N-dimethyltryptamine-induced hyperactivity by monoamine oxidase A inhibitor harmaline in mice and the underlying serotonergic mechanisms. Pharmacol Rep 2016; 68:608-15. [PMID: 26977821 DOI: 10.1016/j.pharep.2016.01.008] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/12/2015] [Revised: 12/17/2015] [Accepted: 01/21/2016] [Indexed: 01/03/2023]
Abstract
BACKGROUND 5-Methoxy-N,N-dimethyltryptamine (5-MeO-DMT) and harmaline are indolealkylamine (IAA) drugs often abused together. Our recent studies have revealed the significant effects of co-administered harmaline, a monoamine oxidase inhibitor (MAOI), on 5-MeO-DMT pharmacokinetics and thermoregulation. This study was to delineate the impact of harmaline and 5-MeO-DMT on home-cage activity in mouse models, as well as the contribution of serotonin (5-HT) receptors. METHODS Home-cage activities of individual animals were monitored automatically in the home cages following implantation of telemetry transmitters and administration of various doses of IAA drugs and 5-HT receptor antagonists. Area under the effect curve (AUEC) of mouse activity values were calculated by trapezoidal rule. RESULTS High dose of harmaline (15mg/kg, ip) alone caused an early-phase (0-45min) hypoactivity in mice that was fully attenuated by 5-HT1A receptor antagonist WAY-100635, whereas a late-phase (45-180min) hyperactivity that was reduced by 5-HT2A receptor antagonist MDL-100907. 5-MeO-DMT (10 and 20mg/kg, ip) alone induced biphasic effects, an early-phase (0-45min) hypoactivity that was completely attenuated by WAY-100635, and a late-phase (45-180min) hyperactivity that was fully suppressed by MDL-100907. Interestingly, co-administration of MAOI harmaline (2-15mg/kg) with a subthreshold dose of 5-MeO-DMT (2mg/kg) induced excessive hyperactivities at late phase (45-180min) that could be abolished by either WAY-100635 or MDL-100907. CONCLUSIONS Co-administration of MAOI with 5-MeO-DMT provokes excessive late-phase hyperactivity, which involves the activation of both 5-HT1A and 5-HT2A receptors.
Collapse
Affiliation(s)
- Xi-Ling Jiang
- Department of Pharmaceutical Sciences, School of Pharmacy and Pharmaceutical Sciences, University at Buffalo, The State University of New York, Buffalo, NY, USA
| | - Hong-Wu Shen
- Department of Pharmaceutical Sciences, School of Pharmacy and Pharmaceutical Sciences, University at Buffalo, The State University of New York, Buffalo, NY, USA
| | - Ai-Ming Yu
- Department of Biochemistry & Molecular Medicine, UC Davis School of Medicine, Sacramento, CA, USA.
| |
Collapse
|
24
|
Jiang XL, Shen HW, Yu AM. Potentiation of 5-methoxy-N,N-dimethyltryptamine-induced hyperthermia by harmaline and the involvement of activation of 5-HT1A and 5-HT2A receptors. Neuropharmacology 2015; 89:342-51. [PMID: 25446678 DOI: 10.1016/j.neuropharm.2014.10.013] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/04/2014] [Revised: 10/08/2014] [Accepted: 10/13/2014] [Indexed: 10/24/2022]
Abstract
5-Methoxy-N,N-dimethyltryptamine (5-MeO-DMT) and harmaline are serotonin (5-HT) analogs often abused together, which alters thermoregulation that may indicate the severity of serotonin toxicity. Our recent studies have revealed that co-administration of monoamine oxidase inhibitor harmaline leads to greater and prolonged exposure to 5-HT agonist 5-MeO-DMT that might be influenced by cytochrome P450 2D6 (CYP2D6) status. This study was to define the effects of harmaline and 5-MeO-DMT on thermoregulation in wild-type and CYP2D6-humanized (Tg-CYP2D6) mice, as well as the involvement of 5-HT receptors. Animal core body temperatures were monitored noninvasively in the home cages after implantation of telemetry transmitters and administration of drugs. Harmaline (5 and 15 mg/kg, i.p.) alone was shown to induce hypothermia that was significantly affected by CYP2D6 status. In contrast, higher doses of 5-MeO-DMT (10 and 20 mg/kg) alone caused hyperthermia. Co-administration of harmaline (2, 5 or 15 mg/kg) remarkably potentiated the hyperthermia elicited by 5-MeO-DMT (2 or 10 mg/kg), which might be influenced by CYP2D6 status at certain dose combination. Interestingly, harmaline-induced hypothermia was only attenuated by 5-HT1A receptor antagonist WAY-100635, whereas 5-MeO-DMT- and harmaline-5-MeO-DMT-induced hyperthermia could be suppressed by either WAY-100635 or 5-HT2A receptor antagonists (MDL-100907 and ketanserin). Moreover, stress-induced hyperthermia under home cage conditions was not affected by WAY-100635 but surprisingly attenuated by MDL-100907 and ketanserin. Our results indicate that co-administration of monoamine oxidase inhibitor largely potentiates 5-MeO-DMT-induced hyperthermia that involves the activation of both 5-HT1A and 5-HT2A receptors. These findings shall provide insights into development of anxiolytic drugs and new strategies to relieve the lethal hyperthermia in serotonin toxicity.
Collapse
Affiliation(s)
- Xi-Ling Jiang
- Department of Pharmaceutical Sciences, School of Pharmacy and Pharmaceutical Sciences, University at Buffalo, The State University of New York, Buffalo, NY 14214-8033, USA
| | | | | |
Collapse
|
25
|
Osório FDL, Sanches RF, Macedo LR, dos Santos RG, Maia-de-Oliveira JP, Wichert-Ana L, de Araujo DB, Riba J, Crippa JA, Hallak JE. Antidepressant effects of a single dose of ayahuasca in patients with recurrent depression: a preliminary report. BRAZILIAN JOURNAL OF PSYCHIATRY 2015; 37:13-20. [DOI: 10.1590/1516-4446-2014-1496] [Citation(s) in RCA: 259] [Impact Index Per Article: 25.9] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/20/2014] [Accepted: 08/08/2014] [Indexed: 11/22/2022]
Affiliation(s)
- Flávia de L. Osório
- Universidade de São Paulo (USP), Brazil; National Science and Technology Institute for Translational Medicine (INCT-TM), Brazil
| | - Rafael F. Sanches
- Universidade de São Paulo (USP), Brazil; Hospital de la Santa Creu i Sant Pau, Spain
| | | | | | | | | | | | - Jordi Riba
- Hospital de la Santa Creu i Sant Pau, Spain; Institut de Recerca, Hospital de la Santa Creu i Sant Pau, Spain; Universitat Autònoma de Barcelona, Spain; CIBERSAM, Spain
| | - José A. Crippa
- Universidade de São Paulo (USP), Brazil; National Science and Technology Institute for Translational Medicine (INCT-TM), Brazil
| | - Jaime E. Hallak
- Universidade de São Paulo (USP), Brazil; National Science and Technology Institute for Translational Medicine (INCT-TM), Brazil
| |
Collapse
|
26
|
Grimsley A, Gallagher R, Hutchison M, Pickup K, Wilson ID, Samuelsson K. Drug-drug interactions and metabolism in cytochrome P450 2C knockout mice: application to troleandomycin and midazolam. Biochem Pharmacol 2013; 86:529-38. [PMID: 23732297 DOI: 10.1016/j.bcp.2013.05.015] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/08/2013] [Revised: 05/22/2013] [Accepted: 05/22/2013] [Indexed: 12/11/2022]
Abstract
Drug-drug interactions (DDIs) may cause serious drug toxicity and delay development of candidate drugs. Screening using human liver microsomes and hepatocytes can help predict DDIs but do not always provide the degree of certainty required for confident progression of a candidate drug. Thus a suitable in vivo test system could be of great value. Here a Cyp2c knockout (KO) mouse was investigated for studying DDIs using midazolam (MDZ) a standard human CYP3A4 substrate and troleandomycin (TAO) a potent human CYP3A4 inhibitor. Pharmacokinetics (PK) and biotransformation of MDZ were investigated following dosing to Cyp2c KO and wild type mice before and after TAO treatment. The noteworthy differences in the metabolism of MDZ in Cyp2c KO compared to wild type mice confirms the important role that Cyp2c enzymes play in the murine metabolism of MDZ in vivo. The impact of Cyp3a inhibition produced a further increase in circulating MDZ concentrations in all individuals from both strains of mice though the impact of the elimination of the Cyp2c pathway in the KO mice on the AUC was less than perhaps expected. We have shown that TAO produces an increase in the MDZ concentration and a reduction in the 1'hydroxymidazolam/midazolam formation ratio but the expected difference in the magnitude of this effect between the wild type and the Cyp2c KO mice was not seen. The magnitude of the TAO effect was also smaller than is reported in humans. Hence further work is required before this animal model could be used to predict clinical interactions.
Collapse
Affiliation(s)
- Aidan Grimsley
- Global DMPK, AstraZeneca UK Ltd., Alderley Park, Macclesfield SK10 4TG, United Kingdom
| | | | | | | | | | | |
Collapse
|
27
|
Cheng J, Zhen Y, Miksys S, Beyoğlu D, Krausz KW, Tyndale RF, Yu A, Idle JR, Gonzalez FJ. Potential role of CYP2D6 in the central nervous system. Xenobiotica 2013; 43:973-84. [PMID: 23614566 DOI: 10.3109/00498254.2013.791410] [Citation(s) in RCA: 48] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/24/2023]
Abstract
1. Cytochrome P450 2D6 (CYP2D6) is a pivotal enzyme responsible for a major drug oxidation polymorphism in human populations. Distribution of CYP2D6 in brain and its role in serotonin metabolism suggest that CYP2D6 may have a function in the central nervous system. 2. To establish an efficient and accurate platform for the study of CYP2D6 in vivo, a human CYP2D6 (Tg-2D6) model was generated by transgenesis in wild-type (WT) C57BL/6 mice using a P1 phage artificial chromosome clone containing the complete human CYP2D locus, including the CYP2D6 gene and 5'- and 3'-flanking sequences. 3. Human CYP2D6 was expressed not only in the liver but also in the brain. The abundance of serotonin and 5-hydroxyindoleacetic acid in brain of Tg-2D6 is higher than in WT mice, either basal levels or after harmaline induction. Metabolomics of brain homogenate and cerebrospinal fluid revealed a significant up-regulation of L-carnitine, acetyl-L-carnitine, pantothenic acid, 2'-deoxycytidine diphosphate (dCDP), anandamide, N-acetylglucosaminylamine and a down-regulation of stearoyl-L-carnitine in Tg-2D6 mice compared with WT mice. Anxiety tests indicate Tg-2D6 mice have a higher capability to adapt to anxiety. 4. Overall, these findings indicate that the Tg-2D6 mouse model may serve as a valuable in vivo tool to determine CYP2D6-involved neurophysiological metabolism and function.
Collapse
Affiliation(s)
- Jie Cheng
- Laboratory of Metabolism, Center for Cancer Research, National Cancer Institute, National Institutes of Health , Bethesda, MD , USA
| | | | | | | | | | | | | | | | | |
Collapse
|
28
|
dos Santos RG. A Critical Evaluation of Reports Associating Ayahuasca with Life-Threatening Adverse Reactions. J Psychoactive Drugs 2013; 45:179-88. [DOI: 10.1080/02791072.2013.785846] [Citation(s) in RCA: 26] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/26/2022]
|
29
|
Guimarães dos Santos R. Safety and Side Effects of Ayahuasca in Humans—An Overview Focusing on Developmental Toxicology. J Psychoactive Drugs 2013; 45:68-78. [DOI: 10.1080/02791072.2013.763564] [Citation(s) in RCA: 38] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/18/2023]
|
30
|
Jiang XL, Shen HW, Mager DE, Yu AM. Pharmacokinetic interactions between monoamine oxidase A inhibitor harmaline and 5-methoxy-N,N-dimethyltryptamine, and the impact of CYP2D6 status. Drug Metab Dispos 2013; 41:975-86. [PMID: 23393220 DOI: 10.1124/dmd.112.050724] [Citation(s) in RCA: 22] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/01/2023] Open
Abstract
5-Methoxy-N,N-dimethyltryptamine (5-MeO-DMT or street name "5-MEO") is a newer designer drug belonging to a group of naturally occurring indolealkylamines. Our recent study has demonstrated that coadministration of monoamine oxidase A (MAO-A) inhibitor harmaline (5 mg/kg) increases systemic exposure to 5-MeO-DMT (2 mg/kg) and active metabolite bufotenine. This study is aimed at delineating harmaline and 5-MeO-DMT pharmacokinetic (PK) interactions at multiple dose levels, as well as the impact of CYP2D6 that affects harmaline PK and determines 5-MeO-DMT O-demethylation to produce bufotenine. Our data revealed that inhibition of MAO-A-mediated metabolic elimination by harmaline (2, 5, and 15 mg/kg) led to a sharp increase in systemic and cerebral exposure to 5-MeO-DMT (2 and 10 mg/kg) at all dose combinations. A more pronounced effect on 5-MeO-DMT PK was associated with greater exposure to harmaline in wild-type mice than CYP2D6-humanized (Tg-CYP2D6) mice. Harmaline (5 mg/kg) also increased blood and brain bufotenine concentrations that were generally higher in Tg-CYP2D6 mice. Surprisingly, greater harmaline dose (15 mg/kg) reduced bufotenine levels. The in vivo inhibitory effect of harmaline on CYP2D6-catalyzed bufotenine formation was confirmed by in vitro study using purified CYP2D6. Given these findings, a unified PK model including the inhibition of MAO-A- and CYP2D6-catalyzed 5-MeO-DMT metabolism by harmaline was developed to describe blood harmaline, 5-MeO-DMT, and bufotenine PK profiles in both wild-type and Tg-CYP2D6 mouse models. This PK model may be further employed to predict harmaline and 5-MeO-DMT PK interactions at various doses, define the impact of CYP2D6 status, and drive harmaline-5-MeO-DMT pharmacodynamics.
Collapse
Affiliation(s)
- Xi-Ling Jiang
- Department of Pharmaceutical Sciences, University at Buffalo, The State University of New York, Buffalo, New York, USA
| | | | | | | |
Collapse
|
31
|
Shen HW, Jiang XL, Gonzalez FJ, Yu AM. Humanized transgenic mouse models for drug metabolism and pharmacokinetic research. Curr Drug Metab 2012; 12:997-1006. [PMID: 22023319 DOI: 10.2174/138920011798062265] [Citation(s) in RCA: 22] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/01/2011] [Revised: 07/16/2011] [Accepted: 07/20/2011] [Indexed: 02/08/2023]
Abstract
Extrapolation of the metabolic, pharmacokinetic and toxicological data obtained from animals to humans is not always straightforward, given the remarkable species difference in drug metabolism that is due in large part to the differences in drug-metabolizing enzymes between animals and humans. Furthermore, genetic variations in drug-metabolizing enzymes may significantly alter pharmacokinetics, drug efficacy and safety. Thus, humanized transgenic mouse lines, in which the human drug-metabolizing enzymes are expressed in mouse tissues in the presence or absence of mouse orthologues, have been developed to address such challenges. These humanized transgenic mice are valuable animal models in understanding the significance of specific human drug-metabolizing enzymes in drug clearance and pharmacokinetics, as well as in predicting potential drug-drug interactions and chemical toxicity in humans. This review, therefore, aims to summarize the development and application of some humanized transgenic mouse models expressing human drug-metabolizing enzymes. The limitations of these genetically modified mouse models are also discussed.
Collapse
Affiliation(s)
- Hong-Wu Shen
- Department of Pharmaceutical Sciences University at Buffalo, The State University of New York, 541 Cooke Hall, Buffalo, NY 14260-1200, USA
| | | | | | | |
Collapse
|
32
|
Cao MR, Li Q, Liu ZL, Liu HH, Wang W, Liao XL, Pan YL, Jiang JW. Harmine induces apoptosis in HepG2 cells via mitochondrial signaling pathway. Hepatobiliary Pancreat Dis Int 2011; 10:599-604. [PMID: 22146623 DOI: 10.1016/s1499-3872(11)60102-1] [Citation(s) in RCA: 45] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/08/2023]
Abstract
BACKGROUND Harmine has antitumor and antinociceptive effects, and inhibits human DNA topoisomerase. However, no detailed data are available on the mechanisms of action of harmine in hepatocellular carcinoma. This study aimed to investigate the effects of harmine on proliferation and apoptosis, and the underlying mechanisms in the human hepatocellular carcinoma cell line HepG2. METHODS The proliferation of HepG2 cells was determined by the cell counting kit-8 (CCK-8) assay and the clone formation test. The morphology of HepG2 cells was examined using fluorescence microscopy after Hoechst 33258 staining. Annexin V/propidium iodide (PI) was used to analyze apoptosis and PI to analyze the cell cycle. Western blotting was used to assess expression of the apoptosis-regulated genes Bcl-2, Bax, Bcl-xl, Mcl-1, caspase-3, and caspase-9. Mitochondrial transmembrane potential (ψm) was determined using JC-1. RESULTS Harmine inhibited the proliferation of HepG2 cells in a dose-dependent manner. Hoechst 33258 staining revealed nuclear fragmentation and chromosomal condensation, cell shrinkage, and attachment loss in HepG2 cells treated with harmine. The percentage of the sub/G1 fraction was increased in a concentration-dependent manner, indicating apoptotic cell death. PI staining showed that harmine changed the cell cycle distribution, by decreasing the proportion of cells in G0/G1 and increasing the proportion in S and G2/M. Harmine induced apoptosis in a concentration-dependent manner, with rates of 20.0%, 32.7% and 64.9%, respectively. JC-1 revealed a decrease in ψm. Apoptosis of HepG2 cells was associated with caspase-3 and caspase-9 activation, down-regulation of Bcl-2, Mcl-1, and Bcl-xl, and no change in Bax. CONCLUSIONS Harmine had an anti-proliferative effect in HepG2 cells by inducing apoptosis. Mitochondrial signal pathways were involved in the apoptosis. The cancer-specific selectivity shown in this study suggested that harmine is a promising novel drug for human hepatocellular carcinoma.
Collapse
Affiliation(s)
- Ming-Rong Cao
- Department of General Surgery, First Affiliated Hospital, Jinan University, Guangzhou 510632, China
| | | | | | | | | | | | | | | |
Collapse
|
33
|
El Gendy MAM, Soshilov AA, Denison MS, El-Kadi AOS. Harmaline and harmalol inhibit the carcinogen-activating enzyme CYP1A1 via transcriptional and posttranslational mechanisms. Food Chem Toxicol 2011; 50:353-62. [PMID: 22037238 DOI: 10.1016/j.fct.2011.10.052] [Citation(s) in RCA: 29] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/18/2011] [Revised: 09/03/2011] [Accepted: 10/14/2011] [Indexed: 01/28/2023]
Abstract
Dioxins are known to cause several human cancers through activation of the aryl hydrocarbon receptor (AhR). Harmaline and harmalol are dihydro-β-carboline compounds present in several medicinal plants such as Peganum harmala. We have previously demonstrated the ability of P. harmala extract to inhibit TCDD-mediated induction of Cyp1a1 in murine hepatoma Hepa 1c1c7 cells. Therefore, the aim of this study is to examine the effect of harmaline and its main metabolite, harmalol, on dioxin-mediated induction of CYP1A1 in human hepatoma HepG2 cells. Our results showed that harmaline and harmalol at concentrations of (0.5-12.5μM) significantly inhibited the dioxin-induced CYP1A1 at mRNA, protein and activity levels in a concentration-dependent manner. The role of AhR was determined by the inhibition of the TCDD-mediated induction of AhR-dependent luciferase activity and the AhR/ARNT/XRE formation by both harmaline and harmalol. In addition, harmaline significantly displaced [(3)H]TCDD in the competitive ligand binding assay. At posttranslational level, both harmaline and harmalol decreased the protein stability of CYP1A1, suggesting that posttranslational modifications are involved. Moreover, the posttranslational modifications of harmaline and harmalol involve ubiquitin-proteasomal pathway and direct inhibitory effects of both compounds on CYP1A1 enzyme. These data suggest that harmaline and harmalol are promising agents for preventing dioxin-mediated effects.
Collapse
Affiliation(s)
- Mohamed A M El Gendy
- Faculty of Pharmacy and Pharmaceutical Sciences, University of Alberta, Edmonton, Alberta, Canada T6G 2N8
| | | | | | | |
Collapse
|
34
|
Zhao T, He YQ, Wang J, Ding KM, Wang CH, Wang ZT. Inhibition of Human Cytochrome P450 Enzymes 3A4 and 2D6 by β-Carboline Alkaloids, Harmine Derivatives. Phytother Res 2011; 25:1671-7. [DOI: 10.1002/ptr.3458] [Citation(s) in RCA: 30] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/03/2010] [Revised: 01/31/2011] [Accepted: 02/02/2011] [Indexed: 10/18/2022]
Affiliation(s)
- Ting Zhao
- Institute of Chinese Materia Medica; Shanghai University of Traditional Chinese Medicine; Shanghai; 201210; China
| | - Yu-qi He
- Institute of Chinese Materia Medica; Shanghai University of Traditional Chinese Medicine; Shanghai; 201210; China
| | - Jun Wang
- Institute of Chinese Materia Medica; Shanghai University of Traditional Chinese Medicine; Shanghai; 201210; China
| | - Ke-min Ding
- Institute of Chinese Materia Medica; Shanghai University of Traditional Chinese Medicine; Shanghai; 201210; China
| | | | | |
Collapse
|
35
|
dos Santos RG. Toxicity of chronic ayahuasca administration to the pregnant rat: how relevant it is regarding the human, ritual use of ayahuasca? ACTA ACUST UNITED AC 2010; 89:533-5; author reply 531-2.. [DOI: 10.1002/bdrb.20272] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/10/2022]
|
36
|
Jiang XL, Gonzalez FJ, Yu AM. Drug-metabolizing enzyme, transporter, and nuclear receptor genetically modified mouse models. Drug Metab Rev 2010; 43:27-40. [PMID: 20854191 DOI: 10.3109/03602532.2010.512294] [Citation(s) in RCA: 30] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022]
Abstract
Determining the in vivo significance of a specific enzyme, transporter, or xenobiotic receptor in drug metabolism and pharmacokinetics may be hampered by gene multiplicity and complexity, levels of expression, and interaction between various components involved. The development of knockout (loss-of-function) and transgenic (gain-of-function) mouse models opens the door to the improved understanding of gene function in a whole-body system. There is also growing interest in the development of humanized mice to overcome species differences in drug metabolism and disposition. This review, therefore, aims to summarize and discuss some successful examples of drug-metabolizing enzyme, transporter, and nuclear-receptor genetically modified mouse models. These genetically modified mouse models have been proven as invaluable models for understanding in vivo function of drug-metabolizing enzymes, transporters, and xenobiotic receptors in drug metabolism and transport, as well as predicting potential drug-drug interaction and toxicity in humans. Nevertheless, concerns remain about interpretation of data obtained from such genetically modified mouse models, in which the expression of related genes is altered significantly.
Collapse
Affiliation(s)
- Xi-Ling Jiang
- Department of Pharmaceutical Sciences, University at Buffalo, The State University of New York, USA
| | | | | |
Collapse
|
37
|
Shen HW, Yu AM. Difference in desipramine metabolic profile between wild-type and CYP2D6-humanized mice. Drug Metab Lett 2010; 3:234-41. [PMID: 19995332 DOI: 10.2174/187231209790218118] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/27/2009] [Accepted: 09/16/2009] [Indexed: 02/04/2023]
Abstract
Desipramine (DMI), a CYP2D6 probe, was used as a model drug to test whether CYP2D6-humanized (Tg-CYP2D6) and wild-type control mice could be used as preclinical animal models to identify the effects of CYP2D6 genotype/phenotype on drug metabolic profiles. After the analyses by liquid chromatography coupled with tandem mass spectrometry, DMI biotransformations were compared in Tg-CYP2D6 and wild-type mouse liver microsomes (MLM), and in human CYP2D6 extensive and poor metabolizer liver microsomes. Furthermore, urinary DMI metabolic profiles in Tg-CYP2D6 and wild-type mice were evaluated. Three metabolites, 2-hydroxyl-, 10-hydroxyl, and N-desmethyl-desipramine were identified in the incubations of DMI with both wild-type and Tg-CYP2D6 MLM, as well as in human CYP2D6 extensive metabolizer liver microsomes. Three additional metabolites were found in mouse urine samples, and their chemical structures were elucidated. Although the ratio of individual metabolites produced in Tg-CYP2D6 MLM was closer to that in human CYP2D6 extensive metabolizer liver microsomes, the urinary DMI metabolic profiles did not show much difference between wild-type and Tg-CYP2D6 mice. The results suggest that other mouse enzymes have significant contribution to DMI metabolism.
Collapse
Affiliation(s)
- Hong-Wu Shen
- Department of Pharmaceutical Sciences, University at Buffalo, The State University of New York, Buffalo, NY 14260-1200, USA
| | | |
Collapse
|
38
|
Shen HW, Wu C, Jiang XL, Yu AM. Effects of monoamine oxidase inhibitor and cytochrome P450 2D6 status on 5-methoxy-N,N-dimethyltryptamine metabolism and pharmacokinetics. Biochem Pharmacol 2010; 80:122-8. [PMID: 20206139 DOI: 10.1016/j.bcp.2010.02.020] [Citation(s) in RCA: 21] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/26/2010] [Revised: 02/23/2010] [Accepted: 02/24/2010] [Indexed: 10/19/2022]
Abstract
5-Methoxy-N,N-dimethyltryptamine (5-MeO-DMT) is a natural psychoactive indolealkylamine drug that has been used for recreational purpose. Our previous study revealed that polymorphic cytochrome P450 2D6 (CYP2D6) catalyzed 5-MeO-DMT O-demethylation to produce active metabolite bufotenine, while 5-MeO-DMT is mainly inactivated through deamination pathway mediated by monoamine oxidase (MAO). This study, therefore, aimed to investigate the impact of CYP2D6 genotype/phenotype status and MAO inhibitor (MAOI) on 5-MeO-DMT metabolism and pharmacokinetics. Enzyme kinetic studies using recombinant CYP2D6 allelic isozymes showed that CYP2D6.2 and CYP2D6.10 exhibited 2.6- and 40-fold lower catalytic efficiency (V(max)/K(m)), respectively, in producing bufotenine from 5-MeO-DMT, compared with wild-type CYP2D6.1. When co-incubated with MAOI pargyline, 5-MeO-DMT O-demethylation in 10 human liver microsomes showed significantly strong correlation with bufuralol 1'-hydroxylase activities (R(2)=0.98; P<0.0001) and CYP2D6 contents (R(2)=0.77; P=0.0007), whereas no appreciable correlations with enzymatic activities of other P450 enzymes. Furthermore, concurrent MAOI harmaline sharply reduced 5-MeO-DMT depletion and increased bufotenine formation in human CYP2D6 extensive metabolizer hepatocytes. In vivo studies in wild-type and CYP2D6-humanized (Tg-CYP2D6) mouse models showed that Tg-CYP2D6 mice receiving the same dose of 5-MeO-DMT (20mg/kg, i.p.) had 60% higher systemic exposure to metabolite bufotenine. In addition, pretreatment of harmaline (5mg/kg, i.p.) led to 3.6- and 4.4-fold higher systemic exposure to 5-MeO-DMT (2mg/kg, i.p.), and 9.9- and 6.1-fold higher systemic exposure to bufotenine in Tg-CYP2D6 and wild-type mice, respectively. These findings indicate that MAOI largely affects 5-MeO-DMT metabolism and pharmacokinetics, as well as bufotenine formation that is mediated by CYP2D6.
Collapse
Affiliation(s)
- Hong-Wu Shen
- Department of Pharmaceutical Sciences, School of Pharmacy and Pharmaceutical Sciences, University at Buffalo, The State University of New York, 541 Cooke Hall, Buffalo, NY 14260-1200, USA
| | | | | | | |
Collapse
|
39
|
Herraiz T, González D, Ancín-Azpilicueta C, Arán VJ, Guillén H. beta-Carboline alkaloids in Peganum harmala and inhibition of human monoamine oxidase (MAO). Food Chem Toxicol 2009; 48:839-45. [PMID: 20036304 DOI: 10.1016/j.fct.2009.12.019] [Citation(s) in RCA: 217] [Impact Index Per Article: 13.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/02/2009] [Revised: 11/21/2009] [Accepted: 12/21/2009] [Indexed: 10/20/2022]
Abstract
Peganum harmala L. is a multipurpose medicinal plant increasingly used for psychoactive recreational purposes (Ayahuasca analog). Harmaline, harmine, harmalol, harmol and tetrahydroharmine were identified and quantified as the main beta-carboline alkaloids in P. harmala extracts. Seeds and roots contained the highest levels of alkaloids with low levels in stems and leaves, and absence in flowers. Harmine and harmaline accumulated in dry seeds at 4.3% and 5.6% (w/w), respectively, harmalol at 0.6%, and tetrahydroharmine at 0.1% (w/w). Roots contained harmine and harmol with 2.0% and 1.4% (w/w), respectively. Seed extracts were potent reversible and competitive inhibitors of human monoamine oxidase (MAO-A) with an IC(50) of 27 microg/l whereas root extracts strongly inhibited MAO-A with an IC(50) of 159 microg/l. In contrast, they were poor inhibitors of MAO-B. Inhibition of MAO-A by seed extracts was quantitatively attributed to harmaline and harmine whereas inhibition by root extracts came from harmine with no additional interferences. Stems and leaves extracts were poor inhibitors of MAO. The potent inhibition of MAO-A by seed and root extracts of P. harmala containing beta-carbolines should contribute to the psychopharmacological and toxicological effects of this plant and could be the basis for its purported antidepressant actions.
Collapse
Affiliation(s)
- T Herraiz
- Spanish Council for Scientific Research, CSIC, Instituto de Fermentaciones Industriales, Madrid, Spain.
| | | | | | | | | |
Collapse
|