<Home — Psychoactive Plant Database



  Psychoactive Plant Database - Neuroactive Phytochemical Collection





Worldwide, there are plants known as psychoactive plants that naturally contain psychedelic active components. They have a high concentration of neuroprotective substances that can interact with the nervous system to produce psychedelic effects. Despite these plants' hazardous potential, recreational use of them is on the rise because of their psychoactive properties. Early neuroscience studies relied heavily on psychoactive plants and plant natural products (NPs), and both recreational and hazardous NPs have contributed significantly to the understanding of almost all neurotransmitter systems. Worldwide, there are many plants that contain psychoactive properties, and people have been using them for ages. Psychoactive plant compounds may significantly alter how people perceive the world.

 

 

1. Inflammopharmacology. 2024 Oct 29. doi: 10.1007/s10787-024-01573-1. Online ahead of print. Anti-inflammatory effects of a methanol extract from Montanoa grandiflora DC. (Asteraceae) leaves on in vitro and in vivo models. Sánchez-Canul M(1), Villa-de la Torre F(1), Borges-Argáez R(2), Huchin-Chan C(3), Valencia-Pacheco G(4), Yáñez-Barrientos E(5), Romero-Hernández M(5), Alonso-Castro AJ(6), Arana-Argáez VE(7). Author information: (1)Laboratorio de Farmacología, Facultad de Química, Universidad Autónoma de Yucatán, Calle 43, No 613 x calle 90, Col. Inalámbrica, CP. 97069, Mérida, Yucatán, México. (2)Centro de Investigación Científica de Yucatán, Unidad de Biotecnología, Mérida, Yucatán, México. (3)Laboratorio de Análisis Clínicos y de Servicio a La Comunidad, Facultad de Química, Universidad Autónoma de Yucatán, Mérida, Yucatán, México. (4)Laboratorio de Hematología, Centro de Investigaciones Regionales "Dr. Hideyo Noguchi", Universidad Autónoma de Yucatán, Mérida, Yucatán, México. (5)Departamento de Química, División de Ciencias Naturales y Exactas, Universidad de Guanajuato, Guanajuato, México. (6)Departamento de Farmacia, División de Ciencias Naturales y Exactas, Universidad de Guanajuato, Guanajuato, México. (7)Laboratorio de Farmacología, Facultad de Química, Universidad Autónoma de Yucatán, Calle 43, No 613 x calle 90, Col. Inalámbrica, CP. 97069, Mérida, Yucatán, México. victor.arana@correo.uady.mx. BACKGROUND: Montanoa grandiflora, a plant species native from Mexico to Central America, locally known as "Teresita" in Yucatán, México, is used to alleviate anxiety, rheumatism, and stomach issues. This study aims to investigate the anti-inflammatory properties of the methanol extract of Montanoa grandiflora leaves (MMG) in experimental models of inflammation. METHODS: Gas chromatography-mass spectroscopy was used to characterize the MMG; cytotoxicity was assessed by MTT assay on murine macrophages and hemolysis assay. The in vitro anti-inflammatory activity was evaluated on LPS-stimulated murine macrophages by measuring of pro- and anti-inflammatory cytokines, NO and H2O2 release. The in vivo anti-inflammatory activity was evaluated using carrageenan-induced mouse paw edema, 12-O-tetradecanoylphorbol 13-acetate induced-ear edema, and 1-fluoro-2,4-dinitrobenzene induced-delayed-type hypersensitivity. In addition, the serum levels of prostaglandins and leukotrienes were assessed. RESULTS: The main compounds found in MMG were terpenes (i.e., β-caryophyllene, (-)-α-cubebene, alloaromadendrene, ( +)-δ-cadinene, β-eudesmol), alkaloid (( ±)-nor-β-hydrastine), cyclic polyol (quinic acid), carbohydrates and their derivatives, and fatty acids (octadecatrienoic acid and octadecanoic acid). MMG did not exhibit cytotoxic or hemolytic activity. However, it demonstrated in vitro anti-inflammatory effects by increasing the production of IL-10, decreasing the levels of TNF-α, IL-1β, IL-6, NO and H2O2. MMG significantly reduced carrageenan-induced paw edema, TPA-induced ear edema, and DNFB-induced delayed-type hypersensitivity in mice with effects comparable to those of standard drugs, as well as serum levels of prostaglandins and leukotrienes. CONCLUSION: The anti-inflammatory activity of MMG is associated with increased IL-10 levels and inhibiting inflammatory cell migration mechanisms, without causing cytotoxic or hemolytic damage in both in vitro and in vivo assays. © 2024. The Author(s), under exclusive licence to Springer Nature Switzerland AG. DOI: 10.1007/s10787-024-01573-1 PMID: 39472421 2. Chem Biodivers. 2024 Oct 10:e202401886. doi: 10.1002/cbdv.202401886. Online ahead of print. In-silico and In-vitro Investigation of Flavonoids and Alkaloids from Artocarpus heterophyllus, Tinospora cordifolia, and Glycosmis pentaphylla as Potential NF-κB Inhibitors in Oral Cancer. Gotsurve KN(1), Das J(2), Kutre S(3), Teja PK(3), Rathod R(4), Chauthe SK(3), Behera SK(5). Author information: (1)NIPER Ahmedabad, Biotechnology, Gandhinagar, Gandhinagar, INDIA. (2)National Institute of Pharmaceutical Education and Research Ahmedabad, Natural Products, Gandhinagar, Gandhinagar, INDIA. (3)National Institute of Pharmaceutical Education and Research Ahmedabad, Natural Products, Gandhinagar, NATIONAL INSTITUTE OF PHARMACEUTICAL EDUCATION AND RESEARCH, Ahmedabad, Gandhina, Gandhinagar, 382355, Gandhinagar, INDIA. (4)National Institute of Pharmaceutical Education and Research Ahmedabad, CIF, Gandhinagar, NATIONAL INSTITUTE OF PHARMACEUTICAL EDUCATION AND RESEARCH, Ahmedabad, Gandhina, Gandhinagar, 382355, Gandhinagar, INDIA. (5)National Institute of Pharmaceutical Education and Research Ahmedabad, Biothechnology, Opposite air force station, Palaj, Gandhinagar, 382355, Gandhinagar, INDIA. Oral cancer is a global health concern, particularly because of its aggressive tissue invasion and metastases, holding 16th position on occurrence among all carcinoma worldwide. The aberrated transcription factor NF-κB1 influences development and spread of oral cancer, making it a potential therapeutic target. This study investigated the therapeutic potential of a few natural compounds from Artocarpus heterophyllus, Tinospora cordifolia, and Glycosmis pentaphylla in the treatment of oral cancer. Two FDA-approved drugs (5-fluorouracil, Docetaxel) and 16 natural compounds, including Artocarpin, Artocarpanone, Cycloartocarpin from Artocarpus heterophyllus; Berberine, Hydrastine, Magnoflorine, Palmatine Chloride, Tetrahydropalmatine from Tinospora cordifolia; and 5-Hydroxyarborinine, 5-Hydroxynoracronycine, 1-Hydroxy-3-methoxy-10-methyl-9-acridone, Des-N-methylacronycine, Des-N-methylnoracronycine, Kokusagenine, Noracronycin, Skimmianine from Glycosmis pentaphylla were examined using in-silico techniques. Among the 16 natural compounds studied, Hydrastine shown the highest binding energy (-6.87 kcal/mol) against NF-κB1, surpassing all other drugs, including the standards 5-fluorouracil (-4.04 kcal/mol) and docetaxel (-2.4 kcal/mol). Further molecular dynamics simulations and in-vitro experiments verified Hydrastine's exceptional anti-cancer activity. The results of in-vitro were well-aligned with the findings of in-silico, revealing considerable cytotoxicity, apoptosis induction, and cell cycle arrest. The findings revealed natural compounds' potential as safer, more effective alternatives to current cancer therapeutics, opening up new avenues for oral cancer treatment. © 2024 Wiley‐VCH GmbH. DOI: 10.1002/cbdv.202401886 PMID: 39387347 3. Front Neurosci. 2023 Oct 24;17:1259742. doi: 10.3389/fnins.2023.1259742. eCollection 2023. Analysis of damage-associated molecular patterns in amyotrophic lateral sclerosis based on ScRNA-seq and bulk RNA-seq data. Shi Y(1), Zhu R(1). Author information: (1)Department of Neurology, The First Affiliated Hospital of China Medical University, Shenyang, China. BACKGROUND: Amyotrophic Lateral Sclerosis (ALS) is a devastating neurodegenerative disorder characterized by the progressive loss of motor neurons. Despite extensive research, the exact etiology of ALS remains elusive. Emerging evidence highlights the critical role of the immune system in ALS pathogenesis and progression. Damage-Associated Molecular Patterns (DAMPs) are endogenous molecules released by stressed or damaged cells, acting as danger signals and activating immune responses. However, their specific involvement in ALS remains unclear. METHODS: We obtained single-cell RNA sequencing (scRNA-seq) data of ALS from the primary motor cortex in the Gene Expression Omnibus (GEO) database. To better understand genes associated with DAMPs, we performed analyses on cell-cell communication and trajectory. The abundance of immune-infiltrating cells was assessed using the single-sample Gene Set Enrichment Analysis (ssGSEA) method. We performed univariate Cox analysis to construct the risk model and utilized the least absolute shrinkage and selection operator (LASSO) analysis. Finally, we identified potential small molecule drugs targeting ALS by screening the Connectivity Map database (CMap) and confirmed their potential through molecular docking analysis. RESULTS: Our study annotated 10 cell types, with the expression of genes related to DAMPs predominantly observed in microglia. Analysis of intercellular communication revealed 12 ligand-receptor pairs in the pathways associated with DAMPs, where microglial cells acted as ligands. Among these pairs, the SPP1-CD44 pair demonstrated the greatest contribution. Furthermore, trajectory analysis demonstrated distinct differentiation fates of different microglial states. Additionally, we constructed a risk model incorporating four genes (TRPM2, ROCK1, HSP90AA1, and HSPA4). The validity of the risk model was supported by multivariate analysis. Moreover, external validation from dataset GSE112681 confirmed the predictive power of the model, which yielded consistent results with datasets GSE112676 and GSE112680. Lastly, the molecular docking analysis suggested that five compounds, namely mead-acid, nifedipine, nifekalant, androstenol, and hydrastine, hold promise as potential candidates for the treatment of ALS. CONCLUSION: Taken together, our study demonstrated that DAMP entities were predominantly observed in microglial cells within the context of ALS. The utilization of a prognostic risk model can accurately predict ALS patient survival. Additionally, genes related to DAMPs may present viable drug targets for ALS therapy. Copyright © 2023 Shi and Zhu. DOI: 10.3389/fnins.2023.1259742 PMCID: PMC10628000 PMID: 37942135 Conflict of interest statement: The authors declare that the research was conducted in the absence of any commercial or financial relationships that could be construed as a potential conflict of interest. 4. Drug Metab Dispos. 2023 Nov;51(11):1483-1489. doi: 10.1124/dmd.123.001360. Epub 2023 Aug 10. Goldenseal-Mediated Inhibition of Intestinal Uptake Transporters Decreases Metformin Systemic Exposure in Mice. Oyanna VO(1), Garcia-Torres KY(1), Bechtold BJ(1), Lynch KD(1), Call MR(1), Horváth M(1), Manwill PK(1), Graf TN(1), Cech NB(1), Oberlies NH(1), Paine MF(1), Clarke JD(2). Author information: (1)Department of Pharmaceutical Sciences, Washington State University, Spokane, Washington (V.O.O., K.Y.G.-T., B.J.B., K.D.L., M.R.C., M.F.P., J.D.C.); Department of Chemistry and Biochemistry, University of North Carolina at Greensboro, North Carolina (P.K.M., T.N.G., N.B.C., N.H.O.); SOLVO Biotechnology, Szeged, Hungary (M.H.); and Center of Excellence for Natural Product Drug Interaction Research, Spokane, Washington (N.B.C., N.H.O., M.F.P., J.D.C.). (2)Department of Pharmaceutical Sciences, Washington State University, Spokane, Washington (V.O.O., K.Y.G.-T., B.J.B., K.D.L., M.R.C., M.F.P., J.D.C.); Department of Chemistry and Biochemistry, University of North Carolina at Greensboro, North Carolina (P.K.M., T.N.G., N.B.C., N.H.O.); SOLVO Biotechnology, Szeged, Hungary (M.H.); and Center of Excellence for Natural Product Drug Interaction Research, Spokane, Washington (N.B.C., N.H.O., M.F.P., J.D.C.) j.clarke@wsu.edu. Goldenseal is a perennial plant native to eastern North America. A recent clinical study reported goldenseal decreased metformin Cmax and area under the blood concentration versus time curve (AUC) by 27% and 23%, respectively, but half-life and renal clearance were unchanged. These observations suggested goldenseal altered processes involved in metformin absorption. The underlying mechanism(s) remain(s) unknown. One mechanism for the decreased metformin systemic exposure is inhibition by goldenseal of intestinal uptake transporters involved in metformin absorption. Goldenseal extract and three goldenseal alkaloids (berberine, (-)-β-hydrastine, hydrastinine) were tested as inhibitors of organic cation transporter (OCT) 3, plasma membrane monoamine transporter (PMAT), and thiamine transporter (THTR) 2 using human embryonic kidney 293 cells overexpressing each transporter. The goldenseal extract, normalized to berberine content, was the strongest inhibitor of each transporter (IC50: 4.9, 13.1, and 5.8 μM for OCT3, PMAT, and THTR2, respectively). A pharmacokinetic study in mice compared the effects of berberine, (-)-β-hydrastine, goldenseal extract, and imatinib (OCT inhibitor) on orally administered metformin. Goldenseal extract and imatinib significantly decreased metformin Cmax by 31% and 25%, respectively, and had no effect on half-life. Berberine and (-)-β-hydrastine had no effect on metformin pharmacokinetics, indicating neither alkaloid alone precipitated the interaction in vivo. A follow-up murine study involving intravenous metformin and oral inhibitors examined the contributions of basolateral enteric/hepatic uptake transporters to the goldenseal-metformin interaction. Goldenseal extract and imatinib had no effect on metformin AUC and half-life, suggesting lack of inhibition of basolateral enteric/hepatic uptake transporters. Results may have implications for patients taking goldenseal with drugs that are substrates for OCT3 and THTR2. SIGNIFICANCE STATEMENT: Goldenseal is used to self-treat respiratory infections and digestive disorders. We investigated potential mechanisms for the clinical pharmacokinetic interaction observed between goldenseal and metformin, specifically inhibition by goldenseal of intestinal uptake transporters (OCT3, PMAT, THTR2) involved in metformin absorption. Goldenseal extract inhibited all three transporters in vitro and decreased metformin systemic exposure in mice. These data may have broader implications for patients co-consuming goldenseal with other drugs that are substrates for these transporters. Copyright © 2023 by The Author(s). DOI: 10.1124/dmd.123.001360 PMCID: PMC10586506 PMID: 37562957 [Indexed for MEDLINE] 5. J Pharmacol Exp Ther. 2023 Dec;387(3):252-264. doi: 10.1124/jpet.123.001681. Epub 2023 Aug 4. An Integrative Approach to Elucidate Mechanisms Underlying the Pharmacokinetic Goldenseal-Midazolam Interaction: Application of In Vitro Assays and Physiologically Based Pharmacokinetic Models to Understand Clinical Observations. Nguyen JT(1), Tian DD(1), Tanna RS(1), Arian CM(1), Calamia JC(1), Rettie AE(1), Thummel KE(1), Paine MF(2). Author information: (1)Department of Pharmaceutical Sciences, College of Pharmacy and Pharmaceutical Sciences, Washington State University, Spokane, Washington (J.T.N., D.-D.T., R.S.T., M.F.P.); Department of Pharmaceutics (C.M.A., J.C.C., K.E.T.) and Department of Medicinal Chemistry (A.E.R.), School of Pharmacy, University of Washington, Seattle, Washington; and Center of Excellence for Natural Product Drug Interaction Research, Spokane, Washington (A.E.R, K.E.T., M.F.P.). (2)Department of Pharmaceutical Sciences, College of Pharmacy and Pharmaceutical Sciences, Washington State University, Spokane, Washington (J.T.N., D.-D.T., R.S.T., M.F.P.); Department of Pharmaceutics (C.M.A., J.C.C., K.E.T.) and Department of Medicinal Chemistry (A.E.R.), School of Pharmacy, University of Washington, Seattle, Washington; and Center of Excellence for Natural Product Drug Interaction Research, Spokane, Washington (A.E.R, K.E.T., M.F.P.) mary.paine@wsu.edu. The natural product goldenseal is a clinical inhibitor of CYP3A activity, as evidenced by a 40%-60% increase in midazolam area under the plasma concentration versus time curve (AUC) after coadministration with goldenseal. The predominant goldenseal alkaloids berberine and (-)-β-hydrastine were previously identified as time-dependent CYP3A inhibitors using human liver microsomes. Whether these alkaloids contribute to the clinical interaction, as well as the primary anatomic site (hepatic vs. intestinal) and mode of CYP3A inhibition (reversible vs. time-dependent), remain uncharacterized. The objective of this study was to mechanistically assess the pharmacokinetic goldenseal-midazolam interaction using an integrated in vitro-in vivo-in silico approach. Using human intestinal microsomes, (-)-β-hydrastine was a more potent time-dependent inhibitor of midazolam 1'-hydroxylation than berberine (KI and kinact: 8.48 μM and 0.041 minutes-1, respectively, vs. >250 μM and ∼0.06 minutes-1, respectively). Both the AUC and Cmax of midazolam increased by 40%-60% after acute (single 3-g dose) and chronic (1 g thrice daily × 6 days) goldenseal administration to healthy adults. These increases, coupled with a modest or no increase (≤23%) in half-life, suggested that goldenseal primarily inhibited intestinal CYP3A. A physiologically based pharmacokinetic interaction model incorporating berberine and (-)-β-hydrastine successfully predicted the goldenseal-midazolam interaction to within 20% of that observed after both chronic and acute goldenseal administration. Simulations implicated (-)-β-hydrastine as the major alkaloid precipitating the interaction, primarily via time-dependent inhibition of intestinal CYP3A, after chronic and acute goldenseal exposure. Results highlight the potential interplay between time-dependent and reversible inhibition of intestinal CYP3A as the mechanism underlying natural product-drug interactions, even after acute exposure to the precipitant. SIGNIFICANCE STATEMENT: Natural products can alter the pharmacokinetics of an object drug, potentially resulting in increased off-target effects or decreased efficacy of the drug. The objective of this work was to evaluate fundamental mechanisms underlying the clinically observed goldenseal-midazolam interaction. Results support the use of an integrated approach involving established in vitro assays, clinical evaluation, and physiologically based pharmacokinetic modeling to elucidate the complex interplay between multiple phytoconstituents and various pharmacokinetic processes driving a drug interaction. Copyright © 2023 by The Author(s). DOI: 10.1124/jpet.123.001681 PMCID: PMC10658920 PMID: 37541764 [Indexed for MEDLINE]