<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. Fitoterapia. 2024 Oct 28;179:106278. doi: 10.1016/j.fitote.2024.106278. Online ahead of print. Rare crocins ameliorate thrombus in zebrafish larvae by regulating lipid accumulation and clotting factors. Xu G(1), Xu P(1), Wang N(1), Qi W(1), Pu Y(2), Kang N(3), Chu J(4), He B(5). Author information: (1)College of Biotechnology and Pharmaceutical Engineering, Nanjing Tech University, Nanjing 211816, China. (2)2011college, Nanjing Tech University, Nanjing 211816, China. (3)School of Pharmaceutical Sciences, Nanjing Tech University, Nanjing 211816, China. (4)School of Pharmaceutical Sciences, Nanjing Tech University, Nanjing 211816, China. Electronic address: cjl2fl@njtech.edu.cn. (5)College of Biotechnology and Pharmaceutical Engineering, Nanjing Tech University, Nanjing 211816, China; School of Pharmaceutical Sciences, Nanjing Tech University, Nanjing 211816, China. Electronic address: bingfanghe@njtech.edu.cn. Crocin-4 is a water-soluble carotenoid that exhibits cardiovascular protection effects through anti-inflammatory and antioxidant effects. However, the pharmacodynamic effects and mechanisms of its analogues crocin-1 and crocin-2' have not been reported. In this study, we evaluated the protective effects of rare crocins on cardiovascular systems. In ox-LDL induced HUVECs model, 0.02, 0.1, 0.5, 1, 2, 3, 4, 5, 6 μg/mL crocin-1 and crocin-2' can increase cell viability by up to 80 %. Meanwhile, rare crocins at concentrations between 25-100 μg/mL crocin-1 and crocin-2' reduced the lipid accumulation by 30 % in cholesterol-induced zebrafish larvae. What's more, the therapeutic potential of rare crocins on thrombosis has also been explored. In vitro experiments, rare crocin-1 and crocin-2' at concentrations of 0.02, 0.05, 0.2, 0.5, 1, 2, 5, 10 μg/mL protected Human Umbilical Vein Endothelial Cells (HUVECs) against lipopolysaccharides-induced oxidative stress and inflammation. In vivo studies revealed that rare crocins at concentrations of 25, 50, 100, 150, 200, and 300 μg/mL exerted significant antithrombotic effect on arachidonic acid (AA)-induced zebrafish and there was nearly no potential risk for the deformity of zebrafish at 300 μg/mL dosages. In brief, rare crocins was viewed as a potentially useful candidate for the treatment of cardiovascular diseases because of its anti-inflammatory, antioxidant, and anticoagulant properties. Copyright © 2024 Elsevier B.V. All rights reserved. DOI: 10.1016/j.fitote.2024.106278 PMID: 39471880 Conflict of interest statement: Declaration of competing interest The authors declare that they have no known competing financial interests or personal relationships that could have appeared to influence the work reported in this paper. 2. Metabolites. 2024 Oct 21;14(10):566. doi: 10.3390/metabo14100566. Metabolomics Approach to Identify Biomarkers of Acute and Subacute Mastitis in Milk Samples: A Pilot Case-Control Study. Quifer-Rada P(1)(2)(3), Aguilar-Camprubí L(1), Samino S(3)(4), Amigó N(3)(4)(5), Soler O(4), Padró-Arocas A(1). Author information: (1)LactApp Women Health, 08014 Barcelona, Spain. (2)Department of Endocrinology & Nutrition, Biomedical Research Institute Sant Pau, Hospital de la Santa Creu i Sant Pau, 08025 Barcelona, Spain. (3)CIBER of Diabetes and Associated Metabolic Diseases, 28029 Madrid, Spain. (4)Biosfer Teslab, 43206 Reus, Spain. (5)Department of Basic Medical Sciences, University Rovira I Virgili, IISPV, 43204 Reus, Spain. Background and aims: Mastitis is one of the main complications during breastfeeding and contributes to the cessation of breastfeeding. However, the etiopathogenesis and diagnosis of mastitis are complex and not yet well defined. We aimed to identify metabolic and lipidic changes in human milk during acute and subacute mastitis in order to detect potential biomarkers of mastitis. Methods: We conducted a pilot case-control study including 14 breastfeeding women with acute mastitis, 32 with subacute mastitis symptoms, and 19 without any mastitis symptoms (control). Milk samples were collected and analyzed by proton nuclear magnetic resonance (H-NMR) for metabolomics analysis. To assess the association between the significant metabolites and lipids and the development of acute and subacute mastitis, multi-adjusted logistic regression models were developed. Results: The NMR-based metabolomics approach was able to identify and quantify a total of 40 metabolites in breast milk samples. After adjusting for confounding variables, acute mastitis was significantly associated with acetate (OR 3.9 IC 1.4-10.8), total cholesterol (OR 14 CI 3.2-62), esterified cholesterol (OR 3.3 CI 1.9-5.8), and sphingomyelin (OR 2.6 CI 1.2-5.8). The other metabolites presented weak association (OR < 2.5). Subacute mastitis was significantly associated with glutamine, lysophosphatidylcholine, phosphatidylcholine, plasmalogen, and total polyunsaturated fatty acids, but only cholesterol showed a strong association (OR > 2.5) with an OR of 2.6 (IC 1.1-6.6). Conclusions: Metabolic alteration in breast milk occurs during a process of both acute and subacute mastitis. Acetate, esterified cholesterol, lysophostidylcholine, and polyunsaturated fatty acids increased in both acute and subacute mastitis. However, according to the multi-adjusted regression logistic models, the candidate biomarkers for acute and subacute mastitis are cholesterol, lysophosphatidylcoholine, phosphatidylcholine, plasmalogen, and polyunsaturated fatty acids. DOI: 10.3390/metabo14100566 PMCID: PMC11509265 PMID: 39452946 Conflict of interest statement: Paola Quifer-Rada, Laia Aguilar-Camprubí and Alba Padró-Arocas are employees of LactApp Women’s Health; Sara Samino, Nuria Amigó and Oria Soler are employees of Biosfer Teslab, The paper reflects the views of the scientists, and not the company. 3. EuroIntervention. 2024 Oct 21;20(20):e1276-e1287. doi: 10.4244/EIJ-D-24-00240. Outcomes with revascularisation versus conservative management of participants with 3-vessel coronary artery disease in the ISCHEMIA trial. Bangalore S(1), Rhodes G(2), Maron DJ(3), Anthopolos R(1), O'Brien SM(2), Jones PG(4), Mark DB(2), Reynolds HR(1), Spertus JA(4), Stone GW(5), White HD(6), Xu Y(1), Fremes SE(7), Hochman JS(1), Ischemia Research Group OBOT. Author information: (1)Department of Medicine, New York University Grossman School of Medicine, New York, NY, USA. (2)Duke Clinical Research Institute, Durham, NC, USA. (3)Department of Medicine, Stanford University, Stanford, CA, USA. (4)Saint Luke's Mid America Heart Institute, University of Missouri-Kansas City, Kansas City, MO, USA. (5)The Zena and Michael A. Wiener Cardiovascular Institute, Icahn School of Medicine at Mount Sinai, New York, NY, USA. (6)Te Whatu Ora Health New Zealand, Te Toka Tumai, Green Lane Cardiovascular Services and University of Auckland, Auckland, New Zealand. (7)University of Toronto, Sunnybrook Health Sciences Centre, Toronto, ON, Canada. BACKGROUND: Whether revascularisation (REV) improves outcomes in patients with three-vessel coronary artery disease (3V-CAD) is uncertain. AIMS: Our objective was to evaluate outcomes with REV (percutaneous coronary intervention [PCI] or coronary artery bypass graft surgery [CABG]) versus medical therapy in patients with 3V-CAD. METHODS: ISCHEMIA participants with 3V-CAD on coronary computed tomography angiography without prior CABG were included. Outcomes following initial invasive management (INV) with REV (PCI or CABG) versus initial conservative management (CON) with medical therapy alone were evaluated. Regression modelling was used to estimate the outcomes if all participants were to undergo prompt REV versus those assigned to CON. Outcomes were cardiovascular (CV) death/myocardial infarction (MI), death, CV death, and quality of life. Bayesian posterior probability for benefit (Pr [benefit]) for 1 percentage point lower 4-year rates with REV versus CON were evaluated. RESULTS: Among 1,236 participants with 3V-CAD (612 INV/624 CON), REV was associated with lower 4-year CV death/MI (adjusted 4-year difference: -4.4, 95% credible interval [CrI] -8.7 to -0.3 percentage points, Pr [benefit]=94.8%) when compared with CON, with similar results for PCI versus CON (-5.8, 95% CrI: -10.8 to -0.5 percentage points, Pr [benefit]=96.4%) and CABG versus CON (-3.7, 95% CrI: -8.8 to 1.5 percentage points, Pr [benefit]=84.7%). Adjusted 4-year REV versus CON differences were as follows: death -1.2 (95% CrI: -4.7 to 2.2) percentage points, CV death -2.3 (95% CrI: -5.5 to 0.8) percentage points, with similar results for PCI and for CABG. The Pr (benefit) for death with REV (PCI or CABG) versus CON was 49-63%. The adjusted 12-month Seattle Angina Questionnaire-7 summary score differences favoured REV: REV versus CON 4.6 (95% CrI: 2.7-6.4) percentage points; PCI versus CON 3.6 (95% CrI: 1.2-5.8) percentage points and CABG versus CON 4.3 (95% CrI: 1.5-6.9) percentage points with high Pr (benefit). CONCLUSIONS: In participants with 3V-CAD, REV (either PCI or CABG) was associated with a lower 4-year CV death/MI rate and improved quality of life, with similar results for PCI versus CON and CABG versus CON. The differences in all-cause mortality between REV and CON were small with wide confidence intervals. (ClinicalTrials.gov: NCT01471522). DOI: 10.4244/EIJ-D-24-00240 PMCID: PMC11472139 PMID: 39432255 [Indexed for MEDLINE] Conflict of interest statement: S. Bangalore reports grants from the National Heart, Lung, and Blood Institute during the conduct of the study; grants and personal fees from Abbott; personal fees from Biotronik, Pfizer, Amgen, and Reata outside of the submitted work. G. Rhodes reports NIH funding. D.J. Maron reports grants from the National Heart, Lung, and Blood Institute during the conduct of the study. R. Anthopolos reports grants from the National Heart, Lung, and Blood Institute during the conduct of the study. S.M. O’Brien reports grants from the National Heart, Lung, and Blood Institute during the conduct of the study. D.B. Mark reports grants from National Heart, Lung, and Blood Institute during the conduct of the study; and grants from HeartFlow and Merck, outside the submitted work. H.R. Reynolds reports grants from the National Heart, Lung, and Blood Institute during the conduct of the study; and non-financial support from Abbott, Siemens, and BioTelemetry, outside of the submitted work. J.A. Spertus reports grants from National Heart, Lung, and Blood Institute during the conduct of the study; personal fees from Bayer, Novartis, AstraZeneca, Amgen, Janssen, and United Healthcare; and grants from American College of Cardiology, outside the submitted work; in addition, he has a patent copyright to the Seattle Angina Questionnaire with royalties paid; is on the Board of Directors for Blue Cross Blue Shield of Kansas City; and reports equity in Health Outcomes Sciences. G.W. Stone has received speaker honoraria from Medtronic, Pulnovo, and Infraredx; has served as a consultant to Valfix, TherOx, Robocath, HeartFlow, Ablative Solutions, Vectorious, Miracor, Neovasc, Abiomed, Ancora, Elucid Bio, Occlutech, CorFlow, Apollo Therapeutics, Impulse Dynamics, Cardiomech, Gore Medical, Amgen, Adona Medical, and Millennia Biopharma; and has equity/options from Ancora, Cagent, Applied Therapeutics, Biostar family of funds, SpectraWAVE, Orchestra Biomed, Aria, Cardiac Success, Valfix, and Xenter; his daughter is an employee at Medtronic; for institutional disclosure, his employer, Mount Sinai Hospital, receives research support from Abbott, Abiomed, Bioventrix, Cardiovascular Systems Inc, Philips, Biosense Webster, Shockwave Medical, Vascular Dynamics, Pulnovo, and V-Wave. H.D. White reports grants from National Heart, Lung, and Blood Institute during the conduct of the study; reports receiving grant support paid to the institution and fees for serving on a steering committee for the ODYSSEY OUTCOMES trial (Evaluation of Cardiovascular Outcomes After an Acute Coronary Syndrome During Treatment With Alirocumab) from Sanofi-Aventis and Regeneron Pharmaceuticals; for the ACCELERATE study (A Study of Evacetrapib in High-Risk Vascular Disease) from Eli Lilly; for the STRENGTH trial (Outcomes Study to Assess Statin Residual Risk Reduction With EpaNova in High CV Risk Patients With Hypertriglyceridemia) from Omthera Pharmaceuticals; for the HEART-FID study (Randomized Placebo-Controlled Trial of FCM as Treatment for Heart Failure With Iron Deficiency) from American Regent; for the CAMELLIA-TIMI study (A Study to Evaluate the Effect of Long-term Treatment With BELVIQ [Lorcaserin HC] on the Incidence of Major Adverse Cardiovascular Events and Conversion to Type 2 Diabetes Mellitus in Obese and Overweight Subjects With Cardiovascular Disease or Multiple Cardiovascular Risk Factors) from Eisai Inc; for the dal-GenE study (Effect of Dalcetrapib vs Placebo on CV Risk in a Genetically Defined Population With a Recent ACS) from DalCor Pharma UK Inc; for the AEGIS-II study from CSL Behring; for the SCORED trial (Effect of Sotagliflozin on Cardiovascular and Renal Events in Patients With Type 2 Diabetes and Moderate Renal Impairment Who Are at Cardiovascular Risk) and the SOLOIST-WHF trial (Effect of Sotagliflozin on Cardiovascular Events in Patients With Type2 Diabetes Post Worsening Heart Failure) from Sanofi-Aventis Australia Pty Ltd; and for the CLEAR Outcomes Study (Evaluation of Major Cardiovascular Events in Patients With, or at High Risk for, Cardiovascular Disease Who Are Statin Intolerant Treated With Bempedoic Acid. [ETC-1002] or Placebo) from Esperion Therapeutics Inc; he was on the advisory board for Genentech, Inc.; and received lecture fees from AstraZeneca. Y. Xu reports grants from the National Heart, Lung, and Blood Institute during the conduct of the study. J.S. Hochman is the PI for the ISCHEMIA trial for which, in addition to support by the National Heart, Lung, and Blood Institute grant, devices and medications were provided by Abbott, Medtronic, Abbott Laboratories (formerly St. Jude Medical, Inc.), Royal Philips NV (formerly Volcano Corporation), Arbor Pharmaceuticals, LLC, AstraZeneca Pharmaceuticals, LP, Merck Sharp & Dohme Corp., Omron Healthcare, Inc., Sunovion Pharmaceuticals, Inc., Espero BioPharma, and Amgen, Inc.; and received financial donations from Arbor Pharmaceuticals LLC and AstraZeneca Pharmaceuticals LP. The other authors have no conflicts of interest to declare. 4. Carbohydr Res. 2024 Oct 16;545:109292. doi: 10.1016/j.carres.2024.109292. Online ahead of print. Picoplatin (II)-loaded chitosan nanocomposites as effective drug delivery systems: Preparation, mechanistic investigation of BSA/5-GMP/GSH binding and biological evaluations. Ahmed NM(1), Ibrahim MM(2), Elmehasseb IM(1), Shaban SY(3). Author information: (1)Chemistry Department, Faculty of Science, Kafrelsheikh University, Kafrelsheikh 33516, Egypt. (2)Department of Chemistry, College of Science, Taif University, P.O. Box 11099, Taif, 21944, Saudi Arabia. (3)Chemistry Department, Faculty of Science, Kafrelsheikh University, Kafrelsheikh 33516, Egypt. Electronic address: shaban.shaban@sci.kfs.edu.eg. The goal of the current study is to improve the characteristics and bioavailability of the drug picoplatin (PPt) by encapsulating it in chitosan nanoparticles (CS NPs) which allows for the targeted delivery of cytotoxic cargo to cancerous tissue, reducing toxic side effects and raising the therapeutic index. When picoplatin was delivered into the CS, it was able to produce a complex with CS (PPt@CS NPs) that had an appropriate particle size of 275 ± 10 nm, a reasonably low PDI of 0.15 ± 0.05, and high stability (ζ = -22.1 ± 0.3 mV). Since almost all pharmaceuticals work by binding to specific proteins or DNA, the in vitro binding mechanism and affinity of bovine serum albumin (BSA), low molecular building units of nucleic acids (5-GMP), and Glutathione (GSH) (considering that cisplatin resistance could be due to a reaction between cisplatin and GSH) to PPt and PPt@CS NPs were examined using stopped-flow and other spectroscopic approaches. Through two reversible processes, a rapid second-order binding followed by a slower first-order isomerization reaction, and a static quenching mechanism, PPt and PPt@CS NPs bind to BSA with relative reactivity of around (PPt)/(PPt@CS NPs) = 1/2.5. The 5-GMP interaction studies demonstrated that, in addition to changing the binding mechanism, PPt's encapsulation in CS increases its rate of reaction through coordination affinity. PPt interacted with 5-GMP via two reversible processes, a rapid second-order binding to phosphate followed by a slower first-order migration to the N7 of pyrimidine moiety. PPt@CS NPs showed weaker binding to GSH compared to PPt and hence PPt@CS NPs exhibits a lower resistance factor. It was also found that the in vitro drug release of PPt@CS NPs in PBS at pH 7.4 was steady, releasing 30 % of the PPt in just 5 h. Nonetheless, 75 % of the release in a pH 5.4 solution containing 10 mM GSH-a solution that mimics the tumor microenvironment-shows that the PPt@CS NPs system is sensitive to GSH and specifically targets malignant tissue. The encapsulation of PPt in CS complex maintained its anticancer activity, as shown by an in vitro cell-survival assay on HepG2 cancer cell lines and also cleavage efficiency toward the minor groove of pBR322 DNA via the hydrolytic way. These findings collectively suggested that inclusion PPt in CS would be an effective strategy to formulate a novel picoplatin formulation intended for use as targeted anticancer treatment. Copyright © 2024 Elsevier Ltd. All rights reserved. DOI: 10.1016/j.carres.2024.109292 PMID: 39427432 Conflict of interest statement: Declaration of competing interest The authors declare that they have no known competing financial interests or personal relationships that could have appeared to influence the work reported in this paper. 5. J Nutr. 2024 Oct 12:S0022-3166(24)01100-3. doi: 10.1016/j.tjnut.2024.10.023. Online ahead of print. A Method to Estimate the Dietary α-Linolenic Acid Requirement Using Nonesterified DHA and Arachidonic Acid Oxylipins and Fatty Acids. Manson A(1), Sidhu KK(1), Fedorova O(1), La HHK(1), Magaji E(1), Nguyen LKL(1), Winter T(1), Aukema HM(2). Author information: (1)Food and Human Nutritional Sciences, University of Manitoba, Winnipeg, MB, Canada; Canadian Centre for Agri-Food Research in Health and Medicine (CCARM), Winnipeg, MB, Canada. (2)Food and Human Nutritional Sciences, University of Manitoba, Winnipeg, MB, Canada; Canadian Centre for Agri-Food Research in Health and Medicine (CCARM), Winnipeg, MB, Canada. Electronic address: harold.aukema@umanitoba.ca. BACKGROUND: The dietary requirement for α-linolenic acid (ALA) remains unclear, as evidenced by the absence of an RDA for this essential fatty acid (FA). In previous studies, we observed that the amount of dietary ALA required to maximize nonesterified (NE) DHA oxylipins appears to be higher than the amount required to maximize tissue esterified DHA levels, which have classically been used to estimate the ALA requirement. Further, we observed that dietary ALA reduces esterified arachidonic acid (ARA) and its NE oxylipins. OBJECTIVES: Since NE oxylipins and FA mediate the biological activities of FA, we examined whether these DHA and ARA pools could be used to determine the dietary ALA requirement. METHODS: Nine groups of 4-wk-old male Sprague-Dawley rats (n = 5) and 10 groups of male and female CD1 mice (n = 6) were provided 0.1-2.5 g ALA and 2 g of linoleic acid per 100 g of AIN93G-based diets. NE DHA and ARA and their oxylipins in serum, liver, kidney, and brain homogenates underwent solid phase extraction and were quantified by HPLC-MS/MS. Breakpoint analysis of transitions from increase to plateau was conducted using piecewise regression. RESULTS: In response to increasing dietary ALA, NE DHA oxylipins, and DHA in serum, liver, and kidney (but not the brain) initially increased rapidly and then plateaued whereas ARA oxylipins and ARA tended to decrease and plateau. Thus, breakpoints were calculated for the ratios of DHA/ARA and hydroxy-DHA/hydroxy-ARA (DHAOH/ARAOH), which consisted of oxylipins synthesized via pathways common to both FA. In serum, liver, and kidney, the highest estimated breakpoint indicated an ALA requirement of ∼0.7 g/100 g diet (1.7% energy), approximately twice that of previous estimations. CONCLUSIONS: This study supports the use of NE DHAOH/ARAOH or DHA/ARA as biochemical indicators of the ALA requirement. Applying this method in rats and mice indicates that the requirement is higher than previously estimated using esterified DHA alone. Copyright © 2024 American Society for Nutrition. Published by Elsevier Inc. All rights reserved. DOI: 10.1016/j.tjnut.2024.10.023 PMID: 39401685 Conflict of interest statement: Conflict of interest statement The authors report no conflict of interest.