1
|
Sosa R, Espinosa-Villafranca P, Saavedra P, Chávez-Hernández ME, Leal-Galicia P, Lago G, Mata F, Mata-Luévanos J, Rodríguez-Serrano LM, Tapia-De-Jesús A, Buenrostro-Jáuregui M. Assessing acute effects of methylphenidate and modafinil on inhibitory capacity, time estimation, attentional lapses, and compulsive-like behavior in rats. Behav Pharmacol 2025; 36:76-96. [PMID: 39883117 DOI: 10.1097/fbp.0000000000000815] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/31/2025]
Abstract
Medications known as 'cognitive enhancers' are increasingly being consumed off-label by healthy people, raising concerns about their safety. The aim of our study was to profile behavioral performance upon oral administration of methylphenidate (2.5 mg/kg) and modafinil (64 mg/kg) - two popular cognitive enhancers - and upon their discontinuation. We modeled cognitively demanding challenges in neurotypical individuals using a behavioral task where Wistar - Lewis rats had to withhold responses for a specified time to obtain food rewards. This task allowed us to extract several measures of behavioral performance associated with clinically meaningful indices, such as compulsive-like responding, incapacity to wait (impulsivity), time estimation (precision and accuracy), and attentional lapses. Our study design involved examining these behavioral indices in subjects administered either methylphenidate, modafinil, or vehicle. We found that subjects administered modafinil obtained fewer rewards and were less efficient in reward pursuing than the vehicle group; this result was likely due to a drug-induced inability to wait. Upon modafinil discontinuation, subjects earned more rewards but did not entirely catch up with the vehicle group. As for methylphenidate, neither favorable nor unfavorable effects were found in our main analyses. However, an exploratory analysis of changes in behavioral performance within sessions suggested that methylphenidate fostered favorable, yet short-lived, effects. We discuss our results in terms of the risks and cost-benefits of doses above or below the effective dose of cognitive enhancement drugs.
Collapse
Affiliation(s)
- Rodrigo Sosa
- Universidad Panamericana, Escuela de Pedagogía y Psicología, Guadalajara, Mexico
| | - Pedro Espinosa-Villafranca
- Max Planck School of Cognition, Max Planck Institute for Human Cognitive and Brain Science, Leipzig, Germany
| | - Pablo Saavedra
- Facultad de Psicología, Universidad Nacional Autónoma de México, Mexico City
| | | | | | - Gustavo Lago
- Departamento de Psicología, Universidad Iberoamericana Ciudad de México
| | - Florencia Mata
- Departamento de Psicología, Universidad Iberoamericana Ciudad de México
| | | | | | | | | |
Collapse
|
2
|
Holborn T, Schifano F, Smith E, Deluca P. The Use of Novel Stimulants in ADHD Self-Medication: A Mixed Methods Analysis. Brain Sci 2025; 15:292. [PMID: 40149813 PMCID: PMC11940814 DOI: 10.3390/brainsci15030292] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/15/2025] [Revised: 03/04/2025] [Accepted: 03/07/2025] [Indexed: 03/29/2025] Open
Abstract
BACKGROUND Numerous individuals suffer from attentional issues, such as ADHD. While medication is considered the first-line treatment, it is unavailable to some. As a result, certain individuals are choosing to self-medicate with novel stimulants, a phenomenon that remains poorly understood. We aimed to investigate which NPSs are being used to self-medicate ADHD, evaluate their perceived effectiveness, and explore the experiences and motivations of those self-medicating. METHODS Data from respondents (n = 225) (mean age [SD] = 29.5 ± 9.6; male = 83%; female = 12%; non-binary = 5%) were collected via an online survey, with nine participants (mean age = 31.4; male = 5; female = 1; non-binary = 3) undertaking further semi-structured interviews and the data being investigated using a framework analysis. RESULTS The most-used NPSs were 4F-MPH and 2-FMA. Some individuals perceived self-medication to be more effective than conventional treatment (p < 0.001). A framework analysis identified the following themes surrounding novel stimulant self-medication: (1) the use of NPS stimulants as a stopgap between treatments; (2) poor access to ADHD treatment; (3) a lack of openness and confidence in psychiatrists and healthcare providers. CONCLUSION Novel stimulants are being used when access to ADHD treatment is poor. Interventions should aim to reduce long treatment wait times and issues surrounding geographical access. Careful consideration should be given before denying stimulant medication to individuals with co-occurring substance use and psychiatric comorbidity. Individuals desire a more patient-centred ADHD treatment with broader pharmacotherapies.
Collapse
Affiliation(s)
- Tayler Holborn
- Institute of Psychiatry, Psychology & Neuroscience, King’s College London, 16 De Crespigny Park, London SE5 8AF, UK; (T.H.)
| | - Fabrizio Schifano
- School of Life and Medical Sciences, University of Hertfordshire, Hatfield AL10 9AB, UK
| | - Emma Smith
- Institute of Psychiatry, Psychology & Neuroscience, King’s College London, 16 De Crespigny Park, London SE5 8AF, UK; (T.H.)
| | - Paolo Deluca
- Institute of Psychiatry, Psychology & Neuroscience, King’s College London, 16 De Crespigny Park, London SE5 8AF, UK; (T.H.)
| |
Collapse
|
3
|
Niżnik Ł, Jabłońska K, Orczyk M, Orzechowska M, Toporowska-Kaźmierak J, Sowińska M, Jasińska J, Jurowski K. Toxicity of New Psychoactive Substance (NPS): Threo-4-methylmethylphenidate (4-Mmph) - Prediction of toxicity using in silico methods. Toxicol In Vitro 2024; 99:105891. [PMID: 38972515 DOI: 10.1016/j.tiv.2024.105891] [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: 03/06/2024] [Revised: 06/28/2024] [Accepted: 07/03/2024] [Indexed: 07/09/2024]
Abstract
This study represents the first application of in silico methods to evaluate the toxicity of 4-methylphenidate (4-Mmph), a new psychoactive substance (NPS). Using advanced in silico toxicology tools, it was feasible to anticipate key aspects of 4-Mmph's toxicological profile, including acute toxicity (LD50), genotoxicity, cardiotoxicity, and possible endocrine disruption. The findings indicate significant acute toxicity with variability among species, a high potential for adverse effects in the gastrointestinal system and lungs, a low genotoxic potential, a significant likelihood of skin irritation, and a notable cardiotoxicity risk associated with hERG channel inhibition. Evaluation of endocrine disruption revealed a low likelihood that 4-Mmph interacts with the estrogen receptor alpha (ER-α), indicating minimal estrogenic activity. These insights, derived from in silico studies, play a crucial role in improving the comprehension of 4-Mmph in forensic and clinical toxicology. These initial toxicological inquiries establish the foundation for future investigations and help formulate risk assessment and management strategies regarding the use and abuse of NPS. This article is part of a larger project funded by the Polish Ministry of Education and Science, titled "Toxicovigilance, Poisoning Prevention, and First Aid in Poisoning with Xenobiotics of Current Clinical Importance in Poland" (Grant Number SKN/SP/570184/2023).
Collapse
Affiliation(s)
- Łukasz Niżnik
- Department of Regulatory and Forensic Toxicology, Institute of Medical Expertises in Łódź, ul. Aleksandrowska 67/93, 91-205 Łódź, Poland
| | - Karolina Jabłońska
- Toxicological Science Club 'Paracelsus', Institute of Medical Sciences, Medical College, Rzeszów University, Al. mjr. W. Kopisto 2a, 35-959 Rzeszów, Poland
| | - Michał Orczyk
- Toxicological Science Club 'Paracelsus', Institute of Medical Sciences, Medical College, Rzeszów University, Al. mjr. W. Kopisto 2a, 35-959 Rzeszów, Poland
| | - Martyna Orzechowska
- Toxicological Science Club 'Paracelsus', Institute of Medical Sciences, Medical College, Rzeszów University, Al. mjr. W. Kopisto 2a, 35-959 Rzeszów, Poland
| | - Joanna Toporowska-Kaźmierak
- Toxicological Science Club 'Paracelsus', Institute of Medical Sciences, Medical College, Rzeszów University, Al. mjr. W. Kopisto 2a, 35-959 Rzeszów, Poland
| | - Marta Sowińska
- Toxicological Science Club 'Paracelsus', Institute of Medical Sciences, Medical College, Rzeszów University, Al. mjr. W. Kopisto 2a, 35-959 Rzeszów, Poland
| | - Judyta Jasińska
- Toxicological Science Club 'Paracelsus', Institute of Medical Sciences, Medical College, Rzeszów University, Al. mjr. W. Kopisto 2a, 35-959 Rzeszów, Poland
| | - Kamil Jurowski
- Department of Regulatory and Forensic Toxicology, Institute of Medical Expertises in Łódź, ul. Aleksandrowska 67/93, 91-205 Łódź, Poland; Laboratory of Innovative Toxicological Research and Analyses, Institute of Medical Sciences, Medical College, Rzeszów University, Al. mjr. W. Kopisto 2a, 35-959 Rzeszów, Poland.
| |
Collapse
|
4
|
Karabulut S, Kaur H, Gauld JW. Uncovering Structure-Activity Relationships of Phenethylamines: Paving the Way for Innovative Mental Health Treatments. ACS Chem Neurosci 2024; 15:972-982. [PMID: 38381069 DOI: 10.1021/acschemneuro.3c00677] [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] [Indexed: 02/22/2024] Open
Abstract
The rapidly evolving psychedelic industry has garnered considerable attention due to 3,4-methylenedioxymethamphetamine-assisted psychotherapy's ground-breaking success in treating moderate-to-severe Post-traumatic Stress Disorder in two Phase 3 clinical trials. This has opened Pandora's box for the development of innovative therapeutic modalities. Of particular interest are the phenethylamines and their ability to inhibit monoamine transporters. In this study, we employed the quantitative structure-activity relationship methodology to develop three vigorous models for the reuptake of serotonin, dopamine, and norepinephrine through monoamine transporters. These models were thoroughly validated using various criteria, including fitting (R2DAT = 0.869, R2SERT = 0.828, and R2NET = 0.887), internal (Q2looDAT = 0.795, Q2looSERT = 0.784, and Q2looNET = 0.820), and external (RMSEextDAT = 0.373, R2extDAT = 0.831, RMSEextSERT = 0.200, R2extSERT = 0.955, RMSEextNET = 0.318, and R2extNET = 0.711) criteria.
Collapse
Affiliation(s)
- Sedat Karabulut
- Department of Chemistry and Biochemistry, University of Windsor, Windsor, Ontario N9B 3P4, Canada
| | - Harpreet Kaur
- Pharmala Biotech, 82 Richmond Street E, Toronto, Ontario M5C 1P1, Canada
| | - James W Gauld
- Department of Chemistry and Biochemistry, University of Windsor, Windsor, Ontario N9B 3P4, Canada
| |
Collapse
|
5
|
Sakamoto M, Suzuki T, Teraoka D, Tanaka K, Saeki Y, Kishimoto K, Nagashima M, Nakajima J, Suzuki J, Inomata A, Moriyasu T, Fukaya H. Analytical characterization and differentiation between threo- and erythro-4-fluoroethylphenidate. Forensic Toxicol 2023; 41:272-286. [PMID: 37097346 DOI: 10.1007/s11419-023-00664-y] [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: 12/05/2022] [Accepted: 04/05/2023] [Indexed: 04/26/2023]
Abstract
PURPOSE Methylphenidate analogs appeared on the drug market during the last years. Its analogs contain two chiral centers and, thus, have potential varying configurations (i.e., threo and erythro forms). This study presents the analytical characterization of 4-fluoroethylphenidate (4-FEP) and its differentiation between threo- and erythro-4-FEP. METHODS Analysis of the samples included high-performance liquid chromatography (HPLC), gas chromatography-electron ionization-mass spectrometry (GC-EI-MS), high-resolution mass spectrometry (HRMS) analyses, nuclear magnetic resonance (NMR) spectroscopy and X-ray crystal structure analysis. RESULTS NMR spectroscopic investigations confirmed the differences between threo- and erythro-4-FEP, and demonstrated that both isomers could be separated using HPLC and GC methods. Two samples obtained from one vendor in 2019 consisted of threo-4-FEP, whereas the other two samples obtained from a different vendor in 2020 consisted of a mixture of threo- and erythro-4-FEP. CONCLUSIONS Several analytical approaches including HPLC, GC-EI-MS, HRMS analyses, NMR spectroscopy and X-ray crystal structure analysis enabled the unambiguous identification of threo- and erythro-4-FEP. The analytical data presented in this article will be useful for identifying threo- and erythro-4-FEP included in illicit products.
Collapse
Affiliation(s)
- Miho Sakamoto
- Tokyo Metropolitan Institute of Public Health, 24-1 Hyakunincho 3-chome, Shinjuku-ku, Tokyo, 169-0073, Japan.
| | - Toshinari Suzuki
- Tokyo Metropolitan Institute of Public Health, 24-1 Hyakunincho 3-chome, Shinjuku-ku, Tokyo, 169-0073, Japan
| | - Daisuke Teraoka
- Tokyo Metropolitan Institute of Public Health, 24-1 Hyakunincho 3-chome, Shinjuku-ku, Tokyo, 169-0073, Japan
| | - Kazue Tanaka
- Tokyo Metropolitan Institute of Public Health, 24-1 Hyakunincho 3-chome, Shinjuku-ku, Tokyo, 169-0073, Japan
| | - Yuki Saeki
- Tokyo Metropolitan Institute of Public Health, 24-1 Hyakunincho 3-chome, Shinjuku-ku, Tokyo, 169-0073, Japan
| | - Kiyoko Kishimoto
- Tokyo Metropolitan Institute of Public Health, 24-1 Hyakunincho 3-chome, Shinjuku-ku, Tokyo, 169-0073, Japan
| | - Machiko Nagashima
- Tokyo Metropolitan Institute of Public Health, 24-1 Hyakunincho 3-chome, Shinjuku-ku, Tokyo, 169-0073, Japan
| | - Jun'ichi Nakajima
- Tokyo Metropolitan Institute of Public Health, 24-1 Hyakunincho 3-chome, Shinjuku-ku, Tokyo, 169-0073, Japan
| | - Jin Suzuki
- Tokyo Metropolitan Institute of Public Health, 24-1 Hyakunincho 3-chome, Shinjuku-ku, Tokyo, 169-0073, Japan
| | - Akiko Inomata
- Tokyo Metropolitan Institute of Public Health, 24-1 Hyakunincho 3-chome, Shinjuku-ku, Tokyo, 169-0073, Japan
| | - Takako Moriyasu
- Tokyo Metropolitan Institute of Public Health, 24-1 Hyakunincho 3-chome, Shinjuku-ku, Tokyo, 169-0073, Japan
- Tokyo Food Sanitation Association Food Research Laboratory, 1-19-10 Tokumaru, Itabashi-ku, Tokyo, 175-0083, Japan
| | - Haruhiko Fukaya
- Tokyo University of Pharmacy and Life Sciences, 1432-1 Horinouchi, Hachioji, Tokyo, 192-0392, Japan
| |
Collapse
|
6
|
Gerostamoulos D, Glowacki L, Pricone M, Crump K, Di Rago M, Joubert S, Lynch MJ, Woodford NW, Drummer OH. Fatal Intoxications from a Combination of 4-Fluoroamphetamine and 25C-NBOMe. J Anal Toxicol 2023; 47:191-196. [PMID: 35975553 DOI: 10.1093/jat/bkac059] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/27/2022] [Revised: 08/05/2022] [Accepted: 08/15/2022] [Indexed: 11/12/2022] Open
Abstract
Six fatalities have occurred from the ingestion of a combination of new psychoactive substances (NPSs), 4-fluoroamphetamine (4FA) and 2-(4-chloro-2,5-dimethoxyphenyl)-N-[(2-methoxyphenyl)methyl]ethanamine (25C-NBOMe) over a 9-month period. Four of these fatalities (one older female and three young males) were from direct adverse effects of drugs, and one each from a fall while being intoxicated and during restraint. All cases were subject to full postmortem examinations that included collection of femoral blood. The four drug-caused fatalities had postmortem blood concentrations for 4FA and 25C-NBOMe of 330-682 ng/L (median 417) and 1.4-12 ng/mL (median 4.3), respectively. The other two cases (both young males) where death was considered to have been caused indirectly by drug intoxication had 4FA and 25C-NBOMe postmortem concentrations of 21 and 123 ng/mL, and 1.8 and 4.5 ng/mL, respectively. None of these cases showed concentrations of drugs that suggested use of high recreational doses. In one drug-caused death, capsules and a brown powder obtained from the scene were found to contain a mixture of these two NPSs. With the exception of one drug-caused death, other drugs were detected; however, the effects of the two NPSs together were regarded as the primary triggers for the deaths. There were no consistent symptoms or pathology in these cases; however, agitation/aggression was observed in two cases prior to their collapse, with seizures in possibly three cases. Pulmonary and/or cerebral edema was noted in three cases. Potentially significant natural disease (a mildly enlarged heart) was only observed in one drug-caused case. These cases illustrate a possible increased risk of sudden death with this combination of drugs, both of which can elevate serotonin concentrations as well as act as strong stimulants. These cases also illustrate the difficulty in detecting NPS in cases where no prior information is available that might suggest their use.
Collapse
Affiliation(s)
- Dimitri Gerostamoulos
- Victorian Institute of Forensic Medicine, 65 Kavanagh Street, Southbank, Victoria 3006, Australia
- Department of Forensic Medicine, Monash University, School of Public Health and Preventive Medicine, 65 Kavanagh Street, Southbank, Victoria 3006, Australia
| | - Linda Glowacki
- Victorian Institute of Forensic Medicine, 65 Kavanagh Street, Southbank, Victoria 3006, Australia
| | - Maria Pricone
- Victorian Institute of Forensic Medicine, 65 Kavanagh Street, Southbank, Victoria 3006, Australia
| | - Kerryn Crump
- Victorian Institute of Forensic Medicine, 65 Kavanagh Street, Southbank, Victoria 3006, Australia
| | - Matthew Di Rago
- Victorian Institute of Forensic Medicine, 65 Kavanagh Street, Southbank, Victoria 3006, Australia
- Department of Forensic Medicine, Monash University, School of Public Health and Preventive Medicine, 65 Kavanagh Street, Southbank, Victoria 3006, Australia
| | - Samantha Joubert
- Victorian Institute of Forensic Medicine, 65 Kavanagh Street, Southbank, Victoria 3006, Australia
| | - Matthew J Lynch
- Victorian Institute of Forensic Medicine, 65 Kavanagh Street, Southbank, Victoria 3006, Australia
- Department of Forensic Medicine, Monash University, School of Public Health and Preventive Medicine, 65 Kavanagh Street, Southbank, Victoria 3006, Australia
| | - Noel W Woodford
- Victorian Institute of Forensic Medicine, 65 Kavanagh Street, Southbank, Victoria 3006, Australia
- Department of Forensic Medicine, Monash University, School of Public Health and Preventive Medicine, 65 Kavanagh Street, Southbank, Victoria 3006, Australia
| | - Olaf H Drummer
- Victorian Institute of Forensic Medicine, 65 Kavanagh Street, Southbank, Victoria 3006, Australia
- Department of Forensic Medicine, Monash University, School of Public Health and Preventive Medicine, 65 Kavanagh Street, Southbank, Victoria 3006, Australia
| |
Collapse
|
7
|
Oh HA, Lee JH, Woo DH. Acute effects of 4-EA-NBOMe, 3, 4-DCMP, LY-2183240 and AB-CHFUPYCA on the excitability of rat cortical primary cultured neurons. Mol Cell Toxicol 2022. [DOI: 10.1007/s13273-022-00224-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/28/2022]
|
8
|
Casiraghi A, Longhena F, Faustini G, Ribaudo G, Suigo L, Camacho-Hernandez GA, Bono F, Brembati V, Newman AH, Gianoncelli A, Straniero V, Bellucci A, Valoti E. Methylphenidate Analogues as a New Class of Potential Disease-Modifying Agents for Parkinson's Disease: Evidence from Cell Models and Alpha-Synuclein Transgenic Mice. Pharmaceutics 2022; 14:1595. [PMID: 36015221 PMCID: PMC9414221 DOI: 10.3390/pharmaceutics14081595] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/17/2022] [Revised: 07/20/2022] [Accepted: 07/26/2022] [Indexed: 12/02/2022] Open
Abstract
Parkinson's disease (PD) is characterized by dopaminergic nigrostriatal neurons degeneration and Lewy body pathology, mainly composed of α-synuclein (αSyn) fibrillary aggregates. We recently described that the neuronal phosphoprotein Synapsin III (Syn III) participates in αSyn pathology in PD brains and is a permissive factor for αSyn aggregation. Moreover, we reported that the gene silencing of Syn III in a human αSyn transgenic (tg) mouse model of PD at a pathological stage, manifesting marked insoluble αSyn deposits and dopaminergic striatal synaptic dysfunction, could reduce αSyn aggregates, restore synaptic functions and motor activities and exert neuroprotective effects. Interestingly, we also described that the monoamine reuptake inhibitor methylphenidate (MPH) can recover the motor activity of human αSyn tg mice through a dopamine (DA) transporter-independent mechanism, which relies on the re-establishment of the functional interaction between Syn III and α-helical αSyn. These findings support that the pathological αSyn/Syn III interaction may constitute a therapeutic target for PD. Here, we studied MPH and some of its analogues as modulators of the pathological αSyn/Syn III interaction. We identified 4-methyl derivative I-threo as a lead candidate modulating αSyn/Syn III interaction and having the ability to reduce αSyn aggregation in vitro and to restore the motility of αSyn tg mice in vivo more efficiently than MPH. Our results support that MPH derivatives may represent a novel class of αSyn clearing agents for PD therapy.
Collapse
Affiliation(s)
- Andrea Casiraghi
- Department of Pharmaceutical Sciences, University of Milan, Via Luigi Mangiagalli 25, 20133 Milano, Italy; (A.C.); (L.S.); (E.V.)
| | - Francesca Longhena
- Department of Molecular and Translational Medicine, University of Brescia, Viale Europa 11, 25123 Brescia, Italy; (F.L.); (G.F.); (G.R.); (F.B.); (V.B.); (A.G.); (A.B.)
| | - Gaia Faustini
- Department of Molecular and Translational Medicine, University of Brescia, Viale Europa 11, 25123 Brescia, Italy; (F.L.); (G.F.); (G.R.); (F.B.); (V.B.); (A.G.); (A.B.)
| | - Giovanni Ribaudo
- Department of Molecular and Translational Medicine, University of Brescia, Viale Europa 11, 25123 Brescia, Italy; (F.L.); (G.F.); (G.R.); (F.B.); (V.B.); (A.G.); (A.B.)
| | - Lorenzo Suigo
- Department of Pharmaceutical Sciences, University of Milan, Via Luigi Mangiagalli 25, 20133 Milano, Italy; (A.C.); (L.S.); (E.V.)
| | - Gisela Andrea Camacho-Hernandez
- Medicinal Chemistry Section, Molecular Targets and Medications Discovery Branch, NIDA-IRP, 333 Cassell Drive, Baltimore, MD 21224, USA; (G.A.C.-H.); (A.H.N.)
| | - Federica Bono
- Department of Molecular and Translational Medicine, University of Brescia, Viale Europa 11, 25123 Brescia, Italy; (F.L.); (G.F.); (G.R.); (F.B.); (V.B.); (A.G.); (A.B.)
| | - Viviana Brembati
- Department of Molecular and Translational Medicine, University of Brescia, Viale Europa 11, 25123 Brescia, Italy; (F.L.); (G.F.); (G.R.); (F.B.); (V.B.); (A.G.); (A.B.)
| | - Amy Hauck Newman
- Medicinal Chemistry Section, Molecular Targets and Medications Discovery Branch, NIDA-IRP, 333 Cassell Drive, Baltimore, MD 21224, USA; (G.A.C.-H.); (A.H.N.)
| | - Alessandra Gianoncelli
- Department of Molecular and Translational Medicine, University of Brescia, Viale Europa 11, 25123 Brescia, Italy; (F.L.); (G.F.); (G.R.); (F.B.); (V.B.); (A.G.); (A.B.)
| | - Valentina Straniero
- Department of Pharmaceutical Sciences, University of Milan, Via Luigi Mangiagalli 25, 20133 Milano, Italy; (A.C.); (L.S.); (E.V.)
| | - Arianna Bellucci
- Department of Molecular and Translational Medicine, University of Brescia, Viale Europa 11, 25123 Brescia, Italy; (F.L.); (G.F.); (G.R.); (F.B.); (V.B.); (A.G.); (A.B.)
| | - Ermanno Valoti
- Department of Pharmaceutical Sciences, University of Milan, Via Luigi Mangiagalli 25, 20133 Milano, Italy; (A.C.); (L.S.); (E.V.)
| |
Collapse
|
9
|
Mohr ALA, Logan BK, Fogarty MF, Krotulski AJ, Papsun DM, Kacinko SL, Huestis MA, Ropero-Miller JD. Reports of Adverse Events Associated with Use of Novel Psychoactive Substances, 2017-2020: A Review. J Anal Toxicol 2022; 46:e116-e185. [PMID: 35445267 PMCID: PMC9282356 DOI: 10.1093/jat/bkac023] [Citation(s) in RCA: 27] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/14/2021] [Revised: 03/17/2022] [Accepted: 04/20/2022] [Indexed: 12/15/2022] Open
Abstract
An important role of modern forensic and clinical toxicologists is to monitor the adverse events of novel psychoactive substances (NPS). Following a prior review from 2013 to 2016, this critical literature review analyzes and evaluates published case reports for NPS from January 2017 through December 2020. The primary objective of this study is to assist in the assessment and interpretation of these cases as well as provide references for confirmation methods. Chemistry, pharmacology, adverse events and user profiles (e.g., polypharmacy) for NPS are provided including case history, clinical symptoms, autopsy findings and analytical results. Literature reviews were performed in PubMed and Google Scholar for publications using search terms such as NPS specific names, general terms (e.g., 'designer drugs' and 'novel psychoactive substances'), drug classes (e.g., 'designer stimulants') and outcome-based terms (e.g., 'overdose' and 'death'). Government and website drug surveillance databases and abstracts published by professional forensic science organizations were also searched. Toxicological data and detailed case information were extracted, tabulated, analyzed and organized by drug category. Case reports included overdose fatalities (378 cases), clinical treatment and hospitalization (771 cases) and driving under the influence of drugs (170 cases) for a total of 1,319 cases providing details of adverse events associated with NPS. Confirmed adverse events with associated toxidromes of more than 60 NPS were reported including synthetic cannabinoid, NPS stimulant, NPS hallucinogen, NPS benzodiazepine and NPS opioid cases. Fifty of these NPS were reported for the first time in January 2017 through December 2020 as compared to the previous 4 years surveyed. This study provides insight and context of case findings described in the literature and in digital government surveillance databases and websites during a recent 4-year period. This review will increase the awareness of adverse events associated with NPS use to better characterize international emerging drug threats.
Collapse
Affiliation(s)
- Amanda L A Mohr
- Center for Forensic Science Research and Education at the Fredric Rieders Family Foundation, 2300 Stratford Ave, Willow Grove, PA 19090, USA
| | - Barry K Logan
- Center for Forensic Science Research and Education at the Fredric Rieders Family Foundation, 2300 Stratford Ave, Willow Grove, PA 19090, USA
- NMS Labs, 200 Welsh Rd, Horsham, PA 19044, USA
| | - Melissa F Fogarty
- Center for Forensic Science Research and Education at the Fredric Rieders Family Foundation, 2300 Stratford Ave, Willow Grove, PA 19090, USA
| | - Alex J Krotulski
- Center for Forensic Science Research and Education at the Fredric Rieders Family Foundation, 2300 Stratford Ave, Willow Grove, PA 19090, USA
| | | | | | - Marilyn A Huestis
- Center for Forensic Science Research and Education at the Fredric Rieders Family Foundation, 2300 Stratford Ave, Willow Grove, PA 19090, USA
- Institute of Emerging Health Professions, Thomas Jefferson University, Philadelphia, PA 19107, USA
| | - Jeri D Ropero-Miller
- RTI International, Center for Forensic Sciences, 3040 East Cornwallis Rd, Research Triangle Park, NC 27709, USA
| |
Collapse
|
10
|
Kang KR, Kim CY, Kim J, Ryu B, Lee SG, Baek J, Kim YJ, Lee JM, Lee Y, Choi SO, Woo DH, Park IH, Chung HM. Establishment of Neurotoxicity Assessment Using Microelectrode Array (MEA) with hiPSC-Derived Neurons and Evaluation of New Psychoactive Substances (NPS). Int J Stem Cells 2022; 15:258-269. [PMID: 35769054 PMCID: PMC9396014 DOI: 10.15283/ijsc21217] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/09/2021] [Revised: 04/01/2022] [Accepted: 04/01/2022] [Indexed: 11/09/2022] Open
Abstract
Background and Objectives Currently, safety pharmacological tests for the central nervous system depend on animal behavioral analysis. However, due to the subjectivity of behavioral analysis and differences between species, there is a limit to appropriate nervous system toxicity assessment, therefore a new neurotoxicity assessment that can simulate the human central nervous system is required. Methods and Results In our study, we developed an in vitro neurotoxicity assessment focusing on neuronal function. To minimize the differences between species and fast screening, hiPSC-derived neurons and a microelectrode array (MEA) that could simultaneously measure the action potentials of the neuronal networks were used. After analyzing the molecular and electrophysiological characters of our neuronal network, we conducted a neurotoxicity assessment on neurotransmitters, neurotoxicants, illicit drugs, and new psychoactive substances (NPS). We found that most substances used in our experiments responded more sensitively to our MEA-based neurotoxicity assessment than to the conventional neurotoxicity assessment. Also, this is the first paper that evaluates various illicit drugs and NPS using MEA-based neurotoxicity assessment using hiPSC-derived neurons. Conclusions Our study expanded the scope of application of neurotoxicity assessment using hiPSC-derived neurons to NPS, and accumulated evaluation data of various toxic substances for hiPSC-derived neurons.
Collapse
Affiliation(s)
- Kyu-Ree Kang
- Department of Stem Cell Biology, School of Medicine, Konkuk University, Seoul, Korea
| | - C-Yoon Kim
- Department of Stem Cell Biology, School of Medicine, Konkuk University, Seoul, Korea.,Department of Physiology, College of Veterinary Medicine, Konkuk University, Seoul, Korea
| | - Jin Kim
- Department of Laboratory Animal Medicine, College of Veterinary Medicine, Seoul National University, Seoul, Korea
| | - Bokyeong Ryu
- Department of Laboratory Animal Medicine, College of Veterinary Medicine, Seoul National University, Seoul, Korea
| | - Seul-Gi Lee
- Department of Stem Cell Biology, School of Medicine, Konkuk University, Seoul, Korea
| | - Jieun Baek
- Department of Stem Cell Biology, School of Medicine, Konkuk University, Seoul, Korea
| | - Ye-Ji Kim
- Drug Abuse Research Group, Research Center of Convergence Toxicology, Korea Institute of Toxicology, Daejeon, Korea
| | - Jin-Moo Lee
- Pharmacological Research Division, Toxicological Evaluation and Research Department, National Institute of Food and Drug Safety Evaluation, Ministry of Food and Drug Safety, Cheongju, Korea
| | - Yootmo Lee
- Pharmacological Research Division, Toxicological Evaluation and Research Department, National Institute of Food and Drug Safety Evaluation, Ministry of Food and Drug Safety, Cheongju, Korea
| | - Sun-Ok Choi
- Pharmacological Research Division, Toxicological Evaluation and Research Department, National Institute of Food and Drug Safety Evaluation, Ministry of Food and Drug Safety, Cheongju, Korea
| | - Dong Ho Woo
- Drug Abuse Research Group, Research Center of Convergence Toxicology, Korea Institute of Toxicology, Daejeon, Korea
| | - Il Hwan Park
- Departments of Thoracis and Cardiovascular Surgery, Yonsei University Wonju College of Medicine, Wonju, Korea
| | - Hyung Min Chung
- Department of Stem Cell Biology, School of Medicine, Konkuk University, Seoul, Korea
| |
Collapse
|
11
|
He K. Pharmacological affinity fingerprints derived from bioactivity data for the identification of designer drugs. J Cheminform 2022; 14:35. [PMID: 35672835 PMCID: PMC9171973 DOI: 10.1186/s13321-022-00607-6] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/02/2022] [Accepted: 05/05/2022] [Indexed: 12/15/2022] Open
Abstract
Facing the continuous emergence of new psychoactive substances (NPS) and their threat to public health, more effective methods for NPS prediction and identification are critical. In this study, the pharmacological affinity fingerprints (Ph-fp) of NPS compounds were predicted by Random Forest classification models using bioactivity data from the ChEMBL database. The binary Ph-fp is the vector consisting of a compound's activity against a list of molecular targets reported to be responsible for the pharmacological effects of NPS. Their performance in similarity searching and unsupervised clustering was assessed and compared to 2D structure fingerprints Morgan and MACCS (1024-bits ECFP4 and 166-bits SMARTS-based MACCS implementation of RDKit). The performance in retrieving compounds according to their pharmacological categorizations is influenced by the predicted active assay counts in Ph-fp and the choice of similarity metric. Overall, the comparative unsupervised clustering analysis suggests the use of a classification model with Morgan fingerprints as input for the construction of Ph-fp. This combination gives satisfactory clustering performance based on external and internal clustering validation indices.
Collapse
Affiliation(s)
- Kedan He
- Physical Sciences, Eastern Connecticut State University, 83 Windham St, Willimantic, CT, 06226, USA.
| |
Collapse
|
12
|
Kolaczynska KE, Ducret P, Trachsel D, Hoener MC, Liechti ME, Luethi D. Pharmacological characterization of 3,4-methylenedioxyamphetamine (MDA) analogs and two amphetamine-based compounds: N,α-DEPEA and DPIA. Eur Neuropsychopharmacol 2022; 59:9-22. [PMID: 35378384 DOI: 10.1016/j.euroneuro.2022.03.006] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/26/2021] [Revised: 02/16/2022] [Accepted: 03/13/2022] [Indexed: 11/04/2022]
Abstract
3,4-methylenedioxyamphetamine (MDA) is a psychoactive compound chemically related to the entactogen MDMA. MDA shares some of the entactogenic effects of MDMA but also exerts stimulant effects and psychedelic properties at higher doses. Here, we examined the pharmacological properties of MDA analogs and related amphetamine-based compounds detected in street drug samples or in sport supplements. We examined the key pharmacological mechanisms including monoamine uptake inhibition and release using human embryonic kidney 293 cells stably transfected with the respective human transporters. Additionally, we assessed monoamine transporter and receptor binding and activation properties. MDA, its fluorinated analogs, as well as the α-ethyl containing BDB and the dimeric amphetamine DPIA inhibited NET with the greatest potency and preferentially inhibited 5-HT vs. dopamine uptake. The β‑methoxy MDA analog 3C-BOH and the amphetamine-based N,α-DEPEA inhibited NET and preferentially inhibited dopamine vs. 5-HT uptake. The test drugs mediated efflux of at least one monoamine with the exception of DPIA. Most compounds bound to 5-HT2A and 5-HT2C receptors (Ki ≤ 10 µM) and several substances activated the 5-HT2A and 5-HT2B receptor as partial or full agonists. Furthermore, several compounds interacted with adrenergic receptors and the trace amine-associated receptor 1 (TAAR1) in the micromolar range. The pharmacological profiles of some fluorinated and nonfluorinated MDA analogs resemble the profile of MDMA. In contrast, 3C-BOH and N,α-DEPEA displayed more pronounced dopaminergic activity similar to amphetamine. Pharmacokinetics and pharmacodynamics studies are necessary to better establish the risks and therapeutic potential of the tested drugs.
Collapse
Affiliation(s)
- Karolina E Kolaczynska
- Division of Clinical Pharmacology and Toxicology, Department of Biomedicine, University Hospital Basel and University of Basel, Basel, Switzerland
| | - Paula Ducret
- Division of Clinical Pharmacology and Toxicology, Department of Biomedicine, University Hospital Basel and University of Basel, Basel, Switzerland
| | | | - Marius C Hoener
- Neuroscience Research, pRED, Roche Innovation Center Basel, F. Hoffmann-La Roche Ltd, Basel, Switzerland
| | - Matthias E Liechti
- Division of Clinical Pharmacology and Toxicology, Department of Biomedicine, University Hospital Basel and University of Basel, Basel, Switzerland.
| | - Dino Luethi
- Division of Clinical Pharmacology and Toxicology, Department of Biomedicine, University Hospital Basel and University of Basel, Basel, Switzerland; Center for Physiology and Pharmacology, Institute of Pharmacology, Medical University of Vienna, Vienna, Austria
| |
Collapse
|
13
|
Motaghinejad M, Motevalian M. Neuroprotective Properties of Minocycline Against Methylphenidate-Induced Neurodegeneration: Possible Role of CREB/BDNF and Akt/GSK3 Signaling Pathways in Rat Hippocampus. Neurotox Res 2022; 40:689-713. [PMID: 35446003 DOI: 10.1007/s12640-021-00454-7] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/24/2021] [Revised: 11/17/2021] [Accepted: 11/29/2021] [Indexed: 12/19/2022]
Abstract
Neurodegeneration is a side effect of methylphenidate (MPH), and minocycline possesses neuroprotective properties. This study aimed to investigate the neuroprotective effects of minocycline against methylphenidate-induced neurodegeneration mediated by signaling pathways of CREB/BDNF and Akt/GSK3. Seven groups of seventy male rats were randomly distributed in seven groups (n = 10). Group 1 received 0.7 ml/rat of normal saline (i.p.), and group 2 was treated with MPH (10 mg/kg, i.p.). Groups 3, 4, 5, and 6 were simultaneously administered MPH (10 mg/kg) and minocycline (10, 20, 30, and 40 mg/kg, i.p.) for 21 days. Minocycline alone (40 mg/kg, i.p.) was administrated to group 7. Open field test (OFT) (on day 22), forced swim test (FST) (on day 24), and elevated plus maze (on day 26) were conducted to analyze the mood-related behaviors; hippocampal oxidative stress, inflammatory, and apoptotic parameters, as well as the levels of protein kinase B (Akt-1), glycogen synthase kinase 3 (GSK3), cAMP response element-binding protein (CREB), and brain-derived neurotrophic factor (BDNF), were also assessed. Furthermore, localization of total CREB, Akt, and GSK3 in the DG and CA1 areas of the hippocampus were measured using immunohistochemistry (IHC). Histological changes in the mentioned areas were also evaluated. Minocycline treatment inhibited MPH-induced mood disorders and decreased lipid peroxidation, oxidized form of glutathione (GSSG), interleukin 1 beta (IL-1β), alpha tumor necrosis factor (TNF-α), Bax, and GSK3 levels. In the contrary, it increased the levels of reduced form of glutathione (GSH), Bcl-2, CREB, BDNF, and Akt-1 and superoxide dismutase (SOD), glutathione peroxidase (GPx), and glutathione reductase (GR) activities in the experimental animals' hippocampus. IHC data showed that minocycline also improved the localization and expression of CREB and Akt positive cells and decreased the GSK3 positive cells in the DG and CA1 regions of the hippocampus of MPH-treated rats. Minocycline also inhibited MPH-induced changes of hippocampal cells' density and shape in both DG and CA1 areas of the hippocampus. According to obtained data, it can be concluded that minocycline probably via activation of the P-CREB/BDNF or Akt/GSK3 signaling pathway can confer its neuroprotective effects against MPH-induced neurodegeneration.
Collapse
Affiliation(s)
- Majid Motaghinejad
- Razi Drug Research Center, Iran University of Medical Sciences, Tehran, Shahid Hemmat High way, Iran Univ. Med. Sci., P.O. Box 14496-14525, Tehran, Iran.
| | - Manijeh Motevalian
- Razi Drug Research Center, Iran University of Medical Sciences, Tehran, Shahid Hemmat High way, Iran Univ. Med. Sci., P.O. Box 14496-14525, Tehran, Iran.
- Department of Pharmacology, School of Medicine, Tehran, Iran University of Medical Sciences, Shahid Hemmat High way, Iran Univ. Med. Sci., P.O. Box 14496-14525, Tehran, Iran.
| |
Collapse
|
14
|
Sahai M, Opacka-Juffry J. Molecular mechanisms of action of stimulant novel psychoactive substances that target the high-affinity transporter for dopamine. Neuronal Signal 2021; 5:NS20210006. [PMID: 34888062 PMCID: PMC8630395 DOI: 10.1042/ns20210006] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/15/2021] [Revised: 10/28/2021] [Accepted: 11/01/2021] [Indexed: 11/17/2022] Open
Abstract
Drug misuse is a significant social and public health problem worldwide. Misused substances exert their neurobehavioural effects through changing neural signalling within the brain, many of them leading to substance dependence and addiction in the longer term. Among drugs with addictive liability, there are illicit classical stimulants such as cocaine and amphetamine, and their more recently available counterparts known as novel psychoactive substances (NPS). Stimulants normally increase dopamine availability in the brain, including the pathway implicated in reward-related behaviour. This pattern is observed in both animal and human brain. The main biological target of stimulants, both classical and NPS, is the dopamine transporter (DAT) implicated in the dopamine-enhancing effects of these drugs. This article aims at reviewing research on the molecular mechanisms underpinning the interactions between stimulant NPS, such as benzofurans, cathinones or piperidine derivatives and DAT, to achieve a greater understanding of the core phenomena that decide about the addictive potential of stimulant NPS. As the methodology is essential in the process of experimental research in this area, we review the applications of in vitro, in vivo and in silico approaches. The latter, including molecular dynamics, attracts the focus of the present review as the method of choice in molecular and atomistic investigations of the mechanisms of addiction of stimulant NPS. Research of this kind is of interest to not only scientists but also health professionals as updated knowledge of NPS, their modes of action and health risks, is needed to tackle the challenges posed by NPS misuse.
Collapse
Affiliation(s)
- Michelle A. Sahai
- School of Life and Health Sciences, University of Roehampton, London SW15 4JD, U.K
| | | |
Collapse
|
15
|
He K. Filter Feature Selection for Unsupervised Clustering of Designer Drugs Using DFT Simulated IR Spectra Data. ACS OMEGA 2021; 6:32151-32165. [PMID: 34870036 PMCID: PMC8638022 DOI: 10.1021/acsomega.1c04945] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 09/07/2021] [Accepted: 11/01/2021] [Indexed: 06/13/2023]
Abstract
The rapid emergence of novel psychoactive substances (NPS) poses new challenges and requirements for forensic testing/analysis techniques. This paper aims to explore the application of unsupervised clustering of NPS compounds' infrared spectra. Two statistical measures, Pearson and Spearman, were used to quantify the spectral similarity and to generate similarity matrices for hierarchical clustering. The correspondence of spectral similarity clustering trees to the commonly used structural/pharmacological categorization was evaluated and compared to the clustering generated using 2D/3D molecular fingerprints. Hybrid model feature selections were applied using different filter-based feature ranking algorithms developed for unsupervised clustering tasks. Since Spearman tends to overestimate the spectral similarity based on the overall pattern of the full spectrum, the clustering result shows the highest degree of improvement from having the nondiscriminative features removed. The loading plots of the first two principal components of the optimal feature subsets confirmed that the most important vibrational bands contributing to the clustering of NPS compounds were selected using non-negative discriminative feature selection (NDFS) algorithms.
Collapse
|
16
|
Haleem DJ, Salman T, Nawaz S, Ikram H. Co-treatment with low doses of buspirone prevents rewarding effects of methylphenidate and upregulates expression of 5-HT1A receptor mRNA in the nucleus accumbens. Behav Brain Res 2021; 418:113660. [PMID: 34752844 DOI: 10.1016/j.bbr.2021.113660] [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: 04/15/2021] [Revised: 10/07/2021] [Accepted: 11/02/2021] [Indexed: 11/18/2022]
Abstract
Accumulating studies consistently show that methylphenidate (MPD), the first line drug for treating Attention-Deficit Hyperactivity Disorder (ADHD), is abused by patients to whom the drug is prescribed. Like other psychostimulants, only low doses of MPD improve cognitive performance while higher doses impair it. Preventing the use of high doses of MPD is important for retaining its therapeutic efficacy. Previously, it has been shown that performance in Morris water maze test is improved in rats treated, orally, with MPD in doses of 2.5 mg/kg; but higher doses (5 mg/kg) impair it. The present study is designed to monitor rewarding effects of 2.5 mg/kg MPD in conditioned place preference (CPP) paradigm and its potential inhibition in buspirone co-treated animals. Our results show that rewarding effects of MPD in CPP paradigm are prevented in rats co-treated with buspirone in doses of 0.1 and 0.3 mg/kg. Animals treated with MPD exhibit a downregulation of 5-HT1A receptor mRNA in the nucleus accumbens which is also prevented in rats co-treated with 0.1 and 0.3 mg/kg but not 1.0 and 2.0 mg/kg buspirone. Administration of buspirone in these doses is not rewarding in CPP test and upregulates 5-HT1A receptor mRNA in the nucleus accumbens. The findings suggest that co-use of low doses of buspirone can prevent rewarding effects of MPD to help retain its therapeutic efficacy.
Collapse
Affiliation(s)
- Darakhshan Jabeen Haleem
- Neuroscience Research Laboratory, Dr Panjwani Center for Molecular Medicine & Drug Research (PCMD), International Center for Chemical and Biological Science (ICCBS), University of Karachi, Karachi 75270, Pakistan; Department of Biochemistry, University of Karachi, Pakistan.
| | - Tabinda Salman
- National Center for Proteomics, University of Karachi, Karachi, Pakistan
| | - Shazia Nawaz
- Neuroscience Research Laboratory, Dr Panjwani Center for Molecular Medicine & Drug Research (PCMD), International Center for Chemical and Biological Science (ICCBS), University of Karachi, Karachi 75270, Pakistan
| | - Huma Ikram
- Department of Biochemistry, University of Karachi, Pakistan
| |
Collapse
|
17
|
Docherty JR, Alsufyani HA. Pharmacology of Drugs Used as Stimulants. J Clin Pharmacol 2021; 61 Suppl 2:S53-S69. [PMID: 34396557 DOI: 10.1002/jcph.1918] [Citation(s) in RCA: 31] [Impact Index Per Article: 7.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/24/2021] [Accepted: 05/22/2021] [Indexed: 12/21/2022]
Abstract
Psychostimulant, cardiovascular, and temperature actions of stimulants involve adrenergic (norepinephrine), dopaminergic (dopamine), and serotonergic (serotonin) pathways. Stimulants such as amphetamine, 3,4-methylenedioxymethamphetamine (MDMA), or mephedrone can act on the neuronal membrane monoamine transporters NET, DAT, and SERT and/or the vesicular monoamine transporter 2 to inhibit reuptake of neurotransmitter or cause release by reverse transport. Stimulants may have additional effects involving pre- and postsynaptic/junctional receptors for norepinephrine, dopamine, and serotonin and other receptors. As a result, stimulants may have a wide range of possible actions. Agents with cocaine or MDMA-like actions can induce serious and potentially fatal adverse events via thermodysregulatory, cardiovascular, or other mechanisms. MDMA-like stimulants may cause hyperthermia that can be life threathening. Recreational users of stimulants should be aware of the dangers of hyperthermia in a rave/club environment.
Collapse
Affiliation(s)
| | - Hadeel A Alsufyani
- Department of Physiology, King Abdulaziz University, Jeddah, Saudi Arabia
| |
Collapse
|
18
|
Caffino L, Mottarlini F, Bilel S, Targa G, Tirri M, Maggi C, Marti M, Fumagalli F. Single Exposure to the Cathinones MDPV and α-PVP Alters Molecular Markers of Neuroplasticity in the Adult Mouse Brain. Int J Mol Sci 2021; 22:7397. [PMID: 34299015 PMCID: PMC8307734 DOI: 10.3390/ijms22147397] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/25/2021] [Revised: 07/06/2021] [Accepted: 07/07/2021] [Indexed: 12/23/2022] Open
Abstract
Synthetic cathinones have gained popularity among young drug users and are widely used in the clandestine market. While the cathinone-induced behavioral profile has been extensively investigated, information on their neuroplastic effects is still rather fragmentary. Accordingly, we have exposed male mice to a single injection of MDPV and α-PVP and sacrificed the animals at different time points (i.e., 30 min, 2 h, and 24 h) to have a rapid readout of the effect of these psychostimulants on neuroplasticity in the frontal lobe and hippocampus, two reward-related brain regions. We found that a single, low dose of MDPV or α-PVP is sufficient to alter the expression of neuroplastic markers in the adult mouse brain. In particular, we found increased expression of the transcription factor Npas4, increased ratio between the vesicular GABA transporter and the vesicular glutamate transporter together with changes in the expression of the neurotrophin Bdnf, confirming the widespread impact of these cathinones on brain plasticity. To sum up, exposure to low dose of cathinones can impair cortical and hippocampal homeostasis, suggesting that abuse of these cathinones at much higher doses, as it occurs in humans, could have an even more profound impact on neuroplasticity.
Collapse
Affiliation(s)
- Lucia Caffino
- Department of Pharmacological and Biomolecular Sciences, Università degli Studi di Milano, Via Balzaretti 9, 20133 Milano, Italy; (L.C.); (F.M.); (G.T.); (C.M.)
| | - Francesca Mottarlini
- Department of Pharmacological and Biomolecular Sciences, Università degli Studi di Milano, Via Balzaretti 9, 20133 Milano, Italy; (L.C.); (F.M.); (G.T.); (C.M.)
| | - Sabrine Bilel
- Section of Legal Medicine and LTTA Center, Department of Translational Medicine, University of Ferrara, 44121 Ferrara, Italy; (S.B.); (M.T.); (M.M.)
| | - Giorgia Targa
- Department of Pharmacological and Biomolecular Sciences, Università degli Studi di Milano, Via Balzaretti 9, 20133 Milano, Italy; (L.C.); (F.M.); (G.T.); (C.M.)
| | - Micaela Tirri
- Section of Legal Medicine and LTTA Center, Department of Translational Medicine, University of Ferrara, 44121 Ferrara, Italy; (S.B.); (M.T.); (M.M.)
| | - Coralie Maggi
- Department of Pharmacological and Biomolecular Sciences, Università degli Studi di Milano, Via Balzaretti 9, 20133 Milano, Italy; (L.C.); (F.M.); (G.T.); (C.M.)
| | - Matteo Marti
- Section of Legal Medicine and LTTA Center, Department of Translational Medicine, University of Ferrara, 44121 Ferrara, Italy; (S.B.); (M.T.); (M.M.)
- Collaborative Center for the Italian National Early Warning System, Department of Anti-Drug Policies, Presidency of the Council of Ministers, 44121 Ferrara, Italy
| | - Fabio Fumagalli
- Department of Pharmacological and Biomolecular Sciences, Università degli Studi di Milano, Via Balzaretti 9, 20133 Milano, Italy; (L.C.); (F.M.); (G.T.); (C.M.)
| |
Collapse
|
19
|
Rudin D, Liechti ME, Luethi D. Molecular and clinical aspects of potential neurotoxicity induced by new psychoactive stimulants and psychedelics. Exp Neurol 2021; 343:113778. [PMID: 34090893 DOI: 10.1016/j.expneurol.2021.113778] [Citation(s) in RCA: 26] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/11/2021] [Revised: 05/25/2021] [Accepted: 05/29/2021] [Indexed: 12/20/2022]
Abstract
New psychoactive stimulants and psychedelics continue to play an important role on the illicit new psychoactive substance (NPS) market. Designer stimulants and psychedelics both affect monoaminergic systems, although by different mechanisms. Stimulant NPS primarily interact with monoamine transporters, either as inhibitors or as substrates. Psychedelic NPS most potently interact with serotonergic receptors and mediate their mind-altering effects mainly through agonism at serotonin 5-hydroxytryptamine-2A (5-HT2A) receptors. Rarely, designer stimulants and psychedelics are associated with potentially severe adverse effects. However, due to the high number of emerging NPS, it is not possible to investigate the toxicity of each individual substance in detail. The brain is an organ particularly sensitive to substance-induced toxicity due to its high metabolic activity. In fact, stimulant and psychedelic NPS have been linked to neurological and cognitive impairments. Furthermore, studies using in vitro cell models or rodents indicate a variety of mechanisms that could potentially lead to neurotoxic damage in NPS users. Cytotoxicity, mitochondrial dysfunction, and oxidative stress may potentially contribute to neurotoxicity of stimulant NPS in addition to altered neurochemistry. Serotonin 5-HT2A receptor-mediated toxicity, oxidative stress, and activation of mitochondrial apoptosis pathways could contribute to neurotoxicity of some psychedelic NPS. However, it remains unclear how well the current preclinical data of NPS-induced neurotoxicity translate to humans.
Collapse
Affiliation(s)
- Deborah Rudin
- Division of Clinical Pharmacology and Toxicology, University Hospital Basel and University of Basel, Basel, Switzerland; Institute of Pharmacology, Medical University of Vienna, Vienna, Austria
| | - Matthias E Liechti
- Division of Clinical Pharmacology and Toxicology, University Hospital Basel and University of Basel, Basel, Switzerland
| | - Dino Luethi
- Division of Clinical Pharmacology and Toxicology, University Hospital Basel and University of Basel, Basel, Switzerland; Institute of Pharmacology, Medical University of Vienna, Vienna, Austria; Institute of Applied Physics, TU Wien, Vienna, Austria.
| |
Collapse
|
20
|
Novel Phenethylamines and Their Potential Interactions With Prescription Drugs: A Systematic Critical Review. Ther Drug Monit 2021; 42:271-281. [PMID: 32022784 DOI: 10.1097/ftd.0000000000000725] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/13/2023]
Abstract
BACKGROUND The novel phenethylamines 4-fluoroamphetamine (4-FA) and 2,5-dimethoxy-4-bromophenethylamine (2C-B) fall in the top 10 most used new psychoactive substances (NPSs) among high-risk substance users. Various phenethylamines and NPS are also highly used in populations with mental disorders, depression, or attention deficit hyperactivity disorder (ADHD). Moreover, NPS use is highly prevalent among men and women with risky sexual behavior. Considering these specific populations and their frequent concurrent use of drugs, such as antidepressants, ADHD medication, and antiretrovirals, reports on potential interactions between these drugs, and phenethylamines 4-FA and 2C-B, were reviewed. METHODS The authors performed a systematic literature review on 4-FA and 2C-B interactions with antidepressants (citalopram, fluoxetine, fluvoxamine, paroxetine, sertraline, duloxetine, bupropion, venlafaxine, phenelzine, moclobemide, and tranylcypromine), ADHD medications (atomoxetine, dexamphetamine, methylphenidate, and modafinil), and antiretrovirals. RESULTS Limited literature exists on the pharmacokinetics and drug-drug interactions of 2C-B and 4-FA. Only one case report indicated a possible interaction between 4-FA and ADHD medication. Although pharmacokinetic interactions between 4-FA and prescription drugs remain speculative, their pharmacodynamic points toward interactions between 4-FA and ADHD medication and antidepressants. The pharmacokinetic and pharmacodynamic profile of 2C-B also points toward such interactions, between 2C-B and prescription drugs such as antidepressants and ADHD medication. CONCLUSIONS A drug-drug (phenethylamine-prescription drug) interaction potential is anticipated, mainly involving monoamine oxidases for 2C-B and 4-FA, with monoamine transporters being more specific to 4-FA.
Collapse
|
21
|
Repantis D, Bovy L, Ohla K, Kühn S, Dresler M. Cognitive enhancement effects of stimulants: a randomized controlled trial testing methylphenidate, modafinil, and caffeine. Psychopharmacology (Berl) 2021; 238:441-451. [PMID: 33201262 PMCID: PMC7826302 DOI: 10.1007/s00213-020-05691-w] [Citation(s) in RCA: 17] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/20/2020] [Accepted: 10/23/2020] [Indexed: 12/14/2022]
Abstract
RATIONAL At all times humans have made attempts to improve their cognitive abilities by different means, among others, with the use of stimulants. Widely available stimulants such as caffeine, but also prescription substances such as methylphenidate and modafinil, are being used by healthy individuals to enhance cognitive performance. OBJECTIVES There is a lack of knowledge on the effects of prescription stimulants when taken by healthy individuals (as compared with patients) and especially on the effects of different substances across different cognitive domains. METHODS We conducted a pilot study with three arms in which male participants received placebo and one of three stimulants (caffeine, methylphenidate, modafinil) and assessed cognitive performance with a test battery that captures various cognitive domains. RESULTS Our study showed some moderate effects of the three stimulants tested. Methylphenidate had positive effects on self-reported fatigue as well as on declarative memory 24 hours after learning; caffeine had a positive effect on sustained attention; there was no significant effect of modafinil in any of the instruments of our test battery. All stimulants were well tolerated, and no trade-off negative effects on other cognitive domains were found. CONCLUSIONS The few observed significant positive effects of the tested stimulants were domain-specific and of rather low magnitude. The results can inform the use of stimulants for cognitive enhancement purposes as well as direct further research to investigate the effects of stimulants on specific cognitive domains that seem most promising, possibly by using tasks that are more demanding.
Collapse
Affiliation(s)
- Dimitris Repantis
- Department of Psychiatry and Psychotherapy, Campus Benjamin Franklin, Charité - Universitätsmedizin Berlin, corporate member of Freie Universität Berlin, Humboldt-Universität zu Berlin, and Berlin Institute of Health, Hindenburgdamm 30, 12203, Berlin, Germany.
- Lise Meitner Group for Environmental Neuroscience, Max Planck Institute for Human Development, Berlin, Germany.
| | - Leonore Bovy
- Donders Institute for Brain, Cognition and Behaviour, Radboud University Medical Center, Nijmegen, The Netherlands
| | - Kathrin Ohla
- Institute of Neuroscience and Medicine (INM-3), Jülich Research Centre, Jülich, Germany
| | - Simone Kühn
- Lise Meitner Group for Environmental Neuroscience, Max Planck Institute for Human Development, Berlin, Germany
- Department of Psychiatry and Psychotherapy, University Medical Center Hamburg-Eppendorf (UKE), Hamburg, Germany
| | - Martin Dresler
- Donders Institute for Brain, Cognition and Behaviour, Radboud University Medical Center, Nijmegen, The Netherlands
| |
Collapse
|
22
|
Microwave-Assisted Synthesis of (Piperidin-1-yl)quinolin-3-yl)methylene)hydrazinecarbothioamides as Potent Inhibitors of Cholinesterases: A Biochemical and In Silico Approach. Molecules 2021; 26:molecules26030656. [PMID: 33513837 PMCID: PMC7866225 DOI: 10.3390/molecules26030656] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/07/2021] [Revised: 01/22/2021] [Accepted: 01/24/2021] [Indexed: 12/17/2022] Open
Abstract
Alzheimer’s disease (AD), a progressive neurodegenerative disorder, characterized by central cognitive dysfunction, memory loss, and intellectual decline poses a major public health problem affecting millions of people around the globe. Despite several clinically approved drugs and development of anti-Alzheimer’s heterocyclic structural leads, the treatment of AD requires safer hybrid therapeutics with characteristic structural and biochemical properties. In this endeavor, we herein report a microwave-assisted synthesis of a library of quinoline thiosemicarbazones endowed with a piperidine moiety, achieved via the condensation of 6/8-methyl-2-(piperidin-1-yl)quinoline-3-carbaldehydes and (un)substituted thiosemicarbazides. The target N-heterocyclic products were isolated in excellent yields. The structures of all the synthesized compounds were fully established using readily available spectroscopic techniques (FTIR, 1H- and 13C-NMR). Anti-Alzheimer potential of the synthesized heterocyclic compounds was evaluated using acetylcholinesterase (AChE) and butyrylcholinesterase (BChE) enzymes. The in vitro biochemical assay results revealed several compounds as potent inhibitors of both enzymes. Among them, five compounds exhibited IC50 values less than 20 μM. N-(3-chlorophenyl)-2-((8-methyl-2-(piperidin-1-yl)quinolin-3-yl)methylene)hydrazine carbothioamide emerged as the most potent dual inhibitor of AChE and BChE with IC50 values of 9.68 and 11.59 μM, respectively. Various informative structure–activity relationship (SAR) analyses were also concluded indicating the critical role of substitution pattern on the inhibitory efficacy of the tested derivatives. In vitro results were further validated through molecular docking analysis where interactive behavior of the potent inhibitors within the active pocket of enzymes was established. Quinoline thiosemicarbazones were also tested for their cytotoxicity using MTT assay against HepG2 cells. Among the 26 novel compounds, there were five cytotoxical and 18 showed proliferative properties.
Collapse
|
23
|
Seeley SL, D'Souza MS, Stoops TS, Rorabaugh BR. Short term methylphenidate treatment does not increase myocardial injury in the ischemic rat heart. Physiol Res 2020; 69:803-812. [PMID: 32469230 DOI: 10.33549/physiolres.934368] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/31/2022] Open
Abstract
Methylphenidate is commonly used for the treatment of attention deficit hyperactivity disorder. The cardiovascular safety of methylphenidate has been a subject of debate with some studies indicating that methylphenidate increases the likelihood of experiencing a myocardial infarction. However, it is unknown whether methylphenidate worsens the extent of injury during an ischemic insult. The purpose of this study was to determine whether short term exposure to methylphenidate increases the extent of myocardial injury during an ischemic insult. Male and female rats received methylphenidate (5 mg/kg/day) or saline for 10 days by oral gavage. Hearts were subjected to 20 min of ischemia and 2 h of reperfusion on a Langendorff isolated heart apparatus on day 11. Cardiac contractile function was monitored via an intraventricular balloon and myocardial injury was assessed by triphenyltetrazolium chloride staining. Methylphenidate significantly increased locomotor activity in male and female rats, confirming absorption of this psychostimulant into the central nervous system. Male hearts had significantly larger infarcts than female hearts, but methylphenidate had no impact on infarct size or postischemic recovery of contractile function in hearts of either sex. These data indicate that methylphenidate does not increase the extent of injury induced by an ischemic insult.
Collapse
Affiliation(s)
- S L Seeley
- Marshall University School of Pharmacy, Huntington, WV, USA.
| | | | | | | |
Collapse
|
24
|
|
25
|
Brenner RG, Oliveri AN, Sinnott-Armstrong W, Levin ED. Effects of sub-chronic methylphenidate on risk-taking and sociability in zebrafish (Danio rerio). NAUNYN-SCHMIEDEBERG'S ARCHIVES OF PHARMACOLOGY 2020; 393:1373-1381. [PMID: 32025747 PMCID: PMC7716188 DOI: 10.1007/s00210-020-01835-z] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/23/2019] [Accepted: 01/30/2020] [Indexed: 12/21/2022]
Abstract
Attention deficit hyperactive disorder (ADHD) is the most common psychiatric disorder in children affecting around 11% of children 4-17 years of age (CDC 2019). Children with ADHD are widely treated with stimulant medications such as methylphenidate (Ritalin®). However, there has been little research on the developmental effects of methylphenidate on risk-taking and sociability. We investigated in zebrafish the potential developmental neurobehavioral toxicity of methylphenidate on these behavioral functions. We chose zebrafish because they provide a model with extensive genetic tools for future mechanistic studies. We studied whether sub-chronic methylphenidate exposure during juvenile development causes neurobehavioral impairments in zebrafish. Methylphenidate diminished responses to environmental stimuli after both acute and sub-chronic dosing. In adult zebrafish, acute methylphenidate impaired avoidance of an approaching visual stimulus modeling a predator and decreased locomotor response to the social visual stimulus of conspecifics. Adult zebrafish dosed acutely with methylphenidate demonstrated behaviors of less retreat from threatening visual stimuli and less approach to conspecifics compared with controls. In a sub-chronic dosing paradigm during development, methylphenidate caused less robust exploration of a novel tank. In the predator avoidance paradigm, sub-chronic dosing that began at an older age (28 dpf) decreased activity levels more than sub-chronic dosing that began at earlier ages (14 dpf and 21 dpf). In the social shoaling task, sub-chronic methylphenidate attenuated reaction to the social stimulus. Acute and developmental methylphenidate exposure decreased response to environmental cues. Additional research is needed to determine critical mechanisms for these effects and to see how these results may be translatable to neurobehavioral toxicity of prescribing Ritalin® to children and adolescents.
Collapse
Affiliation(s)
- Rebecca G Brenner
- Duke Institute for Brain Sciences, Duke University, Durham, NC, USA
- Department of Philosophy, Duke University, Durham, NC, USA
| | - Anthony N Oliveri
- Department of Pharmacology and Cancer Biology, Duke University, Durham, NC, USA
| | - Walter Sinnott-Armstrong
- Duke Institute for Brain Sciences, Duke University, Durham, NC, USA
- Department of Philosophy, Duke University, Durham, NC, USA
| | - Edward D Levin
- Duke Institute for Brain Sciences, Duke University, Durham, NC, USA.
- Department of Pharmacology and Cancer Biology, Duke University, Durham, NC, USA.
- Department of Psychiatry and Behavioral Sciences, Duke University Medical Center, Box 104790, Durham, NC, 27710, USA.
| |
Collapse
|
26
|
Duart-Castells L, Blanco-Gandía MC, Ferrer-Pérez C, Puster B, Pubill D, Miñarro J, Escubedo E, Rodríguez-Arias M. Cross-reinstatement between 3,4-methylenedioxypyrovalerone (MDPV) and cocaine using conditioned place preference. Prog Neuropsychopharmacol Biol Psychiatry 2020; 100:109876. [PMID: 31991149 DOI: 10.1016/j.pnpbp.2020.109876] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/16/2019] [Revised: 01/16/2020] [Accepted: 01/21/2020] [Indexed: 12/12/2022]
Abstract
3,4-Methylenedioxypyrovalerone (MDPV) is a new psychoactive substance (NPS) considered to be a cocaine-like psychostimulant. The substitution of an established illicit drug as cocaine with an NPS is a pattern of use reported among drug users. The aim of this study was to investigate the relationship between cocaine and MDPV in the reinstatement of the conditioned place preference (CPP) paradigm, in order to establish whether there is cross-reinstatement between the two psychostimulants. Four experimental groups of male OF1 mice were subjected to the CPP paradigm: MDPV-MDPV, Cocaine-Cocaine, Cocaine-MDPV, and MDPV-Cocaine. The first drug refers to the substance with which the animals were conditioned (cocaine 10 mg/kg or MDPV 2 mg/kg) and the s to the substance with which preference was reinstated. In parallel, G9a, ΔFosB, CB1 receptor, CDK5, Arc and c-Fos were determined in ventral striatum. MDPV induced CPP at doses from 1 to 4 mg/kg. Although 2 mg/kg MDPV induced a stronger psychostimulant effect than 10 mg/kg cocaine, both doses seemed to be equivalent in their rewarding properties. However, memories associated with MDPV required more time to be extinguished. MDPV and cocaine restore drug-seeking behavior with respect to each other, although relapse into drug-taking is always more pronounced with the conditioning drug. The fact that MDPV-treated mice show increased ΔFosB protein levels correlates with its longer extinction time and points to the activation of neuroplasticity mechanisms that persist for at least 12 days. Moreover, in these animals, a priming-dose of cocaine can trigger significant neuroplasticity, implying a high vulnerability to cocaine abuse.
Collapse
Affiliation(s)
- Leticia Duart-Castells
- Department of Pharmacology, Toxicology and Therapeutic Chemistry, Pharmacology Section and Institute of Biomedicine (IBUB), Faculty of Pharmacy, University of Barcelona, Av. Joan XXIII, 27-31, 08028 Barcelona, Spain
| | - M Carmen Blanco-Gandía
- Department of Psychology and Sociology, University of Zaragoza, C/ Ciudad Escolar s/n, 44003 Teruel, Spain
| | - Carmen Ferrer-Pérez
- Unit of Research Psychobiology of Drug Dependence, Department of Psychobiology, Facultad de Psicología, Universitat de Valencia, Avda. Blasco Ibáñez, 21, 46010 Valencia, Spain
| | - Brigitte Puster
- Department of Pharmacology, Toxicology and Therapeutic Chemistry, Pharmacology Section and Institute of Biomedicine (IBUB), Faculty of Pharmacy, University of Barcelona, Av. Joan XXIII, 27-31, 08028 Barcelona, Spain
| | - David Pubill
- Department of Pharmacology, Toxicology and Therapeutic Chemistry, Pharmacology Section and Institute of Biomedicine (IBUB), Faculty of Pharmacy, University of Barcelona, Av. Joan XXIII, 27-31, 08028 Barcelona, Spain
| | - José Miñarro
- Unit of Research Psychobiology of Drug Dependence, Department of Psychobiology, Facultad de Psicología, Universitat de Valencia, Avda. Blasco Ibáñez, 21, 46010 Valencia, Spain
| | - Elena Escubedo
- Department of Pharmacology, Toxicology and Therapeutic Chemistry, Pharmacology Section and Institute of Biomedicine (IBUB), Faculty of Pharmacy, University of Barcelona, Av. Joan XXIII, 27-31, 08028 Barcelona, Spain.
| | - Marta Rodríguez-Arias
- Unit of Research Psychobiology of Drug Dependence, Department of Psychobiology, Facultad de Psicología, Universitat de Valencia, Avda. Blasco Ibáñez, 21, 46010 Valencia, Spain
| |
Collapse
|
27
|
Ilic M, Holy M, Jaentsch K, Liechti ME, Lubec G, Baumann MH, Sitte HH, Luethi D. Cell-Based Radiotracer Binding and Uptake Inhibition Assays: A Comparison of In Vitro Methods to Assess the Potency of Drugs That Target Monoamine Transporters. Front Pharmacol 2020; 11:673. [PMID: 32508638 PMCID: PMC7248194 DOI: 10.3389/fphar.2020.00673] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/18/2020] [Accepted: 04/24/2020] [Indexed: 01/10/2023] Open
Abstract
High-affinity monoamine transporters are targets for prescribed medications and stimulant drugs of abuse. Therefore, assessing monoamine transporter activity for candidate medications and newly-emerging drugs of abuse provides essential information for industry, academia, and public health. Radiotracer binding and uptake inhibition are the gold standard assays for determining drug–transporter interaction profiles. The combined results from such assays yield a unique biochemical fingerprint for each compound. Over time, different assay methods have been developed to assess transporter activity, and the comparability of data across various assay platforms remains largely unclear. Here, we compare the effects of six well-established stimulants in two different cell-based uptake inhibition assays, one method using adherent cells and the other using suspended cells. Furthermore, we compare the data from transfected cell lines derived from different laboratories and data reported from rat synaptosomes. For transporter inhibitors, IC50 values obtained by the two experimental methods were comparable, but using different transfected cell lines yielded disparate results. For transporter substrates, differences between the two cell lines were less pronounced but the drugs displayed different inhibition potencies when evaluated by the two methods. Our study illustrates the inherent limitations when comparing transporter inhibition data from different laboratories and stresses the importance of including appropriate control experiments with reference compounds when investigating new drugs of interest.
Collapse
Affiliation(s)
- Marija Ilic
- Institute of Pharmacology, Center for Physiology and Pharmacology, Medical University of Vienna, Vienna, Austria.,Department of Pharmaceutical Chemistry, Faculty of Life Sciences, University of Vienna, Vienna, Austria.,Neuroproteomics, Paracelsus Private Medical University, Salzburg, Austria
| | - Marion Holy
- Institute of Pharmacology, Center for Physiology and Pharmacology, Medical University of Vienna, Vienna, Austria
| | - Kathrin Jaentsch
- Institute of Pharmacology, Center for Physiology and Pharmacology, Medical University of Vienna, Vienna, Austria
| | - Matthias E Liechti
- Division of Clinical Pharmacology and Toxicology, Department of Biomedicine, University Hospital Basel and University Basel, Basel, Switzerland
| | - Gert Lubec
- Neuroproteomics, Paracelsus Private Medical University, Salzburg, Austria
| | - Michael H Baumann
- Designer Drug Research Unit, Intramural Research Program, National Institute on Drug Abuse, National Institutes of Health, Baltimore, MD, United States
| | - Harald H Sitte
- Institute of Pharmacology, Center for Physiology and Pharmacology, Medical University of Vienna, Vienna, Austria
| | - Dino Luethi
- Institute of Pharmacology, Center for Physiology and Pharmacology, Medical University of Vienna, Vienna, Austria.,Division of Clinical Pharmacology and Toxicology, Department of Biomedicine, University Hospital Basel and University Basel, Basel, Switzerland
| |
Collapse
|
28
|
Luethi D, Liechti ME. Designer drugs: mechanism of action and adverse effects. Arch Toxicol 2020; 94:1085-1133. [PMID: 32249347 PMCID: PMC7225206 DOI: 10.1007/s00204-020-02693-7] [Citation(s) in RCA: 140] [Impact Index Per Article: 28.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/14/2020] [Accepted: 02/25/2020] [Indexed: 12/18/2022]
Abstract
Psychoactive substances with chemical structures or pharmacological profiles that are similar to traditional drugs of abuse continue to emerge on the recreational drug market. Internet vendors may at least temporarily sell these so-called designer drugs without adhering to legal statutes or facing legal consequences. Overall, the mechanism of action and adverse effects of designer drugs are similar to traditional drugs of abuse. Stimulants, such as amphetamines and cathinones, primarily interact with monoamine transporters and mostly induce sympathomimetic adverse effects. Agonism at μ-opioid receptors and γ-aminobutyric acid-A (GABAA) or GABAB receptors mediates the pharmacological effects of sedatives, which may induce cardiorespiratory depression. Dissociative designer drugs primarily act as N-methyl-D-aspartate receptor antagonists and pose similar health risks as the medically approved dissociative anesthetic ketamine. The cannabinoid type 1 (CB1) receptor is thought to drive the psychoactive effects of synthetic cannabinoids, which are associated with a less desirable effect profile and more severe adverse effects compared with cannabis. Serotonergic 5-hydroxytryptamine-2A (5-HT2A) receptors mediate alterations of perception and cognition that are induced by serotonergic psychedelics. Because of their novelty, designer drugs may remain undetected by routine drug screening, thus hampering evaluations of adverse effects. Intoxication reports suggest that several designer drugs are used concurrently, posing a high risk for severe adverse effects and even death.
Collapse
Affiliation(s)
- Dino Luethi
- Center for Physiology and Pharmacology, Institute of Pharmacology, Medical University of Vienna, Währinger Strasse 13a, 1090, Vienna, Austria.
- Institute of Applied Physics, Vienna University of Technology, Getreidemarkt 9, 1060, Vienna, Austria.
- Division of Clinical Pharmacology and Toxicology, University Hospital Basel and University of Basel, Schanzenstrasse 55, 4056, Basel, Switzerland.
| | - Matthias E Liechti
- Division of Clinical Pharmacology and Toxicology, University Hospital Basel and University of Basel, Schanzenstrasse 55, 4056, Basel, Switzerland.
| |
Collapse
|
29
|
Manier SK, Niedermeier S, Schäper J, Meyer MR. Use of UPLC-HRMS/MS for In Vitro and In Vivo Metabolite Identification of Three Methylphenidate-derived New Psychoactive Substances. J Anal Toxicol 2020; 44:156-162. [PMID: 31355413 DOI: 10.1093/jat/bkz052] [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: 02/14/2019] [Revised: 04/15/2019] [Accepted: 06/09/2019] [Indexed: 11/14/2022] Open
Abstract
The distribution of so-called new psychoactive substances (NPS) as substitute for common drug of abuse was steadily increasing in the last years, but knowledge about their toxicodynamic and toxicokinetic properties is lacking. However, a comprehensive knowledge of their toxicokinetics, particularly their metabolism, is crucial for developing reliable screening procedures and to verify their intake, e.g., in case of intoxications. The aim of this study was therefore to tentatively identify the metabolites of the methylphenidate-derived NPS isopropylphenidate (isopropyl 2-phenyl-2-(2-piperidyl) acetate, IPH), 4-fluoromethylphenidate (methyl 2-(4-fluorophenyl)-2-(piperidin-2-yl) acetate, 4-FMPH) and 3,4-dichloromethylphenidate (methyl 2-(3,4-dichlorophenyl)-2-(piperidin-2-yl) acetate, 3,4-CTMP) using different in vivo and in vitro techniques and ultra-high performance liquid chromatography-high-resolution mass spectrometry (UHPLC-HRMS/MS). Urine samples of male rats were analyzed, and the transfer to human metabolism was done by using pooled human S9 fraction (pS9), which contains the microsomal fraction of liver homogenisate as well as its cytosol. UHPLC-HRMS/MS analysis of rat urine revealed 17 metabolites for IPH (14 phase I and 3 phase II metabolites), 13 metabolites were found for 4-FMPH (12 phase I metabolites and 1 phase II metabolite) and 7 phase I metabolites and no phase II metabolites were found for 3,4-CTMP. pS9 incubations additionally indicated that all investigated substances were primarily hydrolyzed, resulting in the corresponding carboxy metabolites. Finally, these carboxy metabolites should be used as additional analytical targets besides the parent compounds for comprehensive mass spectrometry-based screening procedures.
Collapse
Affiliation(s)
- Sascha K Manier
- Department of Experimental and Clinical Toxicology, Institute of Experimental and Clinical Pharmacology and Toxicology, Center for Molecular Signaling (PZMS), Saarland University, Homburg, Germany
| | - Sophia Niedermeier
- Department of Experimental and Clinical Toxicology, Institute of Experimental and Clinical Pharmacology and Toxicology, Center for Molecular Signaling (PZMS), Saarland University, Homburg, Germany
| | - Jan Schäper
- State Bureau of Criminal Investigation Bavaria, 80636 München, Germany
| | - Markus R Meyer
- Department of Experimental and Clinical Toxicology, Institute of Experimental and Clinical Pharmacology and Toxicology, Center for Molecular Signaling (PZMS), Saarland University, Homburg, Germany
| |
Collapse
|
30
|
Santos‐Toscano R, Guirguis A, Davidson C. How preclinical studies have influenced novel psychoactive substance legislation in the UK and Europe. Br J Clin Pharmacol 2020; 86:452-481. [DOI: 10.1111/bcp.14224] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/20/2019] [Revised: 01/13/2020] [Accepted: 01/14/2020] [Indexed: 12/30/2022] Open
Affiliation(s)
- Raquel Santos‐Toscano
- School of Pharmacy & Biomedical Sciences, Faculty of Clinical & Biomedical Sciences University of Central Lancashire UK
| | - Amira Guirguis
- Swansea University Medical School, Institute of Life Sciences 2, Swansea University Swansea UK
| | - Colin Davidson
- School of Pharmacy & Biomedical Sciences, Faculty of Clinical & Biomedical Sciences University of Central Lancashire UK
| |
Collapse
|
31
|
Luethi D, Kolaczynska KE, Walter M, Suzuki M, Rice KC, Blough BE, Hoener MC, Baumann MH, Liechti ME. Metabolites of the ring-substituted stimulants MDMA, methylone and MDPV differentially affect human monoaminergic systems. J Psychopharmacol 2019; 33:831-841. [PMID: 31038382 PMCID: PMC8269116 DOI: 10.1177/0269881119844185] [Citation(s) in RCA: 20] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
BACKGROUND Amphetamine analogs with a 3,4-methylenedioxy ring-substitution are among the most popular illicit drugs of abuse, exerting stimulant and entactogenic effects. Enzymatic N-demethylation or opening of the 3,4-methylenedioxy ring via O-demethylenation gives rise to metabolites that may be pharmacologically active. Indeed, previous studies in rats show that specific metabolites of 3,4-methylenedioxymethamphetamine (MDMA), 3,4-methylenedioxymethcathinone (methylone) and 3,4-methylenedioxypyrovalerone (MDPV) can interact with monoaminergic systems. AIM Interactions of metabolites of MDMA, methylone and MDPV with human monoaminergic systems were assessed. METHODS The ability of parent drugs and their metabolites to inhibit uptake of tritiated norepinephrine, dopamine and serotonin (5-HT) was assessed in human embryonic kidney 293 cells transfected with human monoamine transporters. Binding affinities and functional activity at monoamine transporters and various receptor subtypes were also determined. RESULTS MDMA and methylone displayed greater potency to inhibit norepinephrine uptake as compared to their effects on dopamine and 5-HT uptake. N-demethylation of MDMA failed to alter uptake inhibition profiles, whereas N-demethylation of methylone decreased overall transporter inhibition potencies. O-demethylenation of MDMA, methylone and MDPV resulted in catechol metabolites that maintained norepinephrine and dopamine uptake inhibition potencies, but markedly reduced activity at 5-HT uptake. O-methylation of the catechol metabolites significantly decreased norepinephrine uptake inhibition, resulting in metabolites lacking significant stimulant properties. CONCLUSIONS Several metabolites of MDMA, methylone and MDPV interact with human transporters and receptors at pharmacologically relevant concentrations. In particular, N-demethylated metabolites of MDMA and methylone circulate in unconjugated form and could contribute to the in vivo activity of the parent compounds in human users.
Collapse
Affiliation(s)
- Dino Luethi
- Division of Clinical Pharmacology and Toxicology, Department of Biomedicine, University Hospital Basel and University of Basel, Basel, Switzerland
| | - Karolina E. Kolaczynska
- Division of Clinical Pharmacology and Toxicology, Department of Biomedicine, University Hospital Basel and University of Basel, Basel, Switzerland
| | - Melanie Walter
- Division of Clinical Pharmacology and Toxicology, Department of Biomedicine, University Hospital Basel and University of Basel, Basel, Switzerland
| | - Masaki Suzuki
- Drug Design and Synthesis Section, Intramural Research Program, National Institute on Drug Abuse, National Institutes of Health, Bethesda, MD, 20892, USA,On leave from the Medicinal Chemistry Research Laboratories, New Drug Research Division , Otsuka Pharmaceutical Co., Ltd., Tokushima, Japan
| | - Kenner C. Rice
- Drug Design and Synthesis Section, Intramural Research Program, National Institute on Drug Abuse, National Institutes of Health, Bethesda, MD, 20892, USA
| | - Bruce E. Blough
- Center for Drug Discovery, Research Triangle Institute, Research Triangle Park, NC, 27709, USA
| | - Marius C. Hoener
- Neuroscience Research, pRED, Roche Innovation Center Basel, F. Hoffmann-La Roche Ltd, Basel, Switzerland
| | - Michael H. Baumann
- Designer Drug Research Unit, Intramural Research Program, National Institute on Drug Abuse, National Institutes of Health, Baltimore, MD, 21224, USA
| | - Matthias E. Liechti
- Division of Clinical Pharmacology and Toxicology, Department of Biomedicine, University Hospital Basel and University of Basel, Basel, Switzerland,Corresponding author: Prof. Dr. med. Matthias E. Liechti, Division of Clinical Pharmacology and Toxicology, University Hospital Basel, Schanzenstrasse 55, CH-4056 Basel, Switzerland. Tel: +41 61 328 68 68; Fax: +41 61 265 45 60;
| |
Collapse
|
32
|
Luethi D, Widmer R, Trachsel D, Hoener MC, Liechti ME. Monoamine receptor interaction profiles of 4-aryl-substituted 2,5-dimethoxyphenethylamines (2C-BI derivatives). Eur J Pharmacol 2019; 855:103-111. [DOI: 10.1016/j.ejphar.2019.05.014] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/25/2019] [Revised: 05/02/2019] [Accepted: 05/03/2019] [Indexed: 10/26/2022]
|
33
|
Rickli A, Hoener MC, Liechti ME. Pharmacological profiles of compounds in preworkout supplements ("boosters"). Eur J Pharmacol 2019; 859:172515. [PMID: 31265842 DOI: 10.1016/j.ejphar.2019.172515] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/05/2019] [Revised: 06/21/2019] [Accepted: 06/28/2019] [Indexed: 02/07/2023]
Abstract
Preworkout supplements ("boosters") are used to enhance physical and mental performance during workouts. These products may contain various chemical substances with undefined pharmacological activity. We investigated whether substances that are contained in commercially available athletic multiple-ingredient preworkout supplements exert amphetamine-type activity at norepinephrine, dopamine, and serotonin transporters (NET, DAT, and SERT, respectively). We assessed the in vitro monoamine transporter inhibition potencies of the substances using human embryonic kidney 293 cells that expressed the human NET, DAT, and SERT. The phenethylamines β-phenethylamine, N-methylphenethylamine, β-methylphenethylamine, N-benzylphenethylamine, N-methyl-β-methylphenethylamine, and methylsynephrine inhibited the NET and less potently the DAT similarly to D-amphetamine. β-phenethylamine was the most potent, with IC50 values of 0.05 and 1.8 μM at the NET and DAT, respectively. These IC50 values were comparable to D-amphetamine (IC50 = 0.09 and 1.3 μM, respectively). The alkylamines 1,3-dimethylbutylamine and 1,3-dimethylamylamine blocked the NET but not the DAT. Most of the phenethylamines interacted with trace amine-associated receptor 1, serotonin 5-hydroxytryptamine-1A receptor, and adrenergic α1A and α2A receptors at submicromolar concentrations. None of the compounds blocked the SERT. In conclusion, products that are used by athletes may contain substances with mainly noradrenergic amphetamine-type properties.
Collapse
Affiliation(s)
- Anna Rickli
- Division of Clinical Pharmacology and Toxicology, Department of Biomedicine, University Hospital Basel and University of Basel, Basel, Switzerland
| | - Marius C Hoener
- Neuroscience Research, pRED, Roche Innovation Center Basel, F. Hoffmann-La Roche Ltd, Basel, Switzerland
| | - Matthias E Liechti
- Division of Clinical Pharmacology and Toxicology, Department of Biomedicine, University Hospital Basel and University of Basel, Basel, Switzerland.
| |
Collapse
|
34
|
Rickli A, Kolaczynska K, Hoener MC, Liechti ME. Pharmacological characterization of the aminorex analogs 4-MAR, 4,4′-DMAR, and 3,4-DMAR. Neurotoxicology 2019; 72:95-100. [DOI: 10.1016/j.neuro.2019.02.011] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/21/2018] [Revised: 01/15/2019] [Accepted: 02/14/2019] [Indexed: 02/08/2023]
|
35
|
Luethi D, Walter M, Zhou X, Rudin D, Krähenbühl S, Liechti ME. Para-Halogenation Affects Monoamine Transporter Inhibition Properties and Hepatocellular Toxicity of Amphetamines and Methcathinones. Front Pharmacol 2019; 10:438. [PMID: 31068823 PMCID: PMC6491784 DOI: 10.3389/fphar.2019.00438] [Citation(s) in RCA: 21] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/06/2019] [Accepted: 04/05/2019] [Indexed: 11/16/2022] Open
Abstract
Halogenated derivatives of amphetamine-type stimulants are appearing on the drug market, often with altered pharmacological profile and sometimes different legal status compared to the non-halogenated substances. The aim of the present study was to investigate the pharmacological profile and hepatocellular toxicity of para-halogenated amphetamines and cathinones. The potential of amphetamine, 4-fluoroamphetamine, 4-chloroamphetamine, methcathinone, 4-fluoromethcathinone, and 4-chloromethcathinone to inhibit the monoamine transporters for norepinephrine, dopamine, and serotonin was determined in transporter-transfected human embryonic kidney 293 cells. Cell membrane integrity, ATP content, oxygen consumption rate, and superoxide levels were measured in human hepatoma HepG2 cells after exposure to the substances for 24 h. All compounds inhibited the norepinephrine transporter at submicromolar concentrations and the dopamine transporter at low micromolar concentrations. The selectivity of the compounds to inhibit the dopamine versus serotonin transporter decreased with increasing size of the para-substituent, resulting in potent serotonin uptake inhibition for the halogenated derivatives. All substances depleted the cellular ATP content at lower concentrations (0.25–2 mM) than cell membrane integrity loss occurred (≥0.5 mM), suggesting mitochondrial toxicity. The amphetamines and 4-chloromethcathinone additionally impaired the mitochondrial respiratory chain, confirming mitochondrial toxicity. The following toxicity rank order for the para-substituents was observed: chloride > fluoride > hydrogen. In conclusion, para-halogenation of stimulants increases the risk for serotonergic neurotoxicity. Furthermore, para-halogenation may increase hepatic toxicity mediated by mitochondrial impairment in susceptible users.
Collapse
Affiliation(s)
- Dino Luethi
- Division of Clinical Pharmacology and Toxicology, Department of Biomedicine, University Hospital Basel, University of Basel, Basel, Switzerland
| | - Melanie Walter
- Division of Clinical Pharmacology and Toxicology, Department of Biomedicine, University Hospital Basel, University of Basel, Basel, Switzerland
| | - Xun Zhou
- Division of Clinical Pharmacology and Toxicology, Department of Biomedicine, University Hospital Basel, University of Basel, Basel, Switzerland
| | - Deborah Rudin
- Division of Clinical Pharmacology and Toxicology, Department of Biomedicine, University Hospital Basel, University of Basel, Basel, Switzerland
| | - Stephan Krähenbühl
- Division of Clinical Pharmacology and Toxicology, Department of Biomedicine, University Hospital Basel, University of Basel, Basel, Switzerland.,Swiss Centre for Applied Human Toxicology (SCAHT), University of Basel, Basel, Switzerland
| | - Matthias E Liechti
- Division of Clinical Pharmacology and Toxicology, Department of Biomedicine, University Hospital Basel, University of Basel, Basel, Switzerland
| |
Collapse
|
36
|
Robins MT, Blaine AT, Ha JE, Brewster AL, van Rijn RM. Repeated Use of the Psychoactive Substance Ethylphenidate Impacts Neurochemistry and Reward Learning in Adolescent Male and Female Mice. Front Neurosci 2019; 13:124. [PMID: 30837836 PMCID: PMC6389692 DOI: 10.3389/fnins.2019.00124] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/06/2018] [Accepted: 02/04/2019] [Indexed: 02/03/2023] Open
Abstract
Schedule II prescription psychostimulants, such as methylphenidate (MPH), can be misused as nootropic drugs, i.e., drugs that enhance focus and cognition. When users are unable to obtain these prescribed medications, they may seek out novel psychoactive substances (NPSs) that are not yet scheduled. An example of a NPS reportedly being abused is ethylphenidate (EPH), a close analog of MPH but with a higher preference for the dopamine transporter compared with the norepinephrine transporter. Therefore, based upon this pharmacological profile and user self-reports, we hypothesized that repeated EPH exposure in adolescent mice may be rewarding and alter cognition. Here, we report that repeated exposure to 15 mg/kg EPH decreased spatial cognitive performance as assessed by the Barnes maze spatial learning task in adolescent male C57Bl/6 mice; however, male mice did not show alterations in the expression of mature BDNF - a protein associated with increased cognitive function - in key brain regions. Acute EPH exposure induced hyperlocomotion at a high dose (15 mg/kg, i.p.), but not a low dose (5 mg/kg, i.p.). Interestingly, mice exhibited significant conditioned place preference at the low EPH dose, suggesting that even non-stimulating doses of EPH are rewarding. In both males and females, repeated EPH exposure increased expression of deltaFosB - a marker associated with increased risk of drug abuse - in the dorsal striatum, nucleus accumbens, and prefrontal cortex. Overall, our results suggest that repeated EPH use in adolescence is psychostimulatory, rewarding, increases crucial brain markers of reward-related behaviors, and may negatively impact spatial performance.
Collapse
Affiliation(s)
- Meridith T Robins
- Department of Medicinal Chemistry and Molecular Pharmacology, Purdue University, West Lafayette, IN, United States.,Purdue Institute for Integrative Neuroscience, Purdue University, West Lafayette, IN, United States
| | - Arryn T Blaine
- Department of Medicinal Chemistry and Molecular Pharmacology, Purdue University, West Lafayette, IN, United States.,Purdue Institute for Integrative Neuroscience, Purdue University, West Lafayette, IN, United States.,Purdue Interdisciplinary Life Sciences Graduate Program, Purdue University, West Lafayette, IN, United States
| | - Jiwon E Ha
- Department of Medicinal Chemistry and Molecular Pharmacology, Purdue University, West Lafayette, IN, United States
| | - Amy L Brewster
- Purdue Institute for Integrative Neuroscience, Purdue University, West Lafayette, IN, United States.,Purdue Interdisciplinary Life Sciences Graduate Program, Purdue University, West Lafayette, IN, United States.,Department of Psychological Sciences, Purdue University, West Lafayette, IN, United States
| | - Richard M van Rijn
- Department of Medicinal Chemistry and Molecular Pharmacology, Purdue University, West Lafayette, IN, United States.,Purdue Institute for Integrative Neuroscience, Purdue University, West Lafayette, IN, United States.,Purdue Interdisciplinary Life Sciences Graduate Program, Purdue University, West Lafayette, IN, United States
| |
Collapse
|
37
|
Zsilla G, Hegyi DE, Baranyi M, Vizi ES. 3,4-Methylenedioxymethamphetamine, mephedrone, and β-phenylethylamine release dopamine from the cytoplasm by means of transporters and keep the concentration high and constant by blocking reuptake. Eur J Pharmacol 2018; 837:72-80. [DOI: 10.1016/j.ejphar.2018.08.037] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/28/2018] [Revised: 08/24/2018] [Accepted: 08/29/2018] [Indexed: 02/02/2023]
|
38
|
Luethi D, Liechti ME. Monoamine Transporter and Receptor Interaction Profiles in Vitro Predict Reported Human Doses of Novel Psychoactive Stimulants and Psychedelics. Int J Neuropsychopharmacol 2018; 21:926-931. [PMID: 29850881 PMCID: PMC6165951 DOI: 10.1093/ijnp/pyy047] [Citation(s) in RCA: 54] [Impact Index Per Article: 7.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/24/2018] [Accepted: 05/24/2018] [Indexed: 12/31/2022] Open
Abstract
BACKGROUND Pharmacological profiles of new psychoactive substances can be established rapidly in vitro and provide information on potential psychoactive effects in humans. The present study investigated whether specific in vitro monoamine transporter and receptor interactions can predict effective psychoactive doses in humans. METHODS We correlated previously assessed in vitro data of stimulants and psychedelics with human doses that are reported on the Internet and in books. RESULTS For stimulants, dopamine and norepinephrine transporter inhibition potency was positively correlated with human doses, whereas serotonin transporter inhibition potency was inversely correlated with human doses. Serotonin 5-hydroxytryptamine-2A (5-HT2A) and 5-HT2C receptor affinity was significantly correlated with psychedelic doses, but 5-HT1A receptor affinity and 5-HT2A and 5-HT2B receptor activation potency were not. CONCLUSIONS The rapid assessment of in vitro pharmacological profiles of new psychoactive substances can help to predict psychoactive doses and effects in humans and facilitate the appropriate scheduling of new psychoactive substances.
Collapse
Affiliation(s)
- Dino Luethi
- Division of Clinical Pharmacology and Toxicology, Department of Biomedicine, University Hospital Basel and University of Basel, Basel, Switzerland
| | - Matthias E Liechti
- Division of Clinical Pharmacology and Toxicology, Department of Biomedicine, University Hospital Basel and University of Basel, Basel, Switzerland,Correspondence: Matthias E. Liechti, ***, Division of Clinical Pharmacology and Toxicology, University Hospital Basel, Schanzenstrasse 55, CH-4056 Basel, Switzerland ()
| |
Collapse
|
39
|
Dolder PC, Strajhar P, Vizeli P, Odermatt A, Liechti ME. Acute effects of lisdexamfetamine and D-amphetamine on social cognition and cognitive performance in a placebo-controlled study in healthy subjects. Psychopharmacology (Berl) 2018; 235:1389-1402. [PMID: 29511807 DOI: 10.1007/s00213-018-4849-0] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/22/2017] [Accepted: 01/31/2018] [Indexed: 01/26/2023]
Abstract
RATIONALE Amphetamines are used as medications but are also misused as cognitive enhancers by healthy subjects and may have additional effects on social cognition. METHODS We investigated the acute effects of single, high, equimolar doses of D-amphetamine (40 mg) and lisdexamfetamine (100 mg) on social cognition and cognitive performance using a randomized, placebo-controlled, double-blind, cross-over design in 24 healthy volunteers. Effects on social cognition were assessed using the Facial Emotion Recognition Task (FERT), Multifaceted Empathy Test (MET), and Sexual Arousal Task (SAT). Cognitive performance was measured using the Digit Symbol Substitution Test (DSST), Digit Span (DS), Stop-Signal Task (SST), and Mackworth Clock Test (MCT). RESULTS D-Amphetamine and lisdexamfetamine had small effects on measures of social cognition. There were no effects on emotion recognition on the FERT. D-Amphetamine increased direct empathy on the MET, but only for positive stimuli. Both amphetamines increased ratings of pleasantness and attractiveness on the SAT in response to sexual but also to neutral stimuli. D-Amphetamine and lisdexamfetamine increased cognitive performance (go-accuracy and vigilance on the SST and MCT, respectively). Lisdexamfetamine increased processing speed on the DSST. Neither drug had an effect on the DS. CONCLUSION Single, high, equimolar doses of D-amphetamine and lisdexamfetamine enhanced certain aspects of cognitive performance in healthy non-sleep-deprived subjects. Both amphetamines also slightly altered aspects of social cognition. Whether these small effects also influence social interaction behavior in amphetamine users remains to be investigated. TRIAL REGISTRATION The study was registered at ClinicalTrials.gov (NCT02668926).
Collapse
Affiliation(s)
- Patrick C Dolder
- Psychopharmacology Research, Division of Clinical Pharmacology and Toxicology, Department of Biomedicine and Department of Clinical Research, University Hospital Basel and University of Basel, Schanzenstrasse 55, 4056, Basel, CH, Switzerland
| | - Petra Strajhar
- Division of Molecular and Systems Toxicology, Department of Pharmaceutical Sciences, University of Basel, Basel, Switzerland
| | - Patrick Vizeli
- Psychopharmacology Research, Division of Clinical Pharmacology and Toxicology, Department of Biomedicine and Department of Clinical Research, University Hospital Basel and University of Basel, Schanzenstrasse 55, 4056, Basel, CH, Switzerland
| | - Alex Odermatt
- Division of Molecular and Systems Toxicology, Department of Pharmaceutical Sciences, University of Basel, Basel, Switzerland
| | - Matthias E Liechti
- Psychopharmacology Research, Division of Clinical Pharmacology and Toxicology, Department of Biomedicine and Department of Clinical Research, University Hospital Basel and University of Basel, Schanzenstrasse 55, 4056, Basel, CH, Switzerland.
| |
Collapse
|
40
|
|