1
|
Łapińska N, Szlęk J, Pacławski A, Mendyk A. Machine Learning Tool for New Selective Serotonin and Serotonin-Norepinephrine Reuptake Inhibitors. Molecules 2025; 30:637. [PMID: 39942741 PMCID: PMC11819831 DOI: 10.3390/molecules30030637] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/06/2024] [Revised: 01/17/2025] [Accepted: 01/24/2025] [Indexed: 02/16/2025] Open
Abstract
Depression, a serious mood disorder, affects about 5% of the population. Currently, there are two groups of antidepressants that are the first-line treatment for depressive disorder: selective serotonin reuptake inhibitors and serotonin-norepinephrine reuptake inhibitors. The aim of the study was to develop Quantitative Structure-Activity Relationship (QSAR) models for serotonin (SERT) and norepinephrine (NET) transporters to predict the affinity and inhibition potential of new molecules. Models were developed using the Automated Machine Learning tool Mljar based on 80% of the dataset according to 10-fold cross-validation and externally validated on the remaining 20% of data. The molecular representation featured two-dimensional Mordred descriptors. For each model, Shapley additive explanations analysis was performed to clarify the influence of the descriptors on the models' predictions. Based on the final QSAR models, the following results were obtained: NET and pIC50 value RMSEtest = 0.678, R2test = 0.640; NET and pKi RMSEtest = 0.590, R2test = 0.709; SERT and pIC50 RMSEtest = 0.645, R2test = 0.678; SERT and pKi value RMSEtest = 0.540, R2test = 0.828. QSAR models for serotonin and norepinephrine transporters have been made available in a new module of the SerotoninAI application to enhance usability for scientists.
Collapse
Affiliation(s)
- Natalia Łapińska
- Department of Pharmaceutical Technology and Biopharmaceutics, Jagiellonian University Medical College, 30-688 Kraków, Poland; (N.Ł.); (A.P.); (A.M.)
| | - Jakub Szlęk
- Department of Pharmaceutical Technology and Biopharmaceutics, Jagiellonian University Medical College, 30-688 Kraków, Poland; (N.Ł.); (A.P.); (A.M.)
- Bioinformatics and In Silico Analysis Laboratory, Center for the Development of Therapies for Civilization and Age-Related Diseases (CDT-CARD), 8 Skawińska St., 31-066 Kraków, Poland
| | - Adam Pacławski
- Department of Pharmaceutical Technology and Biopharmaceutics, Jagiellonian University Medical College, 30-688 Kraków, Poland; (N.Ł.); (A.P.); (A.M.)
| | - Aleksander Mendyk
- Department of Pharmaceutical Technology and Biopharmaceutics, Jagiellonian University Medical College, 30-688 Kraków, Poland; (N.Ł.); (A.P.); (A.M.)
| |
Collapse
|
2
|
Nadal-Gratacós N, Pazos MD, Pubill D, Camarasa J, Escubedo E, Berzosa X, López-Arnau R. Structure-Activity Relationship of Synthetic Cathinones: An Updated Review. ACS Pharmacol Transl Sci 2024; 7:2588-2603. [PMID: 39296271 PMCID: PMC11406692 DOI: 10.1021/acsptsci.4c00299] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/22/2024] [Revised: 07/10/2024] [Accepted: 07/15/2024] [Indexed: 09/21/2024]
Abstract
The escalating prevalence of new psychoactive substances (NPSs) poses a significant public health challenge, evidenced by the vast chemical diversity, with over 500 substances reported annually to the United Nations Office on Drugs and Crime-Early Warning Advisory (UNODC-EWA) in the past five years. Among NPSs, synthetic cathinones are gaining a lot of popularity among users. Notably, synthetic cathinones accounted for approximately 50% of the total quantity of NPSs reported as seized by EU Member States in 2021. Preliminary data from UNODC indicates that a total of 209 synthetic cathinones have been reported to date. As their popularity grows, studying the structure-activity relationship (SAR) of synthetic cathinones is essential. SAR studies elucidate how structural features impact biological effects, aiding in toxicity prediction, regulatory compliance, and forensic identification. Additionally, SAR studies play a pivotal role in guiding drug policies, aiding authorities in categorizing and regulating newly emerging synthetic cathinones, mitigate public health risks and offer valuable insights into potential therapeutic applications. Thus, our Review consolidates recent findings on the effects of different substitutions in the chemical scaffold of synthetic cathinones on their mechanism of action as well as pharmacological and toxicological effects of synthetic cathinones, thus enhancing understanding of the SAR of synthetic cathinones' pharmacology and potential implications.
Collapse
Affiliation(s)
- Núria Nadal-Gratacós
- Department of Pharmacology, Toxicology and Therapeutic Chemistry, Pharmacology Section and Institute of Biomedicine (IBUB), Faculty of Pharmacy, University of Barcelona, 08028 Barcelona, Spain
- Chemical Reactions for Innovative Solutions (CRISOL), IQS School of Engineering, Universitat Ramon Llull, 08017 Barcelona, Spain
| | - Martalu D Pazos
- Department of Pharmacology, Toxicology and Therapeutic Chemistry, Pharmacology Section and Institute of Biomedicine (IBUB), Faculty of Pharmacy, University of Barcelona, 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, 08028 Barcelona, Spain
| | - Jorge Camarasa
- Department of Pharmacology, Toxicology and Therapeutic Chemistry, Pharmacology Section and Institute of Biomedicine (IBUB), Faculty of Pharmacy, University of Barcelona, 08028 Barcelona, Spain
| | - Elena Escubedo
- Department of Pharmacology, Toxicology and Therapeutic Chemistry, Pharmacology Section and Institute of Biomedicine (IBUB), Faculty of Pharmacy, University of Barcelona, 08028 Barcelona, Spain
| | - Xavier Berzosa
- Chemical Reactions for Innovative Solutions (CRISOL), IQS School of Engineering, Universitat Ramon Llull, 08017 Barcelona, Spain
| | - Raúl López-Arnau
- Department of Pharmacology, Toxicology and Therapeutic Chemistry, Pharmacology Section and Institute of Biomedicine (IBUB), Faculty of Pharmacy, University of Barcelona, 08028 Barcelona, Spain
| |
Collapse
|
3
|
Fitzgerald LR, Gannon BM, Walther D, Landavazo A, Hiranita T, Blough BE, Baumann MH, Fantegrossi WE. Structure-activity relationships for locomotor stimulant effects and monoamine transporter interactions of substituted amphetamines and cathinones. Neuropharmacology 2024; 245:109827. [PMID: 38154512 PMCID: PMC10842458 DOI: 10.1016/j.neuropharm.2023.109827] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/05/2023] [Revised: 11/14/2023] [Accepted: 12/20/2023] [Indexed: 12/30/2023]
Abstract
Substitutions to the phenethylamine structure give rise to numerous amphetamines and cathinones, contributing to an ever-growing number of abused novel psychoactive substances. Understanding how various substitutions affect the pharmacology of phenethylamines may help lawmakers and scientists predict the effects of newly emerging drugs. Here, we established structure-activity relationships for locomotor stimulant and monoamine transporter effects of 12 phenethylamines with combinations of para-chloro, β-keto, N-methyl, or N-ethyl additions. Automated photobeam analysis was used to evaluate effects of drugs on ambulatory activity in mice, whereas in vitro assays were used to determine activities at transporters for dopamine (DAT), norepinephrine (NET), and 5-HT (SERT) in rat brain synaptosomes. In mouse studies, all compounds stimulated locomotion, except for 4-chloro-N-ethylcathinone. Amphetamines were more potent stimulants than their β-keto counterparts, while para-chloro amphetamines tended to be more efficacious than unsubstituted amphetamines. Para-chloro compounds also produced lethality at doses on the ascending limbs of their locomotor dose-effect functions. The in vitro assays showed that all compounds inhibited uptake at DAT, NET, and SERT, with most compounds also acting as substrates (i.e., releasers) at these sites. Unsubstituted compounds displayed better potency at DAT and NET relative to SERT. Para-chloro substitution or increased N-alkyl chain length augmented relative potency at SERT, while combined para-chloro and N-ethyl substitutions reduced releasing effects at NET and DAT. These results demonstrate orderly SAR for locomotor stimulant effects, monoamine transporter activities, and lethality induced by phenethylamines. Importantly, 4-chloro compounds produce toxicity in mice that suggests serious risk to humans using these drugs in recreational contexts.
Collapse
Affiliation(s)
- Lauren R Fitzgerald
- Department of Pharmacology and Toxicology, University of Arkansas for Medical Sciences, Little Rock, AR, 72205, USA
| | - Brenda M Gannon
- Department of Pharmacology and Toxicology, University of Arkansas for Medical Sciences, Little Rock, AR, 72205, USA
| | - Donna Walther
- Designer Drug Research Unit, Intramural Research Program, National Institute on Drug Abuse, National Institutes of Health, Baltimore, MD, 21224, USA
| | - Antonio Landavazo
- Center for Drug Discovery, RTI International, Research Triangle Park, NC, 27709, USA
| | - Takato Hiranita
- Department of Pharmacology, University of Texas Health Science Center at San Antonio, San Antonio, TX, 78229, USA
| | - Bruce E Blough
- Center for Drug Discovery, RTI International, Research Triangle Park, NC, 27709, USA
| | - Michael H Baumann
- Designer Drug Research Unit, Intramural Research Program, National Institute on Drug Abuse, National Institutes of Health, Baltimore, MD, 21224, USA
| | - William E Fantegrossi
- Department of Pharmacology and Toxicology, University of Arkansas for Medical Sciences, Little Rock, AR, 72205, USA.
| |
Collapse
|
4
|
Angoa-Perez M, Kuhn DM. The pharmacology and neurotoxicology of synthetic cathinones. ADVANCES IN PHARMACOLOGY (SAN DIEGO, CALIF.) 2023; 99:61-82. [PMID: 38467489 DOI: 10.1016/bs.apha.2023.12.001] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 03/13/2024]
Abstract
The synthetic cathinones are man-made compounds derived from the naturally occurring drug cathinone, which is found in the khat plant. The drugs in this pharmacological class that will be the focus of this chapter include mephedrone, MDPV, methcathinone and methylone. These drugs are colloquially known as "bath salts". This misnomer suggests that these drugs are used for health improvement or that they have legitimate medical uses. The synthetic cathinones are dangerous drugs with powerful pharmacological effects that include high abuse potential, hyperthermia and hyperlocomotion. These drugs also share many of the pharmacological effects of the amphetamine class of drugs including methamphetamine, amphetamine and MDMA and therefore have high potential to cause damage to the central nervous system. The synthetic cathinones are frequently taken in combination with other psychoactive drugs such as alcohol, marijuana and the amphetamine-like stimulants, creating a situation where heightened pharmacological and neurotoxicological effects are likely to occur. Despite the structural features shared by the synthetic cathinones and amphetamine-like stimulants, including their actions at monoamine transporters and receptors, the effects of the synthetic cathinones do not always match those of the amphetamines. In particular, the synthetic cathinones are far less neurotoxic than their amphetamine counterparts, they produce a weaker hyperthermia, and they cause less glial activation. This chapter will briefly review the pharmacology and neurotoxicology of selected synthetic cathinones with the aim of delineating key areas of agreement and disagreement in the literature particularly as it relates to neurotoxicological outcomes.
Collapse
Affiliation(s)
- Mariana Angoa-Perez
- Research and Development Service, John D. Dingell VA Medical Center, Detroit, MI, United States; Department of Psychiatry and Behavioral Neurosciences, Wayne State University School of Medicine, Detroit, MI, United States
| | - Donald M Kuhn
- Research and Development Service, John D. Dingell VA Medical Center, Detroit, MI, United States; Department of Psychiatry and Behavioral Neurosciences, Wayne State University School of Medicine, Detroit, MI, United States.
| |
Collapse
|
5
|
Nguyen VT, Harris AC, Eltit JM. Structural and functional perspectives on interactions between synthetic cathinones and monoamine transporters. ADVANCES IN PHARMACOLOGY (SAN DIEGO, CALIF.) 2023; 99:83-124. [PMID: 38467490 DOI: 10.1016/bs.apha.2023.09.001] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 03/13/2024]
Abstract
Synthetic cathinone derivatives comprise a family of psychoactive compounds structurally related to amphetamine. Over the last decade, clandestine chemists have synthesized a consistent stream of innovative cathinone derivatives to outpace governmental regulatory restrictions. Many of these unregulated substances are produced and distributed as designer drugs. Two of the principal chemical scaffolds exploited to expand the synthetic cathinone family are methcathinone and α-pyrrolidinopentiophenone (or α-pyrrolidinovalerophenone, α-PVP). These compounds' main physiological targets are monoamine transporters, where they promote addiction by potentiating dopaminergic neurotransmission. This chapter describes techniques used to study the pharmacodynamic properties of cathinones at monoamine transporters in vitro. Biochemical techniques described include uptake inhibition and release assays in rat brain synaptosomes and in mammalian expression systems. Electrophysiological techniques include current measurements using the voltage clamp technique. We describe a Ca2+ mobilization assay wherein voltage-gated Ca2+ channels function as reporters to study the action of synthetic cathinones at monoamine transporters. We discuss results from systematic structure-activity relationship studies on simple and complex cathinones at monoamine transporters with an emphasis on identifying structural moieties that modulate potency and selectivity at these transporters. Moreover, different profiles of selectivity at monoamine transporters directly predict compounds associated with behavioral and subjective effects within animals and humans. In conclusion, clarification of the structural aspects of compounds which modulate potency and selectivity at monoamine transporters is critical to identify and predict potential addictive drugs. This knowledge may allow prompt allocation of resources toward drugs that represent the greatest threats after drugs are identified by forensic laboratories.
Collapse
Affiliation(s)
- Vy T Nguyen
- Department of Physiology and Biophysics, School of Medicine, Virginia Commonwealth University, Richmond, VA, United States
| | - Alan C Harris
- Department of Physiology and Biophysics, School of Medicine, Virginia Commonwealth University, Richmond, VA, United States
| | - Jose M Eltit
- Department of Physiology and Biophysics, School of Medicine, Virginia Commonwealth University, Richmond, VA, United States.
| |
Collapse
|
6
|
Davies RA, Nguyen VT, Eltit JM, Glennon RA. Structure-Activity Relationships for a Recently Controlled Synthetic Cathinone Dopamine Transporter Reuptake Inhibitor: α-Pyrrolidinohexiophenone (α-PHP). ACS Chem Neurosci 2023; 14:2527-2536. [PMID: 37406364 PMCID: PMC10670441 DOI: 10.1021/acschemneuro.3c00156] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 07/07/2023] Open
Abstract
α-Pyrrolidinohexiophenone (α-PHP) is the one-carbon unit α-extended homolog of the better-known and widely abused synthetic cathinone central stimulant α-PVP ("flakka"); both are now U.S. Schedule I controlled substances. Structurally, α-PVP and α-PHP possess a common terminal N-pyrrolidine moiety and differ only with respect to the length of their α-alkyl chain. Using a synaptosomal assay, we previously reported that α-PHP is at least as potent as α-PVP as a dopamine transporter (DAT) reuptake inhibitor. A systematic structure-activity study of synthetic cathinones (e.g., α-PHP) as DAT reuptake inhibitors (i.e., transport blockers), a mechanism thought responsible for their abuse liability, has yet to be conducted. Here, we examined a series of 4-substituted α-PHP analogues and found that, with one exception, all behaved as relatively (28- to >300-fold) selective DAT versus serotonin transporter (SERT) reuptake inhibitors with DAT inhibition potencies of most falling within a very narrow (i.e., <3-fold) range. The 4-CF3 analogue of α-PHP was a confirmed "outlier" in that it was at least 80-fold less potent than the other analogues and displayed reduced (i.e., no) DAT vs SERT selectivity. Consideration of various physicochemical properties of the CF3 group, relative to that of the other substituents involved here, provided relatively little insight. Unlike with DAT-releasing agents, as previously reported by us, a QSAR study was precluded because of the limited range of empirical results (with the exception of the 4-CF3 analogue) for DAT reuptake inhibition.
Collapse
Affiliation(s)
- Rachel A. Davies
- Department of Medicinal Chemistry, School of Pharmacy, Virginia Commonwealth University; Richmond, Virginia 23298 U.S
| | - Vy T. Nguyen
- Department of Physiology and Biophysics, School of Medicine, Virginia Commonwealth University; Richmond, Virginia 23298 U.S
| | - Jose M. Eltit
- Department of Physiology and Biophysics, School of Medicine, Virginia Commonwealth University; Richmond, Virginia 23298 U.S
| | - Richard A. Glennon
- Department of Medicinal Chemistry, School of Pharmacy, Virginia Commonwealth University; Richmond, Virginia 23298 U.S
| |
Collapse
|
7
|
Chojnacki MR, Thorndike EB, Partilla JS, Rice KC, Schindler CW, Baumann MH. Neurochemical and Cardiovascular Effects of 4-Chloro Ring-Substituted Synthetic Cathinones in Rats. J Pharmacol Exp Ther 2023; 385:162-170. [PMID: 36669877 PMCID: PMC10201577 DOI: 10.1124/jpet.122.001478] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/18/2022] [Revised: 01/03/2023] [Accepted: 01/12/2023] [Indexed: 01/22/2023] Open
Abstract
Synthetic cathinones are a class of new psychoactive substances that display psychomotor stimulant properties, and novel cathinone analogs continue to emerge in illicit drug markets worldwide. The aim of the present study was to characterize the pharmacology of 4-chloro ring-substituted cathinones that are appearing in illicit drug markets compared with the effects of 4-methylmethcathinone (mephedrone). Synaptosomes were prepared from rat caudate for dopamine transporter (DAT) assays or from whole brain minus caudate and cerebellum for norepinephrine transporter (NET) and serotonin transporter (SERT) assays. Findings from transporter uptake inhibition and release assays showed that mephedrone and 4-chloromethcathinone (4-CMC) function as substrates at DAT, NET, and SERT, with similar potency at all three transporters. In contrast, 4-chloro-α-pyrrolidinopropiophenone (4-CαPPP) was an uptake inhibitor at DAT and NET, with similar potency at each site, but had little activity at SERT. 4-Chloroethcathinone (4-CEC) was a low-potency uptake inhibitor at DAT and NET but a substrate at SERT. In rats implanted with telemetry transmitters, mephedrone and 4-CMC increased blood pressure, heart rate, and locomotor activity to a similar extent. 4-CEC and 4-CαPPP were less potent at increasing blood pressure and had modest stimulatory effects on heart rate and activity. 4-CMC also transiently decreased temperature at the highest dose tested. All three 4-chloro ring-substituted cathinones are biologically active, but only 4-CMC has potency comparable to mephedrone. Collectively, our findings suggest that 4-CMC and other 4-chloro cathinones may have abuse potential and adverse effects in humans that are analogous to those associated with mephedrone. SIGNIFICANCE STATEMENT: The 4-chloro ring-substituted cathinones all produced significant cardiovascular stimulation, with 4-chloromethcathinone (4-CMC) showing potency similar to mephedrone. All of the drugs are likely to be abused given their effects at the dopamine transporter, particularly 4-CMC.
Collapse
Affiliation(s)
- Michael R Chojnacki
- Designer Drug Research Unit (M.R.C., J.S.P., C.W.S., M.H.B.) and Preclinical Pharmacology Section (E.B.T., C.W.S.), National Institute on Drug Abuse Intramural Research Program, Baltimore, Maryland; and Drug Design and Synthesis Section, National Institute on Drug Abuse and National Institute on Alcohol Abuse and Alcoholism Intramural Research Programs, Rockville, Maryland (K.C.R.)
| | - Eric B Thorndike
- Designer Drug Research Unit (M.R.C., J.S.P., C.W.S., M.H.B.) and Preclinical Pharmacology Section (E.B.T., C.W.S.), National Institute on Drug Abuse Intramural Research Program, Baltimore, Maryland; and Drug Design and Synthesis Section, National Institute on Drug Abuse and National Institute on Alcohol Abuse and Alcoholism Intramural Research Programs, Rockville, Maryland (K.C.R.)
| | - John S Partilla
- Designer Drug Research Unit (M.R.C., J.S.P., C.W.S., M.H.B.) and Preclinical Pharmacology Section (E.B.T., C.W.S.), National Institute on Drug Abuse Intramural Research Program, Baltimore, Maryland; and Drug Design and Synthesis Section, National Institute on Drug Abuse and National Institute on Alcohol Abuse and Alcoholism Intramural Research Programs, Rockville, Maryland (K.C.R.)
| | - Kenner C Rice
- Designer Drug Research Unit (M.R.C., J.S.P., C.W.S., M.H.B.) and Preclinical Pharmacology Section (E.B.T., C.W.S.), National Institute on Drug Abuse Intramural Research Program, Baltimore, Maryland; and Drug Design and Synthesis Section, National Institute on Drug Abuse and National Institute on Alcohol Abuse and Alcoholism Intramural Research Programs, Rockville, Maryland (K.C.R.)
| | - Charles W Schindler
- Designer Drug Research Unit (M.R.C., J.S.P., C.W.S., M.H.B.) and Preclinical Pharmacology Section (E.B.T., C.W.S.), National Institute on Drug Abuse Intramural Research Program, Baltimore, Maryland; and Drug Design and Synthesis Section, National Institute on Drug Abuse and National Institute on Alcohol Abuse and Alcoholism Intramural Research Programs, Rockville, Maryland (K.C.R.)
| | - Michael H Baumann
- Designer Drug Research Unit (M.R.C., J.S.P., C.W.S., M.H.B.) and Preclinical Pharmacology Section (E.B.T., C.W.S.), National Institute on Drug Abuse Intramural Research Program, Baltimore, Maryland; and Drug Design and Synthesis Section, National Institute on Drug Abuse and National Institute on Alcohol Abuse and Alcoholism Intramural Research Programs, Rockville, Maryland (K.C.R.)
| |
Collapse
|
8
|
Mayer FP, Niello M, Cintulova D, Sideromenos S, Maier J, Li Y, Bulling S, Kudlacek O, Schicker K, Iwamoto H, Deng F, Wan J, Holy M, Katamish R, Sandtner W, Li Y, Pollak DD, Blakely RD, Mihovilovic MD, Baumann MH, Sitte HH. Serotonin-releasing agents with reduced off-target effects. Mol Psychiatry 2023; 28:722-732. [PMID: 36352123 PMCID: PMC9645344 DOI: 10.1038/s41380-022-01843-w] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/11/2022] [Revised: 10/06/2022] [Accepted: 10/12/2022] [Indexed: 11/10/2022]
Abstract
Increasing extracellular levels of serotonin (5-HT) in the brain ameliorates symptoms of depression and anxiety-related disorders, e.g., social phobias and post-traumatic stress disorder. Recent evidence from preclinical and clinical studies established the therapeutic potential of drugs inducing the release of 5-HT via the 5-HT-transporter. Nevertheless, current 5-HT releasing compounds under clinical investigation carry the risk for abuse and deleterious side effects. Here, we demonstrate that S-enantiomers of certain ring-substituted cathinones show preference for the release of 5-HT ex vivo and in vivo, and exert 5-HT-associated effects in preclinical behavioral models. Importantly, the lead cathinone compounds (1) do not induce substantial dopamine release and (2) display reduced off-target activity at vesicular monoamine transporters and 5-HT2B-receptors, indicative of low abuse-liability and low potential for adverse events. Taken together, our findings identify these agents as lead compounds that may prove useful for the treatment of disorders where elevation of 5-HT has proven beneficial.
Collapse
Affiliation(s)
- Felix P Mayer
- Center for Physiology and Pharmacology, Institute of Pharmacology, Medical University of Vienna, Waehringer Strasse 13a, 1090, Vienna, Austria.
- Department of Biomedical Science, Charles E. Schmidt College of Medicine, Florida Atlantic University, Jupiter, FL, 33458, USA.
| | - Marco Niello
- Center for Physiology and Pharmacology, Institute of Pharmacology, Medical University of Vienna, Waehringer Strasse 13a, 1090, Vienna, Austria
| | | | - Spyridon Sideromenos
- Department of Neurophysiology and Neuropharmacology, Medical University of Vienna, Vienna, Austria
| | - Julian Maier
- Center for Physiology and Pharmacology, Institute of Pharmacology, Medical University of Vienna, Waehringer Strasse 13a, 1090, Vienna, Austria
| | - Yang Li
- Center for Physiology and Pharmacology, Institute of Pharmacology, Medical University of Vienna, Waehringer Strasse 13a, 1090, Vienna, Austria
- Institutes of Brain Science, Fudan University, Shanghai, 200032, China
| | - Simon Bulling
- Center for Physiology and Pharmacology, Institute of Pharmacology, Medical University of Vienna, Waehringer Strasse 13a, 1090, Vienna, Austria
| | - Oliver Kudlacek
- Center for Physiology and Pharmacology, Institute of Pharmacology, Medical University of Vienna, Waehringer Strasse 13a, 1090, Vienna, Austria
| | - Klaus Schicker
- Center for Physiology and Pharmacology, Institute of Pharmacology, Medical University of Vienna, Waehringer Strasse 13a, 1090, Vienna, Austria
| | - Hideki Iwamoto
- Stiles-Nicholson Brain Institute and Department of Biomedical Science, Charles E. Schmidt College of Medicine, Florida Atlantic University, Jupiter, FL, 33458, USA
| | - Fei Deng
- IDG McGovern Institute for Brain Research, Peking University, 100871, Beijing, China
| | - Jinxia Wan
- IDG McGovern Institute for Brain Research, Peking University, 100871, Beijing, China
| | - Marion Holy
- Center for Physiology and Pharmacology, Institute of Pharmacology, Medical University of Vienna, Waehringer Strasse 13a, 1090, Vienna, Austria
| | - Rania Katamish
- Department of Biomedical Science, Charles E. Schmidt College of Medicine, Florida Atlantic University, Jupiter, FL, 33458, USA
| | - Walter Sandtner
- Center for Physiology and Pharmacology, Institute of Pharmacology, Medical University of Vienna, Waehringer Strasse 13a, 1090, Vienna, Austria
| | - Yulong Li
- IDG McGovern Institute for Brain Research, Peking University, 100871, Beijing, China
| | - Daniela D Pollak
- Department of Neurophysiology and Neuropharmacology, Medical University of Vienna, Vienna, Austria
| | - Randy D Blakely
- Department of Biomedical Science, Charles E. Schmidt College of Medicine, Florida Atlantic University, Jupiter, FL, 33458, USA
- Stiles-Nicholson Brain Institute and Department of Biomedical Science, Charles E. Schmidt College of Medicine, Florida Atlantic University, Jupiter, FL, 33458, USA
| | | | - Michael H Baumann
- Designer Drug Research Unit, Intramural Research Program, National Institute on Drug Abuse, National Institutes of Health, Baltimore, MD, 21224, USA
| | - Harald H Sitte
- Center for Physiology and Pharmacology, Institute of Pharmacology, Medical University of Vienna, Waehringer Strasse 13a, 1090, Vienna, Austria.
- AddRess, Center for Addiction Research and Science, Medical University of Vienna, Vienna, Austria.
| |
Collapse
|
9
|
Chen FE, Pitt TA, Okong’o DJ, Wetherbee LG, Fuentes-Rivera JJ, Milner PJ. A Structure-Activity Study of Aromatic Acid Modulators for the Synthesis of Zirconium-Based Metal-Organic Frameworks. CHEMISTRY OF MATERIALS : A PUBLICATION OF THE AMERICAN CHEMICAL SOCIETY 2022; 34:3383-3394. [PMID: 36238710 PMCID: PMC9555823 DOI: 10.1021/acs.chemmater.2c00241] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/11/2023]
Abstract
Acid modulation is among the most widely employed methods for preparing metal-organic frameworks (MOFs) that are both stable and highly crystalline, yet there exist few guiding principles for selecting the optimal modulator for a given system. Using the Zr-based MOFs UiO-66 and UiO-68-Me2 (UiO = Universitetet i Oslo) as representative materials, here we present for the first time an in-depth structure-activity study of acid modulators and identify key principles of modulation for the synthesis of highly crystalline Zr-MOFs. By applying whole pattern fitting of powder X-ray diffraction (PXRD) patterns as a technique for evaluating modulator efficacy, complemented by scanning electron microscopy (SEM), 1H NMR, and thermogravimetric analysis (TGA), we demonstrate that the key to effective modulation is competition between the linker and modulator for coordination to the Zr secondary building units (SBUs). Specifically, we illustrate that a close match in pK a and structure between the linker and modulator favors larger and more well-defined crystallites, particularly with sterically unhindered aromatic acid modulators. Based on our findings, we demonstrate that 5-membered heteroaromatic carboxylic acids are among the most efficient acid modulators identified to date for the synthesis of several representative Zr-MOFs with fcu net topologies. In addition, we find that coordination modulation is superior to exogenous acid modulation at higher modulator concentrations. Finally, we compare 1H NMR and TGA as data-driven methods for quantifying linker deficiencies in modulated MOF syntheses. The guiding principles established herein have critical implications for the scalable and controllable synthesis of highly crystalline and stable MOFs relevant to chemical separations, gas storage, and catalysis.
Collapse
Affiliation(s)
- Faith E. Chen
- Department of Chemistry and Chemical Biology, Cornell University, Ithaca, NY, 14850, United States
| | - Tristan A. Pitt
- Department of Chemistry and Chemical Biology, Cornell University, Ithaca, NY, 14850, United States
| | - Diane J. Okong’o
- Department of Chemistry and Chemical Biology, Cornell University, Ithaca, NY, 14850, United States
| | - Luc G. Wetherbee
- Department of Chemistry and Chemical Biology, Cornell University, Ithaca, NY, 14850, United States
| | - José J. Fuentes-Rivera
- Department of Chemistry and Chemical Biology, Cornell University, Ithaca, NY, 14850, United States
| | - Phillip J. Milner
- Department of Chemistry and Chemical Biology, Cornell University, Ithaca, NY, 14850, United States
| |
Collapse
|
10
|
Steele TWE, Spires Z, Jones CB, Glennon RA, Dukat M, Eltit JM. Non-conserved residues dictate dopamine transporter selectivity for the potent synthetic cathinone and psychostimulant MDPV. Neuropharmacology 2021; 200:108820. [PMID: 34619165 DOI: 10.1016/j.neuropharm.2021.108820] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/13/2021] [Revised: 08/17/2021] [Accepted: 09/29/2021] [Indexed: 10/20/2022]
Abstract
Clandestine chemists are currently exploiting the pyrrolidinophenone scaffold to develop new designer drugs that carry the risk of abuse and overdose. These drugs promote addiction through the rewarding effects of increased dopaminergic neurotransmission. 3,4-Methylenedioxypyrovalerone (MDPV) and its analogs are illicit psychostimulants of this class that are ∼50-fold more potent than cocaine at inhibiting the human dopamine transporter (hDAT). In contrast, MDPV is a weak inhibitor at both the human serotonin transporter (hSERT) and, as it is shown here, the Drosophila melanogaster DAT (dDAT). We studied three conserved residues between hSERT and dDAT that are unique in hDAT (A117, F318, and P323 in dDAT), and one residue that is different in all three transporters (D121 in dDAT). hDAT residues were replaced in the dDAT sequence at these positions using site-directed mutagenesis and stable cell lines were generated expressing these mutant transporters. The potencies of MDPV and two of its analogs were determined using a Ca2+-mobilization assay. In this assay, voltage-gated Ca2+ channels are expressed to sense the membrane electrical depolarization evoked when dopamine is transported through DAT. Each individual mutant slightly improved MDPV's potency, but the combination of all four increased its potency ∼100-fold (2 log units) in inhibiting dDAT activity. Molecular modeling and docking studies were conducted to explore the possible mode of interaction between MDPV and DAT in silico. Two of the studied residues (F318 and P323) are at the entrance of the S1 binding site, whereas the other two (A117 and D121) face the aryl moiety of MDPV when bound to this site. Therefore, these four non-conserved residues can influence MDPV selectivity not only by stabilizing binding, but also by controlling access to its binding site at DAT.
Collapse
Affiliation(s)
- Tyler W E Steele
- Department of Physiology and Biophysics, School of Medicine, Virginia Commonwealth University, USA
| | - Zachary Spires
- Department of Physiology and Biophysics, School of Medicine, Virginia Commonwealth University, USA
| | - Charles B Jones
- Department of Medicinal Chemistry, School of Pharmacy, Virginia Commonwealth University, USA
| | - Richard A Glennon
- Department of Medicinal Chemistry, School of Pharmacy, Virginia Commonwealth University, USA
| | - Małgorzata Dukat
- Department of Medicinal Chemistry, School of Pharmacy, Virginia Commonwealth University, USA
| | - Jose M Eltit
- Department of Physiology and Biophysics, School of Medicine, Virginia Commonwealth University, USA.
| |
Collapse
|
11
|
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
|
12
|
Maier J, Rauter L, Rudin D, Niello M, Holy M, Schmid D, Wilson J, Blough BE, Gannon BM, Murnane KS, Sitte HH. α-PPP and its derivatives are selective partial releasers at the human norepinephrine transporter: A pharmacological characterization of interactions between pyrrolidinopropiophenones and high and low affinity monoamine transporters. Neuropharmacology 2021; 190:108570. [PMID: 33864800 DOI: 10.1016/j.neuropharm.2021.108570] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/18/2021] [Revised: 03/31/2021] [Accepted: 04/12/2021] [Indexed: 12/31/2022]
Abstract
While classical cathinones, such as methcathinone, have been shown to be monoamine releasing agents at human monoamine transporters, the subgroup of α-pyrrolidinophenones has thus far solely been characterized as monoamine transporter reuptake inhibitors. Herein, we report data from previously undescribed α-pyrrolidinopropiophenone (α-PPP) derivatives and compare them with the pharmacologically well-researched α-PVP (α-pyrrolidinovalerophenone). Radiotracer-based in vitro uptake inhibition assays in HEK293 cells show that the investigated α-PPP derivatives inhibit the human high-affinity transporters of dopamine (hDAT) and norepinephrine (hNET) in the low micromolar range, with α-PVP being ten times more potent. Similar to α-PVP, no relevant pharmacological activity was found at the human serotonin transporter (hSERT). Unexpectedly, radiotracer-based in vitro release assays reveal α-PPP, MDPPP and 3Br-PPP, but not α-PVP, to be partial releasing agents at hNET (EC50 values in the low micromolar range). Furthermore, uptake inhibition assays at low-affinity monoamine transporters, i.e., the human organic cation transporters (hOCT) 1-3 and human plasma membrane monoamine transporter (hPMAT), bring to light that all compounds inhibit hOCT1 and 2 (IC50 values in the low micromolar range) while less potently interacting with hPMAT and hOCT3. In conclusion, this study describes (i) three new hybrid compounds that efficaciously block hDAT while being partial releasers at hNET, and (ii) highlights the interactions of α-PPP-derivatives with low-affinity monoamine transporters, giving impetus to further studies investigating the interaction of drugs of abuse with OCT1-3 and PMAT.
Collapse
Affiliation(s)
- Julian Maier
- Medical University of Vienna, Center for Physiology and Pharmacology, Institute of Pharmacology, Währingerstraße 13A, 1090, Vienna, Austria
| | - Laurin Rauter
- Medical University of Vienna, Center for Physiology and Pharmacology, Institute of Pharmacology, Währingerstraße 13A, 1090, Vienna, Austria
| | - Deborah Rudin
- Medical University of Vienna, Center for Physiology and Pharmacology, Institute of Pharmacology, Währingerstraße 13A, 1090, Vienna, Austria
| | - Marco Niello
- Medical University of Vienna, Center for Physiology and Pharmacology, Institute of Pharmacology, Währingerstraße 13A, 1090, Vienna, Austria
| | - Marion Holy
- Medical University of Vienna, Center for Physiology and Pharmacology, Institute of Pharmacology, Währingerstraße 13A, 1090, Vienna, Austria
| | - Diethart Schmid
- Medical University of Vienna, Center for Physiology and Pharmacology, Institute of Physiology, Währingerstraße 13A, 1090, Vienna, Austria
| | - Joseph Wilson
- Research Triangle Institute, Center for Drug Discovery, Research Triangle Park, NC, USA
| | - Bruce E Blough
- Research Triangle Institute, Center for Drug Discovery, Research Triangle Park, NC, USA
| | - Brenda M Gannon
- Mercer University College of Pharmacy, Mercer University Health Sciences Center, Department of Pharmaceutical Sciences, Atlanta, GA, USA; Louisiana State University Health Sciences Center, Shreveport, Department of Pharmacology Toxicology & Neuroscience and Louisiana Addiction Research Center, Shreveport, LA, USA
| | - Kevin S Murnane
- Mercer University College of Pharmacy, Mercer University Health Sciences Center, Department of Pharmaceutical Sciences, Atlanta, GA, USA; Louisiana State University Health Sciences Center, Shreveport, Department of Pharmacology Toxicology & Neuroscience and Louisiana Addiction Research Center, Shreveport, LA, USA
| | - Harald H Sitte
- Medical University of Vienna, Center for Physiology and Pharmacology, Institute of Pharmacology, Währingerstraße 13A, 1090, Vienna, Austria; AddRess Centre for Addiction Research and Science, Medical University of Vienna, Währingerstraße 13A, 1090, Vienna, Austria.
| |
Collapse
|
13
|
Niello M, Cintulová D, Raithmayr P, Holy M, Jäntsch K, Colas C, Ecker GF, Sitte HH, Mihovilovic MD. Effects of Hydroxylated Mephedrone Metabolites on Monoamine Transporter Activity in vitro. Front Pharmacol 2021; 12:654061. [PMID: 33897439 PMCID: PMC8063026 DOI: 10.3389/fphar.2021.654061] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/15/2021] [Accepted: 03/01/2021] [Indexed: 01/11/2023] Open
Abstract
Mephedrone is a largely abused psychostimulant. It elicits the release of monoamines via the high affinity transporters for dopamine (DAT), norepinephrine (NET) and serotonin (SERT). Stereoselective metabolic reactions are involved in the inactivation and the elimination of its chemical structure. However, during these processes, several structures are generated and some of them have been reported to be still pharmacologically active. In this study 1) we have newly synthetized several putative mephedrone metabolites, 2) compared their activity at monoamine transporters, 3) generated quantitative structure activity relationships, and 4) exploited the chemical structure of the putative metabolites to screen a urine sample from a drug user and dissect mephedrone metabolism. We have found that most of the tested metabolites are weak inhibitors of monoamine transporters and that all of them are more potent at DAT and NET in comparison to SERT. The only exception is represented by the COOH-metabolite which shows no pharmacological activity at all three monoamine transporters. The enantioselectivity of mephedrone and its metabolites is present mainly at SERT, with only minor effects at DAT and NET being introduced when the β-keto group is reduced to an OH-group. Importantly, while at DAT the putative metabolites did not show changes in inhibitory potencies, but rather changes in their substrate/blocker profile, at SERT they showed mainly changes in inhibitory potencies. Molecular modeling suggests that the hydrophobic nature of a specific SERT subpocket may be involved in such loss of affinity. Finally, the assessment of the putative metabolites in one urine sample of mephedrone user displayed two previously uncharacterized metabolites, 4-COOH-nor-mephedrone (4-COOH-MC) and dihydro-4- nor-mephedrone (dihydro-4-MC). These results confirm and expand previous studies highlighting the importance of the stereochemistry in the pharmacodynamics of phase-1 metabolites of mephedrone, established their structure-activity relationships at DAT, NET and SERT and pave the way for a systematic dissection of mephedrone metabolic routes. Given the number of structures found having residual and modified pharmacological profiles, these findings may help in understanding the complex subjective effects of administered mephedrone. Moreover, the dissection of mephedrone metabolic routes may help in developing new therapies for treating psychostimulants acute intoxications.
Collapse
Affiliation(s)
- Marco Niello
- Institute of Pharmacology, Medical University, Vienna, Austria
| | | | - Philip Raithmayr
- Department of Pharmaceutical Chemistry, University of Vienna, Vienna, Austria
| | - Marion Holy
- Institute of Pharmacology, Medical University, Vienna, Austria
| | - Kathrin Jäntsch
- Institute of Pharmacology, Medical University, Vienna, Austria
| | - Claire Colas
- Department of Pharmaceutical Chemistry, University of Vienna, Vienna, Austria
| | - Gerhard F. Ecker
- Department of Pharmaceutical Chemistry, University of Vienna, Vienna, Austria
| | - Harald H. Sitte
- Institute of Pharmacology, Medical University, Vienna, Austria
| | | |
Collapse
|
14
|
Jîtcă G, Ősz BE, Tero-Vescan A, Vari CE. Psychoactive Drugs-From Chemical Structure to Oxidative Stress Related to Dopaminergic Neurotransmission. A Review. Antioxidants (Basel) 2021; 10:381. [PMID: 33806320 PMCID: PMC8000782 DOI: 10.3390/antiox10030381] [Citation(s) in RCA: 20] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/22/2021] [Revised: 02/24/2021] [Accepted: 03/01/2021] [Indexed: 12/26/2022] Open
Abstract
Nowadays, more and more young people want to experience illegal, psychoactive substances, without knowing the risks of exposure. Besides affecting social life, psychoactive substances also have an important effect on consumer health. We summarized and analyzed the published literature data with reference to the mechanism of free radical generation and the link between chemical structure and oxidative stress related to dopaminergic neurotransmission. This review presents data on the physicochemical properties, on the ability to cross the blood brain barrier, the chemical structure activity relationship (SAR), and possible mechanisms by which neuronal injuries occur due to oxidative stress as a result of drug abuse such as "bath salts", amphetamines, or cocaine. The mechanisms of action of ingested compounds or their metabolites involve intermediate steps in which free radicals are generated. The brain is strongly affected by the consumption of such substances, facilitating the induction of neurodegenerative diseases. It can be concluded that neurotoxicity is associated with drug abuse. Dependence and oxidative stress are linked to inhibition of neurogenesis and the onset of neuronal death. Understanding the pathological mechanisms following oxidative attack can be a starting point in the development of new therapeutic targets.
Collapse
Affiliation(s)
- George Jîtcă
- Department of Pharmacology and Clinical Pharmacy, Faculty of Pharmacy, George Emil Palade University of Medicine, Pharmacy, Science and Technology of Târgu Mureș, 540142 Târgu Mureș, Romania; (G.J.); (C.E.V.)
| | - Bianca E. Ősz
- Department of Pharmacology and Clinical Pharmacy, Faculty of Pharmacy, George Emil Palade University of Medicine, Pharmacy, Science and Technology of Târgu Mureș, 540142 Târgu Mureș, Romania; (G.J.); (C.E.V.)
| | - Amelia Tero-Vescan
- Department of Biochemistry, Faculty of Pharmacy, George Emil Palade University of Medicine, Pharmacy, Science and Technology of Târgu Mureș, 540142 Târgu Mureș, Romania;
| | - Camil E. Vari
- Department of Pharmacology and Clinical Pharmacy, Faculty of Pharmacy, George Emil Palade University of Medicine, Pharmacy, Science and Technology of Târgu Mureș, 540142 Târgu Mureș, Romania; (G.J.); (C.E.V.)
| |
Collapse
|
15
|
Kovačević S, Banjac MK, Milošević N, Ćurčić J, Marjanović D, Todorović N, Krmar J, Podunavac-Kuzmanović S, Banjac N, Ušćumlić G. Comparative chemometric and quantitative structure-retention relationship analysis of anisotropic lipophilicity of 1-arylsuccinimide derivatives determined in high-performance thin-layer chromatography system with aprotic solvents. J Chromatogr A 2020; 1628:461439. [PMID: 32822979 DOI: 10.1016/j.chroma.2020.461439] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/06/2020] [Revised: 07/26/2020] [Accepted: 07/28/2020] [Indexed: 12/21/2022]
Abstract
Numerous structurally different amides and imides including succinimide derivatives exhibit diverse bioactive potential. The development of new compounds requires rationalization in the design in order to provide structural changes that guarantee favorable physico-chemical properties, pharmacological activity and safety. In the present research, a comprehensive study with comparison of the chromatographic lipophilicity and other physico-chemical properties of five groups of 1-arylsuccinimide derivatives was conducted. The chemometric analysis of their physico-chemical properties was carried out by using unsupervised (hierarchical cluster analysis and principal component analysis) and supervised pattern recognition methods (linear discriminant analysis), while the correlations between the in silico molecular features and chromatographic lipophilicity were examined applying linear and non-linear Quantitative Structure-Retention Relationship (QSRR) approaches. The main aim of the conducted research was to determine similarities and dissimilarities among the studied 1-arylsuccinimides, to point out the molecular features which have significant influence on their lipophilicity, as well as to establish high-quality QSRR models which can be used in prediction of chromatographic lipophilicity of structurally similar 1-arylsuccinimides. This study is a continuation of analysis and determination of the physico-chemical properties of 1-arylsuccinimides which could be important guidelines in further in vitro and eventually in vivo studies of their biological potential.
Collapse
Affiliation(s)
- Strahinja Kovačević
- University of Novi Sad, Faculty of Technology Novi Sad, Bulevar cara Lazara 1, 21000 Novi Sad, Serbia
| | - Milica Karadžić Banjac
- University of Novi Sad, Faculty of Technology Novi Sad, Bulevar cara Lazara 1, 21000 Novi Sad, Serbia.
| | - Nataša Milošević
- University of Novi Sad, Faculty of Medicine, Department of Pharmacy, Hajduk Veljkova 3, 21000 Novi Sad, Serbia
| | - Jelena Ćurčić
- University of Novi Sad, Faculty of Medicine, Department of Pharmacy, Hajduk Veljkova 3, 21000 Novi Sad, Serbia; University Business Academy in Novi Sad, Faculty of Pharmacy Novi Sad, Trg Mladenaca 5, 21000 Novi Sad, Serbia
| | - Dunja Marjanović
- University of Novi Sad, Faculty of Medicine, Department of Pharmacy, Hajduk Veljkova 3, 21000 Novi Sad, Serbia
| | - Nemanja Todorović
- University of Novi Sad, Faculty of Medicine, Department of Pharmacy, Hajduk Veljkova 3, 21000 Novi Sad, Serbia
| | - Jovana Krmar
- University of Belgrade, Faculty of Pharmacy, Department of Drug Analysis, Vojvode Stepe 450, 11221 Belgrade, Serbia
| | | | - Nebojša Banjac
- University of Belgrade, Faculty of Agriculture, 11081 Belgrade-Zemun, Nemanjina 6, Serbia
| | - Gordana Ušćumlić
- University of Belgrade, Faculty of Technology and Metallurgy, Karnegijeva 4, 11000 Belgrade, Serbia
| |
Collapse
|
16
|
Xue W, Fu T, Zheng G, Tu G, Zhang Y, Yang F, Tao L, Yao L, Zhu F. Recent Advances and Challenges of the Drugs Acting on Monoamine Transporters. Curr Med Chem 2020; 27:3830-3876. [DOI: 10.2174/0929867325666181009123218] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/28/2018] [Revised: 09/30/2018] [Accepted: 10/03/2018] [Indexed: 01/06/2023]
Abstract
Background:
The human Monoamine Transporters (hMATs), primarily including hSERT,
hNET and hDAT, are important targets for the treatment of depression and other behavioral disorders
with more than the availability of 30 approved drugs.
Objective:
This paper is to review the recent progress in the binding mode and inhibitory mechanism of
hMATs inhibitors with the central or allosteric binding sites, for the benefit of future hMATs inhibitor
design and discovery. The Structure-Activity Relationship (SAR) and the selectivity for hit/lead compounds
to hMATs that are evaluated by in vitro and in vivo experiments will be highlighted.
Methods:
PubMed and Web of Science databases were searched for protein-ligand interaction, novel
inhibitors design and synthesis studies related to hMATs.
Results:
Literature data indicate that since the first crystal structure determinations of the homologous
bacterial Leucine Transporter (LeuT) complexed with clomipramine, a sizable database of over 100 experimental
structures or computational models has been accumulated that now defines a substantial degree
of structural variability hMATs-ligands recognition. In the meanwhile, a number of novel hMATs
inhibitors have been discovered by medicinal chemistry with significant help from computational models.
Conclusion:
The reported new compounds act on hMATs as well as the structures of the transporters
complexed with diverse ligands by either experiment or computational modeling have shed light on the
poly-pharmacology, multimodal and allosteric regulation of the drugs to transporters. All of the studies
will greatly promote the Structure-Based Drug Design (SBDD) of structurally novel scaffolds with high
activity and selectivity for hMATs.
Collapse
Affiliation(s)
- Weiwei Xue
- Innovative Drug Research and Bioinformatics Group, School of Pharmaceutical Sciences and Chongqing Key Laboratory of Natural Drug Research, Chongqing University, Chongqing 401331, China
| | - Tingting Fu
- Innovative Drug Research and Bioinformatics Group, School of Pharmaceutical Sciences and Chongqing Key Laboratory of Natural Drug Research, Chongqing University, Chongqing 401331, China
| | - Guoxun Zheng
- Innovative Drug Research and Bioinformatics Group, School of Pharmaceutical Sciences and Chongqing Key Laboratory of Natural Drug Research, Chongqing University, Chongqing 401331, China
| | - Gao Tu
- Innovative Drug Research and Bioinformatics Group, School of Pharmaceutical Sciences and Chongqing Key Laboratory of Natural Drug Research, Chongqing University, Chongqing 401331, China
| | - Yang Zhang
- Innovative Drug Research and Bioinformatics Group, School of Pharmaceutical Sciences and Chongqing Key Laboratory of Natural Drug Research, Chongqing University, Chongqing 401331, China
| | - Fengyuan Yang
- Innovative Drug Research and Bioinformatics Group, School of Pharmaceutical Sciences and Chongqing Key Laboratory of Natural Drug Research, Chongqing University, Chongqing 401331, China
| | - Lin Tao
- Key Laboratory of Elemene Class Anti-cancer Chinese Medicine of Zhejiang Province, School of Medicine, Hangzhou Normal University, Hangzhou 310036, China
| | - Lixia Yao
- Department of Health Sciences Research, Mayo Clinic, Rochester, MN 55905, United States
| | - Feng Zhu
- Innovative Drug Research and Bioinformatics Group, School of Pharmaceutical Sciences and Chongqing Key Laboratory of Natural Drug Research, Chongqing University, Chongqing 401331, China
| |
Collapse
|
17
|
Niello M, Cintulova D, Hellsberg E, Jäntsch K, Holy M, Ayatollahi LH, Cozzi NV, Freissmuth M, Sandtner W, Ecker GF, Mihovilovic MD, Sitte HH. para-Trifluoromethyl-methcathinone is an allosteric modulator of the serotonin transporter. Neuropharmacology 2019; 161:107615. [PMID: 31028773 DOI: 10.1016/j.neuropharm.2019.04.021] [Citation(s) in RCA: 23] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/31/2018] [Revised: 04/17/2019] [Accepted: 04/19/2019] [Indexed: 11/15/2022]
Abstract
The transporters for dopamine (DAT) and serotonin (SERT) are important targets in the treatment of psychiatric disorders including major depression, anxiety and attention-deficit hyperactivity disorder. Drugs acting at these transporters can act as inhibitors or as releasers. In addition, it has been recently appreciated that some compounds are less efficacious releasers than amphetamine. Thus, they are classified as partial releasers. Compounds can act on both SERT and DAT or display exquisite selectivity for either SERT or DAT, but the structural basis for selectivity is poorly understood. The trifluoromethyl-substitution of methcathinone in the para-position has been shown to dramatically shift the selectivity of methcathinone (MCAT) towards SERT. Here, we examined MCAT, para-trifluoromethyl-methcathinone (pCF3MCAT) and other analogues to understand (i) the determinants of selectivity and (ii) the effects of the para-CF3-substitution of MCAT on the transport cycle. We systematically tested different para-substituted MCATs by biochemical, computational and electrophysiological approaches: addition of the pCF3group, but not of other substituents with larger van der Waal's volume, lipophilicity or polarity, converted the DAT-selective MCAT into a SERT-selective partial releaser. Electrophysiological and superfusion experiments, together with kinetic modelling, showed that pCF3MCAT, but not MCAT, trapped a fraction of SERTs in an inactive state by occupying the S2-site. These findings define a new mechanism of action for partial releasers, which is distinct from the other two known binding modes underlying partial release. Our observations highlight the fact that the substrate permeation pathway of monoamine transporters supports multiple binding modes, which can be exploited for drug design. This article is part of the issue entitled 'Special Issue on Neurotransmitter Transporters'.
Collapse
Affiliation(s)
- Marco Niello
- Institute of Pharmacology, Medical University, Vienna, Austria
| | | | - Eva Hellsberg
- Department of Pharmaceutical Chemistry, University of Vienna, Vienna, Austria
| | - Kathrin Jäntsch
- Institute of Pharmacology, Medical University, Vienna, Austria
| | - Marion Holy
- Institute of Pharmacology, Medical University, Vienna, Austria
| | | | - Nicholas V Cozzi
- Department of Cell and Regenerative Biology, University of Wisconsin School of Medicine and Public Health, Madison, USA
| | | | - Walter Sandtner
- Institute of Pharmacology, Medical University, Vienna, Austria
| | - Gerhard F Ecker
- Department of Pharmaceutical Chemistry, University of Vienna, Vienna, Austria
| | | | - Harald H Sitte
- Institute of Pharmacology, Medical University, Vienna, Austria.
| |
Collapse
|
18
|
Steele TWE, Eltit JM. Using Ca 2+-channel biosensors to profile amphetamines and cathinones at monoamine transporters: electro-engineering cells to detect potential new psychoactive substances. Psychopharmacology (Berl) 2019; 236:973-988. [PMID: 30448989 PMCID: PMC6525079 DOI: 10.1007/s00213-018-5103-5] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/07/2018] [Accepted: 11/02/2018] [Indexed: 01/20/2023]
Abstract
BACKGROUND The appearance of stimulant-class new psychoactive substances (NPS) is a frequent and significant problem in our society. Cathinone variants are often sold illegally as 3,4-methylenedioxy methamphetamine ("ecstasy") or disguised for legal sale using misleading names such as "bath salts" and carry the risk of promoting disruptive mental states, addiction, and fatal overdose. The principal targets of these recreational drugs are monoamine transporters expressed in catecholaminergic and serotonergic neurons. Some transporter ligands can be transported into cells, where they can promote a massive release of neurotransmitters through reverse transport, and others can block uptake. A ligand's dopamine vs. serotonin transporter selectivity, potency, and activity as a substrate or blocker can help elucidate the abuse liability and subjective effects of a drug. OBJECTIVES Here, we describe the discovery, development, and validation of an emerging methodology for compound activity assessment at monoamine transporters. KEY FINDINGS Substrates generate inward electrical currents through transporters and can depolarize the plasma membrane, whereas blockers work as a "cork in a bottle" and function as antagonists. Voltage-gated Ca2+ channels were co-expressed with monoamine transporters in cultured cells and used to measure fluctuations of the membrane electrical potential. In this system, substrates of monoamine transporters produce reliable dose-dependent Ca2+ signals, while blockers hinder them. DISCUSSION This system constitutes a novel use of voltage-gated Ca2+ channels as biosensors for the purpose of characterizing ligand activity at monoamine transporters using fluorimetry. This approach in combination with in vivo evaluations of drugs' abuse-related effects is a powerful strategy for anticipating potential stimulant-class NPS.
Collapse
Affiliation(s)
- Tyler W E Steele
- Department of Physiology and Biophysics, School of Medicine, Virginia Commonwealth University, 1101 E Marshall St. Rm# 3-038H, Richmond, VA, 23298, USA
| | - Jose M Eltit
- Department of Physiology and Biophysics, School of Medicine, Virginia Commonwealth University, 1101 E Marshall St. Rm# 3-038H, Richmond, VA, 23298, USA.
| |
Collapse
|
19
|
Structure-activity relationships of bath salt components: substituted cathinones and benzofurans at biogenic amine transporters. Psychopharmacology (Berl) 2019; 236:939-952. [PMID: 30397775 PMCID: PMC6500773 DOI: 10.1007/s00213-018-5059-5] [Citation(s) in RCA: 45] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/30/2018] [Accepted: 10/02/2018] [Indexed: 01/05/2023]
Abstract
RATIONALE New psychoactive substances (NPSs), including substituted cathinones and other stimulants, are synthesized, sold on the Internet, and ingested without knowledge of their pharmacological activity and/or toxicity. In vitro pharmacology plays a role in therapeutic drug development, drug-protein in silico interaction modeling, and drug scheduling. OBJECTIVES The goal of this research was to determine mechanisms of action that may indicate NPS abuse liability. METHODS Affinities to displace the radioligand [125I]RTI-55 and potencies to inhibit [3H]neurotransmitter uptake for 22 cathinones, 6 benzofurans and another stimulant were characterized using human embryonic kidney cells stably expressing recombinant human transporters for dopamine, norepinephrine, or serotonin (hDAT, hNET, or hSERT, respectively). Selected compounds were tested for potencies and efficacies at inducing [3H]neurotransmitter release via the transporters. Computational modeling was conducted to explain plausible molecular interactions established by NPS and transporters. RESULTS Most α-pyrrolidinophenones had high hDAT potencies and selectivities in uptake assays, with hDAT/hSERT uptake selectivity ratios of 83-360. Other substituted cathinones varied in their potencies and selectivities, with N-ethyl-hexedrone and N-ethyl-pentylone having highest hDAT potencies and N-propyl-pentedrone having highest hDAT selectivity. 4-Cl-ethcathinone and 3,4-methylenedioxy-N-propylcathinone had higher hSERT selectivity. Benzofurans generally had low hDAT selectivity, especially 1-(2,3-dihydrobenzofuran-5-yl)-N-methylpropan-2-amine, with 25-fold higher hSERT potency. Consistent with this selectivity, the benzofurans were releasers at hSERT. Modeling indicated key amino acids in the transporters' binding pockets that influence drug affinities. CONCLUSIONS The α-pyrrolidinophenones, with high hDAT selectivity, have high abuse potential. Lower hDAT selectivity among benzofurans suggests similarity to methylenedioxymethamphetamine, entactogens with lower stimulant activity.
Collapse
|
20
|
Substrate and inhibitor binding to the serotonin transporter: Insights from computational, crystallographic, and functional studies. Neuropharmacology 2019; 161:107548. [PMID: 30807752 DOI: 10.1016/j.neuropharm.2019.02.030] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/14/2018] [Revised: 02/19/2019] [Accepted: 02/22/2019] [Indexed: 02/02/2023]
Abstract
The serotonin transporter (SERT) belongs to the monoamine transporter family, which also includes the dopamine and norepinephrine transporters. SERT is essential for regulating serotonergic signaling by the reuptake of serotonin from the synaptic cleft back into the presynaptic neuron. Dysregulation of SERT has been implicated in several major psychiatric disorders such as major depressive disorder (MDD). MDD was among the top five leading causes of years lived with disease in 2016 and is characterized as a major global burden. Several drugs have been developed to target SERT for use in the treatment of MDD, and their respective binding modes and locations within SERT have been studied. The elucidation of the first structure of a bacterial SERT homologue in 2005 has accelerated crystallographic, computational, and functional studies to further elucidate drug binding and method of action in SERT. Herein, we aim to highlight and compare these studies with an emphasis on what the different experimental methods conclude on substrate and inhibitor binding modes, and the potential caveats of using the different types of studies are discussed. We focus this review on the binding of cognate substrate and drugs belonging to the different families of antidepressants, including tricyclic antidepressants, selective serotonin reuptake inhibitors, serotonin-norepinephrine reuptake inhibitors, and multimodal drugs, as well as illicit drugs such as cocaine, amphetamines, and ibogaine. This article is part of the issue entitled 'Special Issue on Neurotransmitter Transporters'.
Collapse
|
21
|
Walther D, Shalabi AR, Baumann MH, Glennon RA. Systematic Structure-Activity Studies on Selected 2-, 3-, and 4-Monosubstituted Synthetic Methcathinone Analogs as Monoamine Transporter Releasing Agents. ACS Chem Neurosci 2019; 10:740-745. [PMID: 30354055 DOI: 10.1021/acschemneuro.8b00524] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/29/2022] Open
Abstract
Methcathinone analogs are appearing on the clandestine market at a rate nearly out-pacing the ability of investigators to examine them on an individual basis. To formulate structure-activity relationship (SAR) generalities, we examined the releasing ability of several simple methcathinone analogs at the three monoamine transporters (i.e., the dopamine, norepinephrine, and serotonin transporters, DAT, NET, and SERT, respectively) using in vitro assay methods. The analogs included methcathinone and 14 other compounds monosubstituted at the 2-, 3-, or 4-position. In general, (a) the 2-substituted analogs were less potent than either the 3- or 4-substituted analogs, (b) the 3- and 4-substituted analogs were relatively similar in potency, (c) methcathinone was the most selective as a DAT-releasing agent, and (d) the 3- and 4-CF3 analogs were the least DAT-selective. For the 15 compounds, there was a significant correlation ( r > 0.9) between DAT and NET potency, suggesting relatively similar structure-activity relationships (at least for the compounds examined here). Several of the compounds have appeared on the clandestine market since our studies were initiated, and the present results provide new information on how they might act.
Collapse
Affiliation(s)
- Donna Walther
- Designer Drug Research Unit, Intramural Research Program, National Institute on Drug Abuse, National Institutes of Health, Baltimore, Maryland 21224, United States
| | - Abdelrahman R. Shalabi
- Department of Medicinal Chemistry, School of Pharmacy, Virginia Commonwealth University, Richmond, Virginia 23298, United States
| | - Michael H. Baumann
- Designer Drug Research Unit, Intramural Research Program, National Institute on Drug Abuse, National Institutes of Health, Baltimore, Maryland 21224, United States
| | - Richard A. Glennon
- Department of Medicinal Chemistry, School of Pharmacy, Virginia Commonwealth University, Richmond, Virginia 23298, United States
| |
Collapse
|
22
|
McLaughlin G, Baumann MH, Kavanagh PV, Morris N, Power JD, Dowling G, Twamley B, O’Brien J, Hessman G, Westphal F, Walther D, Brandt SD. Synthesis, analytical characterization, and monoamine transporter activity of the new psychoactive substance 4-methylphenmetrazine (4-MPM), with differentiation from its ortho- and meta- positional isomers. Drug Test Anal 2018; 10:1404-1416. [PMID: 29673128 PMCID: PMC7316143 DOI: 10.1002/dta.2396] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/28/2018] [Revised: 04/12/2018] [Accepted: 04/13/2018] [Indexed: 12/21/2022]
Abstract
The availability of new psychoactive substances (NPS) on the recreational drug market continues to create challenges for scientists in the forensic, clinical and toxicology fields. Phenmetrazine (3-methyl-2-phenylmorpholine) and an array of its analogs form a class of psychostimulants that are well documented in the patent and scientific literature. The present study reports on two phenmetrazine analogs that have been encountered on the NPS market following the introduction of 3-fluorophenmetrazine (3-FPM), namely 4-methylphenmetrazine (4-MPM), and 3-methylphenmetrazine (3-MPM). This study describes the syntheses, analytical characterization, and pharmacological evaluation of the positional isomers of MPM. Analytical characterizations employed various chromatographic, spectroscopic, and mass spectrometric platforms. Pharmacological studies were conducted to assess whether MPM isomers might display stimulant-like effects similar to the parent compound phenmetrazine. The isomers were tested for their ability to inhibit uptake or stimulate release of tritiated substrates at dopamine, norepinephrine and serotonin transporters using in vitro transporter assays in rat brain synaptosomes. The analytical characterization of three vendor samples revealed the presence of 4-MPM in two of the samples and 3-MPM in the third sample, which agreed with the product label. The pharmacological findings suggest that 2-MPM and 3-MPM will exhibit stimulant properties similar to the parent compound phenmetrazine, whereas 4-MPM may display entactogen properties more similar to 3,4-methylenedioxymethamphetamine (MDMA). The combination of test purchases, analytical characterization, targeted organic synthesis, and pharmacological evaluation of NPS and their isomers is an effective approach for the provision of data on these substances as they emerge in the marketplace.
Collapse
Affiliation(s)
- Gavin McLaughlin
- Department of Pharmacology and Therapeutics, School of Medicine, Trinity Centre for Health Sciences, St. James’s Hospital, James’s Street, Dublin 8, D08W9RT, Ireland
- Forensic Science Ireland, Garda Headquarters, Phoenix Park, Dublin 8, D08HN3X, Ireland
| | - Michael H. Baumann
- Designer Drug Research Unit, Intramural Research Program, National Institute on Drug Abuse, National Institutes of Health, 333 Cassell Drive, Suite 4400, Baltimore, MD 21224, USA
| | - Pierce V. Kavanagh
- Department of Pharmacology and Therapeutics, School of Medicine, Trinity Centre for Health Sciences, St. James’s Hospital, James’s Street, Dublin 8, D08W9RT, Ireland
| | - Noreen Morris
- Department of Life & Physical Sciences, Faculty of Science and Health, Athlone Institute of Technology, Dublin Road, Athlone, Co. Westmeath, N37HD68, Ireland
| | - John D. Power
- Department of Pharmacology and Therapeutics, School of Medicine, Trinity Centre for Health Sciences, St. James’s Hospital, James’s Street, Dublin 8, D08W9RT, Ireland
- Forensic Science Ireland, Garda Headquarters, Phoenix Park, Dublin 8, D08HN3X, Ireland
| | - Geraldine Dowling
- Department of Pharmacology and Therapeutics, School of Medicine, Trinity Centre for Health Sciences, St. James’s Hospital, James’s Street, Dublin 8, D08W9RT, Ireland
- Department of Life Sciences, School of Science, Sligo Institute of Technology, Ash Lane, Sligo, F91YW50, Ireland
| | - Brendan Twamley
- School of Chemistry, Trinity College Dublin, College Green, Dublin 2, D02EV57, Ireland
| | - John O’Brien
- School of Chemistry, Trinity College Dublin, College Green, Dublin 2, D02EV57, Ireland
| | - Gary Hessman
- School of Chemistry, Trinity College Dublin, College Green, Dublin 2, D02EV57, Ireland
| | - Folker Westphal
- State Bureau of Criminal Investigation Schleswig-Holstein, Section Narcotics/Toxicology, Mühlenweg 166, D-24116 Kiel, Germany
| | - Donna Walther
- Designer Drug Research Unit, Intramural Research Program, National Institute on Drug Abuse, National Institutes of Health, 333 Cassell Drive, Suite 4400, Baltimore, MD 21224, USA
| | - Simon D. Brandt
- School of Pharmacy and Biomolecular Sciences, Liverpool John Moores University, Byrom Street, Liverpool L3 3AF, UK
| |
Collapse
|
23
|
Battisti UM, Sitta R, Harris A, Sakloth F, Walther D, Ruchala I, Negus SS, Baumann MH, Glennon RA, Eltit JM. Effects of N-Alkyl-4-Methylamphetamine Optical Isomers on Plasma Membrane Monoamine Transporters and Abuse-Related Behavior. ACS Chem Neurosci 2018; 9:1829-1839. [PMID: 29697951 PMCID: PMC6051915 DOI: 10.1021/acschemneuro.8b00138] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022] Open
Abstract
4-Methylamphetamine (4-MA) is an emerging drug of abuse that acts as a substrate at plasma membrane transporters for dopamine (DAT), norepinephrine (NET), and serotonin (SERT), thereby causing nonexocytotic release of monoamine transmitters via reverse transport. Prior studies by us showed that increasing the N-alkyl chain length of N-substituted 4-MA analogues converts 4-MA from a transportable substrate (i.e., releaser) at DAT and NET to a nontransported blocker at these sites. Here, we studied the effects of the individual optical isomers of N-methyl-, N-ethyl-, and N- n-propyl 4-MA on monoamine transporters and abuse-related behavior in rats because action/function might be related to stereochemistry. Uptake inhibition and release assays were conducted in rat brain synaptosomes whereas electrophysiological assessments of drug-transporter interactions were examined using cell-based biosensors. Intracranial-self-stimulation in rats was employed to assess abuse potential in vivo. The experimental evidence demonstrates that S(+) N-methyl 4-MA is a potent and efficacious releaser at DAT, NET, and SERT with the highest abuse potential among the test drugs, whereas R(-) N-methyl 4-MA is a less potent releaser with reduced abuse potential. The S(+)ethyl analogue has decreased efficacy as a releaser at DAT but retains full release activity at NET and SERT with a reduction in abuse-related effects; the R(-)ethyl analogue has a similar profile but is less potent. S(+) N-Propyl 4-MA is a nontransported blocker at DAT and NET but an efficacious releaser at SERT, whereas the R enantiomer is almost inactive. In conclusion, the S enantiomers of the N-alkyl 4-MA analogues are most potent. Lengthening the N-alkyl chain converts compounds from potent nonselective releasers showing abuse-related effects to more selective SERT releasers with no apparent abuse potential.
Collapse
Affiliation(s)
- Umberto M Battisti
- Department of Medicinal Chemistry, School of Pharmacy, Box 980540 , Virginia Commonwealth University , Richmond , Virginia 23298 , United States
| | - Ramsey Sitta
- Department of Physiology and Biophysics, School of Medicine , Virginia Commonwealth University , Richmond , Virginia 23298 United States
| | - Alan Harris
- Department of Physiology and Biophysics, School of Medicine , Virginia Commonwealth University , Richmond , Virginia 23298 United States
| | - Farhana Sakloth
- Department of Pharmacology and Toxicology, School of Medicine , Virginia Commonwealth University , Richmond , Virginia 23298 , United States
| | - Donna Walther
- Designer Drug Research Unit, Intramural Research Program , National Institute on Drug Abuse, National Institutes of Health , Baltimore , Maryland 21224 , United States
| | - Iwona Ruchala
- Department of Physiology and Biophysics, School of Medicine , Virginia Commonwealth University , Richmond , Virginia 23298 United States
| | - S Stevens Negus
- Department of Pharmacology and Toxicology, School of Medicine , Virginia Commonwealth University , Richmond , Virginia 23298 , United States
| | - Michael H Baumann
- Designer Drug Research Unit, Intramural Research Program , National Institute on Drug Abuse, National Institutes of Health , Baltimore , Maryland 21224 , United States
| | - Richard A Glennon
- Department of Medicinal Chemistry, School of Pharmacy, Box 980540 , Virginia Commonwealth University , Richmond , Virginia 23298 , United States
| | - Jose M Eltit
- Department of Physiology and Biophysics, School of Medicine , Virginia Commonwealth University , Richmond , Virginia 23298 United States
| |
Collapse
|
24
|
Maier J, Mayer FP, Luethi D, Holy M, Jäntsch K, Reither H, Hirtler L, Hoener MC, Liechti ME, Pifl C, Brandt SD, Sitte HH. The psychostimulant (±)-cis-4,4'-dimethylaminorex (4,4'-DMAR) interacts with human plasmalemmal and vesicular monoamine transporters. Neuropharmacology 2018; 138:282-291. [PMID: 29908239 DOI: 10.1016/j.neuropharm.2018.06.018] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/26/2018] [Revised: 05/01/2018] [Accepted: 06/12/2018] [Indexed: 10/28/2022]
Abstract
(±)-cis-4,4'-Dimethylaminorex (4,4'-DMAR) is a new psychoactive substance (NPS) that has been associated with 31 fatalities and other adverse events in Europe between June 2013 and February 2014. We used in vitro uptake inhibition and transporter release assays to determine the effects of 4,4'-DMAR on human high-affinity transporters for dopamine (DAT), norepinephrine (NET) and serotonin (SERT). In addition, we assessed its binding affinities to monoamine receptors and transporters. Furthermore, we investigated the interaction of 4,4'-DMAR with the vesicular monoamine transporter 2 (VMAT2) in rat phaeochromocytoma (PC12) cells and synaptic vesicles prepared from human striatum. 4,4'-DMAR inhibited uptake mediated by human DAT, NET or SERT, respectively in the low micromolar range (IC50 values < 2 μM). Release assays identified 4,4'-DMAR as a substrate type releaser, capable of inducing transporter-mediated reverse transport via DAT, NET and SERT. Furthermore, 4,4'-DMAR inhibited both the rat and human isoforms of VMAT2 at a potency similar to 3,4-methylenedioxymethylamphetamine (MDMA). This study identified 4,4'-DMAR as a potent non-selective monoamine releasing agent. In contrast to the known effects of aminorex and 4-methylaminorex, 4,4'-DMAR exerts profound effects on human SERT. The latter finding is consistent with the idea that fatalities associated with its abuse may be linked to monoaminergic toxicity including serotonin syndrome. The activity at VMAT2 suggests that chronic abuse of 4,4'-DMAR may result in long-term neurotoxicity.
Collapse
Affiliation(s)
- Julian Maier
- Medical University of Vienna, Center for Physiology and Pharmacology, Institute of Pharmacology, Währingerstraße 13A, 1090, Vienna, Austria
| | - Felix P Mayer
- Medical University of Vienna, Center for Physiology and Pharmacology, Institute of Pharmacology, Währingerstraße 13A, 1090, Vienna, Austria
| | - Dino Luethi
- University Hospital Basel and University of Basel, Division of Clinical Pharmacology and Toxicology, Department of Biomedicine, Hebelstraße 20, 4031, Basel, Switzerland
| | - Marion Holy
- Medical University of Vienna, Center for Physiology and Pharmacology, Institute of Pharmacology, Währingerstraße 13A, 1090, Vienna, Austria
| | - Kathrin Jäntsch
- Medical University of Vienna, Center for Physiology and Pharmacology, Institute of Pharmacology, Währingerstraße 13A, 1090, Vienna, Austria
| | - Harald Reither
- Medical University of Vienna, Center for Brain Research, Department of Molecular Neurosciences, Spitalgasse 4, 1090, Vienna, Austria
| | - Lena Hirtler
- Medical University of Vienna, Center for Anatomy and Cell Biology, Währingerstraße 13, 1090, Vienna, Austria
| | - Marius C Hoener
- F. Hoffmann - La Roche Ltd., pRED, Roche Innovation Center Basel, Neuroscience Research, Department of Neurosymptomatic Domains, Grenzacherstraße 124, 4070, Basel, Switzerland
| | - Matthias E Liechti
- University Hospital Basel and University of Basel, Division of Clinical Pharmacology and Toxicology, Department of Biomedicine, Hebelstraße 20, 4031, Basel, Switzerland
| | - Christian Pifl
- Medical University of Vienna, Center for Brain Research, Department of Molecular Neurosciences, Spitalgasse 4, 1090, Vienna, Austria
| | - Simon D Brandt
- School of Pharmacy and Biomolecular Sciences, Liverpool John Moores University, Byrom Street, Liverpool, L3 3AF, UK
| | - Harald H Sitte
- Medical University of Vienna, Center for Physiology and Pharmacology, Institute of Pharmacology, Währingerstraße 13A, 1090, Vienna, Austria; Center for Addiction Research and Science, Medical University Vienna, Waehringerstrasse 13A, 1090 Vienna, Austria.
| |
Collapse
|
25
|
Abstract
Synthetic cathinones are derivatives of the naturally occurring compound cathinone, the main psychoactive ingredient in the khat plant Catha edulis. Cathinone is the β-keto analog of amphetamine, and all synthetic cathinones display a β-keto moiety in their structure. Several synthetic cathinones are widely prescribed medications (e.g., bupropion, Wellbutrin®), while others are problematic drugs of abuse (e.g., 4-methylmethcathinone, mephedrone). Similar to amphetamines, synthetic cathinones are psychomotor stimulants that exert their effects by impairing the normal function of plasma membrane transporters for dopamine (DAT), norepinephrine (NET), and 5-HT (SERT). Ring-substituted cathinones like mephedrone are transporter substrates that evoke neurotransmitter release by reversing the normal direction of transporter flux (i.e., releasers), whereas pyrrolidine-containing cathinones like 3,4-methylenedioxypyrovalerone (MDPV) are potent transporter inhibitors that block neurotransmitter uptake (i.e., blockers). Regardless of molecular mechanism, all synthetic cathinones increase extracellular monoamine concentrations in the brain, thereby enhancing cell-to-cell monoamine signaling. Here, we briefly review the mechanisms of action, structure-activity relationships, and in vivo pharmacology of synthetic cathinones. Overall, the findings show that certain synthetic cathinones are powerful drugs of abuse that could pose significant risk to users.
Collapse
|
26
|
Smith DA, Negus SS, Poklis JL, Blough BE, Banks ML. Cocaine-like discriminative stimulus effects of alpha-pyrrolidinovalerophenone, methcathinone and their 3,4-methylenedioxy or 4-methyl analogs in rhesus monkeys. Addict Biol 2017; 22:1169-1178. [PMID: 27060605 DOI: 10.1111/adb.12399] [Citation(s) in RCA: 28] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/03/2015] [Revised: 02/25/2016] [Accepted: 03/01/2016] [Indexed: 01/20/2023]
Abstract
Synthetic cathinones are beta-ketone amphetamine analogs that have emerged as a heterogeneous class of abused compounds that function as either monoamine transporter substrates or inhibitors. Pre-clinical drug discrimination procedures are useful for interrogating structure-activity relationships of abuse-related drug effects; however, in vivo structure-activity relationship comparisons between synthetic cathinones with different mechanisms of action are lacking. The aim of the present study was to determine whether the cocaine-like discriminative stimulus effects of the monoamine transporter inhibitor alpha-pyrrolidinovalerophenone (alpha-PVP) and the monoamine transporter substrate methcathinone were differentially sensitive to 3,4-methylenedioxy and 4-methyl substitutions. Male rhesus monkeys (n = 4) were trained to discriminate intramuscular cocaine (0.32 mg/kg) from saline in a two-key food-reinforced discrimination procedure. Potency and timecourse of cocaine-like discriminative stimulus effects were determined for (±)-alpha-PVP, (±)-methcathinone and their 3,4-methylenedioxy or 4-methyl analogs. Alpha-PVP and methcathinone produced dose- and time-dependent cocaine-like effects. A 3,4-methylenedioxy addition to either alpha-PVP or methcathinone (methylone) did not alter the potency or efficacy to produce cocaine-like effects, but did prolong the time course. A 4-methyl addition to alpha-PVP (pyrovalerone) did not alter the potency or efficacy to produce cocaine-like effects, but did prolong the time course. In contrast, addition of a 4-methyl moiety to methcathinone (4MMC; mephedrone) significantly attenuated efficacy to produce cocaine-like effects. Overall, these results suggest different structural requirements for cocaine-like discriminative stimulus effects of monoamine transporter inhibitor and substrate synthetic cathinone analogs. Given that 4MMC is more hydrophobic than MDMC, these results suggest that hydrophobicity may be an important determinant for limiting monoamine transporter substrate abuse-related behavioral effects.
Collapse
Affiliation(s)
- Douglas A. Smith
- Department of Pharmacology and Toxicology; Virginia Commonwealth University; Richmond VA USA 23298
| | - S. Stevens Negus
- Department of Pharmacology and Toxicology; Virginia Commonwealth University; Richmond VA USA 23298
- Institute for Drug and Alcohol Studies; Virginia Commonwealth University; Richmond VA USA 23298
| | - Justin L. Poklis
- Department of Pharmacology and Toxicology; Virginia Commonwealth University; Richmond VA USA 23298
| | - Bruce E. Blough
- Center for Drug Discovery; Research Triangle Institute; Research Triangle Park NC USA 27709
| | - Matthew L. Banks
- Department of Pharmacology and Toxicology; Virginia Commonwealth University; Richmond VA USA 23298
- Institute for Drug and Alcohol Studies; Virginia Commonwealth University; Richmond VA USA 23298
| |
Collapse
|
27
|
Abstract
The present review briefly explores the neurotoxic properties of methcathinone, mephedrone, methylone, and methylenedioxypyrovalerone (MDPV), four synthetic cathinones most commonly found in "bath salts." Cathinones are β-keto analogs of the commonly abused amphetamines and display pharmacological effects resembling cocaine and amphetamines, but despite their commonalities in chemical structures, synthetic cathinones possess distinct neuropharmacological profiles and produce unique effects. Among the similarities of synthetic cathinones with their non-keto analogs are their targeting of monoamine systems, the release of neurotransmitters, and their stimulant properties. Most of the literature on synthetic cathinones has focused on describing their properties as psychostimulants, their behavioral effects on locomotion, memory, and potential for abuse, whereas descriptions of their neurotoxic properties are not abundant. The biochemical gauges of neurotoxicity induced by non-keto analogs are well studied in humans and experimental animals and include their ability to induce neuroinflammation, oxidative stress, excitotoxicity, temperature alterations as well as dysregulation of neurotransmitter systems and induce changes in monoamine transporters and receptors. These neurotoxicity gauges will serve as parameters to discuss the effects of the four previously mentioned synthetic cathinones alone or in combination with either another cathinone or with some of their non-keto analogs. Bath salts are not a defined combination of drugs and may consist of one synthetic cathinone compound or combinations of more cathinones. Furthermore, this review also presents some of the mechanisms that are thought to underlie this toxicity. A better understanding of the cellular and molecular mechanisms involved in the synthetic cathinones-induced neurotoxicity should contribute to generate modern therapeutic approaches to prevent or attenuate the adverse consequences of use of these drugs in humans.
Collapse
Affiliation(s)
- Mariana Angoa-Pérez
- Research & Development Service, John D. Dingell VA Medical Center, Detroit, MI, 48201, USA.
- Department of Psychiatry and Behavioral Neurosciences, Wayne State University School of Medicine, Detroit, MI, 48201, USA.
| | - John H Anneken
- Research & Development Service, John D. Dingell VA Medical Center, Detroit, MI, 48201, USA
- Department of Psychiatry and Behavioral Neurosciences, Wayne State University School of Medicine, Detroit, MI, 48201, USA
| | - Donald M Kuhn
- Research & Development Service, John D. Dingell VA Medical Center, Detroit, MI, 48201, USA
- Department of Psychiatry and Behavioral Neurosciences, Wayne State University School of Medicine, Detroit, MI, 48201, USA
| |
Collapse
|
28
|
Glennon RA, Dukat M. Synthetic Cathinones: A Brief Overview of Overviews with Applications to the Forensic Sciences. ANNALS OF FORENSIC RESEARCH AND ANALYSIS 2017; 4:1040. [PMID: 30288398 PMCID: PMC6168209] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Subscribe] [Scholar Register] [Indexed: 06/08/2023]
Abstract
Catha edulis, the fresh leaves of which (i.e., khat) are used for their central stimulant actions, has been known for many hundreds of years. S(-)Cathinone was identified as a centrally-active khat constituents >30 years ago. Although khat use was a problem long localized to certain Middle Eastern and certain Eastern African nations, 'synthetic cathinones' (synthetic analogs of cathinone) represent a "new" class of abused substances with growing worldwide appeal. To date, about 150 synthetic cathinones have been identified on the clandestine market, and only a dozen or so have been controlled (U.S. Schedule I). Because these agents do not represent a pharmacologically (i.e., behavioral) or mechanistically homogeneous class of agents, synthetic cathinones are being investigated one agent at a time to understand their actions, mechanisms of action, metabolism, toxicity, and abuse potential - the latter to identify possible modes of overdose treatment and for Scheduling purposes. The available agents might represent only the 'tip of the iceberg'; the potential for many more new synthetic cathinones is very real. Investigation of these agents on an agent-by-agent basis is a daunting task. Attempts are being made to understand these agents as a class, by examination of their structure-activity relationships. Here, we provide an overview of review articles that attempts to shed light on these agents as a class, rather than on an agent-by-agent basis. This article is meant to be a reference resource that might expedite the work of those in this field by directing them to where they can find useful information.
Collapse
Affiliation(s)
- Richard A Glennon
- Department of Medicinal Chemistry, School of Pharmacy, Virginia Commonwealth University, Richmond, Virginia 23298 USA
| | - Małgorzata Dukat
- Department of Medicinal Chemistry, School of Pharmacy, Virginia Commonwealth University, Richmond, Virginia 23298 USA
| |
Collapse
|
29
|
Glennon RA. The 2014 Philip S. Portoghese Medicinal Chemistry Lectureship: The "Phenylalkylaminome" with a Focus on Selected Drugs of Abuse. J Med Chem 2017; 60:2605-2628. [PMID: 28244748 DOI: 10.1021/acs.jmedchem.7b00085] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
The phenylalkylamine, particularly the phenylethylamine, moiety is a common structural feature found embedded in many clinically approved agents. Greater still is its occurrence in drugs of abuse. The simplest phenylethylamine, 2-phenylethylamine itself, is without significant central action when administered at moderate doses, but fairly simple structural modifications profoundly impact its pharmacology and result in large numbers of useful pharmacological tools, agents with therapeutic potential, and in drugs of abuse (e.g., hallucinogens, central stimulants, empathogens), the latter of which are the primary focus here. In vivo drug discrimination techniques and in vitro receptor/transporter methods have been applied to understand the actions of these phenylalkylamines and their mechanisms of action. Thus far, depending upon pendent substituents, certain receptors (e.g., serotonin receptors) and monoamine transporters (i.e., serotonin, dopamine, and norepinephrine transporters) have been implicated as playing major roles in the actions of these abused agents in a complex and, at times, interwoven manner.
Collapse
Affiliation(s)
- Richard A Glennon
- Department of Medicinal Chemistry, School of Pharmacy, Virginia Commonwealth University , Richmond, Virginia 23298, United States
| |
Collapse
|
30
|
Abstract
Until recently, there was rather little interest in the structure-activity relationships (SARs) of cathinone analogs because so few agents were available and because they represented a relatively minor drug abuse problem. Most of the early SAR was formulated on the basis of behavioral (e.g., locomotor and drug discrimination) studies using rodents. With the emergence on the clandestine market in the last few years of a large number of new cathinone analogs, termed "synthetic cathinones", and the realization that they likely act at dopamine, norepinephrine, and/or serotonin transporters as releasing agents (i.e., as substrates) or reuptake inhibitors (i.e., as transport blockers), it has now become possible to better examine their SAR and even their quantitative SAR (QSAR), in a more effective and systematic manner. An SAR picture is beginning to emerge, and key structural features, such as the nature of the terminal amine, the size of the α-substituent, stereochemistry, and the presence and position of aromatic substituents, are being found to impact action (i.e., as releasing agents or reuptake inhibitors) and transporter selectivity.
Collapse
Affiliation(s)
- Richard A Glennon
- Department of Medicinal Chemistry, School of Pharmacy, Virginia Commonwealth University, Richmond, VA, 23298, USA.
| | - Małgorzata Dukat
- Department of Medicinal Chemistry, School of Pharmacy, Virginia Commonwealth University, Richmond, VA, 23298, USA
| |
Collapse
|
31
|
Negus SS, Banks ML. Decoding the Structure of Abuse Potential for New Psychoactive Substances: Structure-Activity Relationships for Abuse-Related Effects of 4-Substituted Methcathinone Analogs. Curr Top Behav Neurosci 2017; 32:119-131. [PMID: 27696217 PMCID: PMC5425248 DOI: 10.1007/7854_2016_18] [Citation(s) in RCA: 30] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
Many cathinone analogs act as substrates or inhibitors at dopamine, norepinephrine, and serotonin transporters (DAT, NET, SERT, respectively). Drug selectivity at DAT vs. SERT is a key determinant of abuse potential for monoamine transporter substrates and inhibitors, such that potency at DAT > SERT is associated with high abuse potential, whereas potency at DAT < SERT is associated with low abuse potential. Quantitative structure-activity relationship (QSAR) studies with a series of 4-substituted methcathinone analogs identified volume of the 4-position substituent on the methcathinone phenyl ring as one structural determinant of both DAT vs. SERT selectivity and abuse-related behavioral effects in an intracranial self-stimulation procedure in rats. Subsequent modeling studies implicated specific amino acids in DAT and SERT that might interact with 4-substituent volume to determine effects produced by this series of cathinone analogs. These studies illustrate use of QSAR analysis to investigate pharmacology of cathinones and function of monoamine transporters.
Collapse
Affiliation(s)
- S Stevens Negus
- Department of Pharmacology and Toxicology, Virginia Commonwealth University, Richmond, VA, USA.
| | - Matthew L Banks
- Department of Pharmacology and Toxicology, Virginia Commonwealth University, Richmond, VA, USA
| |
Collapse
|
32
|
Smith DA, Blough BE, Banks ML. Cocaine-like discriminative stimulus effects of amphetamine, cathinone, methamphetamine, and their 3,4-methylenedioxy analogs in male rhesus monkeys. Psychopharmacology (Berl) 2017; 234:117-127. [PMID: 27709249 PMCID: PMC5203958 DOI: 10.1007/s00213-016-4444-1] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/26/2016] [Accepted: 09/17/2016] [Indexed: 10/20/2022]
Abstract
RATIONALE Synthetic cathinones have emerged as the newest class of abused monoamine transporter substrates. Structurally, these compounds are all beta-ketone amphetamine (cathinone) analogs. Whether synthetic cathinone analogs produce differential behavioral effects from their amphetamine analog counterparts has not been systematically examined. Preclinical drug discrimination procedures have been useful for determining the structure activity relationships (SARs) of abused drugs; however, direct comparisons between amphetamine and cathinone analogs are lacking and, in particular, in non-human primate models. OBJECTIVES The study aim was to determine the potency and time course of (±)-amphetamine, (±)-cathinone, and (±)-methamphetamine and their 3,4-methylenedioxy analogs (±)-MDA, (±)-MDC, and (±)-MDMA, respectively, to produce cocaine-like discriminative stimulus effects. If cathinone analogs have similar behavioral pharmacological properties to their amphetamine counterparts, then we would predict similar potencies and efficacies to produce cocaine-like discriminative stimulus effects. METHODS Male rhesus monkeys (n = 4) were trained to discriminate intramuscular cocaine (0.32 mg/kg) from saline in a two-key food-reinforced discrimination procedure. RESULTS Racemic amphetamine, cathinone, and methamphetamine produced dose-dependent and full substitution, ≥90 % cocaine-appropriate responding, in all monkeys. Addition of 3,4-methylenedioxy moiety attenuated both the potency and efficacy of amphetamine (MDA), cathinone (MDC), and methamphetamine (MDMA) to produce full cocaine-like effects. Moreover, the cocaine-like effects of amphetamine and cathinone were attenuated to a greater extent than those of methamphetamine or previously published methcathinone (Smith et al. 2016). CONCLUSION The presence of an N-methyl group blunted both the potency and the efficacy shift of the 3,4-methylenedioxy addition for both amphetamine and cathinone analogs.
Collapse
Affiliation(s)
- Douglas A. Smith
- Department of Pharmacology & Toxicology, Virginia Commonwealth University, Richmond, VA USA 23298
| | - Bruce. E. Blough
- Center for Drug Discovery, Research Triangle Institute, Research Triangle Park, NC USA
| | - Matthew L. Banks
- Department of Pharmacology & Toxicology, Virginia Commonwealth University, Richmond, VA USA 23298,Corresponding Author: Matthew L. Banks, PharmD, PhD, Department of Pharmacology and Toxicology, Virginia Commonwealth University, 410 N. 12th St., PO Box 980613, Richmond, VA 23298, , Phone: 804-828-8466, Fax: 804-828-2117
| |
Collapse
|
33
|
Kolodziejczyk W, Kar S, Hill GA. Conformational analysis, energy profile, and structural-electronic properties evaluation of mephedrone derivatives employing quantum-mechanical models. Struct Chem 2016. [DOI: 10.1007/s11224-016-0878-7] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/30/2022]
|
34
|
Zdrazil B, Hellsberg E, Viereck M, Ecker GF. From linked open data to molecular interaction: studying selectivity trends for ligands of the human serotonin and dopamine transporter. MEDCHEMCOMM 2016; 7:1819-1831. [PMID: 27891211 PMCID: PMC5100691 DOI: 10.1039/c6md00207b] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 04/13/2016] [Accepted: 07/01/2016] [Indexed: 11/21/2022]
Abstract
Retrieval of congeneric and consistent SAR data sets for protein targets of interest is still a laborious task to do if no appropriate in-house data set is available. However, combining integrated open data sources (such as the Open PHACTS Discovery Platform) with workflow tools now offers the possibility of querying across multiple domains and tailoring the search to the given research question. Starting from two phylogenetically related protein targets of interest (the human serotonin and dopamine transporters), the whole chemical compound space was explored by implementing a scaffold-based clustering of compounds possessing biological measurements for both targets. In addition, potential hERG blocking liabilities were included. The workflow allowed studying the selectivity trends of scaffold series, identifying potentially harmful compound series, and performing SAR, docking studies and molecular dynamics (MD) simulations for a consistent data set of 56 cathinones. This delivered useful insights into driving determinants for hDAT selectivity over hSERT. With respect to the scaffold-based analyses it should be noted that the cathinone data set could be retrieved only when Murcko scaffold analyses were combined with similarity searches such as a common substructure search.
Collapse
Affiliation(s)
- Barbara Zdrazil
- Department of Pharmaceutical Chemistry , Pharmacoinformatics Research Group , University of Vienna , Althanstraße 14 , A-1090 , Austria . ; ; Tel: +43 1 4277 55110
| | - Eva Hellsberg
- Department of Pharmaceutical Chemistry , Pharmacoinformatics Research Group , University of Vienna , Althanstraße 14 , A-1090 , Austria . ; ; Tel: +43 1 4277 55110
| | - Michael Viereck
- Department of Pharmaceutical Chemistry , Pharmacoinformatics Research Group , University of Vienna , Althanstraße 14 , A-1090 , Austria . ; ; Tel: +43 1 4277 55110
| | - Gerhard F Ecker
- Department of Pharmaceutical Chemistry , Pharmacoinformatics Research Group , University of Vienna , Althanstraße 14 , A-1090 , Austria . ; ; Tel: +43 1 4277 55110
| |
Collapse
|
35
|
Hutsell BA, Baumann MH, Partilla JS, Banks ML, Vekariya R, Glennon RA, Negus SS. Abuse-related neurochemical and behavioral effects of cathinone and 4-methylcathinone stereoisomers in rats. Eur Neuropsychopharmacol 2016; 26:288-297. [PMID: 26738428 PMCID: PMC5331761 DOI: 10.1016/j.euroneuro.2015.12.010] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/01/2015] [Revised: 11/13/2015] [Accepted: 12/01/2015] [Indexed: 10/22/2022]
Abstract
Cathinone and many of its analogs produce behavioral effects by promoting transporter-mediated release of the monoamine neurotransmitters dopamine, norepinephrine and/or serotonin. Stereoselectivity is one determinant of neurochemical and behavioral effects of cathinone analogs. This study compared effectiveness of the S(-) and R(+) enantiomers of cathinone and 4-methylcathinone to produce in vitro monoamine release and in vivo abuse-related behavioral effects in rats. For neurochemical studies, drug effects were evaluated on monoamine release through dopamine, norepinephrine, and serotonin transporters (DAT, NET and SERT, respectively) in rat brain synaptosomes. For behavioral studies, drug effects were evaluated on responding for electrical brain stimulation in an intracranial self-stimulation (ICSS) procedure. The cathinone enantiomers differed in potency [S(-)>R(+)], but both enantiomers were >50-fold selective at promoting monoamine release through DAT vs. SERT, and both enantiomers produced ICSS facilitation. The 4-methylcathinone enantiomers also differed in potency [S(-)>R(+)]; however, in neurochemical studies, the decrease in potency from S(-) to R(+)4-methylcathinone was less for DAT than for SERT, and as a result, DAT vs. SERT selectivity was greater for R(+) than for S(-)4-methylcathinone (4.1- vs. 1.2-fold). Moreover, in behavioral studies, S(-)4-methylcathinone produced only ICSS depression, whereas R(+)4-methylcathinone produced ICSS facilitation. This study provides further evidence for stereoselectivity in neurochemical and behavioral actions of cathinone analogs. More importantly, stereoselective 4-methylcathinone effects on ICSS illustrate the potential for diametrically opposite effects of enantiomers in a preclinical behavioral assay of abuse potential.
Collapse
Affiliation(s)
- Blake A Hutsell
- Department of Pharmacology and Toxicology, Virginia Commonwealth University, 410 N. 12th St., PO Box 980613, Richmond, VA 23298, USA
| | - Michael H Baumann
- Designer Drug Research Unit, Intramural Research Program, National Institute on Drug Abuse, National Institutes of Health, Baltimore, MD, USA
| | - John S Partilla
- Designer Drug Research Unit, Intramural Research Program, National Institute on Drug Abuse, National Institutes of Health, Baltimore, MD, USA
| | - Matthew L Banks
- Department of Pharmacology and Toxicology, Virginia Commonwealth University, 410 N. 12th St., PO Box 980613, Richmond, VA 23298, USA; Institute for Drug and Alcohol Studies, Virginia Commonwealth University, USA
| | - Rakesh Vekariya
- Department of Medicinal Chemistry, Virginia Commonwealth University, Richmond, VA, USA
| | - Richard A Glennon
- Department of Medicinal Chemistry, Virginia Commonwealth University, Richmond, VA, USA
| | - S Stevens Negus
- Department of Pharmacology and Toxicology, Virginia Commonwealth University, 410 N. 12th St., PO Box 980613, Richmond, VA 23298, USA; Institute for Drug and Alcohol Studies, Virginia Commonwealth University, USA.
| |
Collapse
|
36
|
Suyama JA, Sakloth F, Kolanos R, Glennon RA, Lazenka MF, Negus SS, Banks ML. Abuse-Related Neurochemical Effects of Para-Substituted Methcathinone Analogs in Rats: Microdialysis Studies of Nucleus Accumbens Dopamine and Serotonin. J Pharmacol Exp Ther 2016; 356:182-90. [PMID: 26645638 PMCID: PMC4702071 DOI: 10.1124/jpet.115.229559] [Citation(s) in RCA: 48] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/23/2015] [Accepted: 10/14/2015] [Indexed: 11/22/2022] Open
Abstract
Methcathinone (MCAT) is a monoamine releaser and parent compound to a new class of designer drugs that includes the synthetic cathinones mephedrone and flephedrone. Using MCAT and a series of para-substituted (or 4-substituted) MCAT analogs, it has been previously shown that expression of abuse-related behavioral effects in rats correlates both with the volume of the para substituent and in vitro neurochemical selectivity to promote monoamine release via the dopamine (DA) versus serotonin (5-HT) transporters in rat brain synaptosomes. The present study used in vivo microdialysis to determine the relationship between these previous measures and the in vivo neurochemical selectivity of these compounds to alter nucleus accumbens (NAc) DA and 5-HT levels. Male Sprague-Dawley rats were implanted with bilateral guide cannulae targeting the NAc. MCAT and five para-substituted analogs (4-F, 4-Cl, 4-Br, 4-CH3, and 4-OCH3) produced dose- and time-dependent increases in NAc DA and/or 5-HT levels. Selectivity was determined as the dose required to increase peak 5-HT levels by 250% divided by the dose required to increase peak DA levels by 250%. This measure of in vivo neurochemical selectivity varied across compounds and correlated with 1) in vivo expression of abuse-related behavioral effects (r = 0.89, P = 0.02); 2) in vitro selectivity to promote monoamine release via DA and 5-HT transporters (r = 0.95, P < 0.01); and 3) molecular volume of the para substituent (r = -0.85, P = 0.03). These results support a relationship between these molecular, neurochemical, and behavioral measures and support a role for molecular structure as a determinant of abuse-related neurochemical and behavioral effects of MCAT analogs.
Collapse
Affiliation(s)
- Julie A Suyama
- Department of Pharmacology and Toxicology (J.A.S., M.F.L., S.S.N., M.L.B.), Department of Medicinal Chemistry (F.S., R.K., R.A.G.), and Institute on Drug and Alcohol Studies (R.A.G., S.S.N., M.L.B.), Virginia Commonwealth University, Richmond, Virginia
| | - Farhana Sakloth
- Department of Pharmacology and Toxicology (J.A.S., M.F.L., S.S.N., M.L.B.), Department of Medicinal Chemistry (F.S., R.K., R.A.G.), and Institute on Drug and Alcohol Studies (R.A.G., S.S.N., M.L.B.), Virginia Commonwealth University, Richmond, Virginia
| | - Renata Kolanos
- Department of Pharmacology and Toxicology (J.A.S., M.F.L., S.S.N., M.L.B.), Department of Medicinal Chemistry (F.S., R.K., R.A.G.), and Institute on Drug and Alcohol Studies (R.A.G., S.S.N., M.L.B.), Virginia Commonwealth University, Richmond, Virginia
| | - Richard A Glennon
- Department of Pharmacology and Toxicology (J.A.S., M.F.L., S.S.N., M.L.B.), Department of Medicinal Chemistry (F.S., R.K., R.A.G.), and Institute on Drug and Alcohol Studies (R.A.G., S.S.N., M.L.B.), Virginia Commonwealth University, Richmond, Virginia
| | - Matthew F Lazenka
- Department of Pharmacology and Toxicology (J.A.S., M.F.L., S.S.N., M.L.B.), Department of Medicinal Chemistry (F.S., R.K., R.A.G.), and Institute on Drug and Alcohol Studies (R.A.G., S.S.N., M.L.B.), Virginia Commonwealth University, Richmond, Virginia
| | - S Stevens Negus
- Department of Pharmacology and Toxicology (J.A.S., M.F.L., S.S.N., M.L.B.), Department of Medicinal Chemistry (F.S., R.K., R.A.G.), and Institute on Drug and Alcohol Studies (R.A.G., S.S.N., M.L.B.), Virginia Commonwealth University, Richmond, Virginia
| | - Matthew L Banks
- Department of Pharmacology and Toxicology (J.A.S., M.F.L., S.S.N., M.L.B.), Department of Medicinal Chemistry (F.S., R.K., R.A.G.), and Institute on Drug and Alcohol Studies (R.A.G., S.S.N., M.L.B.), Virginia Commonwealth University, Richmond, Virginia
| |
Collapse
|
37
|
Pail PB, Costa KM, Leite CE, Campos MM. Comparative pharmacological evaluation of the cathinone derivatives, mephedrone and methedrone, in mice. Neurotoxicology 2015; 50:71-80. [PMID: 26254738 DOI: 10.1016/j.neuro.2015.08.004] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/20/2015] [Revised: 07/20/2015] [Accepted: 08/03/2015] [Indexed: 11/26/2022]
Abstract
Mephedrone and methedrone are cathinone-related compounds, which act as non-selective substrates for monoamine transporters, facilitating a neurotransmitter release. We compared the acute pharmacological effects of mephedrone and methedrone, attempting to further evaluate the action mechanisms of methedrone by responsibly and ethically using mice under approved procedures. The effects of both compounds were examined from 10 to 60 min, in a series of behavioral paradigms, namely open-field, plus-maze, hot-plate and tail suspension tests, whereas neurotransmitter brain tissue levels were determined ex vivo by HPLC. Separate groups were pre-treated with the dopamine (DA) antagonist haloperidol, or the serotonin (5-HT) synthesis inhibitor ρCPA, to further assess the mechanisms underlying methedrone effects. The compounds caused marked hyperlocomotion, displaying dissimilar stereotyped behavior, in an open-field arena. Mephedrone caused anxiolytic-like effects, while methedrone induced anxiogenic-like actions in the elevated plus-maze. Both compounds displayed thermal antinociception, with a reduced immobility time in the tail suspension model. Mephedrone triggered a 2- and 3-fold increment of dopamine and serotonin tissue levels, respectively, in the nucleus accumbens, with a 1.5-fold elevation of tissue dopamine in the frontal cortex. Methedrone caused a 2-fold increment of tissue dopamine in the nucleus accumbens and in the striatum, and a 1.5-fold increment of serotonin tissue levels in the hippocampus and striatum. In vivo methedrone effects were partially inhibited by a pre-treatment with haloperidol or ρCPA. Despite similar actions on locomotion, analgesia, and depression-like behavior, the acute administration of mephedrone and methedrone elicited divergent effects on anxiety-like behavior and stereotyped movements in mice, which might be related to the distinct modulation of brain tissue neurotransmitter levels.
Collapse
Affiliation(s)
- Priscilla B Pail
- PUCRS, Programa de Pós-graduação em Biologia Celular e Molecular, Porto Alegre, RS, Brazil
| | - Kesiane M Costa
- PUCRS, Programa de Pós-graduação em Medicina e Ciências da Saúde, Porto Alegre, RS, Brazil
| | - Carlos E Leite
- PUCRS, Instituto de Toxicologia e Farmacologia, Porto Alegre, RS, Brazil
| | - Maria M Campos
- PUCRS, Programa de Pós-graduação em Medicina e Ciências da Saúde, Porto Alegre, RS, Brazil; PUCRS, Instituto de Toxicologia e Farmacologia, Porto Alegre, RS, Brazil; PUCRS, Faculdade de Odontologia, Porto Alegre, RS, Brazil.
| |
Collapse
|
38
|
Del Bello F, Sakloth F, Partilla JS, Baumann MH, Glennon RA. Ethylenedioxy homologs of N-methyl-(3,4-methylenedioxyphenyl)-2-aminopropane (MDMA) and its corresponding cathinone analog methylenedioxymethcathinone: Interactions with transporters for serotonin, dopamine, and norepinephrine. Bioorg Med Chem 2015; 23:5574-9. [PMID: 26233799 DOI: 10.1016/j.bmc.2015.07.035] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/14/2015] [Revised: 07/07/2015] [Accepted: 07/16/2015] [Indexed: 11/19/2022]
Abstract
N-Methyl-(3,4-methylenedioxyphenyl)-2-aminopropane (MDMA; 'Ecstasy'; 1) and its β-keto analog methylone (MDMC; 2) are popular drugs of abuse. Little is known about their ring-expanded ethylenedioxy homologs. Here, we prepared N-methyl-(3,4-ethylenedioxyphenyl)-2-aminopropane (EDMA; 3), both of its optical isomers, and β-keto EDMA (i.e., EDMC; 4) to examine their effects at transporters for serotonin (SERT), dopamine (DAT), and norepinephrine (NET). In general, ring-expansion of the methylenedioxy group led to a several-fold reduction in potency at all three transporters. With respect to EDMA (3), S(+)3 was 6-fold, 50-fold, and 8-fold more potent than its R(-) enantiomer at SERT, DAT, and NET, respectively. Overall, in the absence of a β-carbonyl group, the ethylenedioxy (i.e., 1,4-dioxane) substituent seems better accommodated at SERT than at DAT and NET.
Collapse
Affiliation(s)
- Fabio Del Bello
- Department of Medicinal Chemistry, School of Pharmacy, Virginia Commonwealth University, Box 980540, Richmond, VA 23298, USA
| | - Farhana Sakloth
- Department of Medicinal Chemistry, School of Pharmacy, Virginia Commonwealth University, Box 980540, Richmond, VA 23298, USA
| | - John S Partilla
- Designer Drug Research Unit, Intramural Research Program, National Institute on Drug Abuse, National Institutes of Health, Baltimore, MD 21224, USA
| | - Michael H Baumann
- Designer Drug Research Unit, Intramural Research Program, National Institute on Drug Abuse, National Institutes of Health, Baltimore, MD 21224, USA
| | - Richard A Glennon
- Department of Medicinal Chemistry, School of Pharmacy, Virginia Commonwealth University, Box 980540, Richmond, VA 23298, USA.
| |
Collapse
|
39
|
Bonano JS, Banks ML, Kolanos R, Sakloth F, Barnier ML, Glennon RA, Cozzi NV, Partilla JS, Baumann MH, Negus SS. Quantitative structure-activity relationship analysis of the pharmacology of para-substituted methcathinone analogues. Br J Pharmacol 2015; 172:2433-44. [PMID: 25438806 DOI: 10.1111/bph.13030] [Citation(s) in RCA: 58] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/22/2014] [Revised: 11/20/2014] [Accepted: 11/27/2014] [Indexed: 02/06/2023] Open
Abstract
BACKGROUND AND PURPOSE Methcathinone (MCAT) is a potent monoamine releaser and parent compound to emerging drugs of abuse including mephedrone (4-CH3 MCAT), the para-methyl analogue of MCAT. This study examined quantitative structure-activity relationships (QSAR) for MCAT and six para-substituted MCAT analogues on (a) in vitro potency to promote monoamine release via dopamine and serotonin transporters (DAT and SERT, respectively), and (b) in vivo modulation of intracranial self-stimulation (ICSS), a behavioural procedure used to evaluate abuse potential. Neurochemical and behavioural effects were correlated with steric (Es ), electronic (σp ) and lipophilic (πp ) parameters of the para substituents. EXPERIMENTAL APPROACH For neurochemical studies, drug effects on monoamine release through DAT and SERT were evaluated in rat brain synaptosomes. For behavioural studies, drug effects were tested in male Sprague-Dawley rats implanted with electrodes targeting the medial forebrain bundle and trained to lever-press for electrical brain stimulation. KEY RESULTS MCAT and all six para-substituted analogues increased monoamine release via DAT and SERT and dose- and time-dependently modulated ICSS. In vitro selectivity for DAT versus SERT correlated with in vivo efficacy to produce abuse-related ICSS facilitation. In addition, the Es values of the para substituents correlated with both selectivity for DAT versus SERT and magnitude of ICSS facilitation. CONCLUSIONS AND IMPLICATIONS Selectivity for DAT versus SERT in vitro is a key determinant of abuse-related ICSS facilitation by these MCAT analogues, and steric aspects of the para substituent of the MCAT scaffold (indicated by Es ) are key determinants of this selectivity.
Collapse
Affiliation(s)
- J S Bonano
- Department of Pharmacology and Toxicology, Virginia Commonwealth University, Richmond, VA, USA
| | | | | | | | | | | | | | | | | | | |
Collapse
|