1
|
Canzian J, Borba JV, Resmim CM, Mohammed KA, Pretzel CW, Adedara IA, Rosemberg DB. The dopamine transporter inhibition using GBR 12909 as a novel pharmacological tool to assess bipolar disorder-like neurobehavioral phenotypes in zebrafish. Behav Brain Res 2025; 477:115302. [PMID: 39442564 DOI: 10.1016/j.bbr.2024.115302] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/18/2024] [Revised: 10/11/2024] [Accepted: 10/19/2024] [Indexed: 10/25/2024]
Abstract
Dopamine (DA) is a neurotransmitter that plays an important role in brain physiology. Changes in DA-mediated signaling have been implicated with the pathophysiology of various neuropsychiatric conditions. Bipolar disorder (BD) is a mental disorder, characterized by alterning between manic/hypomanic and depressive mood. In experimental research, the pharmacological inhibition of DA reuptake using GBR 12909 serves as a tool to elicit BD-like phenotypes. Alternative model organisms, such as the zebrafish (Danio rerio), have been considered important systems for investigating the neurobehavioral changes involved in different neuropsychiatric conditions, including BD. Here, we discuss the use of GBR 12909 as a novel pharmacological strategy to mimic BD-like phenotypes in zebrafish models. We also emphasize the well-conserved DA-mediated signaling in zebrafish and the early expression of dopaminergic biomarkers in the brain, especially focusing on dopamine transporter (DAT), the main target of GBR 12909. Finally, we discuss potential advantages and limitations in the field, the perspectives of using GBR 12909 in BD research, and how distinct validation criteria (i.e., face, predictive, and construct validity) can be assessed in translational approaches using zebrafish-based models.
Collapse
Affiliation(s)
- Julia Canzian
- Laboratory of Experimental Neuropsychobiology, Department of Biochemistry and Molecular Biology, Natural and Exact Sciences Center, Federal University of Santa Maria, 1000 Roraima Avenue, Santa Maria, RS 97105-900, Brazil; Graduate Program in Biological Sciences: Toxicological Biochemistry, Federal University of Santa Maria, 1000 Roraima Avenue, Santa Maria, RS 97105-900, Brazil.
| | - João V Borba
- Laboratory of Experimental Neuropsychobiology, Department of Biochemistry and Molecular Biology, Natural and Exact Sciences Center, Federal University of Santa Maria, 1000 Roraima Avenue, Santa Maria, RS 97105-900, Brazil; Graduate Program in Biological Sciences: Toxicological Biochemistry, Federal University of Santa Maria, 1000 Roraima Avenue, Santa Maria, RS 97105-900, Brazil
| | - Cássio M Resmim
- Laboratory of Experimental Neuropsychobiology, Department of Biochemistry and Molecular Biology, Natural and Exact Sciences Center, Federal University of Santa Maria, 1000 Roraima Avenue, Santa Maria, RS 97105-900, Brazil; Graduate Program in Biological Sciences: Toxicological Biochemistry, Federal University of Santa Maria, 1000 Roraima Avenue, Santa Maria, RS 97105-900, Brazil
| | - Khadija A Mohammed
- Laboratory of Experimental Neuropsychobiology, Department of Biochemistry and Molecular Biology, Natural and Exact Sciences Center, Federal University of Santa Maria, 1000 Roraima Avenue, Santa Maria, RS 97105-900, Brazil; Graduate Program in Biological Sciences: Toxicological Biochemistry, Federal University of Santa Maria, 1000 Roraima Avenue, Santa Maria, RS 97105-900, Brazil
| | - Camilla W Pretzel
- Laboratory of Experimental Neuropsychobiology, Department of Biochemistry and Molecular Biology, Natural and Exact Sciences Center, Federal University of Santa Maria, 1000 Roraima Avenue, Santa Maria, RS 97105-900, Brazil
| | - Isaac A Adedara
- Department of Food Science and Technology, Center of Rural Sciences, Federal University of Santa Maria, Camobi, Santa Maria, RS 97105-900, Brazil
| | - Denis B Rosemberg
- Laboratory of Experimental Neuropsychobiology, Department of Biochemistry and Molecular Biology, Natural and Exact Sciences Center, Federal University of Santa Maria, 1000 Roraima Avenue, Santa Maria, RS 97105-900, Brazil; Graduate Program in Biological Sciences: Toxicological Biochemistry, Federal University of Santa Maria, 1000 Roraima Avenue, Santa Maria, RS 97105-900, Brazil; The International Zebrafish Neuroscience Research Consortium (ZNRC), 309 Palmer Court, Slidell, LA 70458, USA.
| |
Collapse
|
2
|
Poulton A, Gauci N, Khalifa H, Hibbert EJ, Poulton AS. The Impact of Dexamphetamine Treatment for Obesity on Executive Function: A Double-Blind Randomised Controlled Pilot Study. Brain Sci 2024; 14:1274. [PMID: 39766473 PMCID: PMC11674214 DOI: 10.3390/brainsci14121274] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/27/2024] [Revised: 12/16/2024] [Accepted: 12/17/2024] [Indexed: 01/11/2025] Open
Abstract
BACKGROUND Amphetamines increase dopamine levels in mid-brain regions which, in turn, impact top-down executive function. Repeated exposure is linked to substance use disorders. Nonetheless, amphetamines are used to manage attention-deficit/hyperactivity disorder (ADHD) and eating-related disorders. In ADHD, amphetamines upregulate a system characterised by low dopaminergic tone, assisting to improve executive function. A similar process might be at play with eating disorders; however, the effect of amphetamine treatment on executive function in this case has not been thoroughly considered. METHODS Participants (N = 52, Mage = 47.06, SD = 12.29) with a body mass index of 25-60 were randomised to treatment (6-week dexamphetamine titration) or control (placebo) groups. They completed an executive function measure-Barkley Deficits in Executive Functioning Scale (BDEFS-SF)-and response inhibition task-Stop-Signal Task (SST)-at Baseline, throughout titration, at Maintenance, and at Follow-up. Mixed effects models examined whether BDEFS-SF score or the SST variable, stop-signal reaction time (SSRT), changed across sessions as a function of treatment. RESULTS There was no effect of group (p = 0.440), but an effect of session (p = 0.024) on BDEFS-SF, with scores at Time 2 (p = 0.011, 95% CI [0.47, 3.49]) and Maintenance (p = 0.022, 95% CI [-4.89, -0.39]), respectively, higher and lower than other timepoints. There was no group by session interaction (p = 0.659). R2 (conditional) = 0.74; ICC = 0.73. There was an effect of group (p = 0.039) and session (p < 0.001) on SSRT, but no interaction (p = 0.707). Baseline SSRT was significantly longer than the mean of all subsequent timepoints (p < 0.001, 95% CI [16.29, 33.84]). R2 (conditional) = 0.47; ICC = 0.39. CONCLUSIONS There was no discernible impact of amphetamine treatment for obesity on executive function. Our results suggest some variation related to sample size and/or practice effects. Thus, while treatment appears unlikely to render individuals susceptible to substance use disorders, parallels with ADHD might be overstated.
Collapse
Affiliation(s)
- Antoinette Poulton
- Melbourne School of Psychological Sciences, University of Melbourne, Parkville, VIC 3052, Australia
| | - Natalie Gauci
- Nepean Clinical School, Faculty of Medicine and Health, University of Sydney, Sydney, NSW 2751, Australia; (N.G.); (H.K.); (E.J.H.); (A.S.P.)
| | - Hazer Khalifa
- Nepean Clinical School, Faculty of Medicine and Health, University of Sydney, Sydney, NSW 2751, Australia; (N.G.); (H.K.); (E.J.H.); (A.S.P.)
| | - Emily J. Hibbert
- Nepean Clinical School, Faculty of Medicine and Health, University of Sydney, Sydney, NSW 2751, Australia; (N.G.); (H.K.); (E.J.H.); (A.S.P.)
- Charles Perkins Centre-Nepean, Faculty of Medicine and Health, University of Sydney, Sydney, NSW 2003, Australia
- Nepean Hospital, Penrith, NSW 2747, Australia
| | - Alison S. Poulton
- Nepean Clinical School, Faculty of Medicine and Health, University of Sydney, Sydney, NSW 2751, Australia; (N.G.); (H.K.); (E.J.H.); (A.S.P.)
- Charles Perkins Centre-Nepean, Faculty of Medicine and Health, University of Sydney, Sydney, NSW 2003, Australia
- Nepean Hospital, Penrith, NSW 2747, Australia
| |
Collapse
|
3
|
Farr CV, Xiao Y, El-Kasaby A, Schupp M, Hotka M, di Mauro G, Clarke A, Pastor Fernandez M, Sandtner W, Stockner T, Klade C, Maulide N, Freissmuth M. Probing the Chemical Space of Guanidino-Carboxylic Acids to Identify the First Blockers of the Creatine-Transporter-1. Mol Pharmacol 2024; 106:319-333. [PMID: 39322412 DOI: 10.1124/molpharm.124.000995] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/25/2024] [Revised: 08/26/2024] [Accepted: 09/12/2024] [Indexed: 09/27/2024] Open
Abstract
The creatine transporter-1 (CRT-1/SLC6A8) maintains the uphill transport of creatine into cells against a steep concentration gradient. Cellular creatine accumulation is required to support the ATP-buffering by phosphocreatine. More than 60 compounds have been explored in the past for their ability to inhibit cellular creatine uptake, but the number of active compounds is very limited. Here, we show that all currently known inhibitors are full alternative substrates. We analyzed their structure-activity relationship for inhibition of CRT-1 to guide a rational approach to the synthesis of novel creatine transporter ligands. Measurements of both inhibition of [3H]creatine uptake and transport associated currents allowed for differentiating between full and partial substrates and true inhibitors. This combined approach led to a refined understanding of the structural requirements for binding to CRT-1, which translated into the identification of three novel compounds - i.e., compound 1 (2-(N-benzylcarbamimidamido)acetic acid), MIPA572 (=carbamimidoylphenylalanine), and MIPA573 (=carbamimidoyltryptophane) that blocked CRT-1 transport, albeit with low affinity. In addition, we found two new alternative full substrates, namely MIPA574 (carbamimidoylalanine) and GiDi1257 (1-carbamimidoylazetidine-3-carboxylic acid), which was superior in affinity to all known CTR-1 ligands, and one partial substrate, namely GiDi1254 (1-carbamimidoylpiperidine-4-carboxylic acid). SIGNIFICANCE STATEMENT: The creatine transporter-1 (CRT-1) is required to maintain intracellular creatine levels. Inhibition of CRT-1 has been recently proposed as a therapeutic strategy for cancer, but pharmacological tools are scarce. In fact, all available inhibitors are alternative substrates. We tested existing and newly synthesized guanidinocarboxylic acids for CRT-1 inhibition and identified three blockers, one partial and two full substrates of CRT-1. Our results support a refined structural understanding of ligand binding to CRT-1 and provide a proof-of-principle for blockage of CRT-1.
Collapse
Affiliation(s)
- Clemens V Farr
- Institute of Pharmacology and the Gaston H. Glock Research Laboratories for Exploratory Drug Development, Center of Physiology and Pharmacology, Medical University of Vienna, Vienna, Austria (C.V.F., A.E-K., M.H., A.C., W.S., T.S., M.F.); Institute of Organic Chemistry, University of Vienna, Vienna, Austria (Y.X., M.S., G.dM., M.P.F., N.M.); CeMM Research Center for Molecular Medicine of the Austrian Academy of Sciences, Vienna, Austria (Y.X., M.S., N.M.); AOP Orphan Pharmaceuticals GmbH, Vienna, Austria (C.K.); Christian Doppler Laboratory for Entropy-Oriented Drug Design, Institute of Organic Chemistry, University of Vienna, Vienna, Austria (N.M.); and NeGeMac Research Platform for Next Generation Macrocycles, Vienna, Austria (N.M.)
| | - Yi Xiao
- Institute of Pharmacology and the Gaston H. Glock Research Laboratories for Exploratory Drug Development, Center of Physiology and Pharmacology, Medical University of Vienna, Vienna, Austria (C.V.F., A.E-K., M.H., A.C., W.S., T.S., M.F.); Institute of Organic Chemistry, University of Vienna, Vienna, Austria (Y.X., M.S., G.dM., M.P.F., N.M.); CeMM Research Center for Molecular Medicine of the Austrian Academy of Sciences, Vienna, Austria (Y.X., M.S., N.M.); AOP Orphan Pharmaceuticals GmbH, Vienna, Austria (C.K.); Christian Doppler Laboratory for Entropy-Oriented Drug Design, Institute of Organic Chemistry, University of Vienna, Vienna, Austria (N.M.); and NeGeMac Research Platform for Next Generation Macrocycles, Vienna, Austria (N.M.)
| | - Ali El-Kasaby
- Institute of Pharmacology and the Gaston H. Glock Research Laboratories for Exploratory Drug Development, Center of Physiology and Pharmacology, Medical University of Vienna, Vienna, Austria (C.V.F., A.E-K., M.H., A.C., W.S., T.S., M.F.); Institute of Organic Chemistry, University of Vienna, Vienna, Austria (Y.X., M.S., G.dM., M.P.F., N.M.); CeMM Research Center for Molecular Medicine of the Austrian Academy of Sciences, Vienna, Austria (Y.X., M.S., N.M.); AOP Orphan Pharmaceuticals GmbH, Vienna, Austria (C.K.); Christian Doppler Laboratory for Entropy-Oriented Drug Design, Institute of Organic Chemistry, University of Vienna, Vienna, Austria (N.M.); and NeGeMac Research Platform for Next Generation Macrocycles, Vienna, Austria (N.M.)
| | - Manuel Schupp
- Institute of Pharmacology and the Gaston H. Glock Research Laboratories for Exploratory Drug Development, Center of Physiology and Pharmacology, Medical University of Vienna, Vienna, Austria (C.V.F., A.E-K., M.H., A.C., W.S., T.S., M.F.); Institute of Organic Chemistry, University of Vienna, Vienna, Austria (Y.X., M.S., G.dM., M.P.F., N.M.); CeMM Research Center for Molecular Medicine of the Austrian Academy of Sciences, Vienna, Austria (Y.X., M.S., N.M.); AOP Orphan Pharmaceuticals GmbH, Vienna, Austria (C.K.); Christian Doppler Laboratory for Entropy-Oriented Drug Design, Institute of Organic Chemistry, University of Vienna, Vienna, Austria (N.M.); and NeGeMac Research Platform for Next Generation Macrocycles, Vienna, Austria (N.M.)
| | - Matej Hotka
- Institute of Pharmacology and the Gaston H. Glock Research Laboratories for Exploratory Drug Development, Center of Physiology and Pharmacology, Medical University of Vienna, Vienna, Austria (C.V.F., A.E-K., M.H., A.C., W.S., T.S., M.F.); Institute of Organic Chemistry, University of Vienna, Vienna, Austria (Y.X., M.S., G.dM., M.P.F., N.M.); CeMM Research Center for Molecular Medicine of the Austrian Academy of Sciences, Vienna, Austria (Y.X., M.S., N.M.); AOP Orphan Pharmaceuticals GmbH, Vienna, Austria (C.K.); Christian Doppler Laboratory for Entropy-Oriented Drug Design, Institute of Organic Chemistry, University of Vienna, Vienna, Austria (N.M.); and NeGeMac Research Platform for Next Generation Macrocycles, Vienna, Austria (N.M.)
| | - Giovanni di Mauro
- Institute of Pharmacology and the Gaston H. Glock Research Laboratories for Exploratory Drug Development, Center of Physiology and Pharmacology, Medical University of Vienna, Vienna, Austria (C.V.F., A.E-K., M.H., A.C., W.S., T.S., M.F.); Institute of Organic Chemistry, University of Vienna, Vienna, Austria (Y.X., M.S., G.dM., M.P.F., N.M.); CeMM Research Center for Molecular Medicine of the Austrian Academy of Sciences, Vienna, Austria (Y.X., M.S., N.M.); AOP Orphan Pharmaceuticals GmbH, Vienna, Austria (C.K.); Christian Doppler Laboratory for Entropy-Oriented Drug Design, Institute of Organic Chemistry, University of Vienna, Vienna, Austria (N.M.); and NeGeMac Research Platform for Next Generation Macrocycles, Vienna, Austria (N.M.)
| | - Amy Clarke
- Institute of Pharmacology and the Gaston H. Glock Research Laboratories for Exploratory Drug Development, Center of Physiology and Pharmacology, Medical University of Vienna, Vienna, Austria (C.V.F., A.E-K., M.H., A.C., W.S., T.S., M.F.); Institute of Organic Chemistry, University of Vienna, Vienna, Austria (Y.X., M.S., G.dM., M.P.F., N.M.); CeMM Research Center for Molecular Medicine of the Austrian Academy of Sciences, Vienna, Austria (Y.X., M.S., N.M.); AOP Orphan Pharmaceuticals GmbH, Vienna, Austria (C.K.); Christian Doppler Laboratory for Entropy-Oriented Drug Design, Institute of Organic Chemistry, University of Vienna, Vienna, Austria (N.M.); and NeGeMac Research Platform for Next Generation Macrocycles, Vienna, Austria (N.M.)
| | - Miryam Pastor Fernandez
- Institute of Pharmacology and the Gaston H. Glock Research Laboratories for Exploratory Drug Development, Center of Physiology and Pharmacology, Medical University of Vienna, Vienna, Austria (C.V.F., A.E-K., M.H., A.C., W.S., T.S., M.F.); Institute of Organic Chemistry, University of Vienna, Vienna, Austria (Y.X., M.S., G.dM., M.P.F., N.M.); CeMM Research Center for Molecular Medicine of the Austrian Academy of Sciences, Vienna, Austria (Y.X., M.S., N.M.); AOP Orphan Pharmaceuticals GmbH, Vienna, Austria (C.K.); Christian Doppler Laboratory for Entropy-Oriented Drug Design, Institute of Organic Chemistry, University of Vienna, Vienna, Austria (N.M.); and NeGeMac Research Platform for Next Generation Macrocycles, Vienna, Austria (N.M.)
| | - Walter Sandtner
- Institute of Pharmacology and the Gaston H. Glock Research Laboratories for Exploratory Drug Development, Center of Physiology and Pharmacology, Medical University of Vienna, Vienna, Austria (C.V.F., A.E-K., M.H., A.C., W.S., T.S., M.F.); Institute of Organic Chemistry, University of Vienna, Vienna, Austria (Y.X., M.S., G.dM., M.P.F., N.M.); CeMM Research Center for Molecular Medicine of the Austrian Academy of Sciences, Vienna, Austria (Y.X., M.S., N.M.); AOP Orphan Pharmaceuticals GmbH, Vienna, Austria (C.K.); Christian Doppler Laboratory for Entropy-Oriented Drug Design, Institute of Organic Chemistry, University of Vienna, Vienna, Austria (N.M.); and NeGeMac Research Platform for Next Generation Macrocycles, Vienna, Austria (N.M.)
| | - Thomas Stockner
- Institute of Pharmacology and the Gaston H. Glock Research Laboratories for Exploratory Drug Development, Center of Physiology and Pharmacology, Medical University of Vienna, Vienna, Austria (C.V.F., A.E-K., M.H., A.C., W.S., T.S., M.F.); Institute of Organic Chemistry, University of Vienna, Vienna, Austria (Y.X., M.S., G.dM., M.P.F., N.M.); CeMM Research Center for Molecular Medicine of the Austrian Academy of Sciences, Vienna, Austria (Y.X., M.S., N.M.); AOP Orphan Pharmaceuticals GmbH, Vienna, Austria (C.K.); Christian Doppler Laboratory for Entropy-Oriented Drug Design, Institute of Organic Chemistry, University of Vienna, Vienna, Austria (N.M.); and NeGeMac Research Platform for Next Generation Macrocycles, Vienna, Austria (N.M.)
| | - Christoph Klade
- Institute of Pharmacology and the Gaston H. Glock Research Laboratories for Exploratory Drug Development, Center of Physiology and Pharmacology, Medical University of Vienna, Vienna, Austria (C.V.F., A.E-K., M.H., A.C., W.S., T.S., M.F.); Institute of Organic Chemistry, University of Vienna, Vienna, Austria (Y.X., M.S., G.dM., M.P.F., N.M.); CeMM Research Center for Molecular Medicine of the Austrian Academy of Sciences, Vienna, Austria (Y.X., M.S., N.M.); AOP Orphan Pharmaceuticals GmbH, Vienna, Austria (C.K.); Christian Doppler Laboratory for Entropy-Oriented Drug Design, Institute of Organic Chemistry, University of Vienna, Vienna, Austria (N.M.); and NeGeMac Research Platform for Next Generation Macrocycles, Vienna, Austria (N.M.)
| | - Nuno Maulide
- Institute of Pharmacology and the Gaston H. Glock Research Laboratories for Exploratory Drug Development, Center of Physiology and Pharmacology, Medical University of Vienna, Vienna, Austria (C.V.F., A.E-K., M.H., A.C., W.S., T.S., M.F.); Institute of Organic Chemistry, University of Vienna, Vienna, Austria (Y.X., M.S., G.dM., M.P.F., N.M.); CeMM Research Center for Molecular Medicine of the Austrian Academy of Sciences, Vienna, Austria (Y.X., M.S., N.M.); AOP Orphan Pharmaceuticals GmbH, Vienna, Austria (C.K.); Christian Doppler Laboratory for Entropy-Oriented Drug Design, Institute of Organic Chemistry, University of Vienna, Vienna, Austria (N.M.); and NeGeMac Research Platform for Next Generation Macrocycles, Vienna, Austria (N.M.)
| | - Michael Freissmuth
- Institute of Pharmacology and the Gaston H. Glock Research Laboratories for Exploratory Drug Development, Center of Physiology and Pharmacology, Medical University of Vienna, Vienna, Austria (C.V.F., A.E-K., M.H., A.C., W.S., T.S., M.F.); Institute of Organic Chemistry, University of Vienna, Vienna, Austria (Y.X., M.S., G.dM., M.P.F., N.M.); CeMM Research Center for Molecular Medicine of the Austrian Academy of Sciences, Vienna, Austria (Y.X., M.S., N.M.); AOP Orphan Pharmaceuticals GmbH, Vienna, Austria (C.K.); Christian Doppler Laboratory for Entropy-Oriented Drug Design, Institute of Organic Chemistry, University of Vienna, Vienna, Austria (N.M.); and NeGeMac Research Platform for Next Generation Macrocycles, Vienna, Austria (N.M.)
| |
Collapse
|
4
|
Schwartz AC, Stein RA, Gil-Iturbe E, Quick M, Mchaourab HS. Alternating access of a bacterial homolog of neurotransmitter: sodium symporters determined from AlphaFold2 ensembles and DEER spectroscopy. Proc Natl Acad Sci U S A 2024; 121:e2406063121. [PMID: 39302996 PMCID: PMC11459141 DOI: 10.1073/pnas.2406063121] [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: 03/25/2024] [Accepted: 08/16/2024] [Indexed: 09/22/2024] Open
Abstract
Neurotransmitter:sodium symporters (NSSs) play critical roles in neural signaling by regulating neurotransmitter uptake into cells powered by sodium electrochemical gradients. Bacterial NSSs orthologs, including MhsT from Bacillus halodurans, have emerged as model systems to understand the structural motifs of alternating access in NSSs and the extent of conservation of these motifs across the family. Here, we apply a computational/experimental methodology to illuminate the conformational landscape of MhsT alternating access. Capitalizing on our recently developed method, Sampling Protein Ensembles and Conformational Heterogeneity with AlphaFold2 (SPEACH_AF), we derived clusters of MhsT models spanning the transition from inward-facing to outward-facing conformations. Systematic application of double electron-electron resonance (DEER) spectroscopy revealed ligand-dependent movements of multiple structural motifs that underpin MhsT's conformational cycle. Remarkably, comparative DEER analysis in detergent micelles and lipid nanodiscs highlights the profound effect of the environment on the energetics of conformational changes. Through experimentally derived selection of collective variables, we present a model of ion and substrate-powered transport by MhsT consistent with the conformational cycle derived from DEER. Our findings not only advance the understanding of MhsT's function but also uncover motifs of conformational dynamics conserved within the broader context of the NSS family and within the LeuT-fold class of transporters. Importantly, our methodological blueprint introduces an approach that can be applied across a diverse spectrum of transporters to describe their conformational landscapes.
Collapse
Affiliation(s)
| | - Richard A. Stein
- Department of Molecular Physiology and Biophysics, Vanderbilt University, Nashville, TN37232
- Center for Applied AI in Protein Dynamics, Vanderbilt University, Nashville, TN37232
| | - Eva Gil-Iturbe
- Department of Psychiatry, Columbia University Irving Medical Center, New York, NY10032
| | - Matthias Quick
- Department of Psychiatry, Columbia University Irving Medical Center, New York, NY10032
- Department of Physiology and Cellular Biophysics, Columbia University Irving Medical Center, New York, NY10032
| | - Hassane S. Mchaourab
- Department of Molecular Physiology and Biophysics, Vanderbilt University, Nashville, TN37232
- Center for Applied AI in Protein Dynamics, Vanderbilt University, Nashville, TN37232
| |
Collapse
|
5
|
Johnson CB, Walther D, Baggott MJ, Baker LE, Baumann MH. Novel Benzofuran Derivatives Induce Monoamine Release and Substitute for the Discriminative Stimulus Effects of 3,4-Methylenedioxymethamphetamine. J Pharmacol Exp Ther 2024; 391:22-29. [PMID: 38272669 PMCID: PMC11413916 DOI: 10.1124/jpet.123.001837] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/13/2023] [Revised: 12/21/2023] [Accepted: 01/18/2024] [Indexed: 01/27/2024] Open
Abstract
3,4-Methylenedioxymethamphetamine (MDMA) has shown efficacy as a medication adjunct for treating post-traumatic stress disorder (PTSD). However, MDMA is also used in nonmedical contexts that pose risk for cardiovascular and neurologic complications. It is well established that MDMA exerts its effects by stimulating transporter-mediated release of the monoamines 5-hydroxytryptamine (5-HT), norepinephrine, and dopamine. Current research efforts are aimed at developing MDMA-like monoamine releasers with better efficacy and safety profiles. To this end, we investigated neurochemical and behavioral effects of novel analogs of the designer drug 5-(2-methylaminopropyl)benzofuran (5-MAPB). We used in vitro transporter assays in rat brain synaptosomes to examine transmitter uptake inhibition and releasing properties for enantiomers of 5-(2-methylaminobutyl)benzofuran (5-MABB) and 6-(2-methylaminobutyl)benzofuran (6-MABB) compared with MDMA. We then tested these same compounds in male Sprague-Dawley rats trained to discriminate MDMA (1.5 mg/kg) from saline. In vitro results revealed that S isomers of 5- and 6-MABB are efficacious releasing agents at transporters for 5-HT (SERT), norepinephrine (NET), and dopamine (DAT). By contrast, R isomers are efficacious releasers at SERT and partial releasers at NET but lack releasing activity at DAT. In vivo results showed that all compounds produce dose-dependent increases in MDMA-lever responding and full substitution at the highest dose tested. The diminished NET and DAT releasing activities for R isomers of 5- and 6-MABB are associated with reduced potency for inducing behavioral effects. Collectively, these findings indicate that the aminoalkyl benzofuran scaffold may be a viable template for developing compounds with MDMA-like properties. SIGNIFICANCE STATEMENT: Despite the clinical utility of 3,4-methylenedioxymethamphetamine (MDMA), the drug is associated with certain cardiovascular risks and metabolic side effects. Developing a therapeutic alternative with MDMA-like monoamine releasing activity is of interest. Our in vitro and in vivo findings indicate that the aminoalkyl benzofuran scaffold may be useful for developing compounds with MDMA-like properties.
Collapse
Affiliation(s)
- Candace B Johnson
- Department of Psychology, Western Michigan University, Kalamazoo, Michigan (C.B.J., L.E.B.); Designer Drug Research Unit, National Institute on Drug Abuse Intramural Research Program, Baltimore, Maryland (D.W., M.H.B.); and Tactogen Inc., Palo Alto, California (M.J.B.)
| | - Donna Walther
- Department of Psychology, Western Michigan University, Kalamazoo, Michigan (C.B.J., L.E.B.); Designer Drug Research Unit, National Institute on Drug Abuse Intramural Research Program, Baltimore, Maryland (D.W., M.H.B.); and Tactogen Inc., Palo Alto, California (M.J.B.)
| | - Matthew J Baggott
- Department of Psychology, Western Michigan University, Kalamazoo, Michigan (C.B.J., L.E.B.); Designer Drug Research Unit, National Institute on Drug Abuse Intramural Research Program, Baltimore, Maryland (D.W., M.H.B.); and Tactogen Inc., Palo Alto, California (M.J.B.)
| | - Lisa E Baker
- Department of Psychology, Western Michigan University, Kalamazoo, Michigan (C.B.J., L.E.B.); Designer Drug Research Unit, National Institute on Drug Abuse Intramural Research Program, Baltimore, Maryland (D.W., M.H.B.); and Tactogen Inc., Palo Alto, California (M.J.B.)
| | - Michael H Baumann
- Department of Psychology, Western Michigan University, Kalamazoo, Michigan (C.B.J., L.E.B.); Designer Drug Research Unit, National Institute on Drug Abuse Intramural Research Program, Baltimore, Maryland (D.W., M.H.B.); and Tactogen Inc., Palo Alto, California (M.J.B.)
| |
Collapse
|
6
|
Nourani N, Taghvimi A, Bavili-Tabrizi A, Javadzadeh Y, Dastmalchi S. Microextraction Techniques for Sample Preparation of Amphetamines in Urine: A Comprehensive Review. Crit Rev Anal Chem 2024; 54:1304-1319. [PMID: 36093632 DOI: 10.1080/10408347.2022.2113028] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/14/2022]
Abstract
Psychological disorders and dramatic social problems are serious concerns regarding the abuse of amphetamine and its stimulant derivatives worldwide. Consumers of such drugs experience great euphoria along with serious health problems. Determination and quantification of amphetamine-type stimulants are indispensable skills for clinical and forensic laboratories. Analysis of low drug doses in bio-matrices necessitates applications of simple and also effective preparation steps. The preparation procedures not only eliminate adverse matrix effects, but also provide reasonable clean-up and pre-concentration benefits. The current review presents different methods used for sample preparation of amphetamines from urine as the most frequently used biological matrix. The advantages and limitations of various sample preparation methods were discussed focusing on the miniaturized methods.
Collapse
Affiliation(s)
- Nasim Nourani
- Student Research Committee, Tabriz University of Medical Sciences, Tabriz, Iran
- Biotechnology Research Center, Tabriz University of Medical Sciences, Tabriz, Iran
- Department of Medicinal Chemistry, Faculty of Pharmacy, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Arezou Taghvimi
- Biotechnology Research Center, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Ahad Bavili-Tabrizi
- Department of Medicinal Chemistry, Faculty of Pharmacy, Tabriz University of Medical Sciences, Tabriz, Iran
- Pharmaceutical Analysis Research Center and Faculty of Pharmacy, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Yousef Javadzadeh
- Biotechnology Research Center, Tabriz University of Medical Sciences, Tabriz, Iran
- Department of Pharmaceutics, Faculty of Pharmacy, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Siavoush Dastmalchi
- Biotechnology Research Center, Tabriz University of Medical Sciences, Tabriz, Iran
- Department of Medicinal Chemistry, Faculty of Pharmacy, Tabriz University of Medical Sciences, Tabriz, Iran
- Faculty of Pharmacy, Near East University, North Cyprus, Turkey
| |
Collapse
|
7
|
Supplisson S. Dynamic role of GlyT1 as glycine sink or source: Pharmacological implications for the gain control of NMDA receptors. Neuroscience 2024:S0306-4522(24)00350-6. [PMID: 39059742 DOI: 10.1016/j.neuroscience.2024.07.037] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/27/2024] [Revised: 07/03/2024] [Accepted: 07/22/2024] [Indexed: 07/28/2024]
Abstract
Glycine transporter 1 (GlyT1) mediates the termination of inhibitory glycinergic receptor signaling in the spinal cord and brainstem, and is also present diffusely in the forebrain. Here, it regulates the ambient glycine concentration and influences the 'glycine' site occupancy of N-methyl-d-aspartate receptors (NMDARs). GlyT1 is a reversible transporter with a substantial, but not excessive, sodium-motive force for uphill transport. This study investigates its role as a potential source of glycine supply, either by reverse uptake or heteroexchange. Indeed, glutamate alone does not induce NMDAR current in "naive" oocytes co-expressing GluN1/GluN2A and GlyT1, a previously characterized cellular model. However, after substantial intracellular glycine accumulation, GlyT1 reverses its transport mode, and begins to release glycine into the external compartment, allowing NMDAR activation by glutamate alone. These uptake-dependent glutamate currents were blocked by ALX-5407 and potentiated by sarcosine, a specific inhibitor and substrate of GlyT1, respectively, suggesting a higher occupancy of the co-agonist site when GlyT1 functions as a glycine source either by reversed-uptake or by heteroexchange. These two glycine release mechanisms can be distinguished by their voltage dependence, as the reversed-uptake cycle decreases at hyperpolarized potentials, whereas heteroexchange electroneutrality preserves glycine efflux and NMDAR activation at these potentials. These results establish GlyT1-mediated efflux as a positive regulator of NMDAR coagonist site occupancy, and demonstrate the efficacy of sarcosine heteroexchange in enhancing coagonist site occupancy. Because NMDAR facilitation by GlyT1-inhibitors and sarcosine relies on different transport mechanisms, their actions may be a source of variability in reversing NMDAR hypofunction in schizophrenia.
Collapse
Affiliation(s)
- Stéphane Supplisson
- Institut de Biologie de l'ENS (IBENS), Ecole normale supérieure, Université PSL, CNRS, INSERM, Paris, F-75005, France.
| |
Collapse
|
8
|
Jiménez-Torres AC, Porter KD, Hastie JA, Adeniran C, Moukha-Chafiq O, Nguyen TH, Ananthan S, Augelli-Szafran CE, Zhan CG, Zhu J. Effects of SRI-32743, a Novel Quinazoline Structure-Based Compound, on HIV-1 Tat and Cocaine Interaction with Norepinephrine Transporter. Int J Mol Sci 2024; 25:7881. [PMID: 39063123 PMCID: PMC11277056 DOI: 10.3390/ijms25147881] [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: 07/05/2024] [Revised: 07/12/2024] [Accepted: 07/15/2024] [Indexed: 07/28/2024] Open
Abstract
Prolonged exposure to HIV-1 transactivator of transcription (Tat) protein dysregulates monoamine transmission, a physiological change implicated as a key factor in promoting neurocognitive disorders among people living with HIV. We have demonstrated that in vivo expression of Tat in Tat transgenic mice decreases dopamine uptake through both dopamine transporter (DAT) and norepinephrine transporter (NET) in the prefrontal cortex. Further, our novel allosteric inhibitor of monoamine transporters, SRI-32743, has been shown to attenuate Tat-inhibited dopamine transport through DAT and alleviates Tat-potentiated cognitive impairments. The current study reports the pharmacological profiles of SRI-32743 in basal and Tat-induced inhibition of human NET (hNET) function. SRI-32743 exhibited less affinity for hNET binding than desipramine, a classical NET inhibitor, but displayed similar potency for inhibiting hDAT and hNET activity. SRI-32743 concentration-dependently increased hNET affinity for [3H]DA uptake but preserved the Vmax of dopamine transport. SRI-32743 slowed the cocaine-mediated dissociation of [3H]Nisoxetine binding and reduced both [3H]DA and [3H]MPP+ efflux but did not affect d-amphetamine-mediated [3H]DA release through hNET. Finally, we determined that SRI-32743 attenuated a recombinant Tat1-86-induced decrease in [3H]DA uptake via hNET. Our findings demonstrated that SRI-32743 allosterically disrupts the recombinant Tat1-86-hNET interaction, suggesting a potential treatment for HIV-infected individuals with concurrent cocaine abuse.
Collapse
Affiliation(s)
- Ana Catya Jiménez-Torres
- Department of Drug Discovery and Biomedical Sciences, College of Pharmacy, University of South Carolina, 715 Sumter Street, Columbia, SC 29208, USA; (A.C.J.-T.); (K.D.P.); (J.A.H.)
| | - Katherine D. Porter
- Department of Drug Discovery and Biomedical Sciences, College of Pharmacy, University of South Carolina, 715 Sumter Street, Columbia, SC 29208, USA; (A.C.J.-T.); (K.D.P.); (J.A.H.)
| | - Jamison A. Hastie
- Department of Drug Discovery and Biomedical Sciences, College of Pharmacy, University of South Carolina, 715 Sumter Street, Columbia, SC 29208, USA; (A.C.J.-T.); (K.D.P.); (J.A.H.)
| | - Charles Adeniran
- Department of Pharmaceutical Sciences, College of Pharmacy, University of Kentucky, Lexington, KY 40536, USA; (C.A.); (C.-G.Z.)
- Molecular Modeling and Biopharmaceutical Center, College of Pharmacy, University of Kentucky, Lexington, KY 40536, USA
| | - Omar Moukha-Chafiq
- Department of Chemistry, Scientific Platforms Division, Southern Research, Birmingham, AL 35205, USA; (O.M.-C.); (T.H.N.); (S.A.); (C.E.A.-S.)
| | - Theresa H. Nguyen
- Department of Chemistry, Scientific Platforms Division, Southern Research, Birmingham, AL 35205, USA; (O.M.-C.); (T.H.N.); (S.A.); (C.E.A.-S.)
| | - Subramaniam Ananthan
- Department of Chemistry, Scientific Platforms Division, Southern Research, Birmingham, AL 35205, USA; (O.M.-C.); (T.H.N.); (S.A.); (C.E.A.-S.)
| | - Corinne E. Augelli-Szafran
- Department of Chemistry, Scientific Platforms Division, Southern Research, Birmingham, AL 35205, USA; (O.M.-C.); (T.H.N.); (S.A.); (C.E.A.-S.)
| | - Chang-Guo Zhan
- Department of Pharmaceutical Sciences, College of Pharmacy, University of Kentucky, Lexington, KY 40536, USA; (C.A.); (C.-G.Z.)
- Molecular Modeling and Biopharmaceutical Center, College of Pharmacy, University of Kentucky, Lexington, KY 40536, USA
| | - Jun Zhu
- Department of Drug Discovery and Biomedical Sciences, College of Pharmacy, University of South Carolina, 715 Sumter Street, Columbia, SC 29208, USA; (A.C.J.-T.); (K.D.P.); (J.A.H.)
| |
Collapse
|
9
|
El-Kasaby A, Boytsov D, Kasture A, Krumpl G, Hummel T, Freissmuth M, Sandtner W. Allosteric Inhibition and Pharmacochaperoning of the Serotonin Transporter by the Antidepressant Drugs Trazodone and Nefazodone. Mol Pharmacol 2024; 106:56-70. [PMID: 38769018 DOI: 10.1124/molpharm.124.000881] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/23/2024] [Revised: 05/06/2024] [Accepted: 05/07/2024] [Indexed: 05/22/2024] Open
Abstract
The antidepressants trazodone and nefazodone were approved some 4 and 3 decades ago, respectively. Their action is thought to be mediated, at least in part, by inhibition of the serotonin transporter [SERT/solute carrier (SLC)-6A4]. Surprisingly, their mode of action on SERT has not been characterized. Here, we show that, similar to the chemically related drug vilazodone, trazodone and nefazodone are allosteric ligands: trazodone and nefazodone inhibit uptake by and transport-associated currents through SERT in a mixed-competitive and noncompetitive manner, respectively. Contrary to noribogaine and its congeners, all three compounds preferentially interact with the Na+-bound outward-facing state of SERT. Nevertheless, they act as pharmacochaperones and rescue the folding-deficient variant SERT-P601A/G602A. The vast majority of disease-associated point mutations of SLC6 family members impair folding of the encoded transporter proteins. Our findings indicate that their folding defect can be remedied by targeting allosteric sites on SLC6 transporters. SIGNIFICANCE STATEMENT: The serotonin transporter is a member of the solute carrier-6 family and is the target of numerous antidepressants. Trazodone and nefazodone have long been used as antidepressants. Here, this study shows that their inhibition of the serotonin transporter digressed from the competitive mode seen with other antidepressants. Trazodone and nefazodone rescued a folding-deficient variant of the serotonin transporter. This finding demonstrates that folding defects of mutated solute carrier-6 family members can also be corrected by allosteric ligands.
Collapse
Affiliation(s)
- Ali El-Kasaby
- Institute of Pharmacology and the Gaston H. Glock Research Laboratories for Exploratory Drug Development, Centre of Physiology and Pharmacology (A.E.-K., D.B., M.F., W.S.), Medical University of Vienna, Vienna, Austria; Department of Neurobiology, University of Vienna, Vienna, Austria (A.K., T.H.); and MRN Medical Research Network GmbH, Vienna, Austria (G.K.)
| | - Danila Boytsov
- Institute of Pharmacology and the Gaston H. Glock Research Laboratories for Exploratory Drug Development, Centre of Physiology and Pharmacology (A.E.-K., D.B., M.F., W.S.), Medical University of Vienna, Vienna, Austria; Department of Neurobiology, University of Vienna, Vienna, Austria (A.K., T.H.); and MRN Medical Research Network GmbH, Vienna, Austria (G.K.)
| | - Ameya Kasture
- Institute of Pharmacology and the Gaston H. Glock Research Laboratories for Exploratory Drug Development, Centre of Physiology and Pharmacology (A.E.-K., D.B., M.F., W.S.), Medical University of Vienna, Vienna, Austria; Department of Neurobiology, University of Vienna, Vienna, Austria (A.K., T.H.); and MRN Medical Research Network GmbH, Vienna, Austria (G.K.)
| | - Günther Krumpl
- Institute of Pharmacology and the Gaston H. Glock Research Laboratories for Exploratory Drug Development, Centre of Physiology and Pharmacology (A.E.-K., D.B., M.F., W.S.), Medical University of Vienna, Vienna, Austria; Department of Neurobiology, University of Vienna, Vienna, Austria (A.K., T.H.); and MRN Medical Research Network GmbH, Vienna, Austria (G.K.)
| | - Thomas Hummel
- Institute of Pharmacology and the Gaston H. Glock Research Laboratories for Exploratory Drug Development, Centre of Physiology and Pharmacology (A.E.-K., D.B., M.F., W.S.), Medical University of Vienna, Vienna, Austria; Department of Neurobiology, University of Vienna, Vienna, Austria (A.K., T.H.); and MRN Medical Research Network GmbH, Vienna, Austria (G.K.)
| | - Michael Freissmuth
- Institute of Pharmacology and the Gaston H. Glock Research Laboratories for Exploratory Drug Development, Centre of Physiology and Pharmacology (A.E.-K., D.B., M.F., W.S.), Medical University of Vienna, Vienna, Austria; Department of Neurobiology, University of Vienna, Vienna, Austria (A.K., T.H.); and MRN Medical Research Network GmbH, Vienna, Austria (G.K.)
| | - Walter Sandtner
- Institute of Pharmacology and the Gaston H. Glock Research Laboratories for Exploratory Drug Development, Centre of Physiology and Pharmacology (A.E.-K., D.B., M.F., W.S.), Medical University of Vienna, Vienna, Austria; Department of Neurobiology, University of Vienna, Vienna, Austria (A.K., T.H.); and MRN Medical Research Network GmbH, Vienna, Austria (G.K.)
| |
Collapse
|
10
|
Mattingly GW, Carbray JA, Roy P, López FA. Are all ADHD medications created equal? Exploring the differences that enable evening dosing. Postgrad Med 2024; 136:475-486. [PMID: 38904469 DOI: 10.1080/00325481.2024.2370230] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/15/2024] [Accepted: 06/17/2024] [Indexed: 06/22/2024]
Abstract
With more than 30 available stimulant medications, choosing among therapeutic options for attention-deficit/hyperactivity disorder (ADHD) has become increasingly complex and patient specific. All ADHD stimulants owe their action to variants of either amphetamine or methylphenidate, yet formulation and delivery system differences create unique pharmacokinetic and clinical profiles for each medication. A benefit of the diversity within ADHD pharmacotherapy is that it facilitates tailoring treatment to meet patient needs. Historically, there has been a constant among long-acting stimulant options, regardless of formulation, which was morning dosing. The introduction of delayed-release and extended-release methylphenidate (DR/ER-MPH) is the first long-acting stimulant that patients take in the evening, with the clinical effect delayed until awakening in the morning. This paradigm shift has generated questions among clinicians and continued interest in real-world experience and data. This review used available clinical data, real-world evidence, emerging analyses, and clinical experience to evaluate the characteristics of DR/ER-MPH and its clinical utility within the greater context of ADHD medications and to provide clinicians with practical guidance on the use of DR/ER-MPH in children, adolescents, and adults with ADHD.
Collapse
Affiliation(s)
- Gregory W Mattingly
- Department of Psychiatry, Washington University School of Medicine, St. Louis, MO, USA
- Midwest Research Group, St. Charles, MO, USA
- St. Charles Psychiatric Associates, St. Charles, MO, USA
| | - Julie A Carbray
- Department of Psychiatry, University of Illinois Chicago, Chicago, IL, USA
| | - Perry Roy
- Carolina Attention Specialists, Charlotte, NC, USA
| | - Frank A López
- Pediatrix Neurology and Epilepsy Center, Winter Park, FL, USA
| |
Collapse
|
11
|
Bukowski L, Strøm ME, Andersen JL, Maesen JB, Tian L, Sinning S. 5-HT_FAsTR: a versatile, label-free, high-throughput, fluorescence-based microplate assay to quantify serotonin transport and release. Sci Rep 2024; 14:6541. [PMID: 38504103 PMCID: PMC10951269 DOI: 10.1038/s41598-024-56712-z] [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: 10/27/2023] [Accepted: 03/09/2024] [Indexed: 03/21/2024] Open
Abstract
The neurotransmitter serotonin plays a pivotal role in mood and depression. It also acts as a vasoconstrictor within blood vessels and is the main neurotransmitter in the gastrointestinal system. In neurotransmission, released serotonin is taken up by serotonin transporters, which are principal targets of antidepressants and the psychostimulant, ecstasy. The investigation of serotonin transporters have relied almost exclusively on the use of radiolabeled serotonin in heterogenous end-point assays. Here we adapt the genetically encoded fluorescent biosensor, iSeroSnFR, to establish and validate the Serotonin (5-HT) Fluorescence Assay for Transport and Release (5-HT_FAsTR) for functional and pharmacological studies of serotonin transport and release. We demonstrate the applicability of the method for the study of a neuronal, high-affinity, low-capacity serotonin transporter (SERT) as well as an extraneuronal low-affinity, high-capacity organic cation transporter and mutants thereof. 5HT_FAsTR offers an accessible, versatile and reliable semi-homogenous assay format that only relies on a fluorescence plate reader for repeated, real-time measurements of serotonin influx and efflux. 5HT_FAsTR accelerates and democratizes functional characterization and pharmacological studies of serotonin transporters and genetic variants thereof in disease states such as depression, anxiety and ADHD.
Collapse
Affiliation(s)
- Lina Bukowski
- Department of Forensic Medicine, Aarhus University, Palle Juul-Jensens Boulevard 99, 8200, Aarhus N, Denmark
| | - Markus Emanuel Strøm
- Department of Forensic Medicine, Aarhus University, Palle Juul-Jensens Boulevard 99, 8200, Aarhus N, Denmark
| | - Jens Lindengren Andersen
- Department of Forensic Medicine, Aarhus University, Palle Juul-Jensens Boulevard 99, 8200, Aarhus N, Denmark
| | - Jannick Bang Maesen
- Department of Forensic Medicine, Aarhus University, Palle Juul-Jensens Boulevard 99, 8200, Aarhus N, Denmark
| | - Lin Tian
- Department of Biochemistry and Molecular Medicine, University of California, Davis, CA, 95616-8635, USA
- Max Planck Florida Institute for Neuroscience, Jupiter, FL, 33458, USA
| | - Steffen Sinning
- Department of Forensic Medicine, Aarhus University, Palle Juul-Jensens Boulevard 99, 8200, Aarhus N, Denmark.
| |
Collapse
|
12
|
Vaughan RA, Henry LK, Foster JD, Brown CR. Post-translational mechanisms in psychostimulant-induced neurotransmitter efflux. ADVANCES IN PHARMACOLOGY (SAN DIEGO, CALIF.) 2024; 99:1-33. [PMID: 38467478 DOI: 10.1016/bs.apha.2023.10.003] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 03/13/2024]
Abstract
The availability of monoamine neurotransmitters in the brain is under the control of dopamine, norepinephrine, and serotonin transporters expressed on the plasma membrane of monoaminergic neurons. By regulating transmitter levels these proteins mediate crucial functions including cognition, attention, and reward, and dysregulation of their activity is linked to mood and psychiatric disorders of these systems. Amphetamine-based transporter substrates stimulate non-exocytotic transmitter efflux that induces psychomotor stimulation, addiction, altered mood, hallucinations, and psychosis, thus constituting a major component of drug neurochemical and behavioral outcomes. Efflux is under the control of transporter post-translational modifications that synergize with other regulatory events, and this review will summarize our knowledge of these processes and their role in drug mechanisms.
Collapse
Affiliation(s)
- Roxanne A Vaughan
- Department of Biomedical Sciences, University of North Dakota School of Medicine and Health Sciences, Grand Forks, ND, United States.
| | - L Keith Henry
- Department of Biomedical Sciences, University of North Dakota School of Medicine and Health Sciences, Grand Forks, ND, United States
| | - James D Foster
- Department of Biomedical Sciences, University of North Dakota School of Medicine and Health Sciences, Grand Forks, ND, United States
| | - Christopher R Brown
- Department of Biomedical Sciences, University of North Dakota School of Medicine and Health Sciences, Grand Forks, ND, United States
| |
Collapse
|
13
|
Loland CJ, Wellendorph P, Pedersen SF, Gether U. Transmembrane transporter proteins: Capturing transport in motion. Basic Clin Pharmacol Toxicol 2024; 134:203-205. [PMID: 37945540 DOI: 10.1111/bcpt.13960] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/12/2023]
Affiliation(s)
- Claus J Loland
- Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark
| | - Petrine Wellendorph
- Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark
| | - Stine F Pedersen
- Faculty of Science, University of Copenhagen, Copenhagen, Denmark
| | - Ulrik Gether
- Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark
| |
Collapse
|
14
|
Mayer FP, Stewart A, Blakely RD. Leaky lessons learned: Efflux prone dopamine transporter variant reveals sex and circuit specific contributions of D2 receptor signalling to neuropsychiatric disease. Basic Clin Pharmacol Toxicol 2024; 134:206-218. [PMID: 37987120 DOI: 10.1111/bcpt.13964] [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: 07/07/2023] [Revised: 11/14/2023] [Accepted: 11/17/2023] [Indexed: 11/22/2023]
Abstract
Aberrant dopamine (DA) signalling has been implicated in various neuropsychiatric disorders, including attention-deficit/hyperactivity disorder (ADHD), autism spectrum disorder (ASD), schizophrenia, bipolar disorder (BPD) and addiction. The availability of extracellular DA is sculpted by the exocytotic release of vesicular DA and subsequent transporter-mediated clearance, rendering the presynaptic DA transporter (DAT) a crucial regulator of DA neurotransmission. D2-type DA autoreceptors (D2ARs) regulate multiple aspects of DA homeostasis, including (i) DA synthesis, (ii) vesicular release, (iii) DA neuron firing and (iv) the surface expression of DAT and DAT-mediated DA clearance. The DAT Val559 variant, identified in boys with ADHD or ASD, as well as in a girl with BPD, supports anomalous DA efflux (ADE), which we have shown drives tonic activation of D2ARs. Through ex vivo and in vivo studies of the DAT Val559 variant using transgenic knock-in mice, we have uncovered a circuit and sex-specific capacity of D2ARs to regulate DAT, which consequently disrupts DA signalling and behaviour differently in males and females. Our studies reveal the ability of the construct-valid DAT Val559 model to elucidate endogenous mechanisms that support DA signalling, findings that may be of translational and/or therapeutic importance.
Collapse
Affiliation(s)
- Felix P Mayer
- Department of Biomedical Science, Florida Atlantic University, Jupiter, Florida, USA
- Stiles-Nicholson Brain Institute, Florida Atlantic University, Jupiter, Florida, USA
| | - Adele Stewart
- Department of Biomedical Science, Florida Atlantic University, Jupiter, Florida, USA
- Stiles-Nicholson Brain Institute, Florida Atlantic University, Jupiter, Florida, USA
| | - Randy D Blakely
- Department of Biomedical Science, Florida Atlantic University, Jupiter, Florida, USA
- Stiles-Nicholson Brain Institute, Florida Atlantic University, Jupiter, Florida, USA
| |
Collapse
|
15
|
Yu J, Xu Z, Yan W, Shao Z. Elucidating the molecular pharmacology of trace amine-associated receptor 1 to advance antipsychotic drug discovery. Clin Transl Med 2024; 14:e1576. [PMID: 38317588 PMCID: PMC10844839 DOI: 10.1002/ctm2.1576] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/20/2024] [Accepted: 01/24/2024] [Indexed: 02/07/2024] Open
Affiliation(s)
- Jingjing Yu
- Division of Nephrology and Kidney Research InstituteState Key Laboratory of Biotherapy, West China Hospital, Sichuan UniversityChengduChina
| | - Zheng Xu
- Division of Nephrology and Kidney Research InstituteState Key Laboratory of Biotherapy, West China Hospital, Sichuan UniversityChengduChina
| | - Wei Yan
- Division of Nephrology and Kidney Research InstituteState Key Laboratory of Biotherapy, West China Hospital, Sichuan UniversityChengduChina
| | - Zhenhua Shao
- Division of Nephrology and Kidney Research InstituteState Key Laboratory of Biotherapy, West China Hospital, Sichuan UniversityChengduChina
- Frontiers Medical Center, Tianfu Jincheng LaboratoryChengduChina
| |
Collapse
|
16
|
Gradisch R, Schlögl K, Lazzarin E, Niello M, Maier J, Mayer FP, Alves da Silva L, Skopec SMC, Blakely RD, Sitte HH, Mihovilovic MD, Stockner T. Ligand coupling mechanism of the human serotonin transporter differentiates substrates from inhibitors. Nat Commun 2024; 15:417. [PMID: 38195746 PMCID: PMC10776687 DOI: 10.1038/s41467-023-44637-6] [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: 07/17/2023] [Accepted: 12/22/2023] [Indexed: 01/11/2024] Open
Abstract
The presynaptic serotonin transporter (SERT) clears extracellular serotonin following vesicular release to ensure temporal and spatial regulation of serotonergic signalling and neurotransmitter homeostasis. Prescription drugs used to treat neurobehavioral disorders, including depression, anxiety, and obsessive-compulsive disorder, trap SERT by blocking the transport cycle. In contrast, illicit drugs of abuse like amphetamines reverse SERT directionality, causing serotonin efflux. Both processes result in increased extracellular serotonin levels. By combining molecular dynamics simulations with biochemical experiments and using a homologous series of serotonin analogues, we uncovered the coupling mechanism between the substrate and the transporter, which triggers the uptake of serotonin. Free energy analysis showed that only scaffold-bound substrates could initiate SERT occlusion through attractive long-range electrostatic interactions acting on the bundle domain. The associated spatial requirements define substrate and inhibitor properties, enabling additional possibilities for rational drug design approaches.
Collapse
Affiliation(s)
- Ralph Gradisch
- Medical University of Vienna, Institute of Physiology and Pharmacology, Waehringer Straße 13A, 1090, Vienna, Austria
| | - Katharina Schlögl
- TU Wien, Institute of Applied Synthetic Chemistry, Getreidemarkt 9, 1060, Vienna, Austria
| | - Erika Lazzarin
- Medical University of Vienna, Institute of Physiology and Pharmacology, Waehringer Straße 13A, 1090, Vienna, Austria
| | - Marco Niello
- Medical University of Vienna, Institute of Physiology and Pharmacology, Waehringer Straße 13A, 1090, Vienna, Austria
- Genetics of Cognition Laboratory, Neuroscience area, Istituto Italiano di Tecnologia, via Morego, 30, 16163, Genova, Italy
| | - Julian Maier
- Medical University of Vienna, Institute of Physiology and Pharmacology, Waehringer Straße 13A, 1090, Vienna, Austria
| | - Felix P Mayer
- Florida Atlantic University, Department of Biomedical Science, Jupiter, FL, 33458, USA
- Stiles-Nicholson Brain Institute, Jupiter, FL, 33458, USA
- Department of Neuroscience, Faculty of Health and Medical Sciences, University of Copenhagen, DK-2200, Copenhagen, Denmark
| | - Leticia Alves da Silva
- Medical University of Vienna, Institute of Physiology and Pharmacology, Waehringer Straße 13A, 1090, Vienna, Austria
| | - Sophie M C Skopec
- Medical University of Vienna, Institute of Physiology and Pharmacology, Waehringer Straße 13A, 1090, Vienna, Austria
| | - Randy D Blakely
- Florida Atlantic University, Department of Biomedical Science, Jupiter, FL, 33458, USA
- Stiles-Nicholson Brain Institute, Jupiter, FL, 33458, USA
| | - Harald H Sitte
- Medical University of Vienna, Institute of Physiology and Pharmacology, Waehringer Straße 13A, 1090, Vienna, Austria
- Al-Ahliyya Amman University, Hourani Center for Applied Scientific Research, Amman, Jordan
- Medical University of Vienna, Center for Addiction Research and Science, Waehringer Straße 13A, 1090, Vienna, Austria
| | - Marko D Mihovilovic
- TU Wien, Institute of Applied Synthetic Chemistry, Getreidemarkt 9, 1060, Vienna, Austria
| | - Thomas Stockner
- Medical University of Vienna, Institute of Physiology and Pharmacology, Waehringer Straße 13A, 1090, Vienna, Austria.
| |
Collapse
|
17
|
Paul A, Shukla D. Oligomerization of Monoamine Transporters. Subcell Biochem 2024; 104:119-137. [PMID: 38963486 DOI: 10.1007/978-3-031-58843-3_7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 07/05/2024]
Abstract
Transporters of the monoamine transporter (MAT) family regulate the uptake of important neurotransmitters like dopamine, serotonin, and norepinephrine. The MAT family functions using the electrochemical gradient of ions across the membrane and comprises three transporters, dopamine transporter (DAT), serotonin transporter (SERT), and norepinephrine transporter (NET). MAT transporters have been observed to exist in monomeric states to higher-order oligomeric states. Structural features, allosteric modulation, and lipid environment regulate the oligomerization of MAT transporters. NET and SERT oligomerization are regulated by levels of PIP2 present in the membrane. The kink present in TM12 in the MAT family is crucial for dimer interface formation. Allosteric modulation in the dimer interface hinders dimer formation. Oligomerization also influences the transporters' function, trafficking, and regulation. This chapter will focus on recent studies on monoamine transporters and discuss the factors affecting their oligomerization and its impact on their function.
Collapse
Affiliation(s)
- Arnav Paul
- Department of Chemistry, University of Illinois Urbana-Champaign, Urbana, IL, USA
| | - Diwakar Shukla
- Department of Chemical and Biomolecular Engineering, Department of Bioengineering, Center for Biophysics and Quantitative Biology, Department of Plant Biology, University of Illinois at Urbana-Champaign, Urbana, IL, USA.
| |
Collapse
|
18
|
Kukułowicz J, Pietrzak-Lichwa K, Klimończyk K, Idlin N, Bajda M. The SLC6A15-SLC6A20 Neutral Amino Acid Transporter Subfamily: Functions, Diseases, and Their Therapeutic Relevance. Pharmacol Rev 2023; 76:142-193. [PMID: 37940347 DOI: 10.1124/pharmrev.123.000886] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/23/2023] [Revised: 09/07/2023] [Accepted: 11/02/2023] [Indexed: 11/10/2023] Open
Abstract
The neutral amino acid transporter subfamily that consists of six members, consecutively SLC6A15-SLC620, also called orphan transporters, represents membrane, sodium-dependent symporter proteins that belong to the family of solute carrier 6 (SLC6). Primarily, they mediate the transport of neutral amino acids from the extracellular milieu toward cell or storage vesicles utilizing an electric membrane potential as the driving force. Orphan transporters are widely distributed throughout the body, covering many systems; for instance, the central nervous, renal, or intestinal system, supplying cells into molecules used in biochemical, signaling, and building pathways afterward. They are responsible for intestinal absorption and renal reabsorption of amino acids. In the central nervous system, orphan transporters constitute a significant medium for the provision of neurotransmitter precursors. Diseases related with aforementioned transporters highlight their significance; SLC6A19 mutations are associated with metabolic Hartnup disorder, whereas altered expression of SLC6A15 has been associated with a depression/stress-related disorders. Mutations of SLC6A18-SLCA20 cause iminoglycinuria and/or hyperglycinuria. SLC6A18-SLC6A20 to reach the cellular membrane require an ancillary unit ACE2 that is a molecular target for the spike protein of the SARS-CoV-2 virus. SLC6A19 has been proposed as a molecular target for the treatment of metabolic disorders resembling gastric surgery bypass. Inhibition of SLC6A15 appears to have a promising outcome in the treatment of psychiatric disorders. SLC6A19 and SLC6A20 have been suggested as potential targets in the treatment of COVID-19. In this review, we gathered recent advances on orphan transporters, their structure, functions, related disorders, and diseases, and in particular their relevance as therapeutic targets. SIGNIFICANCE STATEMENT: The following review systematizes current knowledge about the SLC6A15-SLCA20 neutral amino acid transporter subfamily and their therapeutic relevance in the treatment of different diseases.
Collapse
Affiliation(s)
- Jędrzej Kukułowicz
- Department of Physicochemical Drug Analysis, Faculty of Pharmacy, Jagiellonian University Medical College, Krakow, Poland
| | - Krzysztof Pietrzak-Lichwa
- Department of Physicochemical Drug Analysis, Faculty of Pharmacy, Jagiellonian University Medical College, Krakow, Poland
| | - Klaudia Klimończyk
- Department of Physicochemical Drug Analysis, Faculty of Pharmacy, Jagiellonian University Medical College, Krakow, Poland
| | - Nathalie Idlin
- Department of Physicochemical Drug Analysis, Faculty of Pharmacy, Jagiellonian University Medical College, Krakow, Poland
| | - Marek Bajda
- Department of Physicochemical Drug Analysis, Faculty of Pharmacy, Jagiellonian University Medical College, Krakow, Poland
| |
Collapse
|
19
|
Mayer FP, Niello M, Bulling S, Zhang YW, Li Y, Kudlacek O, Holy M, Kooti F, Sandtner W, Rudnick G, Schmid D, Sitte HH. Mephedrone induces partial release at human dopamine transporters but full release at human serotonin transporters. Neuropharmacology 2023; 240:109704. [PMID: 37703919 DOI: 10.1016/j.neuropharm.2023.109704] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/19/2022] [Revised: 07/07/2023] [Accepted: 08/31/2023] [Indexed: 09/15/2023]
Abstract
Mephedrone (4-methylmethcathinone) is a cathinone derivative that is recreationally consumed for its energizing and empathogenic effects. The stimulating properties are believed to arise from the ability of mephedrone to interact with the high-affinity transporters for dopamine (DA) (DAT) and norepinephrine (NET), whereas the entactogenic effect presumably relies on its activity at the serotonin (5-HT) transporter (SERT). Early studies found that mephedrone acts as a releaser at NET, DAT and SERT, and thus promotes efflux of the respective monoamines. Evidence linked drug-induced reverse transport of 5-HT via SERT to prosocial effects, whereas activity at DAT is strongly correlated with abuse liability. Consequently, we sought to evaluate the pharmacology of mephedrone at human (h) DAT and SERT, heterologously expressed in human embryonic kidney 293 cells, in further detail. In line with previous studies, we report that mephedrone evokes carrier-mediated release via hDAT and hSERT. We found this effect to be sensitive to the protein kinase C inhibitor GF109203X. Electrophysiological recordings revealed that mephedrone is actively transported by hDAT and hSERT. However, mephedrone acts as a full substrate of hSERT but as a partial substrate of hDAT. Furthermore, when compared to fully efficacious releasing agents at hDAT and hSERT (i.e. S(+)-amphetamine and para-chloroamphetamine, respectively) mephedrone displays greater efficacy as a releaser at hSERT than at hDAT. In summary, this study provides additional insights into the molecular mechanism of action of mephedrone at hDAT and hSERT.
Collapse
Affiliation(s)
- Felix P Mayer
- Institute of Pharmacology, Center for Physiology and Pharmacology, Medical University of Vienna, Waehringer Strasse 13a, A-1090, Vienna, Austria
| | - Marco Niello
- Institute of Pharmacology, Center for Physiology and Pharmacology, Medical University of Vienna, Waehringer Strasse 13a, A-1090, Vienna, Austria
| | - Simon Bulling
- Institute of Pharmacology, Center for Physiology and Pharmacology, Medical University of Vienna, Waehringer Strasse 13a, A-1090, Vienna, Austria
| | - Yuan-Wei Zhang
- Department of Pharmacology, Yale University School of Medicine, 333 Cedar Street, New Haven, CT, 06520-8066, USA
| | - Yang Li
- Institute of Pharmacology, Center for Physiology and Pharmacology, Medical University of Vienna, Waehringer Strasse 13a, A-1090, Vienna, Austria
| | - Oliver Kudlacek
- Institute of Pharmacology, Center for Physiology and Pharmacology, Medical University of Vienna, Waehringer Strasse 13a, A-1090, Vienna, Austria
| | - Marion Holy
- Institute of Pharmacology, Center for Physiology and Pharmacology, Medical University of Vienna, Waehringer Strasse 13a, A-1090, Vienna, Austria
| | - Fatemeh Kooti
- Institute of Pharmacology, Center for Physiology and Pharmacology, Medical University of Vienna, Waehringer Strasse 13a, A-1090, Vienna, Austria
| | - Walter Sandtner
- Institute of Pharmacology, Center for Physiology and Pharmacology, Medical University of Vienna, Waehringer Strasse 13a, A-1090, Vienna, Austria
| | - Gary Rudnick
- Department of Pharmacology, Yale University School of Medicine, 333 Cedar Street, New Haven, CT, 06520-8066, USA
| | - Diethart Schmid
- Institute for Physiology, Center for Physiology and Pharmacology, Medical University of Vienna, Waehringer Strasse 13a, A-1090, Vienna, Austria
| | - Harald H Sitte
- Institute of Pharmacology, Center for Physiology and Pharmacology, Medical University of Vienna, Waehringer Strasse 13a, A-1090, Vienna, Austria; Hourani Center for Applied Scientific Research, Al-Ahliyya Amman University, Amman, Jordan; Center for Addiction Research and Science - AddRess, Medical University of Vienna, Waehringer Strasse 13a, A-1090, Vienna, Austria.
| |
Collapse
|
20
|
Brugnoli FR, Holy M, Niello M, Maier J, Hanreich M, Menzel M, Haberler M, Zulus N, Pickl T, Ivanova C, Muiznieks LD, Garlan B, Sitte HH. Development and validation of an automated microfluidic perfusion platform for parallelized screening of compounds in vitro. Basic Clin Pharmacol Toxicol 2023; 133:535-547. [PMID: 37658634 PMCID: PMC10952622 DOI: 10.1111/bcpt.13940] [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: 06/06/2023] [Revised: 07/28/2023] [Accepted: 08/29/2023] [Indexed: 09/03/2023]
Abstract
Monoamine transporters are of great interest for their role in the physiological activity of the body and their link to mental and behavioural disorders. Currently, static well-plate assays or manual perfusion systems are used to characterize the interaction of psychostimulants, antidepressants and drugs of abuse with the transporters but still suffer from significant drawbacks caused by lack of automation, for example, low reproducibility, non-comparability of results. An automated microfluidic platform was developed to address the need for more standardized procedures for cell-based assays. An automated system was used to control and drive the simultaneous perfusion of 12 channels on a microfluidic chip, establishing a more standardized protocol to perform release assays to study monoamine transporter-mediated substrate efflux. D-Amphetamine, GBR12909 (norepinephrine transporter) and p-chloroamphetamine, paroxetine (serotonin transporter) were used as control compounds to validate the system. The platform was able to produce the expected releasing (D-Amphetamine, p-chloroamphetamine) or inhibiting (GBR12909, paroxetine) profiles for the two transporters. The reduction of manual operation and introduction of automated flow control enabled the implementation of stronger standardized protocols and the possibility of obtaining higher throughput by increasing parallelization.
Collapse
Affiliation(s)
- Francesca R. Brugnoli
- Elvesys ‐ Microfluidic Innovation CenterParisFrance
- Center for Physiology and Pharmacology, Institute of PharmacologyMedical University of ViennaViennaAustria
| | - Marion Holy
- Center for Physiology and Pharmacology, Institute of PharmacologyMedical University of ViennaViennaAustria
| | - Marco Niello
- Center for Physiology and Pharmacology, Institute of PharmacologyMedical University of ViennaViennaAustria
| | - Julian Maier
- Center for Physiology and Pharmacology, Institute of PharmacologyMedical University of ViennaViennaAustria
| | - Marcus Hanreich
- Höhere Technische Bundeslehr‐ und Versuchsanstalt Mödling (HTL Mödling)MödlingAustria
| | - Mario Menzel
- Höhere Technische Bundeslehr‐ und Versuchsanstalt Mödling (HTL Mödling)MödlingAustria
| | - Matthias Haberler
- Höhere Technische Bundeslehr‐ und Versuchsanstalt Mödling (HTL Mödling)MödlingAustria
| | - Niklas Zulus
- Höhere Technische Bundeslehr‐ und Versuchsanstalt Mödling (HTL Mödling)MödlingAustria
| | - Thomas Pickl
- Höhere Technische Bundeslehr‐ und Versuchsanstalt Mödling (HTL Mödling)MödlingAustria
| | | | | | | | - Harald H. Sitte
- Center for Physiology and Pharmacology, Institute of PharmacologyMedical University of ViennaViennaAustria
- Hourani Center for Applied Scientific ResearchAl‐Ahliyya Amman UniversityAmmanJordan
- Center for Addiction Research and Science ‐ AddRessMedical University ViennaViennaAustria
| |
Collapse
|
21
|
Fu T, Zeng S, Zheng Q, Zhu F. The Important Role of Transporter Structures in Drug Disposition, Efficacy, and Toxicity. Drug Metab Dispos 2023; 51:1316-1323. [PMID: 37295948 DOI: 10.1124/dmd.123.001275] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/27/2023] [Revised: 05/27/2023] [Accepted: 06/02/2023] [Indexed: 06/12/2023] Open
Abstract
The ATP-binding cassette (ABC) and solute carrier (SLC) transporters are critical determinants of drug disposition, clinical efficacy, and toxicity as they specifically mediate the influx and efflux of various substrates and drugs. ABC transporters can modulate the pharmacokinetics of many drugs via mediating the translocation of drugs across biologic membranes. SLC transporters are important drug targets involved in the uptake of a broad range of compounds across the membrane. However, high-resolution experimental structures have been reported for a very limited number of transporters, which limits the study of their physiologic functions. In this review, we collected structural information on ABC and SLC transporters and described the application of computational methods in structure prediction. Taking P-glycoprotein (ABCB1) and serotonin transporter (SLC6A4) as examples, we assessed the pivotal role of structure in transport mechanisms, details of ligand-receptor interactions, drug selectivity, the molecular mechanisms of drug-drug interactions, and differences caused by genetic polymorphisms. The data collected contributes toward safer and more effective pharmacological treatments. SIGNIFICANCE STATEMENT: The experimental structure of ATP-binding cassette and solute carrier transporters was collected, and the application of computational methods in structure prediction was described. P-glycoprotein and serotonin transporter were used as examples to reveal the pivotal role of structure in transport mechanisms, drug selectivity, the molecular mechanisms of drug-drug interactions, and differences caused by genetic polymorphisms.
Collapse
Affiliation(s)
- Tingting Fu
- College of Pharmaceutical Sciences, The Second Affiliated Hospital, Zhejiang University School of Medicine, Zhejiang University, Hangzhou, China (F.Z.); School of Pharmaceutical Sciences, Jilin University, Changchun, China (T.F., Q.Z.); College of Pharmaceutical Sciences, Zhejiang University, Hangzhou, China (S.Z., F.Z.); and Innovation Institute for Artificial Intelligence in Medicine of Zhejiang University, Alibaba-Zhejiang University Joint Research Center of Future Digital Healthcare, Hangzhou, China (F.Z.)
| | - Su Zeng
- College of Pharmaceutical Sciences, The Second Affiliated Hospital, Zhejiang University School of Medicine, Zhejiang University, Hangzhou, China (F.Z.); School of Pharmaceutical Sciences, Jilin University, Changchun, China (T.F., Q.Z.); College of Pharmaceutical Sciences, Zhejiang University, Hangzhou, China (S.Z., F.Z.); and Innovation Institute for Artificial Intelligence in Medicine of Zhejiang University, Alibaba-Zhejiang University Joint Research Center of Future Digital Healthcare, Hangzhou, China (F.Z.)
| | - Qingchuan Zheng
- College of Pharmaceutical Sciences, The Second Affiliated Hospital, Zhejiang University School of Medicine, Zhejiang University, Hangzhou, China (F.Z.); School of Pharmaceutical Sciences, Jilin University, Changchun, China (T.F., Q.Z.); College of Pharmaceutical Sciences, Zhejiang University, Hangzhou, China (S.Z., F.Z.); and Innovation Institute for Artificial Intelligence in Medicine of Zhejiang University, Alibaba-Zhejiang University Joint Research Center of Future Digital Healthcare, Hangzhou, China (F.Z.)
| | - Feng Zhu
- College of Pharmaceutical Sciences, The Second Affiliated Hospital, Zhejiang University School of Medicine, Zhejiang University, Hangzhou, China (F.Z.); School of Pharmaceutical Sciences, Jilin University, Changchun, China (T.F., Q.Z.); College of Pharmaceutical Sciences, Zhejiang University, Hangzhou, China (S.Z., F.Z.); and Innovation Institute for Artificial Intelligence in Medicine of Zhejiang University, Alibaba-Zhejiang University Joint Research Center of Future Digital Healthcare, Hangzhou, China (F.Z.)
| |
Collapse
|
22
|
Small C, Cheng MH, Belay SS, Bulloch SL, Zimmerman B, Sorkin A, Block ER. The Alkylamine Stimulant 1,3-Dimethylamylamine Exhibits Substrate-Like Regulation of Dopamine Transporter Function and Localization. J Pharmacol Exp Ther 2023; 386:266-273. [PMID: 37348963 PMCID: PMC10353075 DOI: 10.1124/jpet.122.001573] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/30/2022] [Revised: 02/16/2023] [Accepted: 05/04/2023] [Indexed: 06/24/2023] Open
Abstract
The alkylamine stimulant 1,3-dimethylamylamine (DMAA) is used nonmedically as an appetite suppressant and exercise performance enhancer despite adverse cardiovascular effects that have limited its legal status. There is scant research describing the mechanism of action of DMAA, making it difficult to gauge risks or therapeutic potential. An important molecular target of structurally related phenethylamines, such as amphetamine, for regulating mood, cognition, movement, and the development of substance use disorder is the dopamine transporter, which limits the range and magnitude of dopamine signaling via reuptake from the extracellular space. The present studies were therefore initiated to characterize the effects of DMAA on dopamine transporter function. Specifically, we tested the hypothesis that DMAA exhibits substrate-like effects on dopamine transporter function and trafficking. In transport assays in human embryonic kidney cells, DMAA inhibited dopamine uptake by the human dopamine transporter in a competitive manner. Docking analysis and molecular dynamics simulations supported these findings, revealing that DMAA binds to the S1 substrate binding site and induces a conformational change from outward-facing open to outward-facing closed states, similar to the known substrates. Further supporting substrate-like effects of DMAA, the drug stimulated dopamine transporter endocytosis in a heterologous expression system via cocaine- and protein kinase A-sensitive mechanisms, mirroring findings with amphetamine. Together, these data indicate that DMAA elicits neurologic effects by binding to and regulating function of the dopamine transporter. Furthermore, pharmacologic distinctions from amphetamine reveal structural determinants for regulating transporter conformation and add mechanistic insight for the regulation of dopamine transporter endocytosis. SIGNIFICANCE STATEMENT: The alkylamine stimulant 1,3-dimethylamylamine (DMAA) is used as an appetite suppressant and athletic performance enhancer and is structurally similar to amphetamine, but there is scant research describing its mechanism of action. Characterizing the effects of DMAA on dopamine transporter function supports evaluation of potential risks and therapeutic potential while also revealing mechanistic details of dynamic transporter-substrate interactions.
Collapse
Affiliation(s)
- Cassandra Small
- Science Department, Chatham University, Pittsburgh, Pennsylvania (C.S., S.S.B., S.L.B., B.Z., E.R.B.) and Departments of Computational and Systems Biology (M.H.C.) and Cell Biology (A.S.), School of Medicine, University of Pittsburgh, Pittsburgh, Pennsylvania
| | - Mary Hongying Cheng
- Science Department, Chatham University, Pittsburgh, Pennsylvania (C.S., S.S.B., S.L.B., B.Z., E.R.B.) and Departments of Computational and Systems Biology (M.H.C.) and Cell Biology (A.S.), School of Medicine, University of Pittsburgh, Pittsburgh, Pennsylvania
| | - Saron S Belay
- Science Department, Chatham University, Pittsburgh, Pennsylvania (C.S., S.S.B., S.L.B., B.Z., E.R.B.) and Departments of Computational and Systems Biology (M.H.C.) and Cell Biology (A.S.), School of Medicine, University of Pittsburgh, Pittsburgh, Pennsylvania
| | - Sarah L Bulloch
- Science Department, Chatham University, Pittsburgh, Pennsylvania (C.S., S.S.B., S.L.B., B.Z., E.R.B.) and Departments of Computational and Systems Biology (M.H.C.) and Cell Biology (A.S.), School of Medicine, University of Pittsburgh, Pittsburgh, Pennsylvania
| | - Brooke Zimmerman
- Science Department, Chatham University, Pittsburgh, Pennsylvania (C.S., S.S.B., S.L.B., B.Z., E.R.B.) and Departments of Computational and Systems Biology (M.H.C.) and Cell Biology (A.S.), School of Medicine, University of Pittsburgh, Pittsburgh, Pennsylvania
| | - Alexander Sorkin
- Science Department, Chatham University, Pittsburgh, Pennsylvania (C.S., S.S.B., S.L.B., B.Z., E.R.B.) and Departments of Computational and Systems Biology (M.H.C.) and Cell Biology (A.S.), School of Medicine, University of Pittsburgh, Pittsburgh, Pennsylvania
| | - Ethan R Block
- Science Department, Chatham University, Pittsburgh, Pennsylvania (C.S., S.S.B., S.L.B., B.Z., E.R.B.) and Departments of Computational and Systems Biology (M.H.C.) and Cell Biology (A.S.), School of Medicine, University of Pittsburgh, Pittsburgh, Pennsylvania
| |
Collapse
|
23
|
Schifano F, Vento A, Scherbaum N, Guirguis A. Stimulant and hallucinogenic novel psychoactive substances; an update. Expert Rev Clin Pharmacol 2023; 16:1109-1123. [PMID: 37968919 DOI: 10.1080/17512433.2023.2279192] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/06/2023] [Accepted: 10/31/2023] [Indexed: 11/17/2023]
Abstract
INTRODUCTION The renewed interest in considering a range of stimulants, psychedelics and dissociatives as therapeutics emphasizes the need to draft an updated overview of these drugs' clinical and pharmacological issues. AREAS COVERED The focus here was on: stimulants (e.g. amphetamines, methamphetamine, and pseudoephedrine; phenethylamines; synthetic cathinones; benzofurans; piperazines; aminoindanes; aminorex derivatives; phenmetrazine derivatives; phenidates); classical (e.g. ergolines; tryptamines; psychedelic phenethylamines), and atypical (e.g. PCP/ketamine-like dissociatives) psychedelics.Stimulant and psychedelics are associated with: a) increased central DA levels (psychedelic phenethylamines, synthetic cathinones and stimulants); b) 5-HT receptor subtypes' activation (psychedelic phenethylamines; recent tryptamine and lysergamide derivatives); and c) antagonist activity at NMDA receptors, (phencyclidine-like dissociatives). EXPERT OPINION Clinicians should be regularly informed about the range of NPS and their medical, psychobiological and psychopathological risks both in the acute and long term. Future research should focus on an integrative model in which pro-drug websites' analyses are combined with advanced research approaches, including computational chemistry studies so that in vitro and in vivo preclinical studies of index novel psychoactives can be organized. The future of psychedelic research should focus on identifying robust study designs to convincingly assess the potential therapeutic benefits of psychedelics, molecules likely to present with limited dependence liability levels.
Collapse
Affiliation(s)
- F Schifano
- Psychopharmacology Drug Misuse and Novel Psychoactive Substances Research Unit, School of Life and Medical Sciences, University of Hertfordshire, Hatfield, Herts (UK)
| | - A Vento
- Mental Health Department, Addiction Observatory (Osservatorio sulle dipendenze)- NonProfit Association - Rome, Rome, Italy
| | - N Scherbaum
- LVR-University Hospital, Department of Psychiatry and Psychotherapy, Medical Faculty, University of Duisburg-Essen, Essen, Germany
| | - A Guirguis
- Psychopharmacology Drug Misuse and Novel Psychoactive Substances Research Unit, School of Life and Medical Sciences, University of Hertfordshire, Hatfield, Herts (UK)
- Pharmacy, Swansea University Medical School, Faculty of Medicine, Health and Life Science, Swansea University, Wales, UK
| |
Collapse
|
24
|
Rodrigues CHP, Mariotto LS, Castro JS, Peruquetti PH, Silva-Junior NC, Bruni AT. Acute, chronic, and post-mortem toxicity: a review focused on three different classes of new psychoactive substances. Forensic Toxicol 2023; 41:187-212. [PMID: 36604359 DOI: 10.1007/s11419-022-00657-3] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/27/2022] [Accepted: 12/22/2022] [Indexed: 01/07/2023]
Abstract
PURPOSE New psychoactive substances (NPS) are not controlled under the Single Convention on Narcotic Drugs of 1961 or the 1971 Convention, but they may pose a public health threat. Knowledge of the main properties and toxicological effects of these substances is lacking. According to the current Drugs Law (Law n. 11.343/2006), the Brazilian Surveillance Agency issues directives for forbidden substances in Brazil, and structural classes of synthetic cannabinoids, cathinones, and phenylethylamines are considered illicit drugs. Considering that data on these controlled substances are scattered, the main objective of this work was to collect and organize data to generate relevant information on the toxicological properties of NPS. METHODS We carried out a literature review collecting information on the acute, chronic, and post-mortem toxicity of these classes of NSP. We searched info in five scientific databases considering works from 2017 to 2021 and performed a statistical evaluation of the data. RESULTS Results have shown a general lack of studies in this field given that many NPS have not had their toxicity evaluated. We observed a significant difference in the volume of data concerning acute and chronic/post-mortem toxicity. Moreover, studies on the adverse effects of polydrug use are scarce. CONCLUSIONS More in-depth information about the main threats involving NPS use are needed.
Collapse
Affiliation(s)
- Caio H P Rodrigues
- Department of Chemistry, Faculty of Philosophy, Science and Letters at Ribeirão Preto, University of São Paulo, Ribeirão Preto, SP, 14040-901, Brazil
- INCT Forense - Department of Chemistry, Faculty of Philosophy, Science and Letters at Ribeirão Preto, University of São Paulo, Ribeirão Preto, SP, 14040-901, Brazil
| | - Lívia S Mariotto
- Department of Chemistry, Faculty of Philosophy, Science and Letters at Ribeirão Preto, University of São Paulo, Ribeirão Preto, SP, 14040-901, Brazil
- INCT Forense - Department of Chemistry, Faculty of Philosophy, Science and Letters at Ribeirão Preto, University of São Paulo, Ribeirão Preto, SP, 14040-901, Brazil
| | - Jade S Castro
- Department of Chemistry, Faculty of Philosophy, Science and Letters at Ribeirão Preto, University of São Paulo, Ribeirão Preto, SP, 14040-901, Brazil
- INCT Forense - Department of Chemistry, Faculty of Philosophy, Science and Letters at Ribeirão Preto, University of São Paulo, Ribeirão Preto, SP, 14040-901, Brazil
| | - Paulo H Peruquetti
- Department of Chemistry, Faculty of Philosophy, Science and Letters at Ribeirão Preto, University of São Paulo, Ribeirão Preto, SP, 14040-901, Brazil
| | - Newton C Silva-Junior
- Department of Chemistry, Faculty of Philosophy, Science and Letters at Ribeirão Preto, University of São Paulo, Ribeirão Preto, SP, 14040-901, Brazil
- INCT Forense - Department of Chemistry, Faculty of Philosophy, Science and Letters at Ribeirão Preto, University of São Paulo, Ribeirão Preto, SP, 14040-901, Brazil
| | - Aline T Bruni
- Department of Chemistry, Faculty of Philosophy, Science and Letters at Ribeirão Preto, University of São Paulo, Ribeirão Preto, SP, 14040-901, Brazil.
- INCT Forense - Department of Chemistry, Faculty of Philosophy, Science and Letters at Ribeirão Preto, University of São Paulo, Ribeirão Preto, SP, 14040-901, Brazil.
| |
Collapse
|
25
|
Keighron JD, Bonaventura J, Li Y, Yang JW, DeMarco EM, Hersey M, Cao J, Sandtner W, Michaelides M, Sitte HH, Newman AH, Tanda G. Interactions of calmodulin kinase II with the dopamine transporter facilitate cocaine-induced enhancement of evoked dopamine release. Transl Psychiatry 2023; 13:202. [PMID: 37311803 PMCID: PMC10264427 DOI: 10.1038/s41398-023-02493-4] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/18/2023] [Revised: 05/17/2023] [Accepted: 05/26/2023] [Indexed: 06/15/2023] Open
Abstract
Typical and atypical dopamine uptake inhibitors (DUIs) prefer distinct conformations of the dopamine transporter (DAT) to form ligand-transporter complexes, resulting in markedly different effects on behavior, neurochemistry, and potential for addiction. Here we show that cocaine and cocaine-like typical psychostimulants elicit changes in DA dynamics distinct from those elicited by atypical DUIs, as measured via voltammetry procedures. While both classes of DUIs reduced DA clearance rate, an effect significantly related to their DAT affinity, only typical DUIs elicited a significant stimulation of evoked DA release, an effect unrelated to their DAT affinity, which suggests a mechanism of action other than or in addition to DAT blockade. When given in combination, typical DUIs enhance the stimulatory effects of cocaine on evoked DA release while atypical DUIs blunt them. Pretreatments with an inhibitor of CaMKIIα, a kinase that interacts with DAT and that regulates synapsin phosphorylation and mobilization of reserve pools of DA vesicles, blunted the effects of cocaine on evoked DA release. Our results suggest a role for CaMKIIα in modulating the effects of cocaine on evoked DA release without affecting cocaine inhibition of DA reuptake. This effect is related to a specific DAT conformation stabilized by cocaine. Moreover, atypical DUIs, which prefer a distinct DAT conformation, blunt cocaine's neurochemical and behavioral effects, indicating a unique mechanism underlying their potential as medications for treating psychostimulant use disorder.
Collapse
Affiliation(s)
- Jacqueline D Keighron
- Medication Development Program, National Institute on Drug Abuse, Intramural Research Program, Baltimore, MD, USA
- Department of Biological and Chemical Science, New York Institute of Technology, Old Westbury, NY, USA
| | - Jordi Bonaventura
- Biobehavioral Imaging & Molecular Neuropsychopharmacology Unit, Neuroimaging Research Branch, National Institute on Drug Abuse, Intramural Research Program, Baltimore, MD, USA
- Department of Pathology and Experimental Therapeutics, Institut de Neurociències, Universitat de Barcelona, L'Hospitalet de Llobregat, Catalonia, Spain
| | - Yang Li
- Center for Physiology and Pharmacology, Medical University of Vienna, Vienna, Austria
| | - Jae-Won Yang
- Center for Physiology and Pharmacology, Medical University of Vienna, Vienna, Austria
| | - Emily M DeMarco
- Medication Development Program, National Institute on Drug Abuse, Intramural Research Program, Baltimore, MD, USA
| | - Melinda Hersey
- Medication Development Program, National Institute on Drug Abuse, Intramural Research Program, Baltimore, MD, USA
| | - Jianjing Cao
- Molecular Targets and Medications Discovery Branch, National Institute on Drug Abuse, Intramural Research Program, Baltimore, MD, USA
| | - Walter Sandtner
- Center for Physiology and Pharmacology, Medical University of Vienna, Vienna, Austria
| | - Michael Michaelides
- Biobehavioral Imaging & Molecular Neuropsychopharmacology Unit, Neuroimaging Research Branch, National Institute on Drug Abuse, Intramural Research Program, Baltimore, MD, USA
| | - Harald H Sitte
- Center for Physiology and Pharmacology, Medical University of Vienna, Vienna, Austria
| | - Amy Hauck Newman
- Medication Development Program, National Institute on Drug Abuse, Intramural Research Program, Baltimore, MD, USA
- Molecular Targets and Medications Discovery Branch, National Institute on Drug Abuse, Intramural Research Program, Baltimore, MD, USA
| | - Gianluigi Tanda
- Medication Development Program, National Institute on Drug Abuse, Intramural Research Program, Baltimore, MD, USA.
| |
Collapse
|
26
|
Boytsov D, Schicker K, Hellsberg E, Freissmuth M, Sandtner W. Allosteric modulators of solute carrier function: a theoretical framework. Front Physiol 2023; 14:1166450. [PMID: 37250134 PMCID: PMC10210158 DOI: 10.3389/fphys.2023.1166450] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/15/2023] [Accepted: 04/24/2023] [Indexed: 05/31/2023] Open
Abstract
Large-scale drug screening is currently the basis for the identification of new chemical entities. This is a rather laborious approach, because a large number of compounds must be tested to cover the chemical space in an unbiased fashion. However, the structures of targetable proteins have become increasingly available. Thus, a new era has arguably been ushered in with the advent of methods, which allow for structure-based docking campaigns (i.e., virtual screens). Solute carriers (SLCs) are among the most promising drug targets. This claim is substantiated by the fact that a large fraction of the 400 solute carrier genes is associated with human diseases. The ability to dock large ligand libraries into selected structures of solute carriers has set the stage for rational drug design. In the present study, we show that these structure-based approaches can be refined by taking into account how solute carriers operate. We specifically address the feasibility of targeting solute carriers with allosteric modulators, because their actions differ fundamentally from those of ligands, which bind to the substrate binding site. For the pertinent analysis we used transition state theory in conjunction with the linear free energy relationship (LFER). These provide the theoretical framework to understand how allosteric modulators affect solute carrier function.
Collapse
Affiliation(s)
- D. Boytsov
- Center of Physiology and Pharmacology, Medical University of Vienna, Vienna, Austria
| | - K. Schicker
- Center of Physiology and Pharmacology, Medical University of Vienna, Vienna, Austria
| | - E. Hellsberg
- Computational Structural Biology Unit, National Institute of Neurological Disorders and Stroke (NINDS), National Institutes of Health (NIH), Bethesda, MD, United States
| | - M. Freissmuth
- Center of Physiology and Pharmacology, Medical University of Vienna, Vienna, Austria
| | - W. Sandtner
- Center of Physiology and Pharmacology, Medical University of Vienna, Vienna, Austria
| |
Collapse
|
27
|
Apuy LFM, Barreto MAB, Merino LAH. Efficacy of bupropion and cognitive behavioral therapy in the treatment of methamphetamine use disorder: a systematic review and meta-analysis. REVISTA BRASILEIRA DE PSIQUIATRIA (SAO PAULO, BRAZIL : 1999) 2023; 45. [PMID: 36917815 PMCID: PMC10288480 DOI: 10.47626/1516-4446-2022-2979] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/28/2022] [Accepted: 02/13/2023] [Indexed: 03/06/2023]
Abstract
OBJECTIVES We assessed the efficacy of cognitive behavioral therapy and bupropion compared to cognitive behavioral therapy alone for methamphetamine use disorder. METHODS The selection criteria for this systematic review study with meta-analysis were randomized clinical trials on the efficacy of cognitive behavioral therapy and bupropion in the treatment for methamphetamine use disorder (assessed by urine metabolites). The search was conducted in PubMed, PubMed Central, LILACS, SciELO, Cochrane Library, SCOPUS, Google Scholar, Ovid Medline, Clinicaltrials.gov, and the International Clinical Trials Registry Platform. The primary outcome was relapse. Risk of bias was assessed with the RoB 2 tool. The results of each clinical trial were input into an Excel spreadsheet. We performed a meta-analysis using relative risk and a 95%CI. RESULTS Of the 597 initial articles (498 after removing duplicate records), five were included in the meta-analysis, with an aggregate sample of 539 patients. An overall relative risk of 0.91 (95%CI 0.78-1.05) was estimated for relapse. CONCLUSION Our study limitations included publication bias and heterogeneous populations. We found no evidence that cognitive behavioral therapy and bupropion reduced the risk of relapse compared to cognitive behavioral therapy and placebo.
Collapse
|
28
|
Freissmuth M. Regulated Rapid Round Trips: Endocytotic Cycling of the Dopamine Transporter Shapes Motor Learning. J Biol Chem 2023; 299:104618. [PMID: 36935007 PMCID: PMC10124912 DOI: 10.1016/j.jbc.2023.104618] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 03/15/2023] [Indexed: 03/19/2023] Open
Abstract
The level of dopamine transporters (DATs) in the neuronal plasma membrane shapes learning and motor coordination in mice. Mechanisms underlying the regulated internalization of DAT and its return to the cell surface have been intensively studied in heterologous cells and in neuronal cell bodies. However, whether this cycling also happens in synaptic boutons, or axon terminals, thought to be the major functional site for DAT expression, was an open question that Kearney and colleagues recently addressed in the JBC. They showed that DAT cycling in the presynaptic specialization of dopaminergic neurons is subject to control by a cell-autonomous loop comprising dopamine autoreceptors and metabotropic glutamate receptors. These results should inform future studies in neural development and motor learning.
Collapse
Affiliation(s)
- Michael Freissmuth
- Institute of Pharmacology and the Gaston H. Glock Research Laboratories for Exploratory Drug Development, Center of Physiology and Pharmacology, Medical University of Vienna, Austria.
| |
Collapse
|
29
|
Kuropka P, Zawadzki M, Szpot P. A narrative review of the neuropharmacology of synthetic cathinones-Popular alternatives to classical drugs of abuse. Hum Psychopharmacol 2023; 38:e2866. [PMID: 36866677 DOI: 10.1002/hup.2866] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/01/2022] [Revised: 02/15/2023] [Accepted: 02/16/2023] [Indexed: 03/04/2023]
Abstract
OBJECTIVE To review the literature on the neuropharmacology of synthetic cathinones. METHODS A comprehensive literature search was carried out across multiple databases (mainly PubMed, World Wide Web, and Google Scholar) using relevant keywords. RESULTS Cathinones exhibit a broad toxicological profile, mimicking the effects of a wide variety of 'classic drugs' such as 3,4-methylenedioxymethamphetamine (MDMA), methamphetamine and cocaine. Even small structural changes affect their interactions with key proteins. This article reviews existing knowledge of the mechanisms of action of cathinones at the molecular level, and key findings from research on their structure-activity relationship. The cathinones are also classified according to their chemical structure and neuropharmacological profiles. CONCLUSIONS Synthetic cathinones represent one of the most numerous and widespread groups among new psychoactive substances. Initially developed for therapeutic purposes, they quickly started to be used recreationally. With a rapidly increasing number of new agents entering the market, structure-activity relationship studies are valuable for assessing and predicting the addictive potential and toxicity of new and potential future substances. The neuropharmacological properties of synthetic cathinones are still not fully understood. A full elucidation of the role of some key proteins, including organic cation transporters, requires detailed studies.
Collapse
Affiliation(s)
| | - Marcin Zawadzki
- Institute of Toxicology Research, Borowa, Poland.,Department of Forensic Medicine, Wroclaw Medical University, Wroclaw, Poland
| | - Paweł Szpot
- Institute of Toxicology Research, Borowa, Poland.,Department of Forensic Medicine, Wroclaw Medical University, Wroclaw, Poland
| |
Collapse
|
30
|
Kassim FM, Lahooti SK, Keay EA, Iyyalol R, Rodger J, Albrecht MA, Martin-Iverson MT. Dexamphetamine widens temporal and spatial binding windows in healthy participants. J Psychiatry Neurosci 2023; 48:E90-E98. [PMID: 36918195 PMCID: PMC10019325 DOI: 10.1503/jpn.220149] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/24/2022] [Revised: 09/28/2022] [Accepted: 11/11/2022] [Indexed: 03/16/2023] Open
Abstract
BACKGROUND The pathophysiology of psychosis is complex, but a better understanding of stimulus binding windows (BWs) could help to improve our knowledge base. Previous studies have shown that dopamine release is associated with psychosis and widened BWs. We can probe BW mechanisms using drugs of specific interest to psychosis. Therefore, we were interested in understanding how manipulation of the dopamine or catecholamine systems affect psychosis and BWs. We aimed to investigate the effect of dexamphetamine, as a dopamine-releasing stimulant, on the BWs in a unimodal illusion: the tactile funneling illusion (TFI). METHODS We conducted a randomized, double-blind, counterbalanced placebo-controlled crossover study to investigate funnelling and errors of localization. We administered dexamphetamine (0.45 mg/kg) to 46 participants. We manipulated 5 spatial (5-1 cm) and 3 temporal (0, 500 and 750 ms) conditions in the TFI. RESULTS We found that dexamphetamine increased funnelling illusion (p = 0.009) and increased the error of localization in a delay-dependent manner (p = 0.03). We also found that dexamphetamine significantly increased the error of localization at 500 ms temporal separation and 4 cm spatial separation (p interaction = 0.009; p 500ms|4cm v. baseline = 0.01). LIMITATIONS Although amphetamine-induced models of psychosis are a useful approach to understanding the physiology of psychosis related to dopamine hyperactivity, dexamphetamine is equally effective at releasing noradrenaline and dopamine, and, therefore, we were unable to tease apart the effects of the 2 systems on BWs in our study. CONCLUSION We found that dexamphetamine increases illusory perception on the unimodal TFI in healthy participants, which suggests that dopamine or other catecholamines have a role in increasing tactile spatial and temporal BWs.
Collapse
Affiliation(s)
- Faiz M Kassim
- From the Department of Psychiatry, St. Paul's Hospital Millennium Medical College, Addis Ababa, Ethiopia (Kassim); the Psychopharmacology Unit, School of Biomedical Sciences, University of Western Australia, Perth, WA, Australia (Kassim, Lahooti, Keay, Martin-Iverson); the Psychiatry, Graylands Hospital, Mt Claremont, Perth, WA, Australia (Iyyalol); the Experimental and Regenerative Neurosciences, School of Biological Sciences, University of Western Australia, Crawley, WA, Australia (Rodger); the Brain Plasticity Group, Perron Institute for Neurological and Translational Science, Nedlands, WA, Australia (Rodger); the Western Australian Centre for Road Safety Research, School of Psychological Science, University of Western Australia, Perth, WA, Australia (Albrecht)
| | - Samra Krakonja Lahooti
- From the Department of Psychiatry, St. Paul's Hospital Millennium Medical College, Addis Ababa, Ethiopia (Kassim); the Psychopharmacology Unit, School of Biomedical Sciences, University of Western Australia, Perth, WA, Australia (Kassim, Lahooti, Keay, Martin-Iverson); the Psychiatry, Graylands Hospital, Mt Claremont, Perth, WA, Australia (Iyyalol); the Experimental and Regenerative Neurosciences, School of Biological Sciences, University of Western Australia, Crawley, WA, Australia (Rodger); the Brain Plasticity Group, Perron Institute for Neurological and Translational Science, Nedlands, WA, Australia (Rodger); the Western Australian Centre for Road Safety Research, School of Psychological Science, University of Western Australia, Perth, WA, Australia (Albrecht)
| | - Elizabeth Ann Keay
- From the Department of Psychiatry, St. Paul's Hospital Millennium Medical College, Addis Ababa, Ethiopia (Kassim); the Psychopharmacology Unit, School of Biomedical Sciences, University of Western Australia, Perth, WA, Australia (Kassim, Lahooti, Keay, Martin-Iverson); the Psychiatry, Graylands Hospital, Mt Claremont, Perth, WA, Australia (Iyyalol); the Experimental and Regenerative Neurosciences, School of Biological Sciences, University of Western Australia, Crawley, WA, Australia (Rodger); the Brain Plasticity Group, Perron Institute for Neurological and Translational Science, Nedlands, WA, Australia (Rodger); the Western Australian Centre for Road Safety Research, School of Psychological Science, University of Western Australia, Perth, WA, Australia (Albrecht)
| | - Rajan Iyyalol
- From the Department of Psychiatry, St. Paul's Hospital Millennium Medical College, Addis Ababa, Ethiopia (Kassim); the Psychopharmacology Unit, School of Biomedical Sciences, University of Western Australia, Perth, WA, Australia (Kassim, Lahooti, Keay, Martin-Iverson); the Psychiatry, Graylands Hospital, Mt Claremont, Perth, WA, Australia (Iyyalol); the Experimental and Regenerative Neurosciences, School of Biological Sciences, University of Western Australia, Crawley, WA, Australia (Rodger); the Brain Plasticity Group, Perron Institute for Neurological and Translational Science, Nedlands, WA, Australia (Rodger); the Western Australian Centre for Road Safety Research, School of Psychological Science, University of Western Australia, Perth, WA, Australia (Albrecht)
| | - Jennifer Rodger
- From the Department of Psychiatry, St. Paul's Hospital Millennium Medical College, Addis Ababa, Ethiopia (Kassim); the Psychopharmacology Unit, School of Biomedical Sciences, University of Western Australia, Perth, WA, Australia (Kassim, Lahooti, Keay, Martin-Iverson); the Psychiatry, Graylands Hospital, Mt Claremont, Perth, WA, Australia (Iyyalol); the Experimental and Regenerative Neurosciences, School of Biological Sciences, University of Western Australia, Crawley, WA, Australia (Rodger); the Brain Plasticity Group, Perron Institute for Neurological and Translational Science, Nedlands, WA, Australia (Rodger); the Western Australian Centre for Road Safety Research, School of Psychological Science, University of Western Australia, Perth, WA, Australia (Albrecht)
| | - Matthew A Albrecht
- From the Department of Psychiatry, St. Paul's Hospital Millennium Medical College, Addis Ababa, Ethiopia (Kassim); the Psychopharmacology Unit, School of Biomedical Sciences, University of Western Australia, Perth, WA, Australia (Kassim, Lahooti, Keay, Martin-Iverson); the Psychiatry, Graylands Hospital, Mt Claremont, Perth, WA, Australia (Iyyalol); the Experimental and Regenerative Neurosciences, School of Biological Sciences, University of Western Australia, Crawley, WA, Australia (Rodger); the Brain Plasticity Group, Perron Institute for Neurological and Translational Science, Nedlands, WA, Australia (Rodger); the Western Australian Centre for Road Safety Research, School of Psychological Science, University of Western Australia, Perth, WA, Australia (Albrecht)
| | - Mathew T Martin-Iverson
- From the Department of Psychiatry, St. Paul's Hospital Millennium Medical College, Addis Ababa, Ethiopia (Kassim); the Psychopharmacology Unit, School of Biomedical Sciences, University of Western Australia, Perth, WA, Australia (Kassim, Lahooti, Keay, Martin-Iverson); the Psychiatry, Graylands Hospital, Mt Claremont, Perth, WA, Australia (Iyyalol); the Experimental and Regenerative Neurosciences, School of Biological Sciences, University of Western Australia, Crawley, WA, Australia (Rodger); the Brain Plasticity Group, Perron Institute for Neurological and Translational Science, Nedlands, WA, Australia (Rodger); the Western Australian Centre for Road Safety Research, School of Psychological Science, University of Western Australia, Perth, WA, Australia (Albrecht)
| |
Collapse
|
31
|
Kassim FM, Mark Lim JH, Albrecht MA, Martin-Iverson MT. Dexamphetamine influences funneling illusion based on psychometric score. Hum Psychopharmacol 2023; 38:e2862. [PMID: 36799101 DOI: 10.1002/hup.2862] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/07/2022] [Revised: 02/01/2023] [Accepted: 02/03/2023] [Indexed: 02/18/2023]
Abstract
OBJECTIVES Our team previously showed that like the experience of the rubber hand illusion (RHI) in people with schizophrenia and their offspring¸ dexamphetamine administration to healthy volunteers increases the stimulus binding windows (BWs) in RHI. It is not clear if similar expansions of BWs are present for unimodal illusions. Studies have also shown that subjective or objective effects of amphetamine would be linked to between-person variations in personality measures. Therefore, we aimed to examine the effect of dexamphetamine (DEX), a dopamine-releasing stimulant, on illusory perception using unimodal sensory stimuli (Tactile Funneling Illusion [TFI]) across both temporal and spatial variables. We further examined the relationship between changes in psychometric scores and changes in illusion perception induced by dexamphetamine. METHODS Healthy subjects (N = 20) participated in a randomized, double-blind, counter-balanced, placebo-controlled, cross-over study. The effects of dexamphetamine (0.45 mg/kg, PO, q.d.) on funneling and error of spatial localization (EL) were examined using TFI. Psychotomimetic effects were assessed using a battery of psychological measures. RESULTS Dexamphetamine did not significantly increased the funneling illusion (p = 0.88) or EL (p = 0.5), relative to placebo. However, the degree of change in psychometric scores following dexamphetamine positively correlated with changes in funneling (ρ = 0.48, p = 0.03, n = 20), mainly at 0 ms delay condition (ρ = 0.6, p = 0.004, n = 20). CONCLUSION Unlike multimodal illusions, alteration of BWs does not occur for unimodal illusions after administration of a dopamine-releasing agent. However, our findings indicate that moderate release of dopamine, through its psychotomimetic effect, indirectly influences unimodal illusion.
Collapse
Affiliation(s)
- Faiz M Kassim
- Neuropsychopharmacology Research Unit, School of Biomedical Sciences, University of Western Australia, Perth, Western Australia, Australia
| | - J H Mark Lim
- Neuropsychopharmacology Research Unit, School of Biomedical Sciences, University of Western Australia, Perth, Western Australia, Australia
| | - Matthew A Albrecht
- Western Australian Centre for Road Safety Research, School of Psychological Sciences, University of Western Australia, Perth, Western Australia, Australia
| | - Mathew T Martin-Iverson
- Neuropsychopharmacology Research Unit, School of Biomedical Sciences, University of Western Australia, Perth, Western Australia, Australia
| |
Collapse
|
32
|
Niello M, Sideromenos S, Gradisch R, O´Shea R, Schwazer J, Maier J, Kastner N, Sandtner W, Jäntsch K, Lupica CR, Hoffman AF, Lubec G, Loland CJ, Stockner T, Pollak DD, Baumann MH, Sitte HH. Persistent binding at dopamine transporters determines sustained psychostimulant effects. Proc Natl Acad Sci U S A 2023; 120:e2114204120. [PMID: 36730201 PMCID: PMC9963675 DOI: 10.1073/pnas.2114204120] [Citation(s) in RCA: 9] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/02/2021] [Accepted: 12/28/2022] [Indexed: 02/03/2023] Open
Abstract
Psychostimulants interacting with the dopamine transporter (DAT) can be used illicitly or for the treatment of specific neuropsychiatric disorders. However, they can also produce severe and persistent adverse events. Often, their pharmacological properties in vitro do not fully correlate to their pharmacological profile in vivo. Here, we investigated the pharmacological effects of enantiomers of pyrovalerone, α-pyrrolidinovalerophenone, and 3,4-methylenedioxypyrovalerone as compared to the traditional psychostimulants cocaine and methylphenidate, using a variety of in vitro, computational, and in vivo approaches. We found that in vitro drug-binding kinetics at DAT correlate with the time-course of in vivo psychostimulant action in mice. In particular, a slow dissociation (i.e., slow koff) of S-enantiomers of pyrovalerone analogs from DAT predicts their more persistent in vivo effects when compared to cocaine and methylphenidate. Overall, our findings highlight the critical importance of drug-binding kinetics at DAT for determining the in vivo profile of effects produced by psychostimulant drugs.
Collapse
Affiliation(s)
- Marco Niello
- Center for Physiology and Pharmacology, Institute of Pharmacology, Medical University of Vienna, 1090Vienna, Austria
| | - Spyridon Sideromenos
- Center for Physiology and Pharmacology, Department of Neurophysiology and Neuropharmacology, Medical University of Vienna, 1090Vienna, Austria
| | - Ralph Gradisch
- Center for Physiology and Pharmacology, Institute of Pharmacology, Medical University of Vienna, 1090Vienna, Austria
| | - Ronan O´Shea
- Electrophysiology Research Section, National Institute on Drug Abuse, NIH, Baltimore, MD21224
| | - Jakob Schwazer
- Center for Physiology and Pharmacology, Institute of Pharmacology, Medical University of Vienna, 1090Vienna, Austria
| | - Julian Maier
- Center for Physiology and Pharmacology, Institute of Pharmacology, Medical University of Vienna, 1090Vienna, Austria
| | - Nina Kastner
- Center for Physiology and Pharmacology, Institute of Pharmacology, Medical University of Vienna, 1090Vienna, Austria
| | - Walter Sandtner
- Center for Physiology and Pharmacology, Institute of Pharmacology, Medical University of Vienna, 1090Vienna, Austria
| | - Kathrin Jäntsch
- Center for Physiology and Pharmacology, Institute of Pharmacology, Medical University of Vienna, 1090Vienna, Austria
| | - Carl R. Lupica
- Electrophysiology Research Section, National Institute on Drug Abuse, NIH, Baltimore, MD21224
| | - Alexander F. Hoffman
- Electrophysiology Research Section, National Institute on Drug Abuse, NIH, Baltimore, MD21224
| | - Gert Lubec
- Department of Neuroproteomics, Paracelsus Medical University, 5020Salzburg, Austria
| | - Claus J. Loland
- Department of Neuroscience, Faculty of Health and Medical Sciences, University of Copenhagen, 2200Copenhagen, Denmark
| | - Thomas Stockner
- Center for Physiology and Pharmacology, Institute of Pharmacology, Medical University of Vienna, 1090Vienna, Austria
| | - Daniela D. Pollak
- Center for Physiology and Pharmacology, Department of Neurophysiology and Neuropharmacology, Medical University of Vienna, 1090Vienna, Austria
| | - Michael H. Baumann
- Designer Drug Research Unit, Intramural Research Program, National Institute on Drug Abuse, NIH, Baltimore, MD21224
| | - Harald H. Sitte
- Center for Physiology and Pharmacology, Institute of Pharmacology, Medical University of Vienna, 1090Vienna, Austria
- AddRess, Center for Addiction Research and Science, Medical University of Vienna, 1090Vienna, Austria
| |
Collapse
|
33
|
Tryptophan and Substance Abuse: Mechanisms and Impact. Int J Mol Sci 2023; 24:ijms24032737. [PMID: 36769059 PMCID: PMC9917371 DOI: 10.3390/ijms24032737] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/04/2023] [Revised: 01/22/2023] [Accepted: 01/28/2023] [Indexed: 02/04/2023] Open
Abstract
Addiction, the continuous misuse of addictive material, causes long-term dysfunction in the neurological system. It substantially affects the control strength of reward, memory, and motivation. Addictive substances (alcohol, marijuana, caffeine, heroin, methamphetamine (METH), and nicotine) are highly active central nervous stimulants. Addiction leads to severe health issues, including cardiovascular diseases, serious infections, and pulmonary/dental diseases. Drug dependence may result in unfavorable cognitive impairments that can continue during abstinence and negatively influence recovery performance. Although addiction is a critical global health challenge with numerous consequences and complications, currently, there are no efficient options for treating drug addiction, particularly METH. Currently, novel treatment approaches such as psychological contingency management, cognitive behavioral therapy, and motivational enhancement strategies are of great interest. Herein, we evaluate the devastating impacts of different addictive substances/drugs on users' mental health and the role of tryptophan in alleviating unfavorable side effects. The tryptophan metabolites in the mammalian brain and their potential to treat compulsive abuse of addictive substances are investigated by assessing the functional effects of addictive substances on tryptophan. Future perspectives on developing promising modalities to treat addiction and the role of tryptophan and its metabolites to alleviate drug dependency are discussed.
Collapse
|
34
|
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
|
35
|
Komiyama T. Effects of Genetic Mutation Sites in ADR Genes on Modern Chickens Produced and Domesticated by Artificial Selection. BIOLOGY 2023; 12:biology12020169. [PMID: 36829448 PMCID: PMC9952598 DOI: 10.3390/biology12020169] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 11/07/2022] [Revised: 01/11/2023] [Accepted: 01/19/2023] [Indexed: 01/24/2023]
Abstract
Associations between neurotransmitters, adrenergic receptor (ADR) mutations, and behaviors in chickens produced and domesticated by artificial selection remain unclear. This study investigates the association of neurotransmitters and ADR mutations with egg laying and cockfighting-behaviors associated with significantly different breeding backgrounds-in Shaver Brown and Shamo chickens. Accordingly, the whole sequences of nine ADR genes were determined, and nine amino acid-specific mutation sites from five genes (ADRα1A: S365G, ADRα1D: T440N, ADRα2A: D273E, ADRβ1: N443S, S445N, ADRβ3: R342C, Q404L, and P406S) were extracted. Evolutionary analysis showed that these mutations were not ancestrally derived. These results confirm that the mutations at these sites were artificially selected for domestication and are breed specific. NST population analysis confirmed a difference in the degree of genetic differentiation between the two populations in seven genes. The results further confirm differences in the degree of genetic differentiation between the two populations in Shaver Brown (ADRA1B and ADRA1D) and Shamo (ADRA1A and ADRA2B) chickens, indicating that the ADR gene differs between the two breeds. The effects of artificial selection, guided by the human-driven selection of desirable traits, are reflected in adrenaline gene mutations. Furthermore, certain gene mutations may affect domestication, while others may affect other traits in populations or individuals.
Collapse
Affiliation(s)
- Tomoyoshi Komiyama
- Department of Clinical Pharmacology, Tokai University School of Medicine, Isehara 259-1193, Kanagawa, Japan
| |
Collapse
|
36
|
Bhat S, El-Kasaby A, Kasture A, Boytsov D, Reichelt JB, Hummel T, Sucic S, Pifl C, Freissmuth M, Sandtner W. A mechanism of uncompetitive inhibition of the serotonin transporter. eLife 2023; 12:e82641. [PMID: 36648438 PMCID: PMC9883013 DOI: 10.7554/elife.82641] [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: 08/11/2022] [Accepted: 01/17/2023] [Indexed: 01/18/2023] Open
Abstract
The serotonin transporter (SERT/SLC6A4) is arguably the most extensively studied solute carrier (SLC). During its eponymous action - that is, the retrieval of serotonin from the extracellular space - SERT undergoes a conformational cycle. Typical inhibitors (antidepressant drugs and cocaine), partial and full substrates (amphetamines and their derivatives), and atypical inhibitors (ibogaine analogues) bind preferentially to different states in this cycle. This results in competitive or non-competitive transport inhibition. Here, we explored the action of N-formyl-1,3-bis (3,4-methylenedioxyphenyl)-prop-2-yl-amine (ECSI#6) on SERT: inhibition of serotonin uptake by ECSI#6 was enhanced with increasing serotonin concentration. Conversely, the KM for serotonin was lowered by augmenting ECSI#6. ECSI#6 bound with low affinity to the outward-facing state of SERT but with increased affinity to a potassium-bound state. Electrophysiological recordings showed that ECSI#6 preferentially interacted with the inward-facing state. Kinetic modeling recapitulated the experimental data and verified that uncompetitive inhibition arose from preferential binding of ECSI#6 to the K+-bound, inward-facing conformation of SERT. This binding mode predicted a pharmacochaperoning action of ECSI#6, which was confirmed by examining its effect on the folding-deficient mutant SERT-PG601,602AA: preincubation of HEK293 cells with ECSI#6 restored export of SERT-PG601,602AA from the endoplasmic reticulum and substrate transport. Similarly, in transgenic flies, the administration of ECSI#6 promoted the delivery of SERT-PG601,602AA to the presynaptic specialization of serotonergic neurons. To the best of our knowledge, ECSI#6 is the first example of an uncompetitive SLC inhibitor. Pharmacochaperones endowed with the binding mode of ECSI#6 are attractive, because they can rescue misfolded transporters at concentrations, which cause modest transport inhibition.
Collapse
Affiliation(s)
- Shreyas Bhat
- Institute of Pharmacology and the Gaston H. Glock Research Laboratories for Exploratory Drug Development, Center of Physiology and Pharmacology, Medical University of ViennaViennaAustria
| | - Ali El-Kasaby
- Institute of Pharmacology and the Gaston H. Glock Research Laboratories for Exploratory Drug Development, Center of Physiology and Pharmacology, Medical University of ViennaViennaAustria
| | - Ameya Kasture
- Department of Neurobiology, University of ViennaViennaAustria
| | - Danila Boytsov
- Institute of Pharmacology and the Gaston H. Glock Research Laboratories for Exploratory Drug Development, Center of Physiology and Pharmacology, Medical University of ViennaViennaAustria
| | - Julian B Reichelt
- Institute of Pharmacology and the Gaston H. Glock Research Laboratories for Exploratory Drug Development, Center of Physiology and Pharmacology, Medical University of ViennaViennaAustria
| | - Thomas Hummel
- Department of Neurobiology, University of ViennaViennaAustria
| | - Sonja Sucic
- Institute of Pharmacology and the Gaston H. Glock Research Laboratories for Exploratory Drug Development, Center of Physiology and Pharmacology, Medical University of ViennaViennaAustria
| | - Christian Pifl
- Center for Brain Research, Medical University of ViennaViennaAustria
| | - Michael Freissmuth
- Institute of Pharmacology and the Gaston H. Glock Research Laboratories for Exploratory Drug Development, Center of Physiology and Pharmacology, Medical University of ViennaViennaAustria
| | - Walter Sandtner
- Institute of Pharmacology and the Gaston H. Glock Research Laboratories for Exploratory Drug Development, Center of Physiology and Pharmacology, Medical University of ViennaViennaAustria
| |
Collapse
|
37
|
Pugh CF, DeVree BT, Schmidt SG, Loland CJ. Pharmacological Characterization of Purified Full-Length Dopamine Transporter from Drosophila melanogaster. Cells 2022; 11:cells11233811. [PMID: 36497070 PMCID: PMC9740255 DOI: 10.3390/cells11233811] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/01/2022] [Revised: 11/21/2022] [Accepted: 11/22/2022] [Indexed: 11/29/2022] Open
Abstract
The dopamine transporter (DAT) is a member of the neurotransmitter:sodium symporter (NSS) family, mediating the sodium-driven reuptake of dopamine from the extracellular space thereby terminating dopaminergic neurotransmission. Our current structural understanding of DAT is derived from the resolutions of DAT from Drosophila melanogaster (dDAT). Despite extensive structural studies of purified dDAT in complex with a variety of antidepressants, psychostimulants and its endogenous substrate, dopamine, the molecular pharmacology of purified, full length dDAT is yet to be elucidated. In this study, we functionally characterized purified, full length dDAT in detergent micelles using radioligand binding with the scintillation proximity assay. We elucidate the consequences of Na+ and Cl- binding on [3H]nisoxetine affinity and use this to evaluate the binding profiles of substrates and inhibitors to the transporter. Additionally, the technique allowed us to directly determine a equilibrium binding affinity (Kd) for [3H]dopamine to dDAT. To compare with a more native system, the affinities of specified monoamines and inhibitors was determined on dDAT, human DAT and human norepinephrine transporter expressed in COS-7 cells. With our gathered data, we established a pharmacological profile for purified, full length dDAT that will be useful for subsequent biophysical studies using dDAT as model protein for the mammalian NSS family of proteins.
Collapse
|
38
|
Botha MJ, Kirton SB. In Silico Investigations into the Selectivity of Psychoactive and New Psychoactive Substances in Monoamine Transporters. ACS OMEGA 2022; 7:38311-38321. [PMID: 36340072 PMCID: PMC9631908 DOI: 10.1021/acsomega.2c02714] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 05/02/2022] [Accepted: 10/07/2022] [Indexed: 06/16/2023]
Abstract
New psychoactive substances (NPS) are a group of compounds that mimic the effects of illicit substances. A range of NPS have been shown to interact with the three main classes of monoamine transporters (DAT, NET, and SERT) to differing extents, but it is unclear why these differences arise. To aid in understanding the differences in affinity between the classes of monoamine transporters, several in silico experiments were conducted. Docking experiments showed there was no direct correlation between a range of scoring functions and experimental activity, but Spearman ranking analysis showed a significant correlation (α = 0.1) for DAT, with the affinity ΔG (0.42), αHB (0.40), GoldScore (0.40), and PLP (0.41) scoring functions, and for DAT (0.38) and SERT (0.40) using a consensus scoring approach. Qualitative structure-activity relationship (QSAR) experiments resulted in the generation of robust and predictive three-descriptor models for SERT (r 2 = 0.87, q 2 = 0.8, and test set r 2 = 0.74) and DAT (r 2 = 0.68, q 2 = 0.51, test set r 2 = 0.63). Both QSAR models described similar characteristics for binding, i.e., rigid hydrophobic molecules with a biogenic amine moiety, and were not sufficient to facilitate a deeper understanding of differences in affinity between the monoamine transporters. This contextualizes the observed promiscuity for NPS between the isoforms and highlights the difficulty in the design and development of compounds that are isoform-selective.
Collapse
|
39
|
Development and Validation of a Uplc-Qtof-Ms Method for Blood Analysis of Isomeric Amphetamine-Related Drugs. SEPARATIONS 2022. [DOI: 10.3390/separations9100285] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 03/09/2023] Open
Abstract
The identification of isomeric drugs is gaining increasing importance in forensics and doping control. Isomers vary in terms of safety, effectiveness, and regulation, particularly for amphetamine-related drugs (ARDs). This study developed and validated a pseudo-isocratic UPLC-qTOF-MS analytical method for the identification of isomeric Amphetamine-related drugs (ARDs) in blood following mixed-mode solid-phase extraction (MMSPE). The procedure requires 250 μL of blood to achieve a limit of quantification (LOQ) and detection (LOD) of 20 ng/mL for all analytes. In aged animal blood samples, extraction recoveries of 63–90% and matrix effects of 9–21% were observed. Precision and accuracy for all analytes were within 20% and 89–118%, respectively. The analytical method was developed and validated in accordance with the Scientific Working Group for Forensic Toxicology (SWGTOX) Standard. It has acceptable accuracy and precision for use in doping control and forensic toxicology.
Collapse
|
40
|
The Pharmacological Effects of Phenylephrine are Indirect, Mediated by Noradrenaline Release from the Cytoplasm. Neurochem Res 2022; 47:3272-3284. [PMID: 35945308 PMCID: PMC9546997 DOI: 10.1007/s11064-022-03681-2] [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: 05/05/2022] [Revised: 07/04/2022] [Accepted: 07/06/2022] [Indexed: 10/31/2022]
Abstract
Phenylephrine (PE) is a canonical α1-adrenoceptor-selective agonist. However, unexpected effects of PE have been observed in preclinical and clinical studies, that cannot be easily explained by its actions on α1-adrenoceptors. The probability of the involvement of α2- and β-adrenoceptors in the effect of PE has been raised. In addition, our earlier study observed that PE released noradrenaline (NA) in a [Ca2+]o-independent manner. To elucidate this issue, we have investigated the effects of PE on [3H]NA release and α1-mediated smooth muscle contractions in the mouse vas deferens (MVD) as ex vivo preparation. The release experiments were designed to assess the effects of PE at the presynaptic terminal, whereas smooth muscle isometric contractions in response to electrical field stimulation were used to measure PE effect postsynaptically. Our results show that PE at concentrations between 0.3 and 30 µM significantly enhanced the resting release of [3H]NA in a [Ca2+]o-independent manner. In addition, prazosin did not affect the release of NA evoked by PE. On the contrary, PE-evoked smooth muscle contractions were inhibited by prazosin administration indicating the α1-adrenoceptor-mediated effect. When the function of the NA transporter (NAT) was attenuated with nisoxetine, PE failed to release NA and the contractions were reduced by approximately 88%. The remaining part proved to be prazosin-sensitive. The present work supports the substantial indirect effect of PE which relays on the cytoplasmic release of NA, which might explain the reported side effects for PE.
Collapse
|
41
|
Multimodal detection of dopamine by sniffer cells expressing genetically encoded fluorescent sensors. Commun Biol 2022; 5:578. [PMID: 35689020 PMCID: PMC9187629 DOI: 10.1038/s42003-022-03488-5] [Citation(s) in RCA: 14] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/28/2021] [Accepted: 05/13/2022] [Indexed: 12/21/2022] Open
Abstract
Dopamine supports locomotor control and higher brain functions such as motivation and learning. Consistently, dopaminergic dysfunction is involved in a spectrum of neurological and neuropsychiatric diseases. Detailed data on dopamine dynamics is needed to understand how dopamine signals translate into cellular and behavioral responses, and to uncover pathological disturbances in dopamine-related diseases. Genetically encoded fluorescent dopamine sensors have recently enabled unprecedented monitoring of dopamine dynamics in vivo. However, these sensors' utility for in vitro and ex vivo assays remains unexplored. Here, we present a blueprint for making dopamine sniffer cells for multimodal dopamine detection. We generated sniffer cell lines with inducible expression of seven different dopamine sensors and perform a head-to-head comparison of sensor properties to guide users in sensor selection. In proof-of-principle experiments, we apply the sniffer cells to record endogenous dopamine release from cultured neurons and striatal slices, and for determining tissue dopamine content. Furthermore, we use the sniffer cells to measure dopamine uptake and release via the dopamine transporter as a radiotracer free, high-throughput alternative to electrochemical- and radiotracer-based assays. Importantly, the sniffer cell framework can readily be applied to the growing list of genetically encoded fluorescent neurotransmitter sensors.
Collapse
|
42
|
Kolaczynska KE, Ducret P, Trachsel D, Hoener MC, Liechti ME, Luethi D. Pharmacological characterization of 3,4-methylenedioxyamphetamine (MDA) analogs and two amphetamine-based compounds: N,α-DEPEA and DPIA. Eur Neuropsychopharmacol 2022; 59:9-22. [PMID: 35378384 DOI: 10.1016/j.euroneuro.2022.03.006] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/26/2021] [Revised: 02/16/2022] [Accepted: 03/13/2022] [Indexed: 11/04/2022]
Abstract
3,4-methylenedioxyamphetamine (MDA) is a psychoactive compound chemically related to the entactogen MDMA. MDA shares some of the entactogenic effects of MDMA but also exerts stimulant effects and psychedelic properties at higher doses. Here, we examined the pharmacological properties of MDA analogs and related amphetamine-based compounds detected in street drug samples or in sport supplements. We examined the key pharmacological mechanisms including monoamine uptake inhibition and release using human embryonic kidney 293 cells stably transfected with the respective human transporters. Additionally, we assessed monoamine transporter and receptor binding and activation properties. MDA, its fluorinated analogs, as well as the α-ethyl containing BDB and the dimeric amphetamine DPIA inhibited NET with the greatest potency and preferentially inhibited 5-HT vs. dopamine uptake. The β‑methoxy MDA analog 3C-BOH and the amphetamine-based N,α-DEPEA inhibited NET and preferentially inhibited dopamine vs. 5-HT uptake. The test drugs mediated efflux of at least one monoamine with the exception of DPIA. Most compounds bound to 5-HT2A and 5-HT2C receptors (Ki ≤ 10 µM) and several substances activated the 5-HT2A and 5-HT2B receptor as partial or full agonists. Furthermore, several compounds interacted with adrenergic receptors and the trace amine-associated receptor 1 (TAAR1) in the micromolar range. The pharmacological profiles of some fluorinated and nonfluorinated MDA analogs resemble the profile of MDMA. In contrast, 3C-BOH and N,α-DEPEA displayed more pronounced dopaminergic activity similar to amphetamine. Pharmacokinetics and pharmacodynamics studies are necessary to better establish the risks and therapeutic potential of the tested drugs.
Collapse
Affiliation(s)
- Karolina E Kolaczynska
- Division of Clinical Pharmacology and Toxicology, Department of Biomedicine, University Hospital Basel and University of Basel, Basel, Switzerland
| | - Paula Ducret
- Division of Clinical Pharmacology and Toxicology, Department of Biomedicine, University Hospital Basel and University of Basel, Basel, Switzerland
| | | | - Marius C Hoener
- Neuroscience Research, pRED, Roche Innovation Center Basel, F. Hoffmann-La Roche Ltd, Basel, Switzerland
| | - Matthias E Liechti
- Division of Clinical Pharmacology and Toxicology, Department of Biomedicine, University Hospital Basel and University of Basel, Basel, Switzerland.
| | - Dino Luethi
- Division of Clinical Pharmacology and Toxicology, Department of Biomedicine, University Hospital Basel and University of Basel, Basel, Switzerland; Center for Physiology and Pharmacology, Institute of Pharmacology, Medical University of Vienna, Vienna, Austria
| |
Collapse
|
43
|
Rudin D, McCorvy JD, Glatfelter GC, Luethi D, Szöllősi D, Ljubišić T, Kavanagh PV, Dowling G, Holy M, Jaentsch K, Walther D, Brandt SD, Stockner T, Baumann MH, Halberstadt AL, Sitte HH. (2-Aminopropyl)benzo[β]thiophenes (APBTs) are novel monoamine transporter ligands that lack stimulant effects but display psychedelic-like activity in mice. Neuropsychopharmacology 2022; 47:914-923. [PMID: 34750565 PMCID: PMC8882185 DOI: 10.1038/s41386-021-01221-0] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/17/2021] [Revised: 10/06/2021] [Accepted: 10/21/2021] [Indexed: 01/02/2023]
Abstract
Derivatives of (2-aminopropyl)indole (API) and (2-aminopropyl)benzofuran (APB) are new psychoactive substances which produce stimulant effects in vivo. (2-Aminopropyl)benzo[β]thiophene (APBT) is a novel sulfur-based analog of API and APB that has not been pharmacologically characterized. In the current study, we assessed the pharmacological effects of six APBT positional isomers in vitro, and three of these isomers (3-APBT, 5-APBT, and 6-APBT) were subjected to further investigations in vivo. Uptake inhibition and efflux assays in human transporter-transfected HEK293 cells and in rat brain synaptosomes revealed that APBTs inhibit monoamine reuptake and induce transporter-mediated substrate release. Despite being nonselective transporter releasers like MDMA, the APBT compounds failed to produce locomotor stimulation in C57BL/6J mice. Interestingly, 3-APBT, 5-APBT, and 6-APBT were full agonists at 5-HT2 receptor subtypes as determined by calcium mobilization assays and induced the head-twitch response in C57BL/6J mice, suggesting psychedelic-like activity. Compared to their APB counterparts, ABPT compounds demonstrated that replacing the oxygen atom with sulfur results in enhanced releasing potency at the serotonin transporter and more potent and efficacious activity at 5-HT2 receptors, which fundamentally changed the in vitro and in vivo profile of APBT isomers in the present studies. Overall, our data suggest that APBT isomers may exhibit psychedelic and/or entactogenic effects in humans, with minimal psychomotor stimulation. Whether this unique pharmacological profile of APBT isomers translates into potential therapeutic potential, for instance as candidates for drug-assisted psychotherapy, warrants further investigation.
Collapse
Affiliation(s)
- Deborah Rudin
- Center for Physiology and Pharmacology, Institute of Pharmacology, Medical University of Vienna, Waehringer Strasse 13A, 1090, Vienna, Austria
| | - John D McCorvy
- Department of Cell Biology, Neurobiology & Anatomy, Medical College of Wisconsin, Milwaukee, WI, USA
| | - Grant C Glatfelter
- Designer Drug Research Unit, Intramural Research Program, National Institute on Drug Abuse, National Institutes of Health, Baltimore, MD, USA
| | - Dino Luethi
- Center for Physiology and Pharmacology, Institute of Pharmacology, Medical University of Vienna, Waehringer Strasse 13A, 1090, Vienna, Austria
| | - Dániel Szöllősi
- Center for Physiology and Pharmacology, Institute of Pharmacology, Medical University of Vienna, Waehringer Strasse 13A, 1090, Vienna, Austria
| | - Tea Ljubišić
- Center for Physiology and Pharmacology, Institute of Pharmacology, Medical University of Vienna, Waehringer Strasse 13A, 1090, Vienna, Austria
| | - Pierce V Kavanagh
- Department of Pharmacology and Therapeutics, School of Medicine, Trinity Centre for Health Sciences, St. James Hospital, Dublin, 8, Ireland
| | - Geraldine Dowling
- Department of Pharmacology and Therapeutics, School of Medicine, Trinity Centre for Health Sciences, St. James Hospital, Dublin, 8, Ireland
- Department of Life Sciences, School of Science, Sligo Institute of Technology, Ash Lane, Sligo, Ireland
| | - Marion Holy
- Center for Physiology and Pharmacology, Institute of Pharmacology, Medical University of Vienna, Waehringer Strasse 13A, 1090, Vienna, Austria
| | - Kathrin Jaentsch
- Center for Physiology and Pharmacology, Institute of Pharmacology, Medical University of Vienna, Waehringer Strasse 13A, 1090, Vienna, Austria
| | - Donna Walther
- Designer Drug Research Unit, Intramural Research Program, National Institute on Drug Abuse, National Institutes of Health, Baltimore, MD, USA
| | - Simon D Brandt
- School of Pharmacy and Biomolecular Sciences, Liverpool John Moores University, Liverpool, UK
- Alexander Shulgin Research Institute, Lafayette, CA, USA
| | - Thomas Stockner
- Center for Physiology and Pharmacology, Institute of Pharmacology, Medical University of Vienna, Waehringer Strasse 13A, 1090, Vienna, Austria
| | - Michael H Baumann
- Designer Drug Research Unit, Intramural Research Program, National Institute on Drug Abuse, National Institutes of Health, Baltimore, MD, USA
| | - Adam L Halberstadt
- Department of Psychiatry, University of California San Diego, La Jolla, CA, USA
- Research Service, VA San Diego Healthcare System, La Jolla, CA, USA
| | - Harald H Sitte
- Center for Physiology and Pharmacology, Institute of Pharmacology, Medical University of Vienna, Waehringer Strasse 13A, 1090, Vienna, Austria.
- Center for Addiction Research and Science-AddRess, Medical University Vienna, Waehringer Strasse 13A, 1090, Vienna, Austria.
| |
Collapse
|
44
|
Gradisch R, Szöllősi D, Niello M, Lazzarin E, Sitte HH, Stockner T. Occlusion of the human serotonin transporter is mediated by serotonin-induced conformational changes in the bundle domain. J Biol Chem 2022; 298:101613. [PMID: 35065961 PMCID: PMC8867121 DOI: 10.1016/j.jbc.2022.101613] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/27/2021] [Revised: 01/16/2022] [Accepted: 01/17/2022] [Indexed: 12/04/2022] Open
Abstract
The human serotonin transporter (hSERT) terminates neurotransmission by removing serotonin (5HT) from the synaptic cleft, an essential process for proper functioning of serotonergic neurons. Structures of the hSERT have revealed its molecular architecture in four conformations, including the outward-open and occluded states, and show the transporter's engagement with co-transported ions and the binding mode of inhibitors. In this study, we investigated the molecular mechanism by which the hSERT occludes and sequesters the substrate 5HT. This first step of substrate uptake into cells is a structural change consisting of the transition from the outward-open to the occluded state. Inhibitors such as the antidepressants citalopram, fluoxetine, and sertraline inhibit this step of the transport cycle. Using molecular dynamics simulations, we reached a fully occluded state, in which the transporter-bound 5HT becomes fully shielded from both sides of the membrane by two closed hydrophobic gates. Analysis of 5HT-triggered occlusion showed that bound 5HT serves as an essential trigger for transporter occlusion. Moreover, simulations revealed a complex sequence of steps and showed that movements of bundle domain helices are only partially correlated. 5HT-triggered occlusion is initially dominated by movements of transmembrane helix 1b, while in the final step, only transmembrane helix 6a moves and relaxes an intermediate change in its secondary structure.
Collapse
Affiliation(s)
- Ralph Gradisch
- Institute of Pharmacology, Center for Physiology and Pharmacology, Medical University of Vienna, Vienna, Austria
| | - Dániel Szöllősi
- Institute of Pharmacology, Center for Physiology and Pharmacology, Medical University of Vienna, Vienna, Austria
| | - Marco Niello
- Institute of Pharmacology, Center for Physiology and Pharmacology, Medical University of Vienna, Vienna, Austria
| | - Erika Lazzarin
- Institute of Pharmacology, Center for Physiology and Pharmacology, Medical University of Vienna, Vienna, Austria
| | - Harald H Sitte
- Institute of Pharmacology, Center for Physiology and Pharmacology, Medical University of Vienna, Vienna, Austria
| | - Thomas Stockner
- Institute of Pharmacology, Center for Physiology and Pharmacology, Medical University of Vienna, Vienna, Austria.
| |
Collapse
|
45
|
Schicker K, Farr CV, Boytsov D, Freissmuth M, Sandtner W. Optimizing the Substrate Uptake Rate of Solute Carriers. Front Physiol 2022; 13:817886. [PMID: 35185619 PMCID: PMC8850955 DOI: 10.3389/fphys.2022.817886] [Citation(s) in RCA: 4] [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: 11/18/2021] [Accepted: 01/10/2022] [Indexed: 11/16/2022] Open
Abstract
The diversity in solute carriers arose from evolutionary pressure. Here, we surmised that the adaptive search for optimizing the rate of substrate translocation was also shaped by the ambient extracellular and intracellular concentrations of substrate and co-substrate(s). We explored possible solutions by employing kinetic models, which were based on analytical expressions of the substrate uptake rate, that is, as a function of the microscopic rate constants used to parameterize the transport cycle. We obtained the defining terms for five reaction schemes with identical transport stoichiometry (i.e., Na+: substrate = 2:1). We then utilized an optimization algorithm to find the set of numeric values for the microscopic rate constants, which provided the largest value for the substrate uptake rate: The same optimized rate was achieved by different sets of numerical values for the microscopic rate constants. An in-depth analysis of these sets provided the following insights: (i) In the presence of a low extracellular substrate concentration, a transporter can only cycle at a high rate, if it has low values for both, the Michaelis-Menten constant (KM) for substrate and the maximal substrate uptake rate (Vmax). (ii) The opposite is true for a transporter operating at high extracellular substrate concentrations. (iii) Random order of substrate and co-substrate binding is superior to sequential order, if a transporter is to maintain a high rate of substrate uptake in the presence of accumulating intracellular substrate. Our kinetic models provide a framework to understand how and why the transport cycles of closely related transporters differ.
Collapse
Affiliation(s)
| | | | | | | | - Walter Sandtner
- Center of Physiology and Pharmacology, Medical University of Vienna, Vienna, Austria
| |
Collapse
|
46
|
Ponleitner M, Szöllősi D, El-Kasaby A, Koban F, Freissmuth M, Stockner T. Thermal Unfolding of the Human Serotonin Transporter: Differential Effect by Stabilizing and Destabilizing Mutations and Cholesterol on Thermodynamic and Kinetic Stability. Mol Pharmacol 2022; 101:95-105. [PMID: 34866045 DOI: 10.1124/molpharm.121.000413] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/20/2021] [Accepted: 11/21/2021] [Indexed: 11/22/2022] Open
Abstract
Folding-deficient mutants of solute carrier 6 (SLC6) family members have been linked to human diseases. The serotonin transporter [(SERT)/SLC6A4] is an important drug target in the treatment of depression, anxiety, and obsessive-compulsive disorders and-with structural information in several conformational states-one of the best understood transporters. Here, we surmised that thermal unfolding offered a glimpse on the folding energy landscape of SLC6 transporters. We carried out molecular dynamic (MD) simulations to understand the mechanistic basis for enhanced and reduced stability, respectively, of the thermostabilized variant SERT-Y110A/I291A/T439S, which had previously been used for crystallization of human SERT in the outward-facing state, and of the folding-deficient SERT-P601A/G602A. We also examined the hydrophobic mismatch caused by the absence of cholesterol to explore the contribution of cholesterol to protein stability. When compared with wild type SERT, the thermodynamic and kinetic stability of SERT-Y110A/I291A/T439S was enhanced. In the other instances, changes in these two components were not correlated: the mutations in SERT-P601A/G602A led to a drop in thermodynamic but an increase in kinetic stability. The divergence was even more pronounced after cholesterol depletion, which reduced thermodynamic stability but increased the kinetic stability of wild type SERT to a level comparable to that of SERT-Y110A/I291A/T439S. We conclude that the low cholesterol content of the endoplasmic reticulum facilitates progression of the folding trajectory by reducing the energy difference between folding intermediates and the native state. SIGNIFICANCE STATEMENT: Point mutations in solute carrier 6 (SLC6) family members cause folding diseases. The serotonin transporter [(SERT)/SLC6A4] is a target for antidepressants and the best understood SLC6. This study produced molecular dynamics simulations and examined thermal unfolding of wild type and mutant SERT variants to understand their folding energy landscape. In the folding-deficient SERT-P012A/G602A, changes in kinetic and thermodynamic stability were not correlated. Similarly, cholesterol depletion lowered thermodynamic but enhanced kinetic stability. These observations allow for rationalizing the action of pharmacochaperones.
Collapse
Affiliation(s)
- Markus Ponleitner
- Institute of Pharmacology and the Gaston H. Glock Research Laboratories for Exploratory Drug Development, Center of Physiology and Pharmacology, Medical University of Vienna, Austria
| | - Daniel Szöllősi
- Institute of Pharmacology and the Gaston H. Glock Research Laboratories for Exploratory Drug Development, Center of Physiology and Pharmacology, Medical University of Vienna, Austria
| | - Ali El-Kasaby
- Institute of Pharmacology and the Gaston H. Glock Research Laboratories for Exploratory Drug Development, Center of Physiology and Pharmacology, Medical University of Vienna, Austria
| | - Florian Koban
- Institute of Pharmacology and the Gaston H. Glock Research Laboratories for Exploratory Drug Development, Center of Physiology and Pharmacology, Medical University of Vienna, Austria
| | - Michael Freissmuth
- Institute of Pharmacology and the Gaston H. Glock Research Laboratories for Exploratory Drug Development, Center of Physiology and Pharmacology, Medical University of Vienna, Austria
| | - Thomas Stockner
- Institute of Pharmacology and the Gaston H. Glock Research Laboratories for Exploratory Drug Development, Center of Physiology and Pharmacology, Medical University of Vienna, Austria
| |
Collapse
|
47
|
Marlier D. Doping in Racing Pigeons ( Columba livia domestica): A Review and Actual Situation in Belgium, a Leading Country in This Field. Vet Sci 2022; 9:42. [PMID: 35202294 PMCID: PMC8880243 DOI: 10.3390/vetsci9020042] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/14/2021] [Revised: 01/13/2022] [Accepted: 01/20/2022] [Indexed: 02/05/2023] Open
Abstract
Pigeon racing is a sport in which trained homing pigeons (Columba livia domestica) are released between 60 and 1200 km from their loft and then have to return home as quickly as possible. The first race was held in 1818 in Belgium and since then, Belgium has led the world in pigeon breeding. Unfortunately, as in other sports, doping has become a major issue and doping controls have been implemented. This review provides information about pigeon racing, rules from the Royal Federation Colombophile of Belgium, and laws applicable in Belgium as doping control issues cannot be understood without including them as part of pigeon racing. The main pharmacological data concerning corticoids, non-steroidal anti-inflammatory drugs, anabolic steroids, pain relievers and narcotic analgesics, bronchodilators and β-agonists, drugs acting on the central nervous system and other performance-enhancing drugs, in addition to methods relevant to doping in pigeons are presented. Moreover, the chosen matrix and analytical methods are described.
Collapse
Affiliation(s)
- Didier Marlier
- Bird, Rabbit and Rodent Clinic, Faculty of Veterinary Medicine, University of Liege, B4000 Liege, Belgium
| |
Collapse
|
48
|
Davis S, Zhu J. Substance abuse and neurotransmission. ADVANCES IN PHARMACOLOGY (SAN DIEGO, CALIF.) 2022; 93:403-441. [PMID: 35341573 PMCID: PMC9759822 DOI: 10.1016/bs.apha.2021.10.007] [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: 06/14/2023]
Abstract
The number of people who suffer from a substance abuse disorder has continued to rise over the last decade; particularly, the number of drug-related overdose deaths has sharply increased during the COVID-19 pandemic. Converging lines of clinical observations, supported by imaging and neuropsychological performance testing, have demonstrated that substance abuse-induced dysregulation of neurotransmissions in the brain is critical for development and expression of the addictive properties of abused substances. Recent scientific advances have allowed for better understanding of the neurobiological processes that mediates drugs of abuse and addiction. This chapter presents the past classic concepts and the recent advances in our knowledge about how cocaine, amphetamines, opioids, alcohol, and nicotine alter multiple neurotransmitter systems, which contribute to the behaviors associated with each drug. Additionally, we discuss the interactive effects of HIV-1 or COVID-19 and substance abuse on neurotransmission and neurobiological pathways. Finally, we introduce therapeutic strategies for development of pharmacotherapies for substance abuse disorders.
Collapse
Affiliation(s)
- Sarah Davis
- Department of Drug Discovery and Biomedical Sciences, College of Pharmacy, University of South Carolina, Columbia, SC, United States
| | - Jun Zhu
- Department of Drug Discovery and Biomedical Sciences, College of Pharmacy, University of South Carolina, Columbia, SC, United States.
| |
Collapse
|
49
|
Szöllősi D, Stockner T. Sodium Binding Stabilizes the Outward-Open State of SERT by Limiting Bundle Domain Motions. Cells 2022; 11:cells11020255. [PMID: 35053371 PMCID: PMC8773566 DOI: 10.3390/cells11020255] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/11/2021] [Revised: 12/23/2021] [Accepted: 12/29/2021] [Indexed: 11/16/2022] Open
Abstract
The human serotonin transporter (hSERT) removes the neurotransmitter serotonin from the synaptic cleft by reuptake into the presynaptic nerve terminal. A number of neurologic diseases are associated with dysfunction of the hSERT, and several medications for their treatment are hSERT blockers, including citalopram, fluoxetine, and paroxetine. The substrate transport is energized by the high concentration of external NaCl. We showed through molecular dynamics simulations that the binding of NaCl stabilized the hSERT in the substrate-binding competent conformation, which was characterized by an open access path to the substrate-binding site through the outer vestibule. Importantly, the binding of NaCl reduced the dynamics of the hSERT by decreasing the internal fluctuations of the bundle domain as well as the movement of the bundle domain relative to the scaffold domain. In contrast, the presence of only the bound chloride ion did not reduce the high domain mobility of the apo state.
Collapse
|
50
|
Xu L, Chen LY. Effects of the N-terminal dynamics on the conformational states of human dopamine transporter. Biophys Chem 2022; 283:106765. [PMID: 35101818 PMCID: PMC8898274 DOI: 10.1016/j.bpc.2022.106765] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/21/2021] [Revised: 01/17/2022] [Accepted: 01/21/2022] [Indexed: 11/02/2022]
Abstract
Dopamine transporter mediates the neurotransmitter dopamine homeostasis in a sodium-dependent manner. The transport process involves an alternating access of a substrate to the extracellular and intracellular spaces, which is associated with different conformational states of the transporter. However, the underlying mechanism of modulation of the state transition remains elusive. Here we present a computational simulation study of human dopamine transporter to explore its two end states (outward-facing open and inward-facing open) that have not been determined experimentally. We show that the full-length transporter may tend to adopt the inward-facing open state in its free state. The binding of an amphetamine may not trap the transporter in the outward-facing open state with increasing length of the N-terminal. Furthermore, we identify distinct patterns in the interaction networks between the N-terminal and the intracellular region that could stabilize the state of the transporter, independent of substrate binding and phosphorylation. Our results reveal the essential role of the N-terminal dynamics in modulating the functional states of the dopamine transporter, providing molecular insights into the coupling of conformational transition and substrate passage in neurotransmitter transporters.
Collapse
|