1
|
Zhu R, Sandtner W, Stockner T, Heilinger A, Holy M, Kudlacek O, Wildling L, Saha K, Fröhlich AS, Bindl M, Tziortzouda P, Haider A, Gobl J, Suh SH, Khan JA, Bicher J, Kastner N, Ebner A, Gruber HJ, Freissmuth M, Newman AH, Sitte HH, Hinterdorfer P. Revealing the location and dynamics of a concealed binding site in the dopamine transporter. Nat Commun 2025; 16:4197. [PMID: 40328781 DOI: 10.1038/s41467-025-59511-w] [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: 12/28/2023] [Accepted: 04/24/2025] [Indexed: 05/08/2025] Open
Abstract
The dopamine transporter (DAT) is linked to neuropsychiatric disorders including ADHD, Parkinson's disease, and substance use disorders. Accordingly, DAT is the target of illicit drugs and clinically important medicines. However, the number and function of ligand binding sites in DAT is enigmatic due to conflicting data from available structures and molecular pharmacology. Herein, we design force sensors with DAT ligands and measure their interaction forces with wild-type and mutated DATs, from which two distinct populations of unbinding strengths and off-rates are detected. The high-force population is reduced by V152I and S422A mutations, or by substituting Na+ with K+ or NMDG+. In contrast, several modifications including mutation G386H, acetylation of K92 and K384, mutation K92A, mutation K384A, or protonation of H477 decrease the low-force population. The present data delineate the threshold of binding strength, which may account for certain ligand binding sites to be imperceptible in crystal or cryo-EM structures. Furthermore, the force spectra provide the information on the position and kinetic rates of a herein detected ligand binding site in DAT.
Collapse
Affiliation(s)
- Rong Zhu
- Institute of Biophysics, Johannes Kepler University Linz, Linz, Austria.
| | - Walter Sandtner
- Institute of Pharmacology, Center of Physiology and Pharmacology, Medical University of Vienna, Vienna, Austria
- Gaston H. Glock Research Laboratories for Exploratory Drug Development, Center of Physiology and Pharmacology, Medical University of Vienna, Vienna, Austria
| | - Thomas Stockner
- Institute of Pharmacology, Center of Physiology and Pharmacology, Medical University of Vienna, Vienna, Austria
| | | | - Marion Holy
- Institute of Pharmacology, Center of Physiology and Pharmacology, Medical University of Vienna, Vienna, Austria
| | - Oliver Kudlacek
- Institute of Pharmacology, Center of Physiology and Pharmacology, Medical University of Vienna, Vienna, Austria
| | - Linda Wildling
- Institute of Biophysics, Johannes Kepler University Linz, Linz, Austria
| | - Kusumika Saha
- Institute of Pharmacology, Center of Physiology and Pharmacology, Medical University of Vienna, Vienna, Austria
- Brigham and Women's Hospital, Harvard Medical School, Boston, MA, USA
| | | | - Michael Bindl
- Institute of Biophysics, Johannes Kepler University Linz, Linz, Austria
| | | | - Anna Haider
- Institute of Biophysics, Johannes Kepler University Linz, Linz, Austria
| | - Julia Gobl
- Institute of Biophysics, Johannes Kepler University Linz, Linz, Austria
| | | | - Jawad Akbar Khan
- Institute of Pharmacology, Center of Physiology and Pharmacology, Medical University of Vienna, Vienna, Austria
| | - Julia Bicher
- Institute of Pharmacology, Center of Physiology and Pharmacology, Medical University of Vienna, Vienna, Austria
| | - Nina Kastner
- Institute of Pharmacology, Center of Physiology and Pharmacology, Medical University of Vienna, Vienna, Austria
| | - Andreas Ebner
- Institute of Biophysics, Johannes Kepler University Linz, Linz, Austria
| | - Hermann J Gruber
- Institute of Biophysics, Johannes Kepler University Linz, Linz, Austria
| | - Michael Freissmuth
- Institute of Pharmacology, Center of Physiology and Pharmacology, Medical University of Vienna, Vienna, Austria
- Gaston H. Glock Research Laboratories for Exploratory Drug Development, Center of Physiology and Pharmacology, Medical University of Vienna, Vienna, Austria
| | - Amy Hauck Newman
- Medicinal Chemistry Section, Molecular Targets and Medications Discovery Branch, National Institute on Drug Abuse-Intramural Research Program, Baltimore, MD, USA.
| | - Harald H Sitte
- Institute of Pharmacology, Center of Physiology and Pharmacology, Medical University of Vienna, Vienna, Austria.
- Hourani Center for Applied Scientific Research, Al-Ahliyya Amman University, Amman, Jordan.
- Center for Addiction Research and Science, Medical University Vienna, Vienna, Austria.
| | - Peter Hinterdorfer
- Institute of Biophysics, Johannes Kepler University Linz, Linz, Austria.
| |
Collapse
|
2
|
Wu Y, Cai J, Liu H, Li C, Tang Q, Zhang YW. (-)-Syringaresinol Exerts an Antidepressant-like Activity in Mice by Noncompetitive Inhibition of the Serotonin Transporter. Pharmaceuticals (Basel) 2024; 17:1637. [PMID: 39770479 PMCID: PMC11678425 DOI: 10.3390/ph17121637] [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: 11/11/2024] [Revised: 12/02/2024] [Accepted: 12/03/2024] [Indexed: 01/11/2025] Open
Abstract
BACKGROUND Albizia julibrissin Durazz. is one of the most popular herbs used for depression treatment, but the molecular basis for its mechanism of action has not been fully addressed. Previously, we isolated and identified two lignan glycoside derivatives that were shown to noncompetitively inhibit serotonin transporter (SERT) activity but with a relatively low inhibitory potency compared with those of conventional antidepressants. METHODS We characterized the pharmacological profile of the parental compound of these previously isolated lignan glycosides, (-)-syringaresinol (SYR), in inhibiting SERT by using biochemical, pharmacological, and behavioral approaches. RESULTS SYR, as a potent inhibitor, decreases SERT Vmax but with little change in Km for its fluorescent substrate. SYR was shown to block the conformational conversion essential for substrate transport by stabilizing SERT in an outward-open and inward-closed conformation. In addition, our molecular docking and biochemical validation demonstrated that SYR binds to an allosteric site in SERT and noncompetitively inhibits SERT transport and binding activity. Furthermore, administration of SYR was indicated to exert an antidepressant-like activity and to effectively attenuate chronic unpredictable mild stress (CUMS)-induced abnormalities in behaviors and synaptic protein expression in depressive animal models. CONCLUSIONS This study not only provides molecular insights into the mechanism of action of A. julibrissin in the treatment of depression, but also opens up the possibility of development of a novel class of allosteric site-targeted therapeutic agents with an underlying mechanism of action different from that of conventional antidepressants.
Collapse
Affiliation(s)
- Yingyao Wu
- School of Life Sciences, Guangzhou University, Guangzhou 510006, China
| | - Jianxin Cai
- School of Life Sciences, Guangzhou University, Guangzhou 510006, China
| | - Hanhe Liu
- School of Life Sciences, Guangzhou University, Guangzhou 510006, China
| | - Chan Li
- School of Life Sciences, Guangzhou University, Guangzhou 510006, China
| | - Qingfa Tang
- School of Traditional Chinese Medicine, Southern Medical University, Guangzhou 510515, China
- Guangdong Provincial Key Laboratory of Chinese Medicine Pharmaceutics, Guangzhou 510515, China
| | - Yuan-Wei Zhang
- School of Life Sciences, Guangzhou University, Guangzhou 510006, China
| |
Collapse
|
3
|
Jahanabadi S, Amiri S, Karkeh-Abadi M, Razmi A. Natural psychedelics in the treatment of depression; a review focusing on neurotransmitters. Fitoterapia 2023; 169:105620. [PMID: 37490982 DOI: 10.1016/j.fitote.2023.105620] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/04/2023] [Revised: 07/21/2023] [Accepted: 07/22/2023] [Indexed: 07/27/2023]
Abstract
Natural psychedelic compounds are emerging as potential novel therapeutics in psychiatry. This review will discuss how natural psychedelics exert their neurobiological therapeutic effects, and how different neurotransmission systems mediate the effects of these compounds. Further, current therapeutic strategies for depression, and novel mechanism of action of natural psychedelics in the treatment of depression will be discussed. In this review, our focus will be on N, N-dimethyltryptamine (DMT), reversible type A monoamine oxidase inhibitors, mescaline-containing cacti, psilocybin/psilocin-containing mushrooms, ibogaine, muscimol extracted from Amanita spp. mushrooms and ibotenic acid.
Collapse
Affiliation(s)
- Samane Jahanabadi
- Department of Pharmacology and Toxicology, Faculty of Pharmacy, Shahid Sadoughi University of Medical Sciences, Yazd, Iran; Pharmaceutical Science Research Center, Shahid Sadoughi University of Medical Sciences, Yazd, Iran
| | - Shayan Amiri
- Division of Neurodegenerative Disorders, St Boniface Hospital Albrechtsen Research Centre, Winnipeg, MB, Canada; Department of Pharmacology and Therapeutics, University of Manitoba, Winnipeg, MB, Canada.
| | - Mehdi Karkeh-Abadi
- Medicinal Plants Research Center, Institute of Medicinal Plants, ACECR, Karaj, Iran
| | - Ali Razmi
- Medicinal Plants Research Center, Institute of Medicinal Plants, ACECR, Karaj, Iran.
| |
Collapse
|
4
|
Fonseca ECM, Ferreira LR, Figueiredo PLB, Maia CDSF, Setzer WN, Da Silva JKR. Antidepressant Effects of Essential Oils: A Review of the Past Decade (2012-2022) and Molecular Docking Study of Their Major Chemical Components. Int J Mol Sci 2023; 24:ijms24119244. [PMID: 37298210 DOI: 10.3390/ijms24119244] [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: 03/13/2023] [Revised: 04/19/2023] [Accepted: 04/24/2023] [Indexed: 06/12/2023] Open
Abstract
Depression is a mental disorder that affects more than 300 million people worldwide. The medications available for treatment take a long time to exhibit therapeutic results and present several side effects. Furthermore, there is a decrease in the quality of life of people suffering from this affliction. Essential oils are traditionally used to relieve the symptoms of depression due to the properties of the constituents of these oils to cross the blood-brain barrier acting on depression-related biological receptors associated with reduced toxicity and side effects. In addition, compared to traditional drugs, they have several administration forms. This review provides a comprehensive assessment of studies on plants whose essential oil has exhibit antidepressant activity in the past decade and the mechanism of action of the major components and models tested. An additional in silico study was conducted with the frequent compounds in the composition of these essential oils, providing a molecular approach to the mechanism of action that has been reported in the past decade. This review is valuable for the development of potential antidepressant medications in addition to providing a molecular approach to the antidepressant mechanism of action of the major volatile compounds that have been reported in the past decade.
Collapse
Affiliation(s)
- Emily Christie M Fonseca
- Programa de Pós-Graduação em Farmacologia e Bioquímica, Universidade Federal do Pará, Belém 66075-110, Brazil
| | - Lanalice R Ferreira
- Programa de Pós-Graduação em Química, Universidade Federal do Pará, Belém 66075-110, Brazil
| | - Pablo Luis B Figueiredo
- Laboratório de Química dos Produtos Naturais, Centro de Ciências Biológicas e da Saúde, Universidade do Estado do Pará, Belém 66087-662, Brazil
| | - Cristiane do Socorro F Maia
- Programa de Pós-Graduação em Farmacologia e Bioquímica, Universidade Federal do Pará, Belém 66075-110, Brazil
| | | | - Joyce Kelly R Da Silva
- Programa de Pós-Graduação em Farmacologia e Bioquímica, Universidade Federal do Pará, Belém 66075-110, Brazil
- Programa de Pós-Graduação em Química, Universidade Federal do Pará, Belém 66075-110, Brazil
| |
Collapse
|
5
|
Cao TXD, Filliter C, Montastruc F, Yu OHY, Fergusson E, Rej S, Azoulay L, Renoux C. Selective serotonin reuptake inhibitors and the risk of type 2 diabetes mellitus in youths. J Affect Disord 2022; 318:231-237. [PMID: 36084758 DOI: 10.1016/j.jad.2022.08.094] [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: 01/10/2022] [Revised: 08/24/2022] [Accepted: 08/26/2022] [Indexed: 12/01/2022]
Abstract
BACKGROUND Selective serotonin reuptake inhibitors (SSRIs) have been associated with type 2 diabetes mellitus (T2DM) in youths, possibly via 5-HT2C, H1 receptors and serotonin transporter (SERT). SSRIs have similar affinity for SERT but variable affinity for 5-HT2C and H1. This study assessed whether SSRIs with strong affinity for 5-HT2C and H1 (relative to SERT) were associated with T2DM risk compared with weak-affinity SSRIs. METHODS Using the UK Clinical Practice Research Datalink, we assembled a cohort of patients aged 5-24, newly prescribed a strong-affinity SSRI (citalopram, escitalopram, fluoxetine) or weak affinity (paroxetine, sertraline, fluvoxamine) between 1990 and 2019. We controlled for confounding using standardized mortality ratio weighting, estimated from calendar time-specific propensity scores. We used weighted Cox proportional hazards models to estimate hazard ratios (HRs) of incident T2DM with 95 % confidence intervals (CIs). RESULTS The cohort included 347,368 new users of strong-affinity SSRIs and 131,359 of weak-affinity SSRIs. Strong-affinity SSRIs were not associated with an increased T2DM risk compared with weak-affinity SSRIs (incidence rate 2.8 vs 2.7 per 1000 person-years; HR 1.03, 95 % CI 0.85-1.25). T2DM risk did not vary with duration of use, age or sex. However, the HR was numerically higher in youths with normal or low weight (HR 1.30, 95 % CI 0.85-1.98) and with prior antipsychotic use (HR 1.62, 95 % CI 0.83-3.18). LIMITATIONS Median duration of SSRI use, in line with real-world SSRI prescribing, was relatively short. CONCLUSION T2DM risk did not differ between strong- and weak-affinity SSRIs, providing reassurance for clinicians when choosing between SSRIs in youths.
Collapse
Affiliation(s)
- Thi Xuan Dai Cao
- Department of Epidemiology, Biostatistics, and Occupational Health, McGill University, Montreal, Québec, Canada; Centre for Clinical Epidemiology, Lady Davis Institute, Jewish General Hospital, Montreal, Québec, Canada
| | - Christopher Filliter
- Centre for Clinical Epidemiology, Lady Davis Institute, Jewish General Hospital, Montreal, Québec, Canada
| | - François Montastruc
- Department of Clinical and Medical Pharmacology, Regional Pharmacovigilance Centre, Faculty of Medicine and Toulouse University Hospital, Toulouse, France
| | - Oriana Hoi Yun Yu
- Centre for Clinical Epidemiology, Lady Davis Institute, Jewish General Hospital, Montreal, Québec, Canada; Division of Endocrinology, Jewish General Hospital, Montreal, Québec, Canada
| | - Emma Fergusson
- Oxford Health NHS Trust, Department of Psychiatry, Oxford University, Oxford, United Kingdom
| | - Soham Rej
- Department of Psychiatry, McGill University, Montreal, Québec, Canada
| | - Laurent Azoulay
- Department of Epidemiology, Biostatistics, and Occupational Health, McGill University, Montreal, Québec, Canada; Centre for Clinical Epidemiology, Lady Davis Institute, Jewish General Hospital, Montreal, Québec, Canada; Gerald Bronfman Department of Oncology, McGill University, Montreal, Québec, Canada
| | - Christel Renoux
- Department of Epidemiology, Biostatistics, and Occupational Health, McGill University, Montreal, Québec, Canada; Centre for Clinical Epidemiology, Lady Davis Institute, Jewish General Hospital, Montreal, Québec, Canada; Department of Neurology and Neurosurgery, McGill University, Montreal, Québec, Canada.
| |
Collapse
|
6
|
Liu H, Wu Y, Li C, Tang Q, Zhang YW. Molecular docking and biochemical validation of (-)-syringaresinol-4-O-β-D-apiofuranosyl-(1→2)-β-D-glucopyranoside binding to an allosteric site in monoamine transporters. Front Pharmacol 2022; 13:1018473. [PMID: 36386236 PMCID: PMC9649612 DOI: 10.3389/fphar.2022.1018473] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/01/2022] [Accepted: 10/17/2022] [Indexed: 06/11/2024] Open
Abstract
Albizia julibrissin Durazz is one of the most common herbs used for depression and anxiety treatment, but its mechanism of action as an antidepressant or anxiolytic drug have not been fully understood. We previously isolated and identified one lignan glycoside compound from Albizia Julibrissin Durazz, (-)-syringaresinol-4-O-β-D-apiofuranosyl-(1→2)-β-D-glucopyranoside (SAG), that inhibited all three monoamine transporters with a mechanism of action different from that of the conventional antidepressants. In this study, we generated homology models for human dopamine transporter and human norepinephrine transporter, based on the X-ray structure of Drosophila dopamine transporter, and conducted the molecular docking of SAG to all three human monoamine transporters. Our computational results indicated that SAG binds to an allosteric site (S2) that has been demonstrated to be formed by an aromatic pocket positioned in the scaffold domain in the extracellular vestibule connected to the central site (S1) in these monoamine transporters. In addition, we demonstrated that SAG stabilizes a conformation of serotonin transporter with both the extracellular and cytoplasmic pathways closed. Furthermore, we performed mutagenesis of the residues in both the allosteric and orthosteric sites to biochemically validate SAG binding in all three monoamine transporters. Our results are consistent with the molecular docking calculation and support the association of SAG with the allosteric site. We expect that this herbal molecule could become a lead compound for the development of new therapeutic agents with a novel mechanism of action.
Collapse
Affiliation(s)
- Hanhe Liu
- School of Life Sciences, Guangzhou University, Guangzhou, China
| | - Yingyao Wu
- School of Life Sciences, Guangzhou University, Guangzhou, China
| | - Chan Li
- School of Life Sciences, Guangzhou University, Guangzhou, China
| | - Qingfa Tang
- School of Traditional Chinese Medicine, Southern Medical University, Guangzhou, China
- Guangdong Provincial Key Laboratory of Chinese Medicine Pharmaceutics, Guangzhou, China
| | - Yuan-Wei Zhang
- School of Life Sciences, Guangzhou University, Guangzhou, China
| |
Collapse
|
7
|
Dunham KE, Venton BJ. SSRI antidepressants differentially modulate serotonin reuptake and release in Drosophila. J Neurochem 2022; 162:404-416. [PMID: 35736504 PMCID: PMC9427694 DOI: 10.1111/jnc.15658] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/14/2022] [Revised: 05/27/2022] [Accepted: 06/17/2022] [Indexed: 11/26/2022]
Abstract
Selective serotonin reuptake inhibitor (SSRI) antidepressants are commonly prescribed treatments for depression, but their effects on serotonin reuptake and release are not well understood. Drosophila melanogaster, the fruit fly, expresses the serotonin transporter (dSERT), the major target of SSRIs, but real-time serotonin changes after SSRIs have not been characterized in this model. The goal of this study was to characterize effects of SSRIs on serotonin concentration and reuptake in Drosophila larvae. We applied various doses (0.1-100 μM) of fluoxetine (Prozac), escitalopram (Lexapro), citalopram (Celexa), and paroxetine (Paxil), to ventral nerve cord (VNC) tissue and measured optogenetically-stimulated serotonin release with fast-scan cyclic voltammetry (FSCV). Fluoxetine increased reuptake from 1 to 100 μM, but serotonin concentration only increased at 100 μM. Thus, fluoxetine occupies dSERT and slows clearance but does not affect concentration. Escitalopram and paroxetine increased serotonin concentrations at all doses, but escitalopram increased reuptake more. Citalopram showed lower concentration changes and faster reuptake profiles compared with escitalopram, so the racemic mixture of citalopram does not change reuptake as much as the S-isomer. Dose response curves were constructed to compare dSERT affinities and paroxetine showed the highest affinity and fluoxetine the lowest. These data demonstrate SSRI mechanisms are complex, with separate effects on reuptake or release. Furthermore, dynamic serotonin changes in Drosophila are similar to previous studies in mammals. This work establishes how antidepressants affect serotonin in real-time, which is useful for future studies that will investigate pharmacological effects of SSRIs with different genetic mutations in Drosophila.
Collapse
Affiliation(s)
- Kelly E Dunham
- Department of Chemistry, University of Virginia, Virginia, USA
| | - B Jill Venton
- Department of Chemistry, University of Virginia, Virginia, USA
| |
Collapse
|
8
|
Contreras N, Alvíz-Amador A, Manzur-Villalobos I. In silico study of dimethyltryptamine analogues against 5-HT1B receptor: Molecular docking, dynamic simulations and ADMET prediction. JOURNAL OF HERBMED PHARMACOLOGY 2022. [DOI: 10.34172/jhp.2022.25] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/09/2022] Open
Abstract
Introduction: The 5-HT1B receptor has a potential role in various psychiatric disorders such as depression, anxiety, and post-traumatic stress disorder. The objective of this study was to perform docking and molecular dynamics simulation to evaluate at atomic level the behavior of N,N-dimethyltryptamine (DMT) on 5-HT1B receptor.Methods: In this study, initially, a search for DMT was performed using the PubChem database. Subsequently, molecular docking was executed using AutoDock Vina based in PyRx 0.8 with a 95% analogy. Additionally, ergotamine (ERG) and serotonin were used as control. Then, it ran a total of 100 ns molecular dynamics simulations on 5-HT1B bound with DMT, serotonin, 112814775, and ERG. Finally, pharmacokinetic prediction and IV acute toxicity for analogues and DMT were performed.Results: It was possible to show that 112814775 had the lowest binding energy with the receptor. In addition, 112814775 presented great conformational stability, low mobility, and stiffness compared to the control ligands: ERG, serotonin, and DMT subsequent dynamic analysis. With respect to the free energy calculation, contributions such as Van der Waals, electrostatics, and nonpolar interactions for all systems, were highlighted.Conclusion: 112814775 showed affinities with 5-HT1B receptor and evidenced notable behavior by molecular dynamic simulation according to root-mean-square deviation (RMSD), root-mean-square fluctuation (RMSF), solvent-accessible surface area (SASA), the radius of gyration, number of hydrogen bond, and free energy calculated. These results established the possible relevance of in-silico studies in search of DMT analogues against the 5-HT1B receptor, which may be associated with alterations such as depression and anxiety, and may become future study molecules for the treatment of this type of disorder.
Collapse
Affiliation(s)
- Neyder Contreras
- Pharmacology and Therapeutics Research Group. University of Cartagena, Cartagena D.T y C., Colombia
- GINUMED, Rafael Nuñez University Corporation, Cartagena D.T y C., Colombia
| | - Antistio Alvíz-Amador
- Pharmacology and Therapeutics Research Group. University of Cartagena, Cartagena D.T y C., Colombia
| | | |
Collapse
|
9
|
Ribaudo G, Bortoli M, Witt CE, Parke B, Mena S, Oselladore E, Zagotto G, Hashemi P, Orian L. ROS-Scavenging Selenofluoxetine Derivatives Inhibit In Vivo Serotonin Reuptake. ACS OMEGA 2022; 7:8314-8322. [PMID: 35309454 PMCID: PMC8928538 DOI: 10.1021/acsomega.1c05567] [Citation(s) in RCA: 16] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 10/06/2021] [Accepted: 12/31/2021] [Indexed: 06/14/2023]
Abstract
While the neurochemistry that underpins the behavioral phenotypes of depression is the subject of many studies, oxidative stress caused by the inflammation comorbid with depression has not adequately been addressed. In this study, we described novel antidepressant-antioxidant agents consisting of selenium-modified fluoxetine derivatives to simultaneously target serotonin reuptake (antidepressant action) and oxidative stress. Excitingly, we show that one of these agents (1-F) carries the ability to inhibit serotonin reuptake in vivo in mice. We therefore present a frontier dual strategy that paves the way for the future of antidepressant therapies.
Collapse
Affiliation(s)
- Giovanni Ribaudo
- Dipartimento
di Medicina Molecolare e Traslazionale, Università degli Studi di Brescia, Viale Europa 11, 25123 Brescia, Italy
| | - Marco Bortoli
- Dipartimento
di Scienze Chimiche, Università degli
Studi di Padova Via Marzolo 1, 35131 Padova, Italy
- Institut
de Química Computacional i Catàlisi and Departament
de Química, Universitat de Girona, c/Maria Aurèlia Capmany 69, 17003 Girona, Catalonia, Spain
| | - Colby E. Witt
- Department
of Chemistry and Biochemistry, University
of South Carolina, Columbia South Carolina 29201, United States
| | - Brenna Parke
- Department
of Bioengineering, Imperial College London, London SW7 2AZ, U.K.
| | - Sergio Mena
- Department
of Bioengineering, Imperial College London, London SW7 2AZ, U.K.
| | - Erika Oselladore
- Dipartimento
di Medicina Molecolare e Traslazionale, Università degli Studi di Brescia, Viale Europa 11, 25123 Brescia, Italy
| | - Giuseppe Zagotto
- Dipartimento
di Scienze del Farmaco, Università
degli Studi di Padova, Via Marzolo 5, 35131 Padova, Italy
| | - Parastoo Hashemi
- Department
of Bioengineering, Imperial College London, London SW7 2AZ, U.K.
- Department
of Chemistry and Biochemistry, University
of South Carolina, Columbia South Carolina 29201, United States
| | - Laura Orian
- Dipartimento
di Scienze Chimiche, Università degli
Studi di Padova Via Marzolo 1, 35131 Padova, Italy
| |
Collapse
|
10
|
Chan MC, Selvam B, Young HJ, Procko E, Shukla D. The substrate import mechanism of the human serotonin transporter. Biophys J 2022; 121:715-730. [PMID: 35114149 PMCID: PMC8943754 DOI: 10.1016/j.bpj.2022.01.024] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/29/2021] [Revised: 12/18/2021] [Accepted: 01/25/2022] [Indexed: 11/17/2022] Open
Abstract
The serotonin transporter (SERT) initiates the reuptake of extracellular serotonin in the synapse to terminate neurotransmission. The cryogenic electron microscopy structures of SERT bound to ibogaine and the physiological substrate serotonin resolved in different states have provided a glimpse of the functional conformations at atomistic resolution. However, the conformational dynamics and structural transitions to intermediate states are not fully understood. Furthermore, the molecular basis of how serotonin is recognized and transported remains unclear. In this study, we performed unbiased microsecond-long simulations of the human SERT to investigate the structural dynamics to various intermediate states and elucidated the complete substrate import pathway. Using Markov state models, we characterized a sequential order of conformational-driven ion-coupled substrate binding and transport events and calculated the free energy barriers of conformation transitions associated with the import mechanism. We find that the transition from the occluded to inward-facing state is the rate-limiting step for substrate import and that the substrate decreases the free energy barriers to achieve the inward-facing state. Our study provides insights on the molecular basis of dynamics-driven ion-substrate recognition and transport of SERT that can serve as a model for other closely related neurotransmitter transporters.
Collapse
Affiliation(s)
- Matthew C Chan
- Department of Chemical and Biomolecular Engineering, University of Illinois at Urbana-Champaign, Urbana, Illinois
| | - Balaji Selvam
- Department of Chemical and Biomolecular Engineering, University of Illinois at Urbana-Champaign, Urbana, Illinois
| | - Heather J Young
- Department of Biochemistry, University of Illinois at Urbana-Champaign, Urbana, Illinois
| | - Erik Procko
- Department of Biochemistry, University of Illinois at Urbana-Champaign, Urbana, Illinois; Center for Biophysics and Quantitative Biology, University of Illinois at Urbana-Champaign, Urbana, Illinois; Neuroscience Program, University of Illinois at Urbana-Champaign, Urbana, Illinois; Cancer Center at Illinois, University of Illinois at Urbana-Champaign, Urbana, Illinois
| | - Diwakar Shukla
- Department of Chemical and Biomolecular Engineering, University of Illinois at Urbana-Champaign, Urbana, Illinois; Center for Biophysics and Quantitative Biology, University of Illinois at Urbana-Champaign, Urbana, Illinois; Cancer Center at Illinois, University of Illinois at Urbana-Champaign, Urbana, Illinois; National Center for Supercomputing Applications, University of Illinois at Urbana-Champaign, Urbana, Illinois; Beckman Institute for Advanced Science and Technology, University of Illinois at Urbana-Champaign, Urbana, Illinois; NIH Center for Macromolecular Modeling and Bioinformatics, University of Illinois at Urbana-Champaign, Urbana, Illinois.
| |
Collapse
|
11
|
Sviridova A, Rogovskii V, Kudrin V, Pashenkov M, Boyko A, Melnikov M. The role of 5-HT 2B-receptors in fluoxetine-mediated modulation of Th17- and Th1-cells in multiple sclerosis. J Neuroimmunol 2021; 356:577608. [PMID: 34000471 DOI: 10.1016/j.jneuroim.2021.577608] [Citation(s) in RCA: 13] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/29/2020] [Revised: 05/07/2021] [Accepted: 05/10/2021] [Indexed: 12/12/2022]
Abstract
Fluoxetine is a selective serotonin reuptake inhibitor, which also has an immunomodulatory effect. We investigated the effects of fluoxetine and serotonin (5-HT) on the pro-inflammatory Th17- and Th1-cells in 30 patients with relapsing-remitting MS and 20 healthy subjects. Fluoxetine and 5-HT suppressed IL-17, IFN-γ and GM-CSF production by stimulated СD4+ T-cells in both groups. Blockade of 5-HT2B-receptors decreased the inhibitory effect of fluoxetine on cytokine production in MS patients. Finally, 5-HT2B-receptor activation inhibits IL-17, IFN-γ and GM-CSF production in both groups. These data suggest an anti-inflammatory role for fluoxetine in MS, which could be mediated by the activation of 5-HT2B-receptors.
Collapse
Affiliation(s)
- Anastasiya Sviridova
- Federal Center of Brain research and Neurotechnology of the Federal Medical-Biological Agency of Russia, Department of Neuroimmunology, Moscow, Russia; Pirogov Russian National Research Medical University, Department of Neurology, Neurosurgery and Medical Genetics, Moscow, Russia
| | - Vladimir Rogovskii
- Federal Center of Brain research and Neurotechnology of the Federal Medical-Biological Agency of Russia, Department of Neuroimmunology, Moscow, Russia; Pirogov Russian National Research Medical University, Department of Molecular Pharmacology and Radiobiology, Moscow, Russia
| | - Vladimir Kudrin
- V.V. Zakusov Research Institute of Pharmacology, Laboratory of Neurochemical Pharmacology Moscow, Russia
| | - Mikhail Pashenkov
- National Research Center Institute of Immunology of the Federal Medical-Biological Agency of Russia, Laboratory of Clinical Immunology, Moscow, Russia
| | - Alexey Boyko
- Federal Center of Brain research and Neurotechnology of the Federal Medical-Biological Agency of Russia, Department of Neuroimmunology, Moscow, Russia; Pirogov Russian National Research Medical University, Department of Neurology, Neurosurgery and Medical Genetics, Moscow, Russia
| | - Mikhail Melnikov
- Pirogov Russian National Research Medical University, Department of Neurology, Neurosurgery and Medical Genetics, Moscow, Russia; National Research Center Institute of Immunology of the Federal Medical-Biological Agency of Russia, Laboratory of Clinical Immunology, Moscow, Russia.
| |
Collapse
|
12
|
Rasti AR, Coombe VE, Muzik JR, Kliethermes CL. Pharmacological characterization of the forced swim test in Drosophila melanogaster. INVERTEBRATE NEUROSCIENCE 2020; 20:22. [PMID: 33170389 DOI: 10.1007/s10158-020-00255-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/02/2020] [Accepted: 10/30/2020] [Indexed: 11/30/2022]
Abstract
The forced swim test is commonly used as a preclinical screen of antidepressant medication efficacy in rats and mice. Neckameyer and Nieto-Romero (Stress 18:254-66, 2015) adopted the forced swim test for use with the fruit fly Drosophila melanogaster and showed that behavior in this test is sensitive to several physiologically relevant stressors. However, whether this test might be sensitive to the effects of antidepressant medications or other compounds is unknown. In the current studies, we fed drugs to male and female flies that we expected to either decrease or increase the duration of immobility in the forced swim test, including fluoxetine, desipramine, picrotoxin, reserpine, 3-iodo-tyrosine, and ethanol. Fluoxetine was the only drug tested that affected behavior in this test, and surprisingly, the direction of the effect depended on the duration of feeding. Short-term (30 min) feeding of the drug prior to test resulted in the expected increase in latency to immobility, while a longer feeding duration (20-24 h) decreased this measure. These results suggest that the pharmacological profile of the fly FST is more restricted than that of the rat or mouse FST, and that the duration of drug exposure is an important consideration in pharmacological research using flies.
Collapse
Affiliation(s)
- Aryana R Rasti
- Department of Psychology and Neuroscience, Drake University, 318 Olin Hall, 1344 27th Street, Des Moines, IA, 50311, USA
| | - Victoria E Coombe
- Department of Psychology and Neuroscience, Drake University, 318 Olin Hall, 1344 27th Street, Des Moines, IA, 50311, USA
| | - Jerica R Muzik
- Department of Psychology and Neuroscience, Drake University, 318 Olin Hall, 1344 27th Street, Des Moines, IA, 50311, USA
| | - Christopher L Kliethermes
- Department of Psychology and Neuroscience, Drake University, 318 Olin Hall, 1344 27th Street, Des Moines, IA, 50311, USA.
| |
Collapse
|
13
|
The anti-inflammatory role of SSRI and SNRI in the treatment of depression: a review of human and rodent research studies. Inflammopharmacology 2020; 29:75-90. [PMID: 33164143 DOI: 10.1007/s10787-020-00777-5] [Citation(s) in RCA: 64] [Impact Index Per Article: 12.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/15/2020] [Accepted: 10/31/2020] [Indexed: 12/15/2022]
Abstract
RATIONALE Depression has the topmost prevalence of all psychiatric diseases. It is characterized by a high recurrence rate, disability, and numerous and mostly unclear pathogenic mechanisms. Besides the monoamine or the neurotrophic hypothesis of depression, the inflammatory mechanism has begun to be supported by more and more evidence. At the same time, the current knowledge about the standard treatment of choice, the selective serotonin reuptake inhibitors (SSRIs) and serotonin and noradrenaline reuptake inhibitors (SNRIs), is expanding rapidly, adding more features to the initial ones. OBJECTIVES This review summarizes the in vivo anti-inflammatory effects of SSRIs and SNRIs in the treatment of depression and outlines the particular mechanisms of these effects for each drug separately. In addition, we provide an overview of the inflammation-related theory of depression and the underlying mechanisms. RESULTS SSRIs and SNRIs decrease the neuroinflammation through multiple mechanisms including the reduction of blood or tissue cytokines or regulating complex inflammatory pathways: nuclear factor kappa-light-chain-enhancer of activated B cells (NF-κB), inflammasomes, Toll-like receptor 4 (TLR4), peroxisome proliferator-activated receptor gamma (PPARγ). Also, SSRIs and SNRIs show these effects in association with an antidepressant action. CONCLUSIONS SSRIs and SNRIs have an anti-neuroinflammatory role which might contribute the antidepressant effect.
Collapse
|
14
|
Ortore G, Orlandini E, Betti L, Giannaccini G, Mazzoni MR, Camodeca C, Nencetti S. Focus on Human Monoamine Transporter Selectivity. New Human DAT and NET Models, Experimental Validation, and SERT Affinity Exploration. ACS Chem Neurosci 2020; 11:3214-3232. [PMID: 32991141 PMCID: PMC8015229 DOI: 10.1021/acschemneuro.0c00304] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/30/2022] Open
Abstract
![]()
The most commonly used antidepressant
drugs are the serotonin transporter
inhibitors. Their effects depend strongly on the selectivity for a
single monoamine transporter compared to other amine transporters
or receptors, and the selectivity is roughly influenced by the spatial
protein structure. Here, we provide a computational study on three
human monoamine transporters, i.e., DAT, NET, and SERT. Starting from
the construction of hDAT and hNET models, whose three-dimensional
structure is unknown, and the prediction of the binding pose for 19
known inhibitors, 3D-QSAR models of three human transporters were
built. The training set variability, which was high in structure and
activity profile, was validated using a set of in-house compounds.
Results concern more than one aspect. First of all, hDAT and hNET
three-dimensional structures were built, validated, and compared to
the hSERT one; second, the computational study highlighted the differences
in binding site arrangement statistically correlated to inhibitor
selectivity; third, the profiling of new inhibitors pointed out a
conservation of the inhibitory activity trend between rabbit and human
SERT with a difference of about 1 order of magnitude; fourth, binding
and functional studies confirmed 4-(benzyloxy)-4-phenylpiperidine 20a–d and 21a–d as potent SERT
inhibitors. In particular, one of the compounds (compound 20b) revealed a higher affinity for SERT than paroxetine in human platelets.
Collapse
Affiliation(s)
- Gabriella Ortore
- Department of Pharmacy, University of Pisa, Via Bonanno 6, 56126 Pisa, Italy
| | - Elisabetta Orlandini
- Research Center “E. Piaggio”, University of Pisa, Pisa 56122, Italy
- Department of Earth Sciences, University of Pisa, Via Santa Maria 53-55, 56100 Pisa, Italy
| | - Laura Betti
- Department of Pharmacy, University of Pisa, Via Bonanno 6, 56126 Pisa, Italy
| | - Gino Giannaccini
- Department of Pharmacy, University of Pisa, Via Bonanno 6, 56126 Pisa, Italy
| | - Maria Rosa Mazzoni
- Department of Pharmacy, University of Pisa, Via Bonanno 6, 56126 Pisa, Italy
| | - Caterina Camodeca
- Department of Pharmacy, University of Pisa, Via Bonanno 6, 56126 Pisa, Italy
| | - Susanna Nencetti
- Department of Pharmacy, University of Pisa, Via Bonanno 6, 56126 Pisa, Italy
| |
Collapse
|
15
|
Xue W, Fu T, Zheng G, Tu G, Zhang Y, Yang F, Tao L, Yao L, Zhu F. Recent Advances and Challenges of the Drugs Acting on Monoamine Transporters. Curr Med Chem 2020; 27:3830-3876. [DOI: 10.2174/0929867325666181009123218] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/28/2018] [Revised: 09/30/2018] [Accepted: 10/03/2018] [Indexed: 01/06/2023]
Abstract
Background:
The human Monoamine Transporters (hMATs), primarily including hSERT,
hNET and hDAT, are important targets for the treatment of depression and other behavioral disorders
with more than the availability of 30 approved drugs.
Objective:
This paper is to review the recent progress in the binding mode and inhibitory mechanism of
hMATs inhibitors with the central or allosteric binding sites, for the benefit of future hMATs inhibitor
design and discovery. The Structure-Activity Relationship (SAR) and the selectivity for hit/lead compounds
to hMATs that are evaluated by in vitro and in vivo experiments will be highlighted.
Methods:
PubMed and Web of Science databases were searched for protein-ligand interaction, novel
inhibitors design and synthesis studies related to hMATs.
Results:
Literature data indicate that since the first crystal structure determinations of the homologous
bacterial Leucine Transporter (LeuT) complexed with clomipramine, a sizable database of over 100 experimental
structures or computational models has been accumulated that now defines a substantial degree
of structural variability hMATs-ligands recognition. In the meanwhile, a number of novel hMATs
inhibitors have been discovered by medicinal chemistry with significant help from computational models.
Conclusion:
The reported new compounds act on hMATs as well as the structures of the transporters
complexed with diverse ligands by either experiment or computational modeling have shed light on the
poly-pharmacology, multimodal and allosteric regulation of the drugs to transporters. All of the studies
will greatly promote the Structure-Based Drug Design (SBDD) of structurally novel scaffolds with high
activity and selectivity for hMATs.
Collapse
Affiliation(s)
- Weiwei Xue
- Innovative Drug Research and Bioinformatics Group, School of Pharmaceutical Sciences and Chongqing Key Laboratory of Natural Drug Research, Chongqing University, Chongqing 401331, China
| | - Tingting Fu
- Innovative Drug Research and Bioinformatics Group, School of Pharmaceutical Sciences and Chongqing Key Laboratory of Natural Drug Research, Chongqing University, Chongqing 401331, China
| | - Guoxun Zheng
- Innovative Drug Research and Bioinformatics Group, School of Pharmaceutical Sciences and Chongqing Key Laboratory of Natural Drug Research, Chongqing University, Chongqing 401331, China
| | - Gao Tu
- Innovative Drug Research and Bioinformatics Group, School of Pharmaceutical Sciences and Chongqing Key Laboratory of Natural Drug Research, Chongqing University, Chongqing 401331, China
| | - Yang Zhang
- Innovative Drug Research and Bioinformatics Group, School of Pharmaceutical Sciences and Chongqing Key Laboratory of Natural Drug Research, Chongqing University, Chongqing 401331, China
| | - Fengyuan Yang
- Innovative Drug Research and Bioinformatics Group, School of Pharmaceutical Sciences and Chongqing Key Laboratory of Natural Drug Research, Chongqing University, Chongqing 401331, China
| | - Lin Tao
- Key Laboratory of Elemene Class Anti-cancer Chinese Medicine of Zhejiang Province, School of Medicine, Hangzhou Normal University, Hangzhou 310036, China
| | - Lixia Yao
- Department of Health Sciences Research, Mayo Clinic, Rochester, MN 55905, United States
| | - Feng Zhu
- Innovative Drug Research and Bioinformatics Group, School of Pharmaceutical Sciences and Chongqing Key Laboratory of Natural Drug Research, Chongqing University, Chongqing 401331, China
| |
Collapse
|
16
|
Ribaudo G, Bortoli M, Ongaro A, Oselladore E, Gianoncelli A, Zagotto G, Orian L. Fluoxetine scaffold to design tandem molecular antioxidants and green catalysts. RSC Adv 2020; 10:18583-18593. [PMID: 35518299 PMCID: PMC9053872 DOI: 10.1039/d0ra03509b] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/19/2020] [Accepted: 05/07/2020] [Indexed: 12/12/2022] Open
Abstract
Fluoxetine finds application in the treatment of depression and mood disorders. This selective serotonin-reuptake inhibitor (SSRI) also contrasts oxidative stress by direct ROS scavenging, modulation of the endogenous antioxidant defense system, and/or enhancement of the serotonin antioxidant capacity. We synthesised some fluoxetine analogues incorporating a selenium nucleus, thus expanding its antioxidant potential by enabling a hydroperoxides-inactivating, glutathione peroxidase (GPx)-like activity. Radical scavenging and peroxidatic activity were combined in a water-soluble, drug-like, tandem antioxidant molecule. Selenofluoxetine derivatives were reacted with H2O2 in water, and the mechanistic details of the reaction were unravelled combining nuclear magnetic resonance (NMR), electrospray ionisation-mass spectrometry (ESI-MS) and quantum chemistry calculations. The observed oxidation–elimination process led to the formation of seleninic acid and cinnamylamine in a trans-selective manner. This mechanism is likely to be extended to other substrates for the preparation of unsaturated cinnamylamines. We modified fluoxetine by incorporating a selenium nucleus enabling a hydroperoxide-inactivating, glutathione peroxidase (GPx)-like activity and paving the way for its use as green catalyst.![]()
Collapse
Affiliation(s)
- Giovanni Ribaudo
- Dipartimento di Medicina Molecolare e Traslazionale
- Università degli Studi di Brescia
- 25123 Brescia
- Italy
| | - Marco Bortoli
- Dipartimento di Scienze Chimiche
- Università degli Studi di Padova
- 35131 Padova
- Italy
| | - Alberto Ongaro
- Dipartimento di Medicina Molecolare e Traslazionale
- Università degli Studi di Brescia
- 25123 Brescia
- Italy
| | - Erika Oselladore
- Dipartimento di Scienze del Farmaco
- Università degli Studi di Padova
- 35131 Padova
- Italy
| | - Alessandra Gianoncelli
- Dipartimento di Medicina Molecolare e Traslazionale
- Università degli Studi di Brescia
- 25123 Brescia
- Italy
| | - Giuseppe Zagotto
- Dipartimento di Scienze del Farmaco
- Università degli Studi di Padova
- 35131 Padova
- Italy
| | - Laura Orian
- Dipartimento di Scienze Chimiche
- Università degli Studi di Padova
- 35131 Padova
- Italy
| |
Collapse
|
17
|
Monoamine transporters: structure, intrinsic dynamics and allosteric regulation. Nat Struct Mol Biol 2019; 26:545-556. [PMID: 31270469 DOI: 10.1038/s41594-019-0253-7] [Citation(s) in RCA: 72] [Impact Index Per Article: 12.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/19/2019] [Accepted: 05/14/2019] [Indexed: 12/31/2022]
Abstract
Monoamine transporters (MATs) regulate neurotransmission via the reuptake of dopamine, serotonin and norepinephrine from extra-neuronal regions and thus maintain neurotransmitter homeostasis. As targets of a wide range of compounds, including antidepressants, substances of abuse and drugs for neuropsychiatric and neurodegenerative disorders, their mechanism of action and their modulation by small molecules have long been of broad interest. Recent advances in the structural characterization of dopamine and serotonin transporters have opened the way for structure-based modeling and simulations, which, together with experimental data, now provide mechanistic understanding of their transport function and interactions. Here we review recent progress in the elucidation of the structural dynamics of MATs and their conformational landscape and transitions, as well as allosteric regulation mechanisms.
Collapse
|
18
|
Iyer KA, Alix K, Eltit JM, Solis E, Pan X, Argade MD, Khatri S, De Felice LJ, Sweet DH, Schulte MK, Dukat M. Multi-modal antidepressant-like action of 6- and 7-chloro-2-aminodihydroquinazolines in the mouse tail suspension test. Psychopharmacology (Berl) 2019; 236:2093-2104. [PMID: 30805668 DOI: 10.1007/s00213-019-05203-5] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/27/2018] [Accepted: 02/14/2019] [Indexed: 01/08/2023]
Abstract
RATIONALE 2-Amino-6-chloro-3,4-dihydroquinazoline (e.g., A6CDQ) represents a novel putative antidepressant originally thought to act through a 5-HT3 serotonin receptor antagonist mechanism. Here, we investigated this further by examining a positional isomer of A6CDQ (i.e., A7CDQ). MATERIALS AND METHODS 5-HT3 receptor and transporter activity (uptake-1 and uptake-2) were investigated using a variety of in vitro assays and the in vivo mouse tail suspension test (TST). RESULTS Although A7CDQ binds at 5-HT3 receptors with low affinity (Ki = 1975 nM) compared to A6CDQ (Ki = 80 nM), it retained 5-HT3 receptor antagonist action (IC50 = 5.77 and 0.26 μM, respectively). In the mouse TST A7CDQ produced antidepressant-like actions (ED50 = 0.09 mg/kg) comparable to that of A6CDQ. In addition, A6CDQ was found to be a 5-HT releasing agent (Km = 2.8 μM) at hSERT and a reuptake inhibitor (IC50 = 1.8 μM) at hNET, whereas A7CDQ was a weak reuptake inhibitor (Km = 43.6 μM) at SERT but a releasing agent (EC50 = 3.3 μM) at hNET. Moreover, A6CDQ and A7CDQ were potent inhibitors of uptake-2 (e.g.; OCT3 IC50 = 3.9 and 5.9 μM, respectively). CONCLUSIONS A simple shift of a substituent in a common quinazoline scaffold from one position to another (i.e., a chloro group from the 6- to the 7-position) resulted in a common action in the TST but via a somewhat different mechanism. A6CDQ and A7CDQ might represent the first members of a new class of potential antidepressants with a unique multi-modal mechanism of action.
Collapse
Affiliation(s)
- Kavita A Iyer
- Department of Medicinal Chemistry, School of Pharmacy, Virginia Commonwealth University, PO Box 980540, Richmond, VA, 23298-540, USA
| | - Katie Alix
- Department of Medicinal Chemistry, School of Pharmacy, Virginia Commonwealth University, PO Box 980540, Richmond, VA, 23298-540, USA
| | - Jose M Eltit
- Department of Physiology and Biophysics, School of Medicine, Virginia Commonwealth University, Richmond, VA, 23298, USA
| | - Ernesto Solis
- Department of Physiology and Biophysics, School of Medicine, Virginia Commonwealth University, Richmond, VA, 23298, USA
| | - Xiaolei Pan
- Department of Pharmaceutics, School of Pharmacy, Virginia Commonwealth University, Richmond, VA, 23298, USA
| | - Malaika D Argade
- Department of Medicinal Chemistry, School of Pharmacy, Virginia Commonwealth University, PO Box 980540, Richmond, VA, 23298-540, USA
| | - Shailesh Khatri
- Department of Pharmaceutical Sciences, Philadelphia College of Pharmacy, University of Sciences, Philadelphia, PA, 19104, USA
| | - Louis J De Felice
- Department of Physiology and Biophysics, School of Medicine, Virginia Commonwealth University, Richmond, VA, 23298, USA
| | - Douglas H Sweet
- Department of Pharmaceutics, School of Pharmacy, Virginia Commonwealth University, Richmond, VA, 23298, USA
| | - Marvin K Schulte
- Department of Biomedical and Pharmaceutical Sciences, College of Pharmacy, Kasiska Division of Health Sciences, Idaho State University, Pocatello, ID, 83209, USA
| | - Małgorzata Dukat
- Department of Medicinal Chemistry, School of Pharmacy, Virginia Commonwealth University, PO Box 980540, Richmond, VA, 23298-540, USA.
| |
Collapse
|
19
|
Ladefoged LK, Zeppelin T, Schiøtt B. Molecular modeling of neurological membrane proteins − from binding sites to synapses. Neurosci Lett 2019; 700:38-49. [DOI: 10.1016/j.neulet.2018.05.034] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/28/2018] [Revised: 05/18/2018] [Accepted: 05/22/2018] [Indexed: 01/07/2023]
|
20
|
Substrate and inhibitor binding to the serotonin transporter: Insights from computational, crystallographic, and functional studies. Neuropharmacology 2019; 161:107548. [PMID: 30807752 DOI: 10.1016/j.neuropharm.2019.02.030] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/14/2018] [Revised: 02/19/2019] [Accepted: 02/22/2019] [Indexed: 02/02/2023]
Abstract
The serotonin transporter (SERT) belongs to the monoamine transporter family, which also includes the dopamine and norepinephrine transporters. SERT is essential for regulating serotonergic signaling by the reuptake of serotonin from the synaptic cleft back into the presynaptic neuron. Dysregulation of SERT has been implicated in several major psychiatric disorders such as major depressive disorder (MDD). MDD was among the top five leading causes of years lived with disease in 2016 and is characterized as a major global burden. Several drugs have been developed to target SERT for use in the treatment of MDD, and their respective binding modes and locations within SERT have been studied. The elucidation of the first structure of a bacterial SERT homologue in 2005 has accelerated crystallographic, computational, and functional studies to further elucidate drug binding and method of action in SERT. Herein, we aim to highlight and compare these studies with an emphasis on what the different experimental methods conclude on substrate and inhibitor binding modes, and the potential caveats of using the different types of studies are discussed. We focus this review on the binding of cognate substrate and drugs belonging to the different families of antidepressants, including tricyclic antidepressants, selective serotonin reuptake inhibitors, serotonin-norepinephrine reuptake inhibitors, and multimodal drugs, as well as illicit drugs such as cocaine, amphetamines, and ibogaine. This article is part of the issue entitled 'Special Issue on Neurotransmitter Transporters'.
Collapse
|
21
|
Hayashida KI, Obata H. Strategies to Treat Chronic Pain and Strengthen Impaired Descending Noradrenergic Inhibitory System. Int J Mol Sci 2019; 20:ijms20040822. [PMID: 30769838 PMCID: PMC6412536 DOI: 10.3390/ijms20040822] [Citation(s) in RCA: 55] [Impact Index Per Article: 9.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/12/2019] [Revised: 02/04/2019] [Accepted: 02/12/2019] [Indexed: 12/28/2022] Open
Abstract
Gabapentinoids (gabapentin and pregabalin) and antidepressants (tricyclic antidepressants and serotonin noradrenaline reuptake inhibitors) are often used to treat chronic pain. The descending noradrenergic inhibitory system from the locus coeruleus (LC) to the dorsal horn of the spinal cord plays an important role in the analgesic mechanisms of these drugs. Gabapentinoids activate the LC by inhibiting the release of γ-aminobutyric acid (GABA) and inducing the release of glutamate, thereby increasing noradrenaline levels in the spinal cord. Antidepressants increase noradrenaline levels in the spinal cord by inhibiting reuptake, and accumulating noradrenaline inhibits chronic pain through α2-adrenergic receptors in the spinal cord. Recent animal studies, however, revealed that the function of the descending noradrenergic inhibitory system is impaired in chronic pain states. Other recent studies found that histone deacetylase inhibitors and antidepressants restore the impaired noradrenergic descending inhibitory system acting on noradrenergic neurons in the LC.
Collapse
Affiliation(s)
- Ken-Ichiro Hayashida
- Doctorial Course in Medicine, Organ Function-Oriented Medicine, Akita University Graduate School of Medicine;1-1-1, Hondo, Akita-City, Akita 010-8543, Japan.
| | - Hideaki Obata
- Center for Pain Management and Department of Anesthesiology, Fukushima Medical University; 1 Hikarigaoka, Fukushima-City, Fukushima 960-1295, Japan.
| |
Collapse
|
22
|
Cameron LP, Benson CJ, Dunlap LE, Olson DE. Effects of N, N-Dimethyltryptamine on Rat Behaviors Relevant to Anxiety and Depression. ACS Chem Neurosci 2018; 9:1582-1590. [PMID: 29664276 PMCID: PMC7196340 DOI: 10.1021/acschemneuro.8b00134] [Citation(s) in RCA: 88] [Impact Index Per Article: 12.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/22/2022] Open
Abstract
Depression and anxiety disorders are debilitating diseases resulting in substantial economic costs to society. Traditional antidepressants often take weeks to months to positively affect mood and are ineffective for about 30% of the population. Alternatives, such as ketamine, a dissociative anesthetic capable of producing hallucinations, and the psychoactive tisane ayahuasca, have shown great promise due to their fast-acting nature and effectiveness in treatment-resistant populations. Here, we investigate the effects of N, N-dimethyltryptamine (DMT), the principle hallucinogenic component of ayahuasca, in rodent behavioral assays relevant to anxiety and depression using adult, male, Sprague-Dawley rats. We find that while DMT elicits initial anxiogenic responses in several of these paradigms, its long-lasting effects tend to reduce anxiety by facilitating the extinction of cued fear memory. Furthermore, DMT reduces immobility in the forced swim test, which is a characteristic behavioral response induced by many antidepressants. Our results demonstrate that DMT produces antidepressant and anxiolytic behavioral effects in rodents, warranting further investigation of ayahuasca and classical psychedelics as treatments for depression and post-traumatic stress disorder.
Collapse
Affiliation(s)
- Lindsay P. Cameron
- Neuroscience Graduate Program, University of California, Davis, Davis, California 95618, United States
| | - Charlie J. Benson
- Department of Chemistry, University of California, Davis, Davis, California 95616, United States
| | - Lee E. Dunlap
- Department of Chemistry, University of California, Davis, Davis, California 95616, United States
| | - David E. Olson
- Department of Chemistry, University of California, Davis, Davis, California 95616, United States
- Department of Biochemistry & Molecular Medicine, School of Medicine, University of California, Sacramento, Davis, California 95817, United States
- Center for Neuroscience, University of California, Davis, Davis, California 95618, United States
| |
Collapse
|
23
|
Normann C, Frase S, Haug V, von Wolff G, Clark K, Münzer P, Dorner A, Scholliers J, Horn M, Vo Van T, Seifert G, Serchov T, Biber K, Nissen C, Klugbauer N, Bischofberger J. Antidepressants Rescue Stress-Induced Disruption of Synaptic Plasticity via Serotonin Transporter-Independent Inhibition of L-Type Calcium Channels. Biol Psychiatry 2018; 84:55-64. [PMID: 29174591 DOI: 10.1016/j.biopsych.2017.10.008] [Citation(s) in RCA: 25] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/31/2017] [Revised: 09/29/2017] [Accepted: 10/11/2017] [Indexed: 12/19/2022]
Abstract
BACKGROUND Long-term synaptic plasticity is a basic ability of the brain to dynamically adapt to external stimuli and regulate synaptic strength and ultimately network function. It is dysregulated by behavioral stress in animal models of depression and in humans with major depressive disorder. Antidepressants have been shown to restore disrupted synaptic plasticity in both animal models and humans; however, the underlying mechanism is unclear. METHODS We examined modulation of synaptic plasticity by selective serotonin reuptake inhibitors (SSRIs) in hippocampal brain slices from wild-type rats and serotonin transporter (SERT) knockout mice. Recombinant voltage-gated calcium (Ca2+) channels in heterologous expression systems were used to determine the modulation of Ca2+ channels by SSRIs. We tested the behavioral effects of SSRIs in the chronic behavioral despair model of depression both in the presence and in the absence of SERT. RESULTS SSRIs selectively inhibited hippocampal long-term depression. The inhibition of long-term depression by SSRIs was mediated by a direct block of voltage-activated L-type Ca2+ channels and was independent of SERT. Furthermore, SSRIs protected both wild-type and SERT knockout mice from behavioral despair induced by chronic stress. Finally, long-term depression was facilitated in animals subjected to the behavioral despair model, which was prevented by SSRI treatment. CONCLUSIONS These results showed that antidepressants protected synaptic plasticity and neuronal circuitry from the effects of stress via a modulation of Ca2+ channels and synaptic plasticity independent of SERT. Thus, L-type Ca2+ channels might constitute an important signaling hub for stress response and for pathophysiology and treatment of depression.
Collapse
Affiliation(s)
- Claus Normann
- Department of Psychiatry, University Medical Center Freiburg, University of Freiburg, Freiburg, Germany.
| | - Sibylle Frase
- Department of Psychiatry, University Medical Center Freiburg, University of Freiburg, Freiburg, Germany
| | - Verena Haug
- Department of Psychiatry, University Medical Center Freiburg, University of Freiburg, Freiburg, Germany
| | - Gregor von Wolff
- Department of Psychiatry, University Medical Center Freiburg, University of Freiburg, Freiburg, Germany
| | - Kristin Clark
- Department of Psychiatry, University Medical Center Freiburg, University of Freiburg, Freiburg, Germany
| | - Patrick Münzer
- Institute of Biology, University of Freiburg, Freiburg, Germany
| | - Alexandra Dorner
- Department of Psychiatry, University Medical Center Freiburg, University of Freiburg, Freiburg, Germany
| | - Jonas Scholliers
- Department of Psychiatry, University Medical Center Freiburg, University of Freiburg, Freiburg, Germany
| | - Max Horn
- Department of Psychiatry, University Medical Center Freiburg, University of Freiburg, Freiburg, Germany
| | - Tanja Vo Van
- Department of Psychiatry, University Medical Center Freiburg, University of Freiburg, Freiburg, Germany
| | - Gabriel Seifert
- Department of Psychiatry, University Medical Center Freiburg, University of Freiburg, Freiburg, Germany
| | - Tsvetan Serchov
- Department of Psychiatry, University Medical Center Freiburg, University of Freiburg, Freiburg, Germany
| | - Knut Biber
- Department of Psychiatry, University Medical Center Freiburg, University of Freiburg, Freiburg, Germany
| | - Christoph Nissen
- Department of Psychiatry, University Medical Center Freiburg, University of Freiburg, Freiburg, Germany
| | - Norbert Klugbauer
- Institute of Experimental and Clinical Pharmacology and Toxicology, Faculty of Medicine, University of Freiburg, Freiburg, Germany
| | | |
Collapse
|
24
|
Neiens P, De Simone A, Höfner G, Wanner KT. Simultaneous Multiple MS Binding Assays for the Dopamine, Norepinephrine, and Serotonin Transporters. ChemMedChem 2018; 13:453-463. [PMID: 29451362 DOI: 10.1002/cmdc.201700737] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/24/2017] [Revised: 01/10/2018] [Indexed: 12/17/2022]
Abstract
In this work, we present label-free, mass-spectrometry-based binding assays (MS Binding Assays), targeting the human dopamine, norepinephrine, and serotonin transporters (hDAT, hNET, and hSERT) in simultaneous binding experiments. Using a validated LC-ESI-MS/MS method for quantification of the selective dopamine transporter inhibitor (R,R)-4-(2-benzhydryloxyethyl)-1-(4-fluorobenzyl)piperidin-3-ol ((R,R)-D-84), the selective norepinephrine transporter inhibitor (S,S)-reboxetine, and the selective serotonin reuptake inhibitor (S)-citalopram, binding affinities at the three monoamine transporters could be characterized simultaneously in a single binding experiment. The performed simultaneous saturation and competition experiments yielded results that are in good accordance with those determined in MS Binding Assays addressing the monoamine transporters individually. The results obtained from this study underscore the potential of MS Binding Assays for simultaneous affinity determination at different targets, which is difficult to accomplish with conventional radioligand binding assays.
Collapse
Affiliation(s)
- Patrick Neiens
- Department of Pharmacy-Center of Drug Research, Ludwig-Maximilians-Universität München, Butenandtstr. 5-13, 81377, Munich, Germany
| | - Angela De Simone
- Department for Life Quality Studies, Alma Mater Studiorum-University of Bologna, Corso D'Augusto 237, 47921, Rimini, Italy
| | - Georg Höfner
- Department of Pharmacy-Center of Drug Research, Ludwig-Maximilians-Universität München, Butenandtstr. 5-13, 81377, Munich, Germany
| | - Klaus T Wanner
- Department of Pharmacy-Center of Drug Research, Ludwig-Maximilians-Universität München, Butenandtstr. 5-13, 81377, Munich, Germany
| |
Collapse
|
25
|
Structural basis for recognition of diverse antidepressants by the human serotonin transporter. Nat Struct Mol Biol 2018; 25:170-175. [PMID: 29379174 DOI: 10.1038/s41594-018-0026-8] [Citation(s) in RCA: 100] [Impact Index Per Article: 14.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/08/2017] [Accepted: 12/27/2017] [Indexed: 12/22/2022]
Abstract
Selective serotonin reuptake inhibitors are clinically prescribed antidepressants that act by increasing the local concentrations of neurotransmitters at synapses and in extracellular spaces via blockade of the serotonin transporter. Here we report X-ray structures of engineered thermostable variants of the human serotonin transporter bound to the antidepressants sertraline, fluvoxamine, and paroxetine. The drugs prevent serotonin binding by occupying the central substrate-binding site and stabilizing the transporter in an outward-open conformation. These structures explain how residues within the central site orchestrate binding of chemically diverse inhibitors and mediate transporter drug selectivity.
Collapse
|
26
|
Xue W, Wang P, Tu G, Yang F, Zheng G, Li X, Li X, Chen Y, Yao X, Zhu F. Computational identification of the binding mechanism of a triple reuptake inhibitor amitifadine for the treatment of major depressive disorder. Phys Chem Chem Phys 2018; 20:6606-6616. [DOI: 10.1039/c7cp07869b] [Citation(s) in RCA: 93] [Impact Index Per Article: 13.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/31/2022]
Abstract
A shared binding mode involving eleven key residues at the S1 site of MATs for the binding of amitifadine is identified.
Collapse
|
27
|
Zheng G, Yang F, Fu T, Tu G, Chen Y, Yao X, Xue W, Zhu F. Computational characterization of the selective inhibition of human norepinephrine and serotonin transporters by an escitalopram scaffold. Phys Chem Chem Phys 2018; 20:29513-29527. [DOI: 10.1039/c8cp06232c] [Citation(s) in RCA: 26] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/19/2023]
Abstract
Selective inhibition of human norepinephrine and serotonin transporters has been studied by computational approaches. 4 warm spots in hNET and 4 in hSERT were found to exert a pronounced effect on inhibition by the studied ligands.
Collapse
Affiliation(s)
- Guoxun Zheng
- College of Pharmaceutical Sciences
- Zhejiang University
- Hangzhou 310058
- China
- School of Pharmaceutical Sciences and Collaborative Innovation Center for Brain Science
| | - Fengyuan Yang
- College of Pharmaceutical Sciences
- Zhejiang University
- Hangzhou 310058
- China
- School of Pharmaceutical Sciences and Collaborative Innovation Center for Brain Science
| | - Tingting Fu
- College of Pharmaceutical Sciences
- Zhejiang University
- Hangzhou 310058
- China
- School of Pharmaceutical Sciences and Collaborative Innovation Center for Brain Science
| | - Gao Tu
- College of Pharmaceutical Sciences
- Zhejiang University
- Hangzhou 310058
- China
- School of Pharmaceutical Sciences and Collaborative Innovation Center for Brain Science
| | - Yuzong Chen
- Bioinformatics and Drug Design Group
- Department of Pharmacy
- National University of Singapore
- Singapore 117543
- Singapore
| | - Xiaojun Yao
- State Key Laboratory of Applied Organic Chemistry and Department of Chemistry
- Lanzhou University
- Lanzhou 730000
- China
| | - Weiwei Xue
- School of Pharmaceutical Sciences and Collaborative Innovation Center for Brain Science
- Chongqing University
- Chongqing 401331
- China
| | - Feng Zhu
- College of Pharmaceutical Sciences
- Zhejiang University
- Hangzhou 310058
- China
- School of Pharmaceutical Sciences and Collaborative Innovation Center for Brain Science
| |
Collapse
|
28
|
Analgesic Mechanisms of Antidepressants for Neuropathic Pain. Int J Mol Sci 2017; 18:ijms18112483. [PMID: 29160850 PMCID: PMC5713449 DOI: 10.3390/ijms18112483] [Citation(s) in RCA: 226] [Impact Index Per Article: 28.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/22/2017] [Revised: 11/17/2017] [Accepted: 11/19/2017] [Indexed: 12/16/2022] Open
Abstract
Tricyclic antidepressants and serotonin noradrenaline reuptake inhibitors are used to treat chronic pain, such as neuropathic pain. Why antidepressants are effective for treatment of neuropathic pain and the precise mechanisms underlying their effects, however, remain unclear. The inhibitory effects of these antidepressants for neuropathic pain manifest more quickly than their antidepressive effects, suggesting different modes of action. Recent studies of animal models of neuropathic pain revealed that noradrenaline is extremely important for the inhibition of neuropathic pain. First, increasing noradrenaline in the spinal cord by reuptake inhibition directly inhibits neuropathic pain through α2-adrenergic receptors. Second, increasing noradrenaline acts on the locus coeruleus and improves the function of an impaired descending noradrenergic inhibitory system. Serotonin and dopamine may reinforce the noradrenergic effects to inhibit neuropathic pain. The mechanisms of neuropathic pain inhibition by antidepressants based mainly on experimental findings from animal models of neuropathic pain are discussed in this review.
Collapse
|
29
|
Rannversson H, Andersen J, Bang-Andersen B, Strømgaard K. Mapping the Binding Site for Escitalopram and Paroxetine in the Human Serotonin Transporter Using Genetically Encoded Photo-Cross-Linkers. ACS Chem Biol 2017; 12:2558-2562. [PMID: 28910072 DOI: 10.1021/acschembio.7b00338] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
In spite of the important role of the human serotonin transporter (hSERT) in depression treatment, the molecular details of how antidepressant drugs bind are still not completely understood, in particular those related to potential high- and low-affinity binding sites in hSERT. Here, we utilize amber codon suppression in hSERT to encode the photo-cross-linking unnatural amino acid p-azido-l-phenylalanine into the suggested high- and low-affinity binding sites. We then employ UV-induced cross-linking with azF to map the binding site of escitalopram and paroxetine, two prototypical selective serotonin reuptake inhibitors (SSRIs). We find that the two antidepressant drugs exclusively cross-link to azF incorporated at the high-affinity binding site of hSERT, while cross-linking is not observed at the low-affinity binding site. Combined with previous homology models and recent structural data on hSERT, our results provide important information to understand the molecular details of these clinical relevant binding sites.
Collapse
Affiliation(s)
- Hafsteinn Rannversson
- Center
for Biopharmaceuticals, Department of Drug Design and Pharmacology, University of Copenhagen, Jagtvej 162, 2100 Copenhagen Ø, Denmark
| | - Jacob Andersen
- Center
for Biopharmaceuticals, Department of Drug Design and Pharmacology, University of Copenhagen, Jagtvej 162, 2100 Copenhagen Ø, Denmark
| | | | - Kristian Strømgaard
- Center
for Biopharmaceuticals, Department of Drug Design and Pharmacology, University of Copenhagen, Jagtvej 162, 2100 Copenhagen Ø, Denmark
| |
Collapse
|
30
|
Krout D, Pramod AB, Dahal RA, Tomlinson MJ, Sharma B, Foster JD, Zou MF, Boatang C, Newman AH, Lever JR, Vaughan RA, Henry LK. Inhibitor mechanisms in the S1 binding site of the dopamine transporter defined by multi-site molecular tethering of photoactive cocaine analogs. Biochem Pharmacol 2017; 142:204-215. [PMID: 28734777 DOI: 10.1016/j.bcp.2017.07.015] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/01/2017] [Accepted: 07/17/2017] [Indexed: 10/19/2022]
Abstract
Dopamine transporter (DAT) blockers like cocaine and many other abused and therapeutic drugs bind and stabilize an inactive form of the transporter inhibiting reuptake of extracellular dopamine (DA). The resulting increases in DA lead to the ability of these drugs to induce psychomotor alterations and addiction, but paradoxical findings in animal models indicate that not all DAT antagonists induce cocaine-like behavioral outcomes. How this occurs is not known, but one possibility is that uptake inhibitors may bind at multiple locations or in different poses to stabilize distinct conformational transporter states associated with differential neurochemical endpoints. Understanding the molecular mechanisms governing the pharmacological inhibition of DAT is therefore key for understanding the requisite interactions for behavioral modulation and addiction. Previously, we leveraged complementary computational docking, mutagenesis, peptide mapping, and substituted cysteine accessibility strategies to identify the specific adduction site and binding pose for the crosslinkable, photoactive cocaine analog, RTI 82, which contains a photoactive azide attached at the 2β position of the tropane pharmacophore. Here, we utilize similar methodology with a different cocaine analog N-[4-(4-azido-3-I-iodophenyl)-butyl]-2-carbomethoxy-3-(4-chlorophenyl)tropane, MFZ 2-24, where the photoactive azide is attached to the tropane nitrogen. In contrast to RTI 82, which crosslinked into residue Phe319 of transmembrane domain (TM) 6, our findings show that MFZ 2-24 adducts to Leu80 in TM1 with modeling and biochemical data indicating that MFZ 2-24, like RTI 82, occupies the central S1 binding pocket with the (+)-charged tropane ring nitrogen coordinating with the (-)-charged carboxyl side chain of Asp79. The superimposition of the tropane ring in the three-dimensional binding poses of these two distinct ligands provides strong experimental evidence for cocaine binding to DAT in the S1 site and the importance of the tropane moiety in competitive mechanisms of DA uptake inhibition. These findings set a structure-function baseline for comparison of typical and atypical DAT inhibitors and how their interactions with DAT could lead to the loss of cocaine-like behaviors.
Collapse
Affiliation(s)
- Danielle Krout
- Department of Biomedical Sciences, University of North Dakota School of Medicine and Health Sciences, 1301 North Columbia Road, Grand Forks, ND 58202, USA
| | - Akula Bala Pramod
- Department of Biomedical Sciences, University of North Dakota School of Medicine and Health Sciences, 1301 North Columbia Road, Grand Forks, ND 58202, USA
| | - Rejwi Acharya Dahal
- Department of Biomedical Sciences, University of North Dakota School of Medicine and Health Sciences, 1301 North Columbia Road, Grand Forks, ND 58202, USA
| | - Michael J Tomlinson
- Department of Biomedical Sciences, University of North Dakota School of Medicine and Health Sciences, 1301 North Columbia Road, Grand Forks, ND 58202, USA
| | - Babita Sharma
- Department of Biomedical Sciences, University of North Dakota School of Medicine and Health Sciences, 1301 North Columbia Road, Grand Forks, ND 58202, USA
| | - James D Foster
- Department of Biomedical Sciences, University of North Dakota School of Medicine and Health Sciences, 1301 North Columbia Road, Grand Forks, ND 58202, USA
| | - Mu-Fa Zou
- Medicinal Chemistry Section, National Institute on Drug Abuse - Intramural Research Program, Baltimore, MD 21224, USA
| | - Comfort Boatang
- Medicinal Chemistry Section, National Institute on Drug Abuse - Intramural Research Program, Baltimore, MD 21224, USA
| | - Amy Hauck Newman
- Medicinal Chemistry Section, National Institute on Drug Abuse - Intramural Research Program, Baltimore, MD 21224, USA
| | - John R Lever
- Research Service, Harry S. Truman Memorial Veterans' Hospital, Columbia, MO, 65201, USA; Department of Radiology and Radiopharmaceutical Sciences Institute, University of Missouri, Columbia, MO 65211, USA
| | - Roxanne A Vaughan
- Department of Biomedical Sciences, University of North Dakota School of Medicine and Health Sciences, 1301 North Columbia Road, Grand Forks, ND 58202, USA.
| | - L Keith Henry
- Department of Biomedical Sciences, University of North Dakota School of Medicine and Health Sciences, 1301 North Columbia Road, Grand Forks, ND 58202, USA.
| |
Collapse
|
31
|
Yang F, Fu T, Zhang X, Hu J, Xue W, Zheng G, Li B, Li Y, Yao X, Zhu F. Comparison of computational model and X-ray crystal structure of human serotonin transporter: potential application for the pharmacology of human monoamine transporters. MOLECULAR SIMULATION 2017. [DOI: 10.1080/08927022.2017.1309653] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/15/2022]
Affiliation(s)
- Fengyuan Yang
- Innovative Drug Research and Bioinformatics Group, School of Pharmaceutical Sciences and School of Public Affairs, Chongqing University, Chongqing, P.R. China
| | - Tingting Fu
- Innovative Drug Research and Bioinformatics Group, School of Pharmaceutical Sciences and School of Public Affairs, Chongqing University, Chongqing, P.R. China
| | - Xiaoyu Zhang
- Innovative Drug Research and Bioinformatics Group, School of Pharmaceutical Sciences and School of Public Affairs, Chongqing University, Chongqing, P.R. China
| | - Jie Hu
- Innovative Drug Research and Bioinformatics Group, School of Pharmaceutical Sciences and School of Public Affairs, Chongqing University, Chongqing, P.R. China
| | - Weiwei Xue
- Innovative Drug Research and Bioinformatics Group, School of Pharmaceutical Sciences and School of Public Affairs, Chongqing University, Chongqing, P.R. China
| | - Guoxun Zheng
- Innovative Drug Research and Bioinformatics Group, School of Pharmaceutical Sciences and School of Public Affairs, Chongqing University, Chongqing, P.R. China
| | - Bo Li
- Innovative Drug Research and Bioinformatics Group, School of Pharmaceutical Sciences and School of Public Affairs, Chongqing University, Chongqing, P.R. China
| | - Yinghong Li
- Innovative Drug Research and Bioinformatics Group, School of Pharmaceutical Sciences and School of Public Affairs, Chongqing University, Chongqing, P.R. China
| | - Xiaojun Yao
- State Key Laboratory of Applied Organic Chemistry and Department of Chemistry, Lanzhou University, Lanzhou, P.R. China
| | - Feng Zhu
- Innovative Drug Research and Bioinformatics Group, School of Pharmaceutical Sciences and School of Public Affairs, Chongqing University, Chongqing, P.R. China
| |
Collapse
|
32
|
Andersen J, Ladefoged LK, Kristensen TNB, Munro L, Grouleff J, Stuhr-Hansen N, Kristensen AS, Schiøtt B, Strømgaard K. Interrogating the Molecular Basis for Substrate Recognition in Serotonin and Dopamine Transporters with High-Affinity Substrate-Based Bivalent Ligands. ACS Chem Neurosci 2016; 7:1406-1417. [PMID: 27425420 DOI: 10.1021/acschemneuro.6b00164] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/30/2022] Open
Abstract
The transporters for the neurotransmitters serotonin and dopamine (SERT and DAT, respectively) are targets for drugs used in the treatment of mental disorders and widely used drugs of abuse. Studies of prokaryotic homologues have advanced our structural understanding of SERT and DAT, but it still remains enigmatic whether the human transporters contain one or two high-affinity substrate binding sites. We have designed and employed 24 bivalent ligands possessing a highly systematic combination of substrate moieties (serotonin and/or dopamine) and aliphatic or poly(ethylene glycol) spacers to reveal insight into substrate recognition in SERT and DAT. An optimized bivalent ligand comprising two serotonin moieties binds SERT with 3,800-fold increased affinity compared to that of serotonin, suggesting that the human transporters have two distinct substrate binding sites. We show that the bivalent ligands are inhibitors of SERT and an experimentally validated docking model suggests that the bivalent compounds bind with one substrate moiety in the central binding site (the S1 site), whereas the other substrate moiety binds in a distinct binding site (the S2 site). A systematic study of nonconserved SERT/DAT residues surrounding the proposed binding region showed that nonconserved binding site residues do not contribute to selective recognition of substrates in SERT or DAT. This study provides novel insight into the molecular basis for substrate recognition in human transporters and provides an improved foundation for the development of new drugs targeting SERT and DAT.
Collapse
Affiliation(s)
- Jacob Andersen
- Department
of Drug Design and Pharmacology, University of Copenhagen, DK-2100 Copenhagen, Denmark
| | - Lucy Kate Ladefoged
- Interdisciplinary
Nanoscience Center (iNANO), Department of Chemistry, Aarhus University, DK-8000 Aarhus C, Denmark
| | - Trine N. Bjerre Kristensen
- Interdisciplinary
Nanoscience Center (iNANO), Department of Chemistry, Aarhus University, DK-8000 Aarhus C, Denmark
| | - Lachlan Munro
- Department
of Drug Design and Pharmacology, University of Copenhagen, DK-2100 Copenhagen, Denmark
| | - Julie Grouleff
- Interdisciplinary
Nanoscience Center (iNANO), Department of Chemistry, Aarhus University, DK-8000 Aarhus C, Denmark
| | - Nicolai Stuhr-Hansen
- Department
of Drug Design and Pharmacology, University of Copenhagen, DK-2100 Copenhagen, Denmark
| | - Anders S. Kristensen
- Department
of Drug Design and Pharmacology, University of Copenhagen, DK-2100 Copenhagen, Denmark
| | - Birgit Schiøtt
- Interdisciplinary
Nanoscience Center (iNANO), Department of Chemistry, Aarhus University, DK-8000 Aarhus C, Denmark
| | - Kristian Strømgaard
- Department
of Drug Design and Pharmacology, University of Copenhagen, DK-2100 Copenhagen, Denmark
| |
Collapse
|
33
|
Dupuis A, Wattiez AS, Pinguet J, Richard D, Libert F, Chalus M, Aissouni Y, Sion B, Ardid D, Marin P, Eschalier A, Courteix C. Increasing spinal 5-HT 2A receptor responsiveness mediates anti-allodynic effect and potentiates fluoxetine efficacy in neuropathic rats. Evidence for GABA release. Pharmacol Res 2016; 118:93-103. [PMID: 27663259 DOI: 10.1016/j.phrs.2016.09.021] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/29/2016] [Revised: 09/17/2016] [Accepted: 09/19/2016] [Indexed: 01/13/2023]
Abstract
Antidepressants are one of the first line treatments for neuropathic pain but their use is limited by the incidence and severity of side effects of tricyclics and the weak effectiveness of selective serotonin reuptake inhibitors (SSRIs). Serotonin type 2A (5-HT2A) receptors interact with PDZ proteins that regulate their functionality and SSRI efficacy to alleviate pain. We investigated whether an interfering peptide (TAT-2ASCV) disrupting the interaction between 5-HT2A receptors and associated PDZ proteins would improve the treatment of traumatic neuropathic allodynia. Tactile allodynia was assessed in spinal nerve ligation-induced neuropathic pain in rats using von Frey filaments after acute treatment with TAT-2ASCV and/or 5-HT2A receptor agonist, alone or in combination with repeated treatment with fluoxetine. In vivo microdialysis was performed in order to examine the involvement of GABA in TAT-2ASCV/fluoxetine treatment-associated analgesia. TAT-2ASCV (100ng, single i.t. injection) improved SNL-induced tactile allodynia by increasing 5-HT2A receptor responsiveness to endogenous 5-HT. Fluoxetine alone (10mg/kg, five i.p. injections) slightly increased tactile thresholds and its co-administration with TAT-2ASCV (100ng, single i.t. injection) further enhanced the anti-allodynic effect. This effect depends on the integrity of descending serotonergic bulbospinal pathways and spinal release of GABA. The anti-allodynic effect of fluoxetine can be enhanced by disrupting 5-HT2A receptor-PDZ protein interactions. This enhancement depends on 5-HT2A receptor activation, spinal GABA release and GABAA receptor activation.
Collapse
Affiliation(s)
- Amandine Dupuis
- INSERM, U 1107, Neuro-Dol, F-63001 Clermont-Ferrand, France; Clermont Université, Université d'Auvergne, Pharmacologie Fondamentale et Clinique de la Douleur, BP 10448, F-63000 Clermont-Ferrand, France
| | - Anne-Sophie Wattiez
- INSERM, U 1107, Neuro-Dol, F-63001 Clermont-Ferrand, France; Clermont Université, Université d'Auvergne, Pharmacologie Fondamentale et Clinique de la Douleur, BP 10448, F-63000 Clermont-Ferrand, France
| | - Jérémy Pinguet
- INSERM, U 1107, Neuro-Dol, F-63001 Clermont-Ferrand, France; Clermont Université, Université d'Auvergne, Pharmacologie Fondamentale et Clinique de la Douleur, BP 10448, F-63000 Clermont-Ferrand, France; CHU Clermont-Ferrand, Service de Pharmacologie, Clermont-Ferrand, France
| | - Damien Richard
- INSERM, U 1107, Neuro-Dol, F-63001 Clermont-Ferrand, France; Clermont Université, Université d'Auvergne, Pharmacologie Fondamentale et Clinique de la Douleur, BP 10448, F-63000 Clermont-Ferrand, France; CHU Clermont-Ferrand, Service de Pharmacologie, Clermont-Ferrand, France
| | - Frédéric Libert
- INSERM, U 1107, Neuro-Dol, F-63001 Clermont-Ferrand, France; Clermont Université, Université d'Auvergne, Pharmacologie Fondamentale et Clinique de la Douleur, BP 10448, F-63000 Clermont-Ferrand, France; CHU Clermont-Ferrand, Service de Pharmacologie, Clermont-Ferrand, France
| | - Maryse Chalus
- INSERM, U 1107, Neuro-Dol, F-63001 Clermont-Ferrand, France; Clermont Université, Université d'Auvergne, Pharmacologie Fondamentale et Clinique de la Douleur, BP 10448, F-63000 Clermont-Ferrand, France
| | - Youssef Aissouni
- INSERM, U 1107, Neuro-Dol, F-63001 Clermont-Ferrand, France; Clermont Université, Université d'Auvergne, Pharmacologie Fondamentale et Clinique de la Douleur, BP 10448, F-63000 Clermont-Ferrand, France
| | - Benoit Sion
- INSERM, U 1107, Neuro-Dol, F-63001 Clermont-Ferrand, France; Clermont Université, Université d'Auvergne, Pharmacologie Fondamentale et Clinique de la Douleur, BP 10448, F-63000 Clermont-Ferrand, France
| | - Denis Ardid
- INSERM, U 1107, Neuro-Dol, F-63001 Clermont-Ferrand, France; Clermont Université, Université d'Auvergne, Pharmacologie Fondamentale et Clinique de la Douleur, BP 10448, F-63000 Clermont-Ferrand, France
| | - Philippe Marin
- Institut de Génomique Fonctionnelle, CNRS, UMR 5203, INSERM U1191, Université de Montpellier, F-34094 Montpellier Cedex 5, France
| | - Alain Eschalier
- INSERM, U 1107, Neuro-Dol, F-63001 Clermont-Ferrand, France; Clermont Université, Université d'Auvergne, Pharmacologie Fondamentale et Clinique de la Douleur, BP 10448, F-63000 Clermont-Ferrand, France; CHU Clermont-Ferrand, Service de Pharmacologie, Clermont-Ferrand, France
| | - Christine Courteix
- INSERM, U 1107, Neuro-Dol, F-63001 Clermont-Ferrand, France; Clermont Université, Université d'Auvergne, Pharmacologie Fondamentale et Clinique de la Douleur, BP 10448, F-63000 Clermont-Ferrand, France.
| |
Collapse
|
34
|
Zheng G, Xue W, Wang P, Yang F, Li B, Li X, Li Y, Yao X, Zhu F. Exploring the Inhibitory Mechanism of Approved Selective Norepinephrine Reuptake Inhibitors and Reboxetine Enantiomers by Molecular Dynamics Study. Sci Rep 2016; 6:26883. [PMID: 27230580 PMCID: PMC4882549 DOI: 10.1038/srep26883] [Citation(s) in RCA: 40] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/10/2015] [Accepted: 05/09/2016] [Indexed: 12/28/2022] Open
Abstract
Selective norepinephrine reuptake inhibitors (sNRIs) provide an effective class of approved antipsychotics, whose inhibitory mechanism could facilitate the discovery of privileged scaffolds with enhanced drug efficacy. However, the crystal structure of human norepinephrine transporter (hNET) has not been determined yet and the inhibitory mechanism of sNRIs remains elusive. In this work, multiple computational methods were integrated to explore the inhibitory mechanism of approved sNRIs (atomoxetine, maprotiline, reboxetine and viloxazine), and 3 lines of evidences were provided to verify the calculation results. Consequently, a binding mode defined by interactions between three chemical moieties in sNRIs and eleven residues in hNET was identified as shared by approved sNRIs. In the meantime, binding modes of reboxetine's enantiomers with hNET were compared. 6 key residues favoring the binding of (S, S)-reboxetine over that of (R, R)-reboxetine were discovered. This is the first study reporting that those 11 residues are the common determinants for the binding of approved sNRIs. The identified binding mode shed light on the inhibitory mechanism of approved sNRIs, which could help identify novel scaffolds with improved drug efficacy.
Collapse
Affiliation(s)
- Guoxun Zheng
- Innovative Drug Research and Bioinformatics Group, Innovative Drug Research Centre and School of Pharmaceutical Sciences, Chongqing University, Chongqing 401331, China
| | - Weiwei Xue
- Innovative Drug Research and Bioinformatics Group, Innovative Drug Research Centre and School of Pharmaceutical Sciences, Chongqing University, Chongqing 401331, China
| | - Panpan Wang
- Innovative Drug Research and Bioinformatics Group, Innovative Drug Research Centre and School of Pharmaceutical Sciences, Chongqing University, Chongqing 401331, China
| | - Fengyuan Yang
- Innovative Drug Research and Bioinformatics Group, Innovative Drug Research Centre and School of Pharmaceutical Sciences, Chongqing University, Chongqing 401331, China
| | - Bo Li
- Innovative Drug Research and Bioinformatics Group, Innovative Drug Research Centre and School of Pharmaceutical Sciences, Chongqing University, Chongqing 401331, China
| | - Xiaofeng Li
- Innovative Drug Research and Bioinformatics Group, Innovative Drug Research Centre and School of Pharmaceutical Sciences, Chongqing University, Chongqing 401331, China
| | - Yinghong Li
- Innovative Drug Research and Bioinformatics Group, Innovative Drug Research Centre and School of Pharmaceutical Sciences, Chongqing University, Chongqing 401331, China
| | - Xiaojun Yao
- State Key Laboratory of Applied Organic Chemistry and Department of Chemistry, Lanzhou University, Lanzhou 730000, China
| | - Feng Zhu
- Innovative Drug Research and Bioinformatics Group, Innovative Drug Research Centre and School of Pharmaceutical Sciences, Chongqing University, Chongqing 401331, China
| |
Collapse
|
35
|
Grouleff J, Søndergaard S, Koldsø H, Schiøtt B. Properties of an inward-facing state of LeuT: conformational stability and substrate release. Biophys J 2016; 108:1390-1399. [PMID: 25809252 DOI: 10.1016/j.bpj.2015.02.010] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/24/2014] [Revised: 02/02/2015] [Accepted: 02/03/2015] [Indexed: 01/12/2023] Open
Abstract
The leucine transporter (LeuT) is a bacterial homolog of the human monoamine transporters, which are important pharmaceutical targets. There are no high-resolution structures of the human transporters available; however, LeuT has been crystallized in several different conformational states. Recently, an inward-facing conformation of LeuT was solved revealing an unexpectedly large movement of transmembrane helix 1a (TM1a). We have performed molecular dynamics simulations of the mutated and wild-type transporter, with and without the cocrystallized Fab antibody fragment, to investigate the properties of this inward-facing conformation in relation to transport by LeuT within the membrane environment. In all of the simulations, local conformational changes with respect to the crystal structure are consistently observed, especially in TM1a. Umbrella sampling revealed a soft potential for TM1a tilting. Furthermore, simulations of inward-facing LeuT with Na(+) ions and substrate bound suggest that one of the Na(+) ion binding sites is fully disrupted. Release of alanine and the second Na(+) ion is also observed, giving insight into the final stage of the translocation process in atomistic detail.
Collapse
Affiliation(s)
- Julie Grouleff
- Center for Insoluble Protein Structures and Interdisciplinary Nanoscience Center, Department of Chemistry, Aarhus University, Aarhus, Denmark
| | - Siri Søndergaard
- Center for Insoluble Protein Structures and Interdisciplinary Nanoscience Center, Department of Chemistry, Aarhus University, Aarhus, Denmark
| | - Heidi Koldsø
- Center for Insoluble Protein Structures and Interdisciplinary Nanoscience Center, Department of Chemistry, Aarhus University, Aarhus, Denmark
| | - Birgit Schiøtt
- Center for Insoluble Protein Structures and Interdisciplinary Nanoscience Center, Department of Chemistry, Aarhus University, Aarhus, Denmark.
| |
Collapse
|
36
|
Synthesis and inhibitory evaluation of 3-linked imipramines for the exploration of the S2 site of the human serotonin transporter. Bioorg Med Chem 2016; 24:2725-38. [PMID: 27160055 DOI: 10.1016/j.bmc.2016.04.039] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/19/2015] [Revised: 04/16/2016] [Accepted: 04/20/2016] [Indexed: 01/10/2023]
Abstract
The human serotonin transporter is the primary target of several antidepressant drugs, and the importance of a primary, high affinity binding site (S1) for antidepressant binding is well documented. The existence of a lower affinity, secondary binding site (S2) has, however, been debated. Herein we report the synthesis of 3-position coupled imipramine ligands from clomipramine using a copper free Sonogashira reaction. Ligand design was inspired by results from docking and steered molecular dynamics simulations, and the ligands were utilized in a structure-activity relationship study of the positional relationship between the S1 and S2 sites. The computer simulations suggested that the S2 site does indeed exist although with lower affinity for imipramine than observed within the S1 site. Additionally, it was possible to dock the 3-linked imipramine analogs into positions which occupy the S1 and the S2 site simultaneously. The structure activity relationship study showed that the shortest ligands were the most potent, and mutations enlarging the proposed S2 site were found to affect the larger ligands positively, while the smaller ligands were mostly unaffected.
Collapse
|
37
|
Genetically encoded photocrosslinkers locate the high-affinity binding site of antidepressant drugs in the human serotonin transporter. Nat Commun 2016; 7:11261. [PMID: 27089947 PMCID: PMC4838859 DOI: 10.1038/ncomms11261] [Citation(s) in RCA: 46] [Impact Index Per Article: 5.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/23/2015] [Accepted: 03/07/2016] [Indexed: 02/02/2023] Open
Abstract
Despite the well-established role of the human serotonin transporter (hSERT) in the treatment of depression, the molecular details of antidepressant drug binding are still not fully understood. Here we utilize amber codon suppression in a membrane-bound transporter protein to encode photocrosslinking unnatural amino acids (UAAs) into 75 different positions in hSERT. UAAs are incorporated with high specificity, and functionally active transporters have similar transport properties and pharmacological profiles compared with wild-type transporters. We employ ultraviolet-induced crosslinking with p-azido-L-phenylalanine (azF) at selected positions in hSERT to map the binding site of imipramine, a prototypical tricyclic antidepressant, and vortioxetine, a novel multimodal antidepressant. We find that the two antidepressants crosslink with azF incorporated at different positions within the central substrate-binding site of hSERT, while no crosslinking is observed at the vestibular-binding site. Taken together, our data provide direct evidence for defining the high-affinity antidepressant binding site in hSERT. Molecular details of how antidepressant drugs bind to the human serotonin transporter are not currently clear. Here, the authors introduce photo-cross-linkers into the protein and map the binding site of several antidepressants.
Collapse
|
38
|
Davis BA, Nagarajan A, Forrest LR, Singh SK. Mechanism of Paroxetine (Paxil) Inhibition of the Serotonin Transporter. Sci Rep 2016; 6:23789. [PMID: 27032980 PMCID: PMC4817154 DOI: 10.1038/srep23789] [Citation(s) in RCA: 40] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/19/2016] [Accepted: 03/15/2016] [Indexed: 11/11/2022] Open
Abstract
The serotonin transporter (SERT) is an integral membrane protein that exploits preexisting sodium-, chloride-, and potassium ion gradients to catalyze the thermodynamically unfavorable movement of synaptic serotonin into the presynaptic neuron. SERT has garnered significant clinical attention partly because it is the target of multiple psychoactive agents, including the antidepressant paroxetine (Paxil), the most potent selective serotonin reuptake inhibitor known. However, the binding site and orientation of paroxetine in SERT remain controversial. To provide molecular insight, we constructed SERT homology models based on the Drosophila melanogaster dopamine transporter and docked paroxetine to these models. We tested the predicted binding configurations with a combination of radioligand binding and flux assays on wild-type and mutant SERTs. Our data suggest that the orientation of paroxetine, specifically its fluorophenyl ring, in SERT’s substrate binding site directly depends on this pocket’s charge distribution, and thereby provide an avenue toward understanding and enhancing high-affinity antidepressant activity.
Collapse
Affiliation(s)
- Bruce A Davis
- Department of Cellular and Molecular Physiology, Yale University School of Medicine, 333 Cedar Street, New Haven, CT 06520 USA
| | - Anu Nagarajan
- Computational Structural Biology Section, National Institute of Neurological Disorders and Stroke, 35 Convent Drive, Bethesda, MD 20892 USA
| | - Lucy R Forrest
- Computational Structural Biology Section, National Institute of Neurological Disorders and Stroke, 35 Convent Drive, Bethesda, MD 20892 USA
| | - Satinder K Singh
- Department of Cellular and Molecular Physiology, Yale University School of Medicine, 333 Cedar Street, New Haven, CT 06520 USA
| |
Collapse
|
39
|
Larsen MAB, Plenge P, Andersen J, Eildal JNN, Kristensen AS, Bøgesø KP, Gether U, Strømgaard K, Bang-Andersen B, Loland CJ. Structure-activity relationship studies of citalopram derivatives: examining substituents conferring selectivity for the allosteric site in the 5-HT transporter. Br J Pharmacol 2016; 173:925-36. [PMID: 26699847 DOI: 10.1111/bph.13411] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/30/2015] [Revised: 12/10/2015] [Accepted: 12/14/2015] [Indexed: 01/05/2023] Open
Abstract
BACKGROUND AND PURPOSE The 5-HT transporter (SERT) is a target for antidepressant drugs. SERT possesses two binding sites: the orthosteric (S1) binding site, which is the presumed target for current SERT inhibitors, and an allosteric (S2) site for which potential therapeutic effects are unknown. The antidepressant drug citalopram displays high-affinity S1 binding and low-affinity S2 binding. To elucidate a possible therapeutic role of allosteric inhibition of SERT, a drug that specifically targets the allosteric site is required. The purpose of this study was to find a compound having higher selectivity towards the S2 site. EXPERIMENTAL APPROACH We performed a systematic structure-activity relationship study based on the scaffold of citalopram and the structurally closely related congener, talopram, which shows low-affinity S1 binding in SERT. The role of the four chemical substituents, which distinguish citalopram from talopram in conferring selectivity towards the S1 and S2 site, respectively, was assessed by determining the binding of 14 citalopram/talopram analogous to the S1 and S2 binding sites in SERT using membranes of COS7 cells transiently expressing SERT. KEY RESULTS The structure-activity relationship study revealed that dimethyl citalopram possesses the highest affinity for the allosteric site relative to the S1 site in SERT and has approximately twofold selectivity for the allosteric site relative to the S1 site in SERT. CONCLUSIONS AND IMPLICATIONS The compound could be a useful lead for future synthesis of drugs with high affinity and high selectivity towards the allosteric binding site.
Collapse
Affiliation(s)
- M Andreas B Larsen
- Department of Neuroscience and Pharmacology, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark.,Department of Immunology and Microbiology, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark
| | - Per Plenge
- Department of Neuroscience and Pharmacology, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark
| | - Jacob Andersen
- Department of Drug Design and Pharmacology, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark
| | - Jonas N N Eildal
- Department of Drug Design and Pharmacology, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark
| | - Anders S Kristensen
- Department of Drug Design and Pharmacology, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark
| | - Klaus P Bøgesø
- Neuroscience Drug Discovery, H. Lundbeck A/S, Valby, Denmark
| | - Ulrik Gether
- Department of Neuroscience and Pharmacology, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark
| | - Kristian Strømgaard
- Department of Drug Design and Pharmacology, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark
| | - Benny Bang-Andersen
- Department of Drug Design and Pharmacology, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark.,Neuroscience Drug Discovery, H. Lundbeck A/S, Valby, Denmark
| | - Claus J Loland
- Department of Neuroscience and Pharmacology, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark
| |
Collapse
|
40
|
Xue W, Wang P, Li B, Li Y, Xu X, Yang F, Yao X, Chen YZ, Xu F, Zhu F. Identification of the inhibitory mechanism of FDA approved selective serotonin reuptake inhibitors: an insight from molecular dynamics simulation study. Phys Chem Chem Phys 2016; 18:3260-71. [DOI: 10.1039/c5cp05771j] [Citation(s) in RCA: 49] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/28/2022]
Abstract
The binding mode shared by 4 FDA approved SSRIs treating major depression was identified by integrating multiple computational methods.
Collapse
|
41
|
Andersen J, Ladefoged LK, Wang D, Kristensen TNB, Bang-Andersen B, Kristensen AS, Schiøtt B, Strømgaard K. Binding of the multimodal antidepressant drug vortioxetine to the human serotonin transporter. ACS Chem Neurosci 2015; 6:1892-900. [PMID: 26389667 DOI: 10.1021/acschemneuro.5b00225] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022] Open
Abstract
Selective inhibitors of the human serotonin transporter (hSERT) have been first-line treatment against depression for several decades. Recently, vortioxetine was approved as a new therapeutic option for the treatment of depression. Vortioxetine represents a new class of antidepressant drugs with a multimodal pharmacological profile that in addition to potent inhibition of hSERT include agonistic or antagonistic effects at different serotonin receptors. We used a combination of computational, chemical, and biological methods to decipher the molecular basis for high affinity binding of vortioxetine in hSERT. X-ray crystal structures of the bacterial amino acid transporter LeuT and the Drosophila melanogaster dopamine transporter were used to build homology models of hSERT. Comparative modeling and ligand docking suggest that vortioxetine can adopt several distinct binding modes within the central binding site of hSERT. To distinguish between the identified binding modes, we determined the effect of 57 functional hSERT point mutants on vortioxetine potency and characterized seven structurally related analogs of vortioxetine in a subset of the point mutants. This allowed us to determine the orientation of vortioxetine within the central binding site and showed that only one of the proposed binding modes is functionally relevant. The findings provide important new insight about the molecular basis for high affinity recognition of vortioxetine in hSERT, which is essential for future structure-based drug discovery of novel multimodal drugs with fine-tuned selectivity across different transporter and receptor proteins in the human brain.
Collapse
Affiliation(s)
- Jacob Andersen
- Department of Drug Design and Pharmacology, University of Copenhagen, Universitetsparken 2, DK-2100 Copenhagen, Denmark
| | - Lucy Kate Ladefoged
- Center for Insoluble Protein Structures (inSPIN) and Interdisciplinary
Nanoscience Center (iNANO), Department of Chemistry, Aarhus University, Langelandsgade
140, DK-8000 Aarhus
C, Denmark
| | - Danyang Wang
- Department of Drug Design and Pharmacology, University of Copenhagen, Universitetsparken 2, DK-2100 Copenhagen, Denmark
| | - Trine N. Bjerre Kristensen
- Center for Insoluble Protein Structures (inSPIN) and Interdisciplinary
Nanoscience Center (iNANO), Department of Chemistry, Aarhus University, Langelandsgade
140, DK-8000 Aarhus
C, Denmark
| | - Benny Bang-Andersen
- Department of Drug Design and Pharmacology, University of Copenhagen, Universitetsparken 2, DK-2100 Copenhagen, Denmark
- Lundbeck Research Denmark, H. Lundbeck A/S, Ottiliavej 9, DK-2500 Valby, Denmark
| | - Anders S. Kristensen
- Department of Drug Design and Pharmacology, University of Copenhagen, Universitetsparken 2, DK-2100 Copenhagen, Denmark
| | - Birgit Schiøtt
- Center for Insoluble Protein Structures (inSPIN) and Interdisciplinary
Nanoscience Center (iNANO), Department of Chemistry, Aarhus University, Langelandsgade
140, DK-8000 Aarhus
C, Denmark
| | - Kristian Strømgaard
- Department of Drug Design and Pharmacology, University of Copenhagen, Universitetsparken 2, DK-2100 Copenhagen, Denmark
| |
Collapse
|
42
|
Andersen J, Ringsted KB, Bang-Andersen B, Strømgaard K, Kristensen AS. Binding site residues control inhibitor selectivity in the human norepinephrine transporter but not in the human dopamine transporter. Sci Rep 2015; 5:15650. [PMID: 26503701 PMCID: PMC4621520 DOI: 10.1038/srep15650] [Citation(s) in RCA: 25] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/06/2015] [Accepted: 10/01/2015] [Indexed: 02/08/2023] Open
Abstract
The transporters for norepinephrine and dopamine (NET and DAT, respectively) constitute the molecular targets for recreational drugs and therapeutics used in the treatment of psychiatric disorders. Despite a strikingly similar amino acid sequence and predicted topology between these transporters, some inhibitors display a high degree of selectivity between NET and DAT. Here, a systematic mutational analysis of non-conserved residues within the extracellular entry pathway and the high affinity binding site in NET and DAT was performed to examine their role for selective inhibitor recognition. Changing the six diverging residues in the central binding site of NET to the complementary residues in DAT transferred a DAT-like pharmacology to NET, showing that non-conserved binding site residues in NET are critical determinants for inhibitor selectivity. In contrast, changing the equivalent residues in the central site of DAT to the corresponding residues in NET had modest effects on the same inhibitors, suggesting that non-conserved binding site residues in DAT play a minor role for selective inhibitor recognition. Our data points towards distinct structural determinants governing inhibitor selectivity in NET and DAT, and provide important new insight into the molecular basis for NET/DAT selectivity of therapeutic and recreational drugs.
Collapse
Affiliation(s)
- Jacob Andersen
- Department of Drug Design and Pharmacology, University of Copenhagen, Jagtvej 162, DK-2100 Copenhagen, Denmark
| | - Kristoffer B Ringsted
- Department of Drug Design and Pharmacology, University of Copenhagen, Jagtvej 162, DK-2100 Copenhagen, Denmark
| | - Benny Bang-Andersen
- Department of Drug Design and Pharmacology, University of Copenhagen, Jagtvej 162, DK-2100 Copenhagen, Denmark.,Lundbeck Research Denmark, H. Lundbeck A/S, Ottiliavej 9, DK-2500 Valby, Denmark
| | - Kristian Strømgaard
- Department of Drug Design and Pharmacology, University of Copenhagen, Jagtvej 162, DK-2100 Copenhagen, Denmark
| | - Anders S Kristensen
- Department of Drug Design and Pharmacology, University of Copenhagen, Jagtvej 162, DK-2100 Copenhagen, Denmark
| |
Collapse
|
43
|
Grouleff J, Ladefoged LK, Koldsø H, Schiøtt B. Monoamine transporters: insights from molecular dynamics simulations. Front Pharmacol 2015; 6:235. [PMID: 26528185 PMCID: PMC4607855 DOI: 10.3389/fphar.2015.00235] [Citation(s) in RCA: 52] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/01/2015] [Accepted: 10/01/2015] [Indexed: 01/31/2023] Open
Abstract
The human monoamine transporters (MATs) facilitate the reuptake of the neurotransmitters serotonin, dopamine, and norepinephrine from the synaptic cleft. Imbalance in monoaminergic neurotransmission is linked to various diseases including major depression, attention deficit hyperactivity disorder, schizophrenia, and Parkinson's disease. Inhibition of the MATs is thus an important strategy for treatment of such diseases. The MATs are sodium-coupled transport proteins belonging to the neurotransmitter/Na(+) symporter (NSS) family, and the publication of the first high-resolution structure of a NSS family member, the bacterial leucine transporter LeuT, in 2005, proved to be a major stepping stone for understanding this family of transporters. Structural data allows for the use of computational methods to study the MATs, which in turn has led to a number of important discoveries. The process of substrate translocation across the membrane is an intrinsically dynamic process. Molecular dynamics simulations, which can provide atomistic details of molecular motion on ns to ms timescales, are therefore well-suited for studying transport processes. In this review, we outline how molecular dynamics simulations have provided insight into the large scale motions associated with transport of the neurotransmitters, as well as the presence of external and internal gates, the coupling between ion and substrate transport, and differences in the conformational changes induced by substrates and inhibitors.
Collapse
Affiliation(s)
- Julie Grouleff
- Center for Insoluble Protein Structures and Interdisciplinary Nanoscience Center, Department of Chemistry, Aarhus University Aarhus, Denmark
| | - Lucy Kate Ladefoged
- Center for Insoluble Protein Structures and Interdisciplinary Nanoscience Center, Department of Chemistry, Aarhus University Aarhus, Denmark
| | - Heidi Koldsø
- Department of Biochemistry, University of Oxford Oxford, UK
| | - Birgit Schiøtt
- Center for Insoluble Protein Structures and Interdisciplinary Nanoscience Center, Department of Chemistry, Aarhus University Aarhus, Denmark
| |
Collapse
|
44
|
Söderhielm PC, Andersen J, Munro L, Nielsen AT, Kristensen AS. Substrate and Inhibitor-Specific Conformational Changes in the Human Serotonin Transporter Revealed by Voltage-Clamp Fluorometry. Mol Pharmacol 2015; 88:676-88. [PMID: 26174773 DOI: 10.1124/mol.115.099911] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/13/2015] [Accepted: 07/13/2015] [Indexed: 01/05/2023] Open
Abstract
The serotonin transporter (SERT) regulates neurotransmission by the biogenic monoamine neurotransmitter serotonin (5-HT, 5-hydroxytryptamine) in the central nervous system, and drugs inhibiting SERT are widely used for the treatment of a variety of central nervous system diseases. The conformational dynamics of SERT transport function and inhibition is currently poorly understood. We used voltage-clamp fluorometry to study conformational changes in human SERT (hSERT) during 5-HT transport and inhibitor binding. Cys residues were introduced at 12 positions in hSERT to enable covalent attachment of a rhodamine-based fluorophore. Transport-associated changes in fluorescence from fluorophore-labeled hSERT expressed in Xenopus oocytes could be robustly detected at four positions in hSERT: endogenous Cys109 in the top of transmembrane domain (TM) 1b, Cys substituted for Thr323 in the top of TM6, Ala419 in the interface between TM8 and extracellular loop (EL) 4, and Leu481 in EL5. The reporter positions were used for time-resolved measurement of conformational changes during 5-HT transport and binding of cocaine and the selective serotonin reuptake inhibitors fluoxetine and escitalopram. At all reporter positions, fluorescence changes observed upon substrate application were distinctly different from those observed upon inhibitor application, with respect to relative amplitude or direction. Furthermore, escitalopram, fluoxetine, and cocaine induced a very similar pattern of fluorescent changes overall, which included movements within or around TM1b, EL4, and EL5. Taken together, our data lead us to suggest that competitive inhibitors stabilize hSERT in a state that is different from the apo outward-open conformation as well as inward-facing conformations.
Collapse
Affiliation(s)
- Pella C Söderhielm
- Department of Drug Design and Pharmacology, University of Copenhagen, Copenhagen, Denmark
| | - Jacob Andersen
- Department of Drug Design and Pharmacology, University of Copenhagen, Copenhagen, Denmark
| | - Lachlan Munro
- Department of Drug Design and Pharmacology, University of Copenhagen, Copenhagen, Denmark
| | - Anne T Nielsen
- Department of Drug Design and Pharmacology, University of Copenhagen, Copenhagen, Denmark
| | - Anders S Kristensen
- Department of Drug Design and Pharmacology, University of Copenhagen, Copenhagen, Denmark
| |
Collapse
|
45
|
Mortensen OV, Kortagere S. Designing modulators of monoamine transporters using virtual screening techniques. Front Pharmacol 2015; 6:223. [PMID: 26483692 PMCID: PMC4586420 DOI: 10.3389/fphar.2015.00223] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/30/2015] [Accepted: 09/17/2015] [Indexed: 12/15/2022] Open
Abstract
The plasma-membrane monoamine transporters (MATs), including the serotonin (SERT), norepinephrine (NET) and dopamine (DAT) transporters, serve a pivotal role in limiting monoamine-mediated neurotransmission through the reuptake of their respective monoamine neurotransmitters. The transporters are the main target of clinically used psychostimulants and antidepressants. Despite the availability of several potent and selective MAT substrates and inhibitors the continuing need for therapeutic drugs to treat brain disorders involving aberrant monoamine signaling provides a compelling reason to identify novel ways of targeting and modulating the MATs. Designing novel modulators of MAT function have been limited by the lack of three dimensional structure information of the individual MATs. However, crystal structures of LeuT, a bacterial homolog of MATs, in a substrate-bound occluded, substrate-free outward-open, and an apo inward-open state and also with competitive and non-competitive inhibitors have been determined. In addition, several structures of the Drosophila DAT have also been resolved. Together with computational modeling and experimental data gathered over the past decade, these structures have dramatically advanced our understanding of several aspects of SERT, NET, and DAT transporter function, including some of the molecular determinants of ligand interaction at orthosteric substrate and inhibitor binding pockets. In addition progress has been made in the understanding of how allosteric modulation of MAT function can be achieved. Here we will review all the efforts up to date that has been made through computational approaches employing structural models of MATs to design small molecule modulators to the orthosteric and allosteric sites using virtual screening techniques.
Collapse
Affiliation(s)
- Ole V Mortensen
- Department of Pharmacology and Physiology, Drexel University College of Medicine , Philadelphia, PA, USA
| | - Sandhya Kortagere
- Department of Microbiology and Immunology, Drexel University College of Medicine , Philadelphia, PA, USA
| |
Collapse
|
46
|
Koldsø H, Grouleff J, Schiøtt B. Insights to ligand binding to the monoamine transporters-from homology modeling to LeuBAT and dDAT. Front Pharmacol 2015; 6:208. [PMID: 26441663 PMCID: PMC4585151 DOI: 10.3389/fphar.2015.00208] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/01/2015] [Accepted: 09/07/2015] [Indexed: 11/20/2022] Open
Abstract
Understanding of drug binding to the human biogenic amine transporters (BATs) is essential to explain the mechanism of action of these pharmaceuticals but more importantly to be able to develop new and improved compounds to be used in the treatment of depression or drug addiction. Until recently no high resolution structure was available of the BATs and homology modeling was a necessity. Various studies have revealed experimentally validated binding modes of numerous ligands to the BATs using homology modeling. Here we examine and discuss the similarities between the binding models of substrates, antidepressants, psychostimulants, and mazindol in homology models of the human BATs and the recently published crystal structures of the Drosophila dopamine transporter and the engineered protein, LeuBAT. The comparison reveals that careful computational modeling combined with experimental data can be utilized to predict binding of molecules to proteins that agree very well with crystal structures.
Collapse
Affiliation(s)
- Heidi Koldsø
- Department of Biochemistry, University of Oxford , Oxford, UK ; inSPIN and iNANO Centers, Department of Chemistry, Aarhus University , Aarhus C, Denmark
| | - Julie Grouleff
- inSPIN and iNANO Centers, Department of Chemistry, Aarhus University , Aarhus C, Denmark
| | - Birgit Schiøtt
- inSPIN and iNANO Centers, Department of Chemistry, Aarhus University , Aarhus C, Denmark
| |
Collapse
|
47
|
Hasenhuetl PS, Schicker K, Koenig X, Li Y, Sarker S, Stockner T, Sucic S, Sitte HH, Freissmuth M, Sandtner W. Ligand Selectivity among the Dopamine and Serotonin Transporters Specified by the Forward Binding Reaction. Mol Pharmacol 2015; 88:12-8. [PMID: 25873594 DOI: 10.1124/mol.115.099036] [Citation(s) in RCA: 26] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/18/2015] [Accepted: 04/14/2015] [Indexed: 11/08/2023] Open
Abstract
The membrane transporters for the monoamines serotonin (SERT) and dopamine (DAT) are prominent targets of various psychoactive substances, including competitive inhibitors, such as tricyclic antidepressants, methylphenidate, and cocaine. Upon rapid application of a substrate, SERT and DAT display an inwardly directed current comprised of a peak and a steady-state component. Binding of a competitive inhibitor to the transporter leads to reduction of the peak current amplitude because occupancy of the transporter by an inhibitor prevents the induction of the peak current by the substrate. We show that the inhibitory effect on the peak current can be used to study the association rate constant (k(on)), dissociation rate constant (k(off)), and equilibrium dissociation constant (K(D)) of chemically distinct SERT and DAT inhibitors, with high temporal precision and without the need of high-affinity radioligands as surrogates. We exemplify our approach by measuring the kinetics of cocaine, methylphenidate, and desipramine binding to SERT and DAT. Our analysis revealed that the selectivity of methylphenidate and desipramine for DAT and SERT, respectively, can be accounted for by their rate of association and not by the residence time in their respective binding sites.
Collapse
Affiliation(s)
- Peter S Hasenhuetl
- Institute of Pharmacology, Center of Physiology and Pharmacology, Medical University of Vienna, Vienna, Austria (P.S.H., K.S., X.K., Y.L., Su.S., T.S., So.S., H.H.S., M.F., W.S.)
| | - Klaus Schicker
- Institute of Pharmacology, Center of Physiology and Pharmacology, Medical University of Vienna, Vienna, Austria (P.S.H., K.S., X.K., Y.L., Su.S., T.S., So.S., H.H.S., M.F., W.S.)
| | - Xaver Koenig
- Institute of Pharmacology, Center of Physiology and Pharmacology, Medical University of Vienna, Vienna, Austria (P.S.H., K.S., X.K., Y.L., Su.S., T.S., So.S., H.H.S., M.F., W.S.)
| | - Yang Li
- Institute of Pharmacology, Center of Physiology and Pharmacology, Medical University of Vienna, Vienna, Austria (P.S.H., K.S., X.K., Y.L., Su.S., T.S., So.S., H.H.S., M.F., W.S.)
| | - Subhodeep Sarker
- Institute of Pharmacology, Center of Physiology and Pharmacology, Medical University of Vienna, Vienna, Austria (P.S.H., K.S., X.K., Y.L., Su.S., T.S., So.S., H.H.S., M.F., W.S.)
| | - Thomas Stockner
- Institute of Pharmacology, Center of Physiology and Pharmacology, Medical University of Vienna, Vienna, Austria (P.S.H., K.S., X.K., Y.L., Su.S., T.S., So.S., H.H.S., M.F., W.S.)
| | - Sonja Sucic
- Institute of Pharmacology, Center of Physiology and Pharmacology, Medical University of Vienna, Vienna, Austria (P.S.H., K.S., X.K., Y.L., Su.S., T.S., So.S., H.H.S., M.F., W.S.)
| | - Harald H Sitte
- Institute of Pharmacology, Center of Physiology and Pharmacology, Medical University of Vienna, Vienna, Austria (P.S.H., K.S., X.K., Y.L., Su.S., T.S., So.S., H.H.S., M.F., W.S.)
| | - Michael Freissmuth
- Institute of Pharmacology, Center of Physiology and Pharmacology, Medical University of Vienna, Vienna, Austria (P.S.H., K.S., X.K., Y.L., Su.S., T.S., So.S., H.H.S., M.F., W.S.)
| | - Walter Sandtner
- Institute of Pharmacology, Center of Physiology and Pharmacology, Medical University of Vienna, Vienna, Austria (P.S.H., K.S., X.K., Y.L., Su.S., T.S., So.S., H.H.S., M.F., W.S.)
| |
Collapse
|
48
|
Guedj F, Pennings JLA, Ferres MA, Graham LC, Wick HC, Miczek KA, Slonim DK, Bianchi DW. The fetal brain transcriptome and neonatal behavioral phenotype in the Ts1Cje mouse model of Down syndrome. Am J Med Genet A 2015; 167A:1993-2008. [PMID: 25975229 DOI: 10.1002/ajmg.a.37156] [Citation(s) in RCA: 26] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/26/2014] [Accepted: 04/27/2015] [Indexed: 11/07/2022]
Abstract
Human fetuses with Down syndrome demonstrate abnormal brain growth and reduced neurogenesis. Despite the prenatal onset of the phenotype, most therapeutic trials have been conducted in adults. Here, we present evidence for fetal brain molecular and neonatal behavioral alterations in the Ts1Cje mouse model of Down syndrome. Embryonic day 15.5 brain hemisphere RNA from Ts1Cje embryos (n = 5) and wild type littermates (n = 5) was processed and hybridized to mouse gene 1.0 ST arrays. Bioinformatic analyses were implemented to identify differential gene and pathway regulation during Ts1Cje fetal brain development. In separate experiments, the Fox scale, ultrasonic vocalization and homing tests were used to investigate behavioral deficits in Ts1Cje pups (n = 29) versus WT littermates (n = 64) at postnatal days 3-21. Ts1Cje fetal brains displayed more differentially regulated genes (n = 71) than adult (n = 31) when compared to their age-matched euploid brains. Ts1Cje embryonic brains showed up-regulation of cell cycle markers and down-regulation of the solute-carrier amino acid transporters. Several cellular processes were dysregulated at both stages, including apoptosis, inflammation, Jak/Stat signaling, G-protein signaling, and oxidoreductase activity. In addition, early behavioral deficits in surface righting, cliff aversion, negative geotaxis, forelimb grasp, ultrasonic vocalization, and the homing tests were observed. The Ts1Cje mouse model exhibits abnormal gene expression during fetal brain development, and significant neonatal behavioral deficits in the pre-weaning period. In combination with human studies, this suggests that the Down syndrome phenotype manifests prenatally and provides a rationale for prenatal therapy to improve perinatal brain development and postnatal neurocognition.
Collapse
Affiliation(s)
- Faycal Guedj
- Mother Infant Research Institute, Tufts Medical Center and the Floating Hospital for Children, Boston, Massachusetts
| | - Jeroen L A Pennings
- Center for Health Protection (GZB), National Institute for Public Health and the Environment (RIVM), Bilthoven, The Netherlands
| | - Millie A Ferres
- Mother Infant Research Institute, Tufts Medical Center and the Floating Hospital for Children, Boston, Massachusetts
| | - Leah C Graham
- Mother Infant Research Institute, Tufts Medical Center and the Floating Hospital for Children, Boston, Massachusetts
| | - Heather C Wick
- Department of Computer Science, Tufts University, Medford, Massachusetts
| | - Klaus A Miczek
- Department of Psychology, Tufts University, Medford, Massachusetts
| | - Donna K Slonim
- Department of Computer Science, Tufts University, Medford, Massachusetts
| | - Diana W Bianchi
- Mother Infant Research Institute, Tufts Medical Center and the Floating Hospital for Children, Boston, Massachusetts
| |
Collapse
|
49
|
Rannversson H, Wilson P, Kristensen KB, Sinning S, Kristensen AS, Strømgaard K, Andersen J. Importance of the Extracellular Loop 4 in the Human Serotonin Transporter for Inhibitor Binding and Substrate Translocation. J Biol Chem 2015; 290:14582-94. [PMID: 25903124 DOI: 10.1074/jbc.m114.629071] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/01/2014] [Indexed: 12/20/2022] Open
Abstract
The serotonin transporter (SERT) terminates serotonergic neurotransmission by performing reuptake of released serotonin, and SERT is the primary target for antidepressants. SERT mediates the reuptake of serotonin through an alternating access mechanism, implying that a central substrate site is connected to both sides of the membrane by permeation pathways, of which only one is accessible at a time. The coordinated conformational changes in SERT associated with substrate translocation are not fully understood. Here, we have identified a Leu to Glu mutation at position 406 (L406E) in the extracellular loop 4 (EL4) of human SERT, which induced a remarkable gain-of-potency (up to >40-fold) for a range of SERT inhibitors. The effects were highly specific for L406E relative to six other mutations in the same position, including the closely related L406D mutation, showing that the effects induced by L406E are not simply charge-related effects. Leu(406) is located >10 Å from the central inhibitor binding site indicating that the mutation affects inhibitor binding in an indirect manner. We found that L406E decreased accessibility to a residue in the cytoplasmic pathway. The shift in equilibrium to favor a more outward-facing conformation of SERT can explain the reduced turnover rate and increased association rate of inhibitor binding we found for L406E. Together, our findings show that EL4 allosterically can modulate inhibitor binding within the central binding site, and substantiates that EL4 has an important role in controlling the conformational equilibrium of human SERT.
Collapse
Affiliation(s)
- Hafsteinn Rannversson
- From the Department of Drug Design and Pharmacology, University of Copenhagen, DK-2100 Copenhagen and
| | - Pamela Wilson
- From the Department of Drug Design and Pharmacology, University of Copenhagen, DK-2100 Copenhagen and
| | - Kristina Birch Kristensen
- the Translational Neuropsychiatry Unit, Department of Clinical Medicine, Aarhus University, DK-8240 Risskov, Denmark
| | - Steffen Sinning
- the Translational Neuropsychiatry Unit, Department of Clinical Medicine, Aarhus University, DK-8240 Risskov, Denmark
| | - Anders Skov Kristensen
- From the Department of Drug Design and Pharmacology, University of Copenhagen, DK-2100 Copenhagen and
| | - Kristian Strømgaard
- From the Department of Drug Design and Pharmacology, University of Copenhagen, DK-2100 Copenhagen and
| | - Jacob Andersen
- From the Department of Drug Design and Pharmacology, University of Copenhagen, DK-2100 Copenhagen and
| |
Collapse
|
50
|
Hertz L, Rothman DL, Li B, Peng L. Chronic SSRI stimulation of astrocytic 5-HT2B receptors change multiple gene expressions/editings and metabolism of glutamate, glucose and glycogen: a potential paradigm shift. Front Behav Neurosci 2015; 9:25. [PMID: 25750618 PMCID: PMC4335176 DOI: 10.3389/fnbeh.2015.00025] [Citation(s) in RCA: 30] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/28/2014] [Accepted: 01/23/2015] [Indexed: 11/13/2022] Open
Abstract
It is firmly believed that the mechanism of action of SSRIs in major depression is to inhibit the serotonin transporter, SERT, and increase extracellular concentration of serotonin. However, this undisputed observation does not prove that SERT inhibition is the mechanism, let alone the only mechanism, by which SSRI's exert their therapeutic effects. It has recently been demonstrated that 5-HT2B receptor stimulation is needed for the antidepressant effect of fluoxetine in vivo. The ability of all five currently used SSRIs to stimulate the 5-HT2B receptor equipotentially in cultured astrocytes has been known for several years, and increasing evidence has shown the importance of astrocytes and astrocyte-neuronal interactions for neuroplasticity and complex brain activity. This paper reviews acute and chronic effects of 5-HT2B receptor stimulation in cultured astrocytes and in astrocytes freshly isolated from brains of mice treated with fluoxetine for 14 days together with effects of anti-depressant therapy on turnover of glutamate and GABA and metabolism of glucose and glycogen. It is suggested that these events are causally related to the mechanism of action of SSRIs and of interest for development of newer antidepressant drugs.
Collapse
Affiliation(s)
- Leif Hertz
- Laboratory of Brain Metabolic Diseases, Institute of Metabolic Disease Research and Drug Development, China Medical University Shenyang, China
| | - Douglas L Rothman
- Magnetic Resonance Research Center, Diagnostic Radiology and Biomedical Engineering, Yale University New Haven, CT, USA
| | - Baoman Li
- Laboratory of Brain Metabolic Diseases, Institute of Metabolic Disease Research and Drug Development, China Medical University Shenyang, China
| | - Liang Peng
- Laboratory of Brain Metabolic Diseases, Institute of Metabolic Disease Research and Drug Development, China Medical University Shenyang, China
| |
Collapse
|