1
|
Zhang H, Yin YL, Dai A, Zhang T, Zhang C, Wu C, Hu W, He X, Pan B, Jin S, Yuan Q, Wang MW, Yang D, Xu HE, Jiang Y. Dimerization and antidepressant recognition at noradrenaline transporter. Nature 2024; 630:247-254. [PMID: 38750358 DOI: 10.1038/s41586-024-07437-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/25/2023] [Accepted: 04/17/2024] [Indexed: 06/07/2024]
Abstract
The noradrenaline transporter has a pivotal role in regulating neurotransmitter balance and is crucial for normal physiology and neurobiology1. Dysfunction of noradrenaline transporter has been implicated in numerous neuropsychiatric diseases, including depression and attention deficit hyperactivity disorder2. Here we report cryo-electron microscopy structures of noradrenaline transporter in apo and substrate-bound forms, and as complexes with six antidepressants. The structures reveal a noradrenaline transporter dimer interface that is mediated predominantly by cholesterol and lipid molecules. The substrate noradrenaline binds deep in the central binding pocket, and its amine group interacts with a conserved aspartate residue. Our structures also provide insight into antidepressant recognition and monoamine transporter selectivity. Together, these findings advance our understanding of noradrenaline transporter regulation and inhibition, and provide templates for designing improved antidepressants to treat neuropsychiatric disorders.
Collapse
Affiliation(s)
- Heng Zhang
- State Key Laboratory of Drug Research, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai, China
| | | | - Antao Dai
- State Key Laboratory of Chemical Biology, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai, China
- National Center for Drug Screening, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai, China
| | - Tianwei Zhang
- Lingang Laboratory, Shanghai, China
- School of Life Science and Technology, ShanghaiTech University, Shanghai, China
| | - Chao Zhang
- National Center for Drug Screening, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai, China
- School of Pharmacy, Fudan University, Shanghai, China
| | - Canrong Wu
- State Key Laboratory of Drug Research, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai, China
| | - Wen Hu
- State Key Laboratory of Drug Research, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai, China
- Shanghai Advanced Electron Microscope Center, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai, China
| | - Xinheng He
- State Key Laboratory of Drug Research, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai, China
- University of Chinese Academy of Sciences, Beijing, China
| | | | | | - Qingning Yuan
- State Key Laboratory of Drug Research, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai, China
- Shanghai Advanced Electron Microscope Center, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai, China
| | - Ming-Wei Wang
- Research Center for Deepsea Bioresources, Sanya, China
- Department of Pharmacology, School of Basic Medical Sciences, Fudan University, Shanghai, China
- School of Pharmacy, Hainan Medical University, Haikou, China
| | - Dehua Yang
- State Key Laboratory of Chemical Biology, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai, China.
- National Center for Drug Screening, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai, China.
- University of Chinese Academy of Sciences, Beijing, China.
| | - H Eric Xu
- State Key Laboratory of Drug Research, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai, China.
- School of Life Science and Technology, ShanghaiTech University, Shanghai, China.
- Shanghai Advanced Electron Microscope Center, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai, China.
| | - Yi Jiang
- Lingang Laboratory, Shanghai, China.
- School of Life Science and Technology, ShanghaiTech University, Shanghai, China.
| |
Collapse
|
2
|
Vaughan RA, Henry LK, Foster JD, Brown CR. Post-translational mechanisms in psychostimulant-induced neurotransmitter efflux. ADVANCES IN PHARMACOLOGY (SAN DIEGO, CALIF.) 2024; 99:1-33. [PMID: 38467478 DOI: 10.1016/bs.apha.2023.10.003] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 03/13/2024]
Abstract
The availability of monoamine neurotransmitters in the brain is under the control of dopamine, norepinephrine, and serotonin transporters expressed on the plasma membrane of monoaminergic neurons. By regulating transmitter levels these proteins mediate crucial functions including cognition, attention, and reward, and dysregulation of their activity is linked to mood and psychiatric disorders of these systems. Amphetamine-based transporter substrates stimulate non-exocytotic transmitter efflux that induces psychomotor stimulation, addiction, altered mood, hallucinations, and psychosis, thus constituting a major component of drug neurochemical and behavioral outcomes. Efflux is under the control of transporter post-translational modifications that synergize with other regulatory events, and this review will summarize our knowledge of these processes and their role in drug mechanisms.
Collapse
Affiliation(s)
- Roxanne A Vaughan
- Department of Biomedical Sciences, University of North Dakota School of Medicine and Health Sciences, Grand Forks, ND, United States.
| | - L Keith Henry
- Department of Biomedical Sciences, University of North Dakota School of Medicine and Health Sciences, Grand Forks, ND, United States
| | - James D Foster
- Department of Biomedical Sciences, University of North Dakota School of Medicine and Health Sciences, Grand Forks, ND, United States
| | - Christopher R Brown
- Department of Biomedical Sciences, University of North Dakota School of Medicine and Health Sciences, Grand Forks, ND, United States
| |
Collapse
|
3
|
Mayer FP, Niello M, Bulling S, Zhang YW, Li Y, Kudlacek O, Holy M, Kooti F, Sandtner W, Rudnick G, Schmid D, Sitte HH. Mephedrone induces partial release at human dopamine transporters but full release at human serotonin transporters. Neuropharmacology 2023; 240:109704. [PMID: 37703919 DOI: 10.1016/j.neuropharm.2023.109704] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/19/2022] [Revised: 07/07/2023] [Accepted: 08/31/2023] [Indexed: 09/15/2023]
Abstract
Mephedrone (4-methylmethcathinone) is a cathinone derivative that is recreationally consumed for its energizing and empathogenic effects. The stimulating properties are believed to arise from the ability of mephedrone to interact with the high-affinity transporters for dopamine (DA) (DAT) and norepinephrine (NET), whereas the entactogenic effect presumably relies on its activity at the serotonin (5-HT) transporter (SERT). Early studies found that mephedrone acts as a releaser at NET, DAT and SERT, and thus promotes efflux of the respective monoamines. Evidence linked drug-induced reverse transport of 5-HT via SERT to prosocial effects, whereas activity at DAT is strongly correlated with abuse liability. Consequently, we sought to evaluate the pharmacology of mephedrone at human (h) DAT and SERT, heterologously expressed in human embryonic kidney 293 cells, in further detail. In line with previous studies, we report that mephedrone evokes carrier-mediated release via hDAT and hSERT. We found this effect to be sensitive to the protein kinase C inhibitor GF109203X. Electrophysiological recordings revealed that mephedrone is actively transported by hDAT and hSERT. However, mephedrone acts as a full substrate of hSERT but as a partial substrate of hDAT. Furthermore, when compared to fully efficacious releasing agents at hDAT and hSERT (i.e. S(+)-amphetamine and para-chloroamphetamine, respectively) mephedrone displays greater efficacy as a releaser at hSERT than at hDAT. In summary, this study provides additional insights into the molecular mechanism of action of mephedrone at hDAT and hSERT.
Collapse
Affiliation(s)
- Felix P Mayer
- Institute of Pharmacology, Center for Physiology and Pharmacology, Medical University of Vienna, Waehringer Strasse 13a, A-1090, Vienna, Austria
| | - Marco Niello
- Institute of Pharmacology, Center for Physiology and Pharmacology, Medical University of Vienna, Waehringer Strasse 13a, A-1090, Vienna, Austria
| | - Simon Bulling
- Institute of Pharmacology, Center for Physiology and Pharmacology, Medical University of Vienna, Waehringer Strasse 13a, A-1090, Vienna, Austria
| | - Yuan-Wei Zhang
- Department of Pharmacology, Yale University School of Medicine, 333 Cedar Street, New Haven, CT, 06520-8066, USA
| | - Yang Li
- Institute of Pharmacology, Center for Physiology and Pharmacology, Medical University of Vienna, Waehringer Strasse 13a, A-1090, Vienna, Austria
| | - Oliver Kudlacek
- Institute of Pharmacology, Center for Physiology and Pharmacology, Medical University of Vienna, Waehringer Strasse 13a, A-1090, Vienna, Austria
| | - Marion Holy
- Institute of Pharmacology, Center for Physiology and Pharmacology, Medical University of Vienna, Waehringer Strasse 13a, A-1090, Vienna, Austria
| | - Fatemeh Kooti
- Institute of Pharmacology, Center for Physiology and Pharmacology, Medical University of Vienna, Waehringer Strasse 13a, A-1090, Vienna, Austria
| | - Walter Sandtner
- Institute of Pharmacology, Center for Physiology and Pharmacology, Medical University of Vienna, Waehringer Strasse 13a, A-1090, Vienna, Austria
| | - Gary Rudnick
- Department of Pharmacology, Yale University School of Medicine, 333 Cedar Street, New Haven, CT, 06520-8066, USA
| | - Diethart Schmid
- Institute for Physiology, Center for Physiology and Pharmacology, Medical University of Vienna, Waehringer Strasse 13a, A-1090, Vienna, Austria
| | - Harald H Sitte
- Institute of Pharmacology, Center for Physiology and Pharmacology, Medical University of Vienna, Waehringer Strasse 13a, A-1090, Vienna, Austria; Hourani Center for Applied Scientific Research, Al-Ahliyya Amman University, Amman, Jordan; Center for Addiction Research and Science - AddRess, Medical University of Vienna, Waehringer Strasse 13a, A-1090, Vienna, Austria.
| |
Collapse
|
4
|
Luethi D, Maier J, Rudin D, Szöllősi D, Angenoorth TJF, Stankovic S, Schittmayer M, Burger I, Yang JW, Jaentsch K, Holy M, Das AK, Brameshuber M, Camacho-Hernandez GA, Casiraghi A, Newman AH, Kudlacek O, Birner-Gruenberger R, Stockner T, Schütz GJ, Sitte HH. Phosphatidylinositol 4,5-bisphosphate (PIP 2) facilitates norepinephrine transporter dimerization and modulates substrate efflux. Commun Biol 2022; 5:1259. [PMID: 36396757 PMCID: PMC9672106 DOI: 10.1038/s42003-022-04210-1] [Citation(s) in RCA: 12] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/03/2022] [Accepted: 11/01/2022] [Indexed: 11/19/2022] Open
Abstract
The plasmalemmal norepinephrine transporter (NET) regulates cardiovascular sympathetic activity by clearing extracellular norepinephrine in the synaptic cleft. Here, we investigate the subunit stoichiometry and function of NET using single-molecule fluorescence microscopy and flux assays. In particular, we show the effect of phosphatidylinositol 4,5-bisphosphate (PIP2) on NET oligomerization and efflux. NET forms monomers (~60%) and dimers (~40%) at the plasma membrane. PIP2 depletion results in a decrease in the average oligomeric state and decreases NET-mediated substrate efflux while not affecting substrate uptake. Mutation of the putative PIP2 binding residues R121, K334, and R440 to alanines does not affect NET dimerization but results in decreased substrate efflux that is not altered upon PIP2 depletion; this indicates that PIP2 interactions with these residues affect NET-mediated efflux. A dysregulation of norepinephrine and PIP2 signaling have both been implicated in neuropsychiatric and cardiovascular diseases. This study provides evidence that PIP2 directly regulates NET organization and function.
Collapse
Affiliation(s)
- Dino Luethi
- Institute of Pharmacology, Center for Physiology and Pharmacology, Medical University of Vienna, Waehringer Strasse 13A, 1090, Vienna, Austria
- Institute of Applied Physics, TU Wien, Lehargasse 6, 1060, Vienna, Austria
| | - Julian Maier
- Institute of Pharmacology, Center for Physiology and Pharmacology, Medical University of Vienna, Waehringer Strasse 13A, 1090, Vienna, Austria
| | - Deborah Rudin
- Institute of Pharmacology, Center for Physiology and Pharmacology, Medical University of Vienna, Waehringer Strasse 13A, 1090, Vienna, Austria
| | - Dániel Szöllősi
- Institute of Pharmacology, Center for Physiology and Pharmacology, Medical University of Vienna, Waehringer Strasse 13A, 1090, Vienna, Austria
| | - Thomas J F Angenoorth
- Institute of Pharmacology, Center for Physiology and Pharmacology, Medical University of Vienna, Waehringer Strasse 13A, 1090, Vienna, Austria
| | - Stevan Stankovic
- Institute of Pharmacology, Center for Physiology and Pharmacology, Medical University of Vienna, Waehringer Strasse 13A, 1090, Vienna, Austria
| | - Matthias Schittmayer
- Institute of Chemical Technologies and Analytics, TU Wien, Getreidemarkt 9, 1060, Vienna, Austria
| | - Isabella Burger
- Institute of Chemical Technologies and Analytics, TU Wien, Getreidemarkt 9, 1060, Vienna, Austria
| | - Jae-Won Yang
- Institute of Pharmacology, Center for Physiology and Pharmacology, Medical University of Vienna, Waehringer Strasse 13A, 1090, Vienna, Austria
| | - Kathrin Jaentsch
- Institute of Pharmacology, Center for Physiology and Pharmacology, Medical University of Vienna, Waehringer Strasse 13A, 1090, Vienna, Austria
| | - Marion Holy
- Institute of Pharmacology, Center for Physiology and Pharmacology, Medical University of Vienna, Waehringer Strasse 13A, 1090, Vienna, Austria
| | - Anand Kant Das
- Institute of Applied Physics, TU Wien, Lehargasse 6, 1060, Vienna, Austria
- Physics Program, New York University Abu Dhabi, Saadiyat Island, 129188, Abu Dhabi, United Arab Emirates
| | - Mario Brameshuber
- Institute of Applied Physics, TU Wien, Lehargasse 6, 1060, Vienna, Austria
| | - Gisela Andrea Camacho-Hernandez
- Medicinal Chemistry Section, Molecular Targets and Medications Discovery Branch, National Institute on Drug Abuse - Intramural Research Program, Baltimore, MD, 21224, USA
| | - Andrea Casiraghi
- Medicinal Chemistry Section, Molecular Targets and Medications Discovery Branch, National Institute on Drug Abuse - Intramural Research Program, Baltimore, MD, 21224, USA
- Department of Pharmaceutical Sciences, University of Milan, Via Luigi Mangiagalli 25, 20133, Milan, Italy
| | - Amy Hauck Newman
- Medicinal Chemistry Section, Molecular Targets and Medications Discovery Branch, National Institute on Drug Abuse - Intramural Research Program, Baltimore, MD, 21224, USA
| | - Oliver Kudlacek
- Institute of Pharmacology, Center for Physiology and Pharmacology, Medical University of Vienna, Waehringer Strasse 13A, 1090, Vienna, Austria
| | - Ruth Birner-Gruenberger
- Institute of Chemical Technologies and Analytics, TU Wien, Getreidemarkt 9, 1060, Vienna, Austria
- Diagnostic and Research Institute of Pathology, Medical University of Graz, Neue Stiftingtalstrasse 6, 8010, Graz, Austria
| | - Thomas Stockner
- Institute of Pharmacology, Center for Physiology and Pharmacology, Medical University of Vienna, Waehringer Strasse 13A, 1090, Vienna, Austria
| | - Gerhard J Schütz
- Institute of Applied Physics, TU Wien, Lehargasse 6, 1060, Vienna, Austria.
| | - Harald H Sitte
- Institute of Pharmacology, Center for Physiology and Pharmacology, Medical University of Vienna, Waehringer Strasse 13A, 1090, Vienna, Austria.
| |
Collapse
|
5
|
Maier J, Rauter L, Rudin D, Niello M, Holy M, Schmid D, Wilson J, Blough BE, Gannon BM, Murnane KS, Sitte HH. α-PPP and its derivatives are selective partial releasers at the human norepinephrine transporter: A pharmacological characterization of interactions between pyrrolidinopropiophenones and high and low affinity monoamine transporters. Neuropharmacology 2021; 190:108570. [PMID: 33864800 DOI: 10.1016/j.neuropharm.2021.108570] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/18/2021] [Revised: 03/31/2021] [Accepted: 04/12/2021] [Indexed: 12/31/2022]
Abstract
While classical cathinones, such as methcathinone, have been shown to be monoamine releasing agents at human monoamine transporters, the subgroup of α-pyrrolidinophenones has thus far solely been characterized as monoamine transporter reuptake inhibitors. Herein, we report data from previously undescribed α-pyrrolidinopropiophenone (α-PPP) derivatives and compare them with the pharmacologically well-researched α-PVP (α-pyrrolidinovalerophenone). Radiotracer-based in vitro uptake inhibition assays in HEK293 cells show that the investigated α-PPP derivatives inhibit the human high-affinity transporters of dopamine (hDAT) and norepinephrine (hNET) in the low micromolar range, with α-PVP being ten times more potent. Similar to α-PVP, no relevant pharmacological activity was found at the human serotonin transporter (hSERT). Unexpectedly, radiotracer-based in vitro release assays reveal α-PPP, MDPPP and 3Br-PPP, but not α-PVP, to be partial releasing agents at hNET (EC50 values in the low micromolar range). Furthermore, uptake inhibition assays at low-affinity monoamine transporters, i.e., the human organic cation transporters (hOCT) 1-3 and human plasma membrane monoamine transporter (hPMAT), bring to light that all compounds inhibit hOCT1 and 2 (IC50 values in the low micromolar range) while less potently interacting with hPMAT and hOCT3. In conclusion, this study describes (i) three new hybrid compounds that efficaciously block hDAT while being partial releasers at hNET, and (ii) highlights the interactions of α-PPP-derivatives with low-affinity monoamine transporters, giving impetus to further studies investigating the interaction of drugs of abuse with OCT1-3 and PMAT.
Collapse
Affiliation(s)
- Julian Maier
- Medical University of Vienna, Center for Physiology and Pharmacology, Institute of Pharmacology, Währingerstraße 13A, 1090, Vienna, Austria
| | - Laurin Rauter
- Medical University of Vienna, Center for Physiology and Pharmacology, Institute of Pharmacology, Währingerstraße 13A, 1090, Vienna, Austria
| | - Deborah Rudin
- Medical University of Vienna, Center for Physiology and Pharmacology, Institute of Pharmacology, Währingerstraße 13A, 1090, Vienna, Austria
| | - Marco Niello
- Medical University of Vienna, Center for Physiology and Pharmacology, Institute of Pharmacology, Währingerstraße 13A, 1090, Vienna, Austria
| | - Marion Holy
- Medical University of Vienna, Center for Physiology and Pharmacology, Institute of Pharmacology, Währingerstraße 13A, 1090, Vienna, Austria
| | - Diethart Schmid
- Medical University of Vienna, Center for Physiology and Pharmacology, Institute of Physiology, Währingerstraße 13A, 1090, Vienna, Austria
| | - Joseph Wilson
- Research Triangle Institute, Center for Drug Discovery, Research Triangle Park, NC, USA
| | - Bruce E Blough
- Research Triangle Institute, Center for Drug Discovery, Research Triangle Park, NC, USA
| | - Brenda M Gannon
- Mercer University College of Pharmacy, Mercer University Health Sciences Center, Department of Pharmaceutical Sciences, Atlanta, GA, USA; Louisiana State University Health Sciences Center, Shreveport, Department of Pharmacology Toxicology & Neuroscience and Louisiana Addiction Research Center, Shreveport, LA, USA
| | - Kevin S Murnane
- Mercer University College of Pharmacy, Mercer University Health Sciences Center, Department of Pharmaceutical Sciences, Atlanta, GA, USA; Louisiana State University Health Sciences Center, Shreveport, Department of Pharmacology Toxicology & Neuroscience and Louisiana Addiction Research Center, Shreveport, LA, USA
| | - Harald H Sitte
- Medical University of Vienna, Center for Physiology and Pharmacology, Institute of Pharmacology, Währingerstraße 13A, 1090, Vienna, Austria; AddRess Centre for Addiction Research and Science, Medical University of Vienna, Währingerstraße 13A, 1090, Vienna, Austria.
| |
Collapse
|
6
|
Gether U, Sitte HH. It takes two-or more-to tango: Revisiting the role of dopamine transporter oligomerization. J Biol Chem 2021; 296:100629. [PMID: 34237901 PMCID: PMC8100069 DOI: 10.1016/j.jbc.2021.100629] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/30/2022] Open
Abstract
The dopamine transporter utilizes the transmembrane sodium gradient to mediate reuptake of dopamine from the extracellular space. The dopamine transporter can form dimers and possibly also higher order structures in the plasma membrane, and this oligomerization has been implicated in both trafficking and transport. However, we still do not fully understand its biological importance. A study by Sorkina et al. now describes a series of small molecules that link transporter conformation to oligomerization and endocytosis, providing an interesting step forward in an intricate dance.
Collapse
Affiliation(s)
- Ulrik Gether
- Department of Neuroscience, Faculty of Health and Medical Sciences, Maersk Tower 7.5, University of Copenhagen, Copenhagen, Denmark.
| | - Harald H Sitte
- Center for Physiology and Pharmacology, Institute of Pharmacology, Medical University Vienna, Vienna, Austria
| |
Collapse
|
7
|
Transporter tandems: precise tools for normalizing active transporter in the plasma membrane. Biochem J 2021; 477:4191-4206. [PMID: 33073844 DOI: 10.1042/bcj20200666] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/20/2020] [Revised: 10/12/2020] [Accepted: 10/19/2020] [Indexed: 12/12/2022]
Abstract
The transport efficiency (TE) describes the performance of a transport protein for a specific substrate. To compare the TE of different transporters, the number of active transporters in the plasma membrane must be monitored, as it may vary for each transporter and experiment. Available methods, like LC-MS quantification of tryptic peptides, fail to discriminate inactive intracellular transporters or, like cell-surface biotinylation followed by affinity chromatography and Western blotting, are imprecise and very laborious. We wanted to normalize active transporters by the activity of a second transporter. A transporter tandem, generated by joining two transporter cDNAs into a single open reading frame, should guarantee a 1 : 1 stoichiometry. Here we created a series of tandems with different linkers between the human ergothioneine (ET) transporter ETT (gene symbol SLC22A4) and organic cation transporter OCT2 (SLC22A2). The linker sequence strongly affected the expression strength. The stoichiometry was validated by absolute peptide quantification and untargeted peptide analysis. Compared with wild-type ETT, the normalized ET clearance of the natural variant L503F was higher (f = 1.34); G462E was completely inactive. The general usefulness of the tandem strategy was demonstrated by linking several transporters with ETT; every construct was active in both parts. Transporter tandems can be used - without membrane isolation or protein quantification - as precise tools for transporter number normalization, to identify, for example, relevant transporters for a drug. It is necessary, however, to find suitable linkers, to check the order of transporters, and to verify the absence of functional interference by saturation kinetics.
Collapse
|
8
|
Jayaraman K, Das AK, Luethi D, Szöllősi D, Schütz GJ, Reith MEA, Sitte HH, Stockner T. SLC6 transporter oligomerization. J Neurochem 2020; 157:919-929. [PMID: 32767560 PMCID: PMC8247324 DOI: 10.1111/jnc.15145] [Citation(s) in RCA: 29] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/08/2020] [Revised: 07/31/2020] [Accepted: 07/31/2020] [Indexed: 12/18/2022]
Abstract
Transporters of the solute carrier 6 (SLC6) family mediate the reuptake of neurotransmitters such as dopamine, norepinephrine, serotonin, GABA, and glycine. SLC6 family members are 12 transmembrane helix‐spanning proteins that operate using the transmembrane sodium gradient for transport. These transporters assume various quaternary arrangements ranging from monomers to complex stoichiometries with multiple subunits. Dopamine and serotonin transporter oligomerization has been implicated in trafficking of newly formed proteins from the endoplasmic reticulum to the plasma membrane with a pre‐fixed assembly. Once at the plasma membrane, oligomers are kept fixed in their quaternary assembly by interaction with phosphoinositides. While it remains unclear how oligomer formation precisely affects physiological transporter function, it has been shown that oligomerization supports the activity of release‐type psychostimulants. Most recently, single molecule microscopy experiments unveiled that the stoichiometry differs between individual members of the SLC6 family. The present overview summarizes our understanding of the influence of plasma membrane constituents on transporter oligomerization, describes the known interfaces between protomers and discusses open questions. ![]()
Collapse
Affiliation(s)
- Kumaresan Jayaraman
- Institute of Pharmacology, Center for Physiology and Pharmacology, Medical University of Vienna, Vienna, Austria
| | - Anand K Das
- Institute of Applied Physics, Vienna University of Technology, Vienna, Austria
| | - Dino Luethi
- Institute of Pharmacology, Center for Physiology and Pharmacology, Medical University of Vienna, Vienna, Austria.,Institute of Applied Physics, Vienna University of Technology, Vienna, Austria
| | - Dániel Szöllősi
- Institute of Pharmacology, Center for Physiology and Pharmacology, Medical University of Vienna, Vienna, Austria
| | - Gerhard J Schütz
- Institute of Applied Physics, Vienna University of Technology, Vienna, Austria
| | - Maarten E A Reith
- Department of Psychiatry, New York University School of Medicine, New York City, NY, USA
| | - Harald H Sitte
- Institute of Pharmacology, Center for Physiology and Pharmacology, Medical University of Vienna, Vienna, Austria
| | - Thomas Stockner
- Institute of Pharmacology, Center for Physiology and Pharmacology, Medical University of Vienna, Vienna, Austria
| |
Collapse
|
9
|
Khan JA, Sohail A, Jayaraman K, Szöllősi D, Sandtner W, Sitte HH, Stockner T. The Amino Terminus of LeuT Changes Conformation in an Environment Sensitive Manner. Neurochem Res 2020; 45:1387-1398. [PMID: 31858375 PMCID: PMC7260283 DOI: 10.1007/s11064-019-02928-9] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/07/2019] [Revised: 12/04/2019] [Accepted: 12/05/2019] [Indexed: 12/21/2022]
Abstract
Neurotransmitter:sodium symporters are highly expressed in the human brain and catalyze the uptake of substrate through the plasma membrane by using the electrochemical gradient of sodium as the energy source. The bacterial homolog LeuT, a small amino acid transporter isolated from the bacteria Aquifex aeolicus, is the founding member of the family and has been crystallized in three conformations. The N-terminus is structurally well defined and strongly interacts with the transporter core in the outward-facing conformations. However, it could not be resolved in the inward-facing conformation, which indicates enhanced mobility. Here we investigate conformations and dynamics of the N-terminus, by combining molecular dynamics simulations with experimental verification using distance measurements and accessibility studies. We found strongly increased dynamics of the N-terminus, but also that helix TM1A is subject to enhanced mobility. TM1A moves towards the transporter core in the membrane environment, reaching a conformation that is closer to the structure of LeuT with wild type sequence, indicating that the mutation introduced to create the inward-facing structure might have altered the position of helix TM1A. The mobile N-terminus avoids entering the open vestibule of the inward-facing state, as accessibility studies do not show any reduction of quenching by iodide of a fluorophore attached to the N-terminus.
Collapse
Affiliation(s)
- Jawad A Khan
- Institute of Pharmacology, Center for Physiology and Pharmacology, Medical University of Vienna, Waehringerstr. 13a, 1090, Vienna, Austria
| | - Azmat Sohail
- Institute of Pharmacology, Center for Physiology and Pharmacology, Medical University of Vienna, Waehringerstr. 13a, 1090, Vienna, Austria
| | - Kumaresan Jayaraman
- Institute of Pharmacology, Center for Physiology and Pharmacology, Medical University of Vienna, Waehringerstr. 13a, 1090, Vienna, Austria
- Institute for Pharmaceutical and Medicinal Chemistry, Heinrich Heine University Düsseldorf, Düsseldorf, Germany
| | - Dániel Szöllősi
- Institute of Pharmacology, Center for Physiology and Pharmacology, Medical University of Vienna, Waehringerstr. 13a, 1090, Vienna, Austria
| | - Walter Sandtner
- Institute of Pharmacology, Center for Physiology and Pharmacology, Medical University of Vienna, Waehringerstr. 13a, 1090, Vienna, Austria
| | - Harald H Sitte
- Institute of Pharmacology, Center for Physiology and Pharmacology, Medical University of Vienna, Waehringerstr. 13a, 1090, Vienna, Austria
| | - Thomas Stockner
- Institute of Pharmacology, Center for Physiology and Pharmacology, Medical University of Vienna, Waehringerstr. 13a, 1090, Vienna, Austria.
| |
Collapse
|
10
|
Diallinas G, Martzoukou O. Transporter membrane traffic and function: lessons from a mould. FEBS J 2019; 286:4861-4875. [DOI: 10.1111/febs.15078] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/02/2019] [Revised: 08/26/2019] [Accepted: 09/30/2019] [Indexed: 12/16/2022]
Affiliation(s)
- George Diallinas
- Department of Biology National and Kapodistrian University of Athens Greece
| | - Olga Martzoukou
- Department of Biology National and Kapodistrian University of Athens Greece
| |
Collapse
|
11
|
Pyle E, Guo C, Hofmann T, Schmidt C, Ribiero O, Politis A, Byrne B. Protein–Lipid Interactions Stabilize the Oligomeric State of BOR1p from Saccharomyces cerevisiae. Anal Chem 2019; 91:13071-13079. [DOI: 10.1021/acs.analchem.9b03271] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/06/2023]
Affiliation(s)
- Euan Pyle
- Department of Life Sciences, Imperial College London, Exhibition Road, London SW7 2AZ, U.K
- Department of Chemistry, Kings College London, 7 Trinity Street, London SE1 1DB, U.K
| | - Chengzhi Guo
- Department of Life Sciences, Imperial College London, Exhibition Road, London SW7 2AZ, U.K
| | - Tommy Hofmann
- Interdisciplinary Research Center HALOmem, Charles Tanford Protein Centre, Martin Luther University Halle-Wittenberg, Kurt-Mothes-Str 3a, D-06120 Halle, Germany
| | - Carla Schmidt
- Interdisciplinary Research Center HALOmem, Charles Tanford Protein Centre, Martin Luther University Halle-Wittenberg, Kurt-Mothes-Str 3a, D-06120 Halle, Germany
| | - Orquidea Ribiero
- Department of Life Sciences, Imperial College London, Exhibition Road, London SW7 2AZ, U.K
| | - Argyris Politis
- Department of Chemistry, Kings College London, 7 Trinity Street, London SE1 1DB, U.K
| | - Bernadette Byrne
- Department of Life Sciences, Imperial College London, Exhibition Road, London SW7 2AZ, U.K
| |
Collapse
|
12
|
Abstract
Cell nutrition, detoxification, signalling, homeostasis and response to drugs, processes related to cell growth, differentiation and survival are all mediated by plasma membrane (PM) proteins called transporters. Despite their distinct fine structures, mechanism of function, energetic requirements, kinetics and substrate specificities, all transporters are characterized by a main hydrophobic body embedded in the PM as a series of tightly packed, often intertwined, α-helices that traverse the lipid bilayer in a zigzag mode, connected with intracellular or extracellular loops and hydrophilic N- and C-termini. Whereas longstanding genetic, biochemical and biophysical evidence suggests that specific transmembrane segments, and also their connecting loops, are responsible for substrate recognition and transport dynamics, emerging evidence also reveals the functional importance of transporter N- and C-termini, in respect to transport catalysis, substrate specificity, subcellular expression, stability and signalling. This review highlights selected prototypic examples of transporters in which their termini play important roles in their functioning.
Collapse
Affiliation(s)
- Emmanuel Mikros
- Faculty of Pharmacy, National and Kapodistrian University of Athens, Panepistimioupolis, 15771 Athens, Greece
| | - George Diallinas
- Department of Biology, National and Kapodistrian University of Athens, Panepistimioupolis, 15781 Athens, Greece
| |
Collapse
|
13
|
Cheng MH, Ponzoni L, Sorkina T, Lee JY, Zhang S, Sorkin A, Bahar I. Trimerization of dopamine transporter triggered by AIM-100 binding: Molecular mechanism and effect of mutations. Neuropharmacology 2019; 161:107676. [PMID: 31228486 DOI: 10.1016/j.neuropharm.2019.107676] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/26/2018] [Revised: 06/17/2019] [Accepted: 06/18/2019] [Indexed: 10/26/2022]
Abstract
Recent work demonstrated the propensity of dopamine transporters (DATs) to form trimers or higher oligomers, enhanced upon binding a furopyrimidine, AIM-100. AIM-100 binding promotes DAT endocytosis and thereby moderates dopaminergic transmission. Despite the neurobiological significance of these events, the molecular mechanisms that underlie the stabilization of DAT trimer and the key interactions that modulate the trimerization of DAT, and not serotonin transporter SERT, remain unclear. In the present study, we determined three structural models, termed trimer-W238, -C306 and -Y303, for possible trimerization of DATs . To this aim, we used structural data resolved for DAT and its structural homologs that share the LeuT fold, advanced computational modeling and simulations, site-directed mutagenesis experiments and live-cell imaging assays. The models are in accord with the versatility of LeuT fold to stabilize dimeric or higher order constructs. Selected residues show a high propensity to occupy interfacial regions. Among them, D231-W238 in the extracellular loop EL2, including the intersubunit salt-bridge forming pair D231/D232-R237 (not present in SERT) (in trimer-W238), the loop EL3 (trimers-C306 and -Y303), and W497 on the intracellularly exposed IL5 loop (trimer-C306) and its spatial neighbors (e.g. K525) near the C-terminus are computationally predicted and experimentally confirmed to play important roles in enabling the correct folding and/or oligomerization of DATs in the presence of AIM-100. The study suggests the possibility of controlling the effective transport of dopamine by altering the oligomerization state of DAT upon small molecule binding, as a possible intervention strategy to modulate dopaminergic signaling. This article is part of the issue entitled 'Special Issue on Neurotransmitter Transporters'.
Collapse
Affiliation(s)
- Mary Hongying Cheng
- Department of Computational and Systems Biology, School of Medicine, University of Pittsburgh, Pittsburgh, PA, USA
| | - Luca Ponzoni
- Department of Computational and Systems Biology, School of Medicine, University of Pittsburgh, Pittsburgh, PA, USA
| | - Tatiana Sorkina
- Department of Cell Biology, School of Medicine, University of Pittsburgh, Pittsburgh, PA, USA
| | - Ji Young Lee
- Department of Computational and Systems Biology, School of Medicine, University of Pittsburgh, Pittsburgh, PA, USA
| | - She Zhang
- Department of Computational and Systems Biology, School of Medicine, University of Pittsburgh, Pittsburgh, PA, USA
| | - Alexander Sorkin
- Department of Cell Biology, School of Medicine, University of Pittsburgh, Pittsburgh, PA, USA
| | - Ivet Bahar
- Department of Computational and Systems Biology, School of Medicine, University of Pittsburgh, Pittsburgh, PA, USA.
| |
Collapse
|
14
|
Bolland DE, Moritz AE, Stanislowski DJ, Vaughan RA, Foster JD. Palmitoylation by Multiple DHHC Enzymes Enhances Dopamine Transporter Function and Stability. ACS Chem Neurosci 2019; 10:2707-2717. [PMID: 30965003 PMCID: PMC6746250 DOI: 10.1021/acschemneuro.8b00558] [Citation(s) in RCA: 21] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023] Open
Abstract
The dopamine transporter (DAT) is a plasma membrane protein that mediates the reuptake of extracellular dopamine (DA) and controls the spatiotemporal dynamics of dopaminergic neurotransmission. The transporter is subject to fine control that tailors clearance of transmitter to physiological demands, and dysregulation of reuptake induced by psychostimulant drugs, transporter polymorphisms, and signaling defects may impact transmitter tone in disease states. We previously demonstrated that DAT undergoes complex regulation by palmitoylation, with acute inhibition of the modification leading to rapid reduction of transport activity and sustained inhibition of the modification leading to transporter degradation and reduced expression. Here, to examine mechanisms and outcomes related to increased modification, we coexpressed DAT with palmitoyl acyltransferases (PATs), also known as DHHC enzymes, which catalyze palmitate addition to proteins. Of 12 PATs tested, DAT palmitoylation was stimulated by DHHC2, DHHC3, DHHC8, DHHC15, and DHHC17, with others having no effect. Increased modification was localized to previously identified palmitoylation site Cys580 and resulted in upregulation of transport kinetics and elevated transporter expression mediated by reduced degradation. These findings confirm palmitoylation as a regulator of multiple DAT properties crucial for appropriate DA homeostasis and identify several potential PAT pathways linked to these effects. Defects in palmitoylation processes thus represent possible mechanisms of transport imbalances in DA disorders.
Collapse
Affiliation(s)
| | | | - Daniel J. Stanislowski
- Department of Biomedical Sciences, University of North Dakota, School of Medicine and Health Sciences, Grand Forks, ND 58202
| | - Roxanne A. Vaughan
- Department of Biomedical Sciences, University of North Dakota, School of Medicine and Health Sciences, Grand Forks, ND 58202
| | - James D. Foster
- Department of Biomedical Sciences, University of North Dakota, School of Medicine and Health Sciences, Grand Forks, ND 58202
| |
Collapse
|
15
|
Stereochemistry of phase-1 metabolites of mephedrone determines their effectiveness as releasers at the serotonin transporter. Neuropharmacology 2019; 148:199-209. [DOI: 10.1016/j.neuropharm.2018.12.032] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/29/2018] [Revised: 12/17/2018] [Accepted: 12/31/2018] [Indexed: 12/20/2022]
|
16
|
Hasenhuetl PS, Bhat S, Freissmuth M, Sandtner W. Functional Selectivity and Partial Efficacy at the Monoamine Transporters: A Unified Model of Allosteric Modulation and Amphetamine-Induced Substrate Release. Mol Pharmacol 2019; 95:303-312. [PMID: 30567955 DOI: 10.1124/mol.118.114793] [Citation(s) in RCA: 23] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/02/2018] [Accepted: 12/13/2018] [Indexed: 12/13/2022] Open
Abstract
All clinically approved drugs targeting the plasmalemmal transporters for dopamine, norepinephrine, and serotonin act either as competitive uptake inhibitors or as amphetamine-like releasers. Monoamine transporter (MAT) ligands that allosterically affect MAT-mediated substrate uptake, release, or both were recently discovered. Their modes of action have not yet been explained in a unified framework. Here, we go beyond competitive inhibitors and classic amphetamines and introduce concepts for partial efficacy at and allosteric modulation of MATs. After we elaborate on a kinetic account for amphetamine action, we provide an explanation for partial release (i.e., the observation that some amphetamines are less efficacious than others in inducing monoamine efflux). We then elucidate mechanisms of allosteric inhibition and stimulation of MATs, which can be functionally selective for either substrate uptake or amphetamine-induced release. These concepts are integrated into a parsimonious kinetic framework, which relies exclusively on physiologic transport modes (without any deviation from an alternating access mechanism). The model posits cooperative substrate and Na+ binding and functional selectivity by conformational selection (i.e., preference of the allosteric modulators for the substrate-loaded or substrate-free states of the transporter). Thus, current knowledge about the kinetics of monoamine transport is sufficiently detailed to provide a quantitative description of the releasing action of amphetamines, of substrate uptake, and of selective modulation thereof by allosteric modulators.
Collapse
Affiliation(s)
- Peter S Hasenhuetl
- Institute of Pharmacology, Gaston H. Glock Research Laboratories for Exploratory Drug Development, Center of Physiology and Pharmacology, Medical University of Vienna, Vienna, Austria
| | - Shreyas Bhat
- Institute of Pharmacology, Gaston H. Glock Research Laboratories for Exploratory Drug Development, Center of Physiology and Pharmacology, Medical University of Vienna, Vienna, Austria
| | - Michael Freissmuth
- Institute of Pharmacology, Gaston H. Glock Research Laboratories for Exploratory Drug Development, Center of Physiology and Pharmacology, Medical University of Vienna, Vienna, Austria
| | - Walter Sandtner
- Institute of Pharmacology, Gaston H. Glock Research Laboratories for Exploratory Drug Development, Center of Physiology and Pharmacology, Medical University of Vienna, Vienna, Austria
| |
Collapse
|
17
|
Das AK, Kudlacek O, Baumgart F, Jaentsch K, Stockner T, Sitte HH, Schütz GJ. Dopamine transporter forms stable dimers in the live cell plasma membrane in a phosphatidylinositol 4,5-bisphosphate-independent manner. J Biol Chem 2019; 294:5632-5642. [PMID: 30705091 PMCID: PMC6462504 DOI: 10.1074/jbc.ra118.006178] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/08/2018] [Revised: 01/28/2019] [Indexed: 01/01/2023] Open
Abstract
The human dopamine transporter (hDAT) regulates the level of the neurotransmitter dopamine (DA) in the synaptic cleft and recycles DA for storage in the presynaptic vesicular pool. Many neurotransmitter transporters exist as oligomers, but the physiological role of oligomerization remains unclear; for example, it has been speculated to be a prerequisite for amphetamine-induced release and protein trafficking. Previous studies point to an oligomeric quaternary structure of hDAT; however, the exact stoichiometry and the fraction of co-existing oligomeric states are not known. Here, we used single-molecule brightness analysis to quantify the degree of oligomerization of heterologously expressed hDAT fused to monomeric GFP (mGFP–hDAT) in Chinese hamster ovary (CHO) cells. We observed that monomers and dimers of mGFP–hDAT co-exist and that higher-order molecular complexes of mGFP–hDAT are absent at the plasma membrane. The mGFP–hDAT dimers were stable over several minutes, and the fraction of dimers was independent of the mGFP–hDAT surface density. Furthermore, neither oxidation nor depletion of cholesterol had any effect on the fraction of dimers. Unlike for the human serotonin transporter (hSERT), in which direct binding of phosphatidylinositol 4,5-bisphosphate (PIP2) stabilized the oligomers, the stability of mGFP–hDAT dimers was PIP2 independent.
Collapse
Affiliation(s)
- Anand Kant Das
- From the Institute of Applied Physics, TU Wien, Getreidemarkt 9, A-1060, Vienna and
| | - Oliver Kudlacek
- the Center for Physiology and Pharmacology, Institute of Pharmacology, Medical University Vienna, Waehringerstrasse 13a, A-1090 Vienna, Austria
| | - Florian Baumgart
- From the Institute of Applied Physics, TU Wien, Getreidemarkt 9, A-1060, Vienna and
| | - Kathrin Jaentsch
- the Center for Physiology and Pharmacology, Institute of Pharmacology, Medical University Vienna, Waehringerstrasse 13a, A-1090 Vienna, Austria
| | - Thomas Stockner
- the Center for Physiology and Pharmacology, Institute of Pharmacology, Medical University Vienna, Waehringerstrasse 13a, A-1090 Vienna, Austria
| | - Harald H Sitte
- the Center for Physiology and Pharmacology, Institute of Pharmacology, Medical University Vienna, Waehringerstrasse 13a, A-1090 Vienna, Austria
| | - Gerhard J Schütz
- From the Institute of Applied Physics, TU Wien, Getreidemarkt 9, A-1060, Vienna and
| |
Collapse
|
18
|
Abstract
There is a plethora of amphetamine derivatives exerting stimulant, euphoric, anti-fatigue, and hallucinogenic effects; all structural properties allowing these effects are contained within the amphetamine structure. In the first part of this review, the interaction of amphetamine with the dopamine transporter (DAT), crucially involved in its behavioral effects, is covered, as well as the role of dopamine synthesis, the vesicular monoamine transporter VMAT2, and organic cation 3 transporter (OCT3). The second part deals with requirements in amphetamine's effect on the kinases PKC, CaMKII, and ERK, whereas the third part focuses on where we are in developing anti-amphetamine therapeutics. Thus, treatments are discussed that target DAT, VMAT2, PKC, CaMKII, and OCT3. As is generally true for the development of therapeutics for substance use disorder, there are multiple preclinically promising specific compounds against (meth)amphetamine, for which further development and clinical trials are badly needed.
Collapse
Affiliation(s)
- Maarten E A Reith
- Department of Psychiatry, New York University School of Medicine, New York, NY, USA.
| | - Margaret E Gnegy
- Department of Pharmacology, University of Michigan School of Medicine, Ann Arbor, MI, USA
| |
Collapse
|
19
|
Maier J, Mayer FP, Luethi D, Holy M, Jäntsch K, Reither H, Hirtler L, Hoener MC, Liechti ME, Pifl C, Brandt SD, Sitte HH. The psychostimulant (±)-cis-4,4'-dimethylaminorex (4,4'-DMAR) interacts with human plasmalemmal and vesicular monoamine transporters. Neuropharmacology 2018; 138:282-291. [PMID: 29908239 DOI: 10.1016/j.neuropharm.2018.06.018] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/26/2018] [Revised: 05/01/2018] [Accepted: 06/12/2018] [Indexed: 10/28/2022]
Abstract
(±)-cis-4,4'-Dimethylaminorex (4,4'-DMAR) is a new psychoactive substance (NPS) that has been associated with 31 fatalities and other adverse events in Europe between June 2013 and February 2014. We used in vitro uptake inhibition and transporter release assays to determine the effects of 4,4'-DMAR on human high-affinity transporters for dopamine (DAT), norepinephrine (NET) and serotonin (SERT). In addition, we assessed its binding affinities to monoamine receptors and transporters. Furthermore, we investigated the interaction of 4,4'-DMAR with the vesicular monoamine transporter 2 (VMAT2) in rat phaeochromocytoma (PC12) cells and synaptic vesicles prepared from human striatum. 4,4'-DMAR inhibited uptake mediated by human DAT, NET or SERT, respectively in the low micromolar range (IC50 values < 2 μM). Release assays identified 4,4'-DMAR as a substrate type releaser, capable of inducing transporter-mediated reverse transport via DAT, NET and SERT. Furthermore, 4,4'-DMAR inhibited both the rat and human isoforms of VMAT2 at a potency similar to 3,4-methylenedioxymethylamphetamine (MDMA). This study identified 4,4'-DMAR as a potent non-selective monoamine releasing agent. In contrast to the known effects of aminorex and 4-methylaminorex, 4,4'-DMAR exerts profound effects on human SERT. The latter finding is consistent with the idea that fatalities associated with its abuse may be linked to monoaminergic toxicity including serotonin syndrome. The activity at VMAT2 suggests that chronic abuse of 4,4'-DMAR may result in long-term neurotoxicity.
Collapse
Affiliation(s)
- Julian Maier
- Medical University of Vienna, Center for Physiology and Pharmacology, Institute of Pharmacology, Währingerstraße 13A, 1090, Vienna, Austria
| | - Felix P Mayer
- Medical University of Vienna, Center for Physiology and Pharmacology, Institute of Pharmacology, Währingerstraße 13A, 1090, Vienna, Austria
| | - Dino Luethi
- University Hospital Basel and University of Basel, Division of Clinical Pharmacology and Toxicology, Department of Biomedicine, Hebelstraße 20, 4031, Basel, Switzerland
| | - Marion Holy
- Medical University of Vienna, Center for Physiology and Pharmacology, Institute of Pharmacology, Währingerstraße 13A, 1090, Vienna, Austria
| | - Kathrin Jäntsch
- Medical University of Vienna, Center for Physiology and Pharmacology, Institute of Pharmacology, Währingerstraße 13A, 1090, Vienna, Austria
| | - Harald Reither
- Medical University of Vienna, Center for Brain Research, Department of Molecular Neurosciences, Spitalgasse 4, 1090, Vienna, Austria
| | - Lena Hirtler
- Medical University of Vienna, Center for Anatomy and Cell Biology, Währingerstraße 13, 1090, Vienna, Austria
| | - Marius C Hoener
- F. Hoffmann - La Roche Ltd., pRED, Roche Innovation Center Basel, Neuroscience Research, Department of Neurosymptomatic Domains, Grenzacherstraße 124, 4070, Basel, Switzerland
| | - Matthias E Liechti
- University Hospital Basel and University of Basel, Division of Clinical Pharmacology and Toxicology, Department of Biomedicine, Hebelstraße 20, 4031, Basel, Switzerland
| | - Christian Pifl
- Medical University of Vienna, Center for Brain Research, Department of Molecular Neurosciences, Spitalgasse 4, 1090, Vienna, Austria
| | - Simon D Brandt
- School of Pharmacy and Biomolecular Sciences, Liverpool John Moores University, Byrom Street, Liverpool, L3 3AF, UK
| | - Harald H Sitte
- Medical University of Vienna, Center for Physiology and Pharmacology, Institute of Pharmacology, Währingerstraße 13A, 1090, Vienna, Austria; Center for Addiction Research and Science, Medical University Vienna, Waehringerstrasse 13A, 1090 Vienna, Austria.
| |
Collapse
|
20
|
Jayaraman K, Morley AN, Szöllősi D, Wassenaar TA, Sitte HH, Stockner T. Dopamine transporter oligomerization involves the scaffold domain, but spares the bundle domain. PLoS Comput Biol 2018; 14:e1006229. [PMID: 29874235 PMCID: PMC6005636 DOI: 10.1371/journal.pcbi.1006229] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/13/2018] [Revised: 06/18/2018] [Accepted: 05/24/2018] [Indexed: 12/11/2022] Open
Abstract
The human dopamine transporter (hDAT) is located on presynaptic neurons, where it plays an essential role in limiting dopaminergic signaling by temporarily curtailing high neurotransmitter concentration through rapid re-uptake. Transport by hDAT is energized by transmembrane ionic gradients. Dysfunction of this transporter leads to disease states, such as Parkinson’s disease, bipolar disorder or depression. It has been shown that hDAT and other members of the monoamine transporter family exist in oligomeric forms at the plasma membrane. Several residues are known to be involved in oligomerization, but interaction interfaces, oligomer orientation and the quarternary arrangement in the plasma membrane remain poorly understood. Here we examine oligomeric forms of hDAT using a direct approach, by following dimerization of two randomly-oriented hDAT transporters in 512 independent simulations, each being 2 μs in length. We employed the DAFT (docking assay for transmembrane components) approach, which is an unbiased molecular dynamics simulation method to identify oligomers, their conformations and populations. The overall ensemble of a total of >1 ms simulation time revealed a limited number of symmetric and asymmetric dimers. The identified dimer interfaces include all residues known to be involved in dimerization. Importantly, we find that the surface of the bundle domain is largely excluded from engaging in dimeric interfaces. Such an interaction would typically lead to inhibition by stabilization of one conformation, while substrate transport relies on a large scale rotation between the inward-facing and the outward-facing state. The human dopamine transporter efficiently removes the neurotransmitter dopamine from the synaptic cleft. Alteration of dopamine transporter function is associated with several neurological diseases, including mood disorders or attention-deficit hyperactivity disorder, but is also a major player in addiction and drug abuse. Functional studies have revealed that not only is transporter oligomerization involved in surface expression and endocytosis, but, more importantly, in reverse transport (efflux) of dopamine that is triggered by amphetamine-like drugs of abuse. Structural knowledge of transporter oligomerization is largely missing. We performed a large scale comprehensive computational study on transporter oligomerization to reveal dimer geometries and the residues involved in the interfaces. The dimer conformations we find in our dataset are fully consistent with all available experimental data in the literature, but also show novel interfaces. We further verified all dimer geometries by free energy calculations. Our results identified an unpredicted—but for the mechanism of substrate transport essential—property: the bundle domain, which moves during the transport cycle, is excluded from contributing to dimer interfaces, thereby allowing for unrestrained movements necessary to translocate substrates through the membrane.
Collapse
Affiliation(s)
- Kumaresan Jayaraman
- Medical University of Vienna Center for Physiology and Pharmacology, Institute of Pharmacology, Vienna, Austria
| | - Alex N. Morley
- Medical University of Vienna Center for Physiology and Pharmacology, Institute of Pharmacology, Vienna, Austria
| | - Daniel Szöllősi
- Medical University of Vienna Center for Physiology and Pharmacology, Institute of Pharmacology, Vienna, Austria
| | - Tsjerk A. Wassenaar
- Groningen Biomolecular Sciences and Biotechnology Institute and Zernike Institute for Advanced Materials, University of Groningen, Groningen, The Netherlands
| | - Harald H. Sitte
- Medical University of Vienna Center for Physiology and Pharmacology, Institute of Pharmacology, Vienna, Austria
| | - Thomas Stockner
- Medical University of Vienna Center for Physiology and Pharmacology, Institute of Pharmacology, Vienna, Austria
- * E-mail:
| |
Collapse
|
21
|
Fischer J, Kleinau G, Rutz C, Zwanziger D, Khajavi N, Müller A, Rehders M, Brix K, Worth CL, Führer D, Krude H, Wiesner B, Schülein R, Biebermann H. Evidence of G-protein-coupled receptor and substrate transporter heteromerization at a single molecule level. Cell Mol Life Sci 2018; 75:2227-2239. [PMID: 29290039 PMCID: PMC11105501 DOI: 10.1007/s00018-017-2728-1] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/29/2017] [Revised: 11/17/2017] [Accepted: 12/11/2017] [Indexed: 10/18/2022]
Abstract
G-protein-coupled receptors (GPCRs) can constitute complexes with non-GPCR integral membrane proteins, while such interaction has not been demonstrated at a single molecule level so far. We here investigated the potential interaction between the thyrotropin receptor (TSHR) and the monocarboxylate transporter 8 (MCT8), a member of the major facilitator superfamily (MFS), using fluorescence cross-correlation spectroscopy (FCCS). Both the proteins are expressed endogenously on the basolateral plasma membrane of the thyrocytes and are involved in stimulation of thyroid hormone production and release. Indeed, we demonstrate strong interaction between both the proteins which causes a suppressed activation of Gq/11 by TSH-stimulated TSHR. Thus, we provide not only evidence for a novel interaction between the TSHR and MCT8, but could also prove this interaction on a single molecule level. Moreover, this interaction forces biased signaling at the TSHR. These results are of general interest for both the GPCR and the MFS research fields.
Collapse
Affiliation(s)
- Jana Fischer
- Institut für Experimentelle Pädiatrische Endokrinologie, Charité - Universitätsmedizin Berlin, 13353, Berlin, Germany
| | - Gunnar Kleinau
- Institut für Experimentelle Pädiatrische Endokrinologie, Charité - Universitätsmedizin Berlin, 13353, Berlin, Germany
- Group Protein X-ray Crystallography and Signal Transduction, Institute of Medical Physics and Biophysics, Charité - Universitätsmedizin Berlin, 10117, Berlin, Germany
| | - Claudia Rutz
- Protein Trafficking Group, Leibniz-Forschungsinstitut für Molekulare Pharmakologie, Robert-Rössle-Str. 10, 13125, Berlin, Germany
| | - Denise Zwanziger
- Division of Laboratory Research, Department of Endocrinology, Diabetology and Metabolism, University Hospital Essen, University Duisburg-Essen, 45147, Essen, Germany
| | - Noushafarin Khajavi
- Institut für Experimentelle Pädiatrische Endokrinologie, Charité - Universitätsmedizin Berlin, 13353, Berlin, Germany
| | - Anne Müller
- Institut für Experimentelle Pädiatrische Endokrinologie, Charité - Universitätsmedizin Berlin, 13353, Berlin, Germany
| | - Maren Rehders
- Department of Life Sciences and Chemistry, Jacobs University Bremen, 28759, Bremen, Germany
| | - Klaudia Brix
- Department of Life Sciences and Chemistry, Jacobs University Bremen, 28759, Bremen, Germany
| | - Catherine L Worth
- Structural Bioinformatics and Protein Design Group, Leibniz-Forschungsinstitut für Molekulare Pharmakologie, 13125, Berlin, Germany
| | - Dagmar Führer
- Division of Laboratory Research, Department of Endocrinology, Diabetology and Metabolism, University Hospital Essen, University Duisburg-Essen, 45147, Essen, Germany
| | - Heiko Krude
- Institut für Experimentelle Pädiatrische Endokrinologie, Charité - Universitätsmedizin Berlin, 13353, Berlin, Germany
| | - Burkhard Wiesner
- Protein Trafficking Group, Leibniz-Forschungsinstitut für Molekulare Pharmakologie, Robert-Rössle-Str. 10, 13125, Berlin, Germany
- Cellular Imaging Group, Leibniz-Forschungsinstitut für Molekulare Pharmakologie, 13125, Berlin, Germany
| | - Ralf Schülein
- Protein Trafficking Group, Leibniz-Forschungsinstitut für Molekulare Pharmakologie, Robert-Rössle-Str. 10, 13125, Berlin, Germany.
| | - Heike Biebermann
- Institut für Experimentelle Pädiatrische Endokrinologie, Charité - Universitätsmedizin Berlin, 13353, Berlin, Germany.
| |
Collapse
|
22
|
Hasenhuetl PS, Bhat S, Mayer FP, Sitte HH, Freissmuth M, Sandtner W. A kinetic account for amphetamine-induced monoamine release. J Gen Physiol 2018; 150:431-451. [PMID: 29439119 PMCID: PMC5839721 DOI: 10.1085/jgp.201711915] [Citation(s) in RCA: 24] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/02/2017] [Accepted: 01/18/2018] [Indexed: 01/14/2023] Open
Abstract
This study on serotonin transporters shows that amphetamine-induced monoamine release requires cooperative substrate and cosubstrate binding. A kinetic model is presented that can account qualitatively and quantitatively for the releasing action of amphetamines. The plasmalemmal monoamine transporters for dopamine, norepinephrine, and serotonin (SERT) are targets for amphetamines. In vivo, amphetamines elicit most, if not all, of their actions by triggering monoamine efflux. This is thought to be accomplished by an amphetamine-induced switch from the forward-transport to the substrate-exchange mode. The mechanism underlying this switch has remained elusive; available kinetic models posit that substrates and cosubstrate Na+ ions bind either in a random or in a sequential order. Neither can account for all reported experimental observations. We used electrophysiological recordings to interrogate crucial conformational transitions associated with the binding of five different substrates (serotonin, para-chloroamphetamine, and the high-affinity naphthyl-propan-amines PAL-287, PAL-1045, and PAL-1046) to human SERT expressed in HEK293 cells; specifically, we determined the relaxation kinetics of SERT from a substrate-loaded to a substrate-free state at various intracellular and extracellular Na+ concentrations. These rates and their dependence on intracellular and extracellular Na+ concentrations differed considerably between substrates. We also examined the effect of K+ on substrate affinity and found that K+ enhanced substrate dissociation. A kinetic model was developed, which allowed for random, but cooperative, binding of substrate and Na+ (or K+). The synthetic data generated by this model recapitulated the experimental observations. More importantly, the cooperative binding model accounted for the releasing action of amphetamines without any digression from alternating access. To the best of our knowledge, this model is the first to provide a mechanistic framework for amphetamine-induced monoamine release and to account for the findings that some substrates are less efficacious than others in promoting the substrate-exchange mode.
Collapse
Affiliation(s)
- Peter S Hasenhuetl
- Institute of Pharmacology and the Gaston H. Glock Research Laboratories for Exploratory Drug Development, Center of Physiology and Pharmacology, Medical University of Vienna, Vienna, Austria
| | - Shreyas Bhat
- Institute of Pharmacology and the Gaston H. Glock Research Laboratories for Exploratory Drug Development, Center of Physiology and Pharmacology, Medical University of Vienna, Vienna, Austria
| | - Felix P Mayer
- Institute of Pharmacology and the Gaston H. Glock Research Laboratories for Exploratory Drug Development, Center of Physiology and Pharmacology, Medical University of Vienna, Vienna, Austria
| | - Harald H Sitte
- Institute of Pharmacology and the Gaston H. Glock Research Laboratories for Exploratory Drug Development, Center of Physiology and Pharmacology, Medical University of Vienna, Vienna, Austria
| | - Michael Freissmuth
- Institute of Pharmacology and the Gaston H. Glock Research Laboratories for Exploratory Drug Development, Center of Physiology and Pharmacology, Medical University of Vienna, Vienna, Austria
| | - Walter Sandtner
- Institute of Pharmacology and the Gaston H. Glock Research Laboratories for Exploratory Drug Development, Center of Physiology and Pharmacology, Medical University of Vienna, Vienna, Austria
| |
Collapse
|
23
|
Karam CS, Javitch JA. Phosphorylation of the Amino Terminus of the Dopamine Transporter: Regulatory Mechanisms and Implications for Amphetamine Action. ADVANCES IN PHARMACOLOGY (SAN DIEGO, CALIF.) 2017; 82:205-234. [PMID: 29413521 DOI: 10.1016/bs.apha.2017.09.002] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
Amphetamines (AMPHs) are potent psychostimulants that are widely used and abused, with profound medical and societal impact. Their actions at dopaminergic neurons are thought to mediate their therapeutic efficacy as well as their liability for abuse and dependence. AMPHs target the dopamine transporter (DAT), the plasmalemmal membrane protein that mediates the inactivation of released dopamine (DA) through its reuptake. AMPHs act as substrates for DAT and are known to cause mobilization of dopamine (DA) to the cell exterior via DAT-mediated reverse transport (efflux). It has become increasingly evident that the mechanisms that regulate AMPH-induced DA efflux are distinct from those that regulate DA uptake. Central to these mechanisms is the phosphorylation of the DAT amino (N)-terminus, which has been repeatedly demonstrated to facilitate DAT-mediated DA efflux, without impacting other aspects of DAT physiology. This review aims to summarize the current status of knowledge regarding DAT N-terminal phosphorylation and its regulation by protein modulators and the membrane microenvironment. A better understanding of these mechanisms may lead to the identification of novel therapeutic approaches that interfere selectively with the pharmacological effects of AMPHs without altering the physiological function of DAT.
Collapse
Affiliation(s)
- Caline S Karam
- College of Physicians & Surgeons, Columbia University, New York, NY, United States; Division of Molecular Therapeutics, New York State Psychiatric Institute, New York, NY, United States
| | - Jonathan A Javitch
- College of Physicians & Surgeons, Columbia University, New York, NY, United States; Division of Molecular Therapeutics, New York State Psychiatric Institute, New York, NY, United States.
| |
Collapse
|
24
|
Luethi D, Kaeser PJ, Brandt SD, Krähenbühl S, Hoener MC, Liechti ME. Pharmacological profile of methylphenidate-based designer drugs. Neuropharmacology 2017; 134:133-140. [PMID: 28823611 DOI: 10.1016/j.neuropharm.2017.08.020] [Citation(s) in RCA: 38] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/10/2017] [Revised: 08/14/2017] [Accepted: 08/16/2017] [Indexed: 12/13/2022]
Abstract
BACKGROUND Methylphenidate-based designer drugs are new psychoactive substances (NPS) that are used outside medical settings and their pharmacology is largely unexplored. The aim of the present study was to characterize the pharmacology of methylphenidate-based substances in vitro. METHODS We determined the potencies of the methylphenidate-based NPS N-benzylethylphenidate, 3,4-dichloroethylphenidate, 3,4-dichloromethylphenidate, ethylnaphthidate, ethylphenidate, 4-fluoromethylphenidate, isopropylphenidate, 4-methylmethylphenidate, methylmorphenate, and propylphenidate and the potencies of the related compounds cocaine and modafinil with respect to norepinephrine, dopamine, and serotonin transporter inhibition in transporter-transfected human embryonic kidney 293 cells. We also investigated monoamine efflux and monoamine receptor and transporter binding affinities. Furthermore, we assessed the cell integrity under assay conditions. RESULTS All methylphenidate-based substances inhibited the norepinephrine and dopamine transporters 4 to >1000-fold more potently than the serotonin transporter. Similar to methylphenidate and cocaine, methylphenidate-based NPS did not elicit transporter-mediated efflux of monoamines. Besides binding to monoamine transporters, several test drugs had affinity for adrenergic, serotonergic, and rat trace amine-associated receptors but not for dopaminergic or mouse trace amine-associated receptors. No cytotoxicity was observed after drug treatment at assay concentrations. CONCLUSION Methylphenidate-based substances had pharmacological profiles similar to methylphenidate and cocaine. The predominant actions on dopamine transporters vs. serotonin transporters may be relevant when considering abuse liability. This article is part of the Special Issue entitled 'Designer Drugs and Legal Highs.'
Collapse
Affiliation(s)
- Dino Luethi
- Division of Clinical Pharmacology and Toxicology, Department of Biomedicine, University Hospital Basel and University of Basel, Basel, Switzerland
| | - Philine J Kaeser
- Division of Clinical Pharmacology and Toxicology, Department of Biomedicine, University Hospital Basel and University of Basel, Basel, Switzerland
| | - Simon D Brandt
- School of Pharmacy and Biomolecular Sciences, Liverpool John Moores University, Liverpool, United Kingdom
| | - Stephan Krähenbühl
- Division of Clinical Pharmacology and Toxicology, Department of Biomedicine, University Hospital Basel and University of Basel, Basel, Switzerland
| | - Marius C Hoener
- Neuroscience Research, pRED, Roche Innovation Center Basel, F. Hoffmann-La Roche Ltd, Basel, Switzerland
| | - Matthias E Liechti
- Division of Clinical Pharmacology and Toxicology, Department of Biomedicine, University Hospital Basel and University of Basel, Basel, Switzerland.
| |
Collapse
|
25
|
Transporter oligomerization: form and function. Biochem Soc Trans 2017; 44:1737-1744. [PMID: 27913684 PMCID: PMC5134999 DOI: 10.1042/bst20160217] [Citation(s) in RCA: 34] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/25/2016] [Revised: 09/16/2016] [Accepted: 09/19/2016] [Indexed: 12/22/2022]
Abstract
Transporters are integral membrane proteins with central roles in the efficient movement of molecules across biological membranes. Many transporters exist as oligomers in the membrane. Depending on the individual transport protein, oligomerization can have roles in membrane trafficking, function, regulation and turnover. For example, our recent studies on UapA, a nucleobase ascorbate transporter, from Aspergillus nidulans, have revealed both that dimerization of this protein is essential for correct trafficking to the membrane and the structural basis of how one UapA protomer can affect the function of the closely associated adjacent protomer. Here, we review the roles of oligomerization in many particularly well-studied transporters and transporter families.
Collapse
|
26
|
Mayer FP, Luf A, Nagy C, Holy M, Schmid R, Freissmuth M, Sitte HH. Application of a Combined Approach to Identify New Psychoactive Street Drugs and Decipher Their Mechanisms at Monoamine Transporters. Curr Top Behav Neurosci 2017; 32:333-350. [PMID: 28025810 DOI: 10.1007/7854_2016_63] [Citation(s) in RCA: 27] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
Psychoactive compounds can cause acute and long-term health problems and lead to addiction. In addition to well-studied and legally controlled compounds like cocaine, new psychoactive substances (NPS) are appearing in street drug markets as replacement strategies and legal alternatives. NPS are effectively marketed as "designer drugs" or "research chemicals" without any knowledge of their underlying pharmacological mode of action and their potential toxicological effects and obviously devoid of any registration process. As of 2016, the knowledge of structure-activity relationships for most NPS is scarce, and predicting detailed pharmacological activity of newly emerging drugs is a challenging task. Therefore, it is important to combine different approaches and employ biological test systems that are superior to mere chemical analysis in recognizing novel and potentially harmful street drugs. In this chapter, we provide a detailed description of techniques to decipher the molecular mechanism of action of NPS that target the high-affinity transporters for dopamine, norepinephrine, and serotonin. In addition, this chapter provides insights into a combined approach to identify and characterize new psychoactive street drugs of unknown content in a collaboration with the Austrian prevention project "checkit!."
Collapse
Affiliation(s)
- Felix P Mayer
- Center of Physiology and Pharmacology, Institute of Pharmacology, Medical University Vienna, Waehringerstrasse 13a, A-1090, Vienna, Austria
| | - Anton Luf
- Clinical Department of Laboratory Medicine, Medical University of Vienna, Waehringer Guertel 10-20, 1090, Vienna, Austria
| | - Constanze Nagy
- checkit! - Suchthilfe Wien GmbH, Gumpendorfer Gürtel 8, 1060, Vienna, Austria
| | - Marion Holy
- Center of Physiology and Pharmacology, Institute of Pharmacology, Medical University Vienna, Waehringerstrasse 13a, A-1090, Vienna, Austria
| | - Rainer Schmid
- Clinical Department of Laboratory Medicine, Medical University of Vienna, Waehringer Guertel 10-20, 1090, Vienna, Austria
| | - Michael Freissmuth
- Center of Physiology and Pharmacology, Institute of Pharmacology, Medical University Vienna, Waehringerstrasse 13a, A-1090, Vienna, Austria
| | - Harald H Sitte
- Center of Physiology and Pharmacology, Institute of Pharmacology, Medical University Vienna, Waehringerstrasse 13a, A-1090, Vienna, Austria.
- Center for Addiction Research and Science - Medical University Vienna, Waehringerstrasse 13A, 1090, Vienna, Austria.
| |
Collapse
|
27
|
Zestos AG, Kennedy RT. Microdialysis Coupled with LC-MS/MS for In Vivo Neurochemical Monitoring. AAPS JOURNAL 2017; 19:1284-1293. [PMID: 28660399 DOI: 10.1208/s12248-017-0114-4] [Citation(s) in RCA: 58] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/31/2017] [Accepted: 06/13/2017] [Indexed: 10/19/2022]
Abstract
Microdialysis is a powerful sampling technique used to monitor small molecules in vivo. Despite the many applications of microdialysis sampling, it is limited by the method of analyzing the resulting samples. An emerging technique for analysis of microdialysis samples is liquid chromatography-tandem mass spectrometry (LC-MS/MS). This technique is highly versatile, allowing multiplexed analysis of neurotransmitters, metabolites, and neuropeptides. Using LC-MS/MS for polar neurotransmitters is hampered by weak retention reverse phase LC columns. Several derivatization reagents have been utilized to enhance separation and resolution of neurochemicals in dialysate samples including benzoyl chloride (BzCl), dansyl chloride, formaldehyde, ethylchloroformate, and propionic anhydride. BzCl reacts with amine and phenol groups so that many neurotransmitters can be labeled. Besides improving separation on reverse phase columns, this reagent also increases sensitivity. It is available in a heavy form so that it can be used to make stable-isotope labeled internal standard for improved quantification. Using BzCl with LC-MS/MS has allowed for measuring as many as 70 neurochemicals in a single assay. With slightly different conditions, LC-MS/MS has also been used for monitoring endocannabinoids. LC-MS/MS is also useful for neuropeptide assay because it allows for highly sensitive, sequence specific measurement of most peptides. These advances have allowed for multiplexed neurotransmitter measurements in behavioral, circuit analysis, and drug effect studies.
Collapse
Affiliation(s)
- Alexander G Zestos
- Department of Chemistry, University of Michigan, 930 N. University, Ann Arbor, Michigan, 48109-1055, USA.,Department of Pharmacology, University of Michigan, 2301 MSRB III, 1150 W. Medical Center Dr., Ann Arbor, Michigan, 48109-1055, USA.,Department of Chemistry, American University, 4400 Massachusetts Avenue, NW, Washington, District of Columbia, 20016, USA
| | - Robert T Kennedy
- Department of Chemistry, University of Michigan, 930 N. University, Ann Arbor, Michigan, 48109-1055, USA. .,Department of Pharmacology, University of Michigan, 2301 MSRB III, 1150 W. Medical Center Dr., Ann Arbor, Michigan, 48109-1055, USA.
| |
Collapse
|
28
|
Cheng MH, Garcia-Olivares J, Wasserman S, DiPietro J, Bahar I. Allosteric modulation of human dopamine transporter activity under conditions promoting its dimerization. J Biol Chem 2017; 292:12471-12482. [PMID: 28584050 DOI: 10.1074/jbc.m116.763565] [Citation(s) in RCA: 24] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/16/2016] [Revised: 05/12/2017] [Indexed: 12/27/2022] Open
Abstract
The human dopamine (DA) transporter (hDAT) is a key regulator of neurotransmission and a target for antidepressants and addictive drugs. Despite the recent resolution of dDAT structures from Drosophila melanogaster, complete understanding of its mechanism of function and even information on its biological assembly is lacking. The resolved dDAT structures are monomeric, but growing evidence suggests that hDAT might function as a multimer, and its oligomerization may be relevant to addictive drug effects. Here, using structure-based computations, we examined the possible mechanisms of hDAT dimerization and its dynamics in a lipid bilayer. Using a combination of site-directed mutagenesis, DA-uptake, and cross-linking experiments that exploited the capacity of Cys-306 to form intermonomeric disulfide bridges in the presence of an oxidizing agent, we tested the effects of mutations at transmembrane segment (TM) 6 and 12 helices in HEK293 cells. The most probable structural model for hDAT dimer suggested by computations and experiments differed from the dimeric structure resolved for the bacterial homolog, LeuT, presumably because of a kink at TM12 preventing favorable monomer packing. Instead, TM2, TM6, and TM11 line the dimer interface. Molecular dynamics simulations of the dimeric hDAT indicated that the two subunits tend to undergo cooperative structural changes, both on local (extracellular gate opening/closure) and global (transition between outward-facing and inward-facing states) scales. These observations suggest that hDAT transport properties may be allosterically modulated under conditions promoting dimerization. Our study provides critical insights into approaches for examining the oligomerization of neurotransmitter transporters and sheds light on their drug modulation.
Collapse
Affiliation(s)
- Mary Hongying Cheng
- Department of Computational and Systems Biology, School of Medicine, University of Pittsburgh, Pittsburgh, Pennsylvania 15260
| | - Jennie Garcia-Olivares
- Laboratory of Molecular and Cellular Neurobiology, National Institute of Mental Health, Bethesda, Maryland 20892
| | - Steven Wasserman
- Laboratory of Molecular and Cellular Neurobiology, National Institute of Mental Health, Bethesda, Maryland 20892
| | - Jennifer DiPietro
- Laboratory of Molecular and Cellular Neurobiology, National Institute of Mental Health, Bethesda, Maryland 20892
| | - Ivet Bahar
- Department of Computational and Systems Biology, School of Medicine, University of Pittsburgh, Pittsburgh, Pennsylvania 15260.
| |
Collapse
|
29
|
Rastedt DE, Vaughan RA, Foster JD. Palmitoylation mechanisms in dopamine transporter regulation. J Chem Neuroanat 2017; 83-84:3-9. [PMID: 28115272 DOI: 10.1016/j.jchemneu.2017.01.002] [Citation(s) in RCA: 40] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/16/2016] [Revised: 12/21/2016] [Accepted: 01/17/2017] [Indexed: 02/01/2023]
Abstract
The neurotransmitter dopamine (DA) plays a key role in several biological processes including reward, mood, motor activity and attention. Synaptic DA homeostasis is controlled by the dopamine transporter (DAT) which transports extracellular DA into the presynaptic neuron after release and regulates its availability to receptors. Many neurological disorders such as schizophrenia, bipolar disorder, Parkinson disease and attention-deficit hyperactivity disorder are associated with imbalances in DA homeostasis that may be related to DAT dysfunction. DAT is also a target of psychostimulant and therapeutic drugs that inhibit DA reuptake and lead to elevated dopaminergic neurotransmission. We have recently demonstrated the acute and chronic modulation of DA reuptake activity and DAT stability through S-palmitoylation, the linkage of a 16-carbon palmitate group to cysteine via a thioester bond. This review summarizes the properties and regulation of DAT palmitoylation and describes how it serves to affect various transporter functions. Better understanding of the role of palmitoylation in regulation of DAT function may lead to identification of therapeutic targets for modulation of DA homeostasis in the treatment of dopaminergic disorders.
Collapse
Affiliation(s)
- Danielle E Rastedt
- Department of Biomedical Sciences, University of North Dakota, School of Medicine and Health Sciences, Grand Forks, ND 58202, United States
| | - Roxanne A Vaughan
- Department of Biomedical Sciences, University of North Dakota, School of Medicine and Health Sciences, Grand Forks, ND 58202, United States
| | - James D Foster
- Department of Biomedical Sciences, University of North Dakota, School of Medicine and Health Sciences, Grand Forks, ND 58202, United States.
| |
Collapse
|
30
|
Kern C, Erdem FA, El-Kasaby A, Sandtner W, Freissmuth M, Sucic S. The N Terminus Specifies the Switch between Transport Modes of the Human Serotonin Transporter. J Biol Chem 2017; 292:3603-3613. [PMID: 28104804 PMCID: PMC5339746 DOI: 10.1074/jbc.m116.771360] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/06/2016] [Revised: 01/18/2017] [Indexed: 10/25/2022] Open
Abstract
The serotonin transporter (SERT) and other monoamine transporters operate in either a forward transport mode where the transporter undergoes a full transport cycle or an exchange mode where the transporter seesaws through half-cycles. Amphetamines trigger the exchange mode, leading to substrate efflux. This efflux was proposed to rely on the N terminus, which was suggested to adopt different conformations in the inward facing, outward facing and amphetamine-bound states. This prediction was verified by tryptic digestion of SERT-expressing membranes: in the absence of Na+, the N terminus was rapidly digested. Amphetamine conferred protection against cleavage, suggesting a relay between the conformational states of the hydrophobic core and the N terminus. We searched for a candidate segment that supported the conformational switch by serial truncation removing 22 (ΔN22), 32 (ΔN32), or 42 (ΔN42) N-terminal residues. This did not affect surface expression, inhibitor binding, and substrate influx. However, amphetamine-induced efflux by SERT-ΔN32 or SERT-ΔN42 (but not by SERT-ΔN22) was markedly diminished. We examined the individual steps in the transport cycle by recording transporter-associated currents: the recovery rate of capacitive peak, but not of steady state, currents was significantly lower for SERT-ΔN32 than that of wild type SERT and SERT-ΔN22. Thus, the exchange mode of SERT-ΔN32 was selectively impaired. Our observations show that the N terminus affords the switch between transport modes. The findings are consistent with a model where the N terminus acts as a lever to support amphetamine-induced efflux by SERT.
Collapse
Affiliation(s)
- Carina Kern
- From the Institute of Pharmacology, Center of Physiology and Pharmacology, Medical University of Vienna, A-1090 Vienna, Austria
| | - Fatma Asli Erdem
- From the Institute of Pharmacology, Center of Physiology and Pharmacology, Medical University of Vienna, A-1090 Vienna, Austria
| | - Ali El-Kasaby
- From the Institute of Pharmacology, Center of Physiology and Pharmacology, Medical University of Vienna, A-1090 Vienna, Austria
| | - Walter Sandtner
- From the Institute of Pharmacology, Center of Physiology and Pharmacology, Medical University of Vienna, A-1090 Vienna, Austria
| | - Michael Freissmuth
- From the Institute of Pharmacology, Center of Physiology and Pharmacology, Medical University of Vienna, A-1090 Vienna, Austria
| | - Sonja Sucic
- From the Institute of Pharmacology, Center of Physiology and Pharmacology, Medical University of Vienna, A-1090 Vienna, Austria
| |
Collapse
|
31
|
Anderluh A, Hofmaier T, Klotzsch E, Kudlacek O, Stockner T, Sitte HH, Schütz GJ. Direct PIP 2 binding mediates stable oligomer formation of the serotonin transporter. Nat Commun 2017; 8:14089. [PMID: 28102201 PMCID: PMC5253637 DOI: 10.1038/ncomms14089] [Citation(s) in RCA: 58] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/19/2016] [Accepted: 11/28/2016] [Indexed: 12/30/2022] Open
Abstract
The human serotonin transporter (hSERT) mediates uptake of serotonin from the synaptic cleft and thereby terminates serotonergic signalling. We have previously found by single-molecule microscopy that SERT forms stable higher-order oligomers of differing stoichiometry at the plasma membrane of living cells. Here, we report that SERT oligomer assembly at the endoplasmic reticulum (ER) membrane follows a dynamic equilibration process, characterized by rapid exchange of subunits between different oligomers, and by a concentration dependence of the degree of oligomerization. After trafficking to the plasma membrane, however, the SERT stoichiometry is fixed. Stabilization of the oligomeric SERT complexes is mediated by the direct binding to phosphoinositide phosphatidylinositol-4,5-biphosphate (PIP2). The observed spatial decoupling of oligomer formation from the site of oligomer operation provides cells with the ability to define protein quaternary structures independent of protein density at the cell surface.
Collapse
Affiliation(s)
- Andreas Anderluh
- Institute of Applied Physics, TU Wien, Wiedner Hauptstrasse 8-10, Vienna 1040, Austria
| | - Tina Hofmaier
- Center for Physiology and Pharmacology, Institute of Pharmacology, Medical University Vienna, Waehringerstrasse 13A, Vienna 1090, Austria
| | - Enrico Klotzsch
- EMBL Australia Node in Single Molecule Science, School of Medical Sciences, ARC Centre of Excellence in Advanced Molecular Imaging, University of New South Wales, Sydney, New South Wales 2052, Australia
| | - Oliver Kudlacek
- Center for Physiology and Pharmacology, Institute of Pharmacology, Medical University Vienna, Waehringerstrasse 13A, Vienna 1090, Austria
| | - Thomas Stockner
- Center for Physiology and Pharmacology, Institute of Pharmacology, Medical University Vienna, Waehringerstrasse 13A, Vienna 1090, Austria
| | - Harald H. Sitte
- Center for Physiology and Pharmacology, Institute of Pharmacology, Medical University Vienna, Waehringerstrasse 13A, Vienna 1090, Austria
| | - Gerhard J. Schütz
- Institute of Applied Physics, TU Wien, Wiedner Hauptstrasse 8-10, Vienna 1040, Austria
| |
Collapse
|
32
|
Abstract
Products containing psychoactive synthetic cathinones, such as mephedrone and 3,4-methylenedioxypyrovalerone (MDPV) are prevalent in our society. Synthetic cathinones are structurally similar to methamphetamine, and numerous synthetics have biological activity at dopamine, serotonin, and norepinephrine transporters. Importantly, monoamine transporters co-transport sodium ions along with their substrate, and movement of substrates and ions through the transporter can generate measurable ionic currents. Here we review how electrophysiological information has enabled us to determine how synthetic cathinones affect transporter-mediated currents in cells that express these transporters. Specifically, drugs that act as transporter substrates induce inward depolarizing currents when cells are held near their resting membrane potential, whereas drugs that act as transporter blockers induce apparent outward currents by blocking an inherent inward leak current. We have employed the two-electrode voltage-clamp technique in Xenopus laevis oocytes overexpressing monoamine transporters to determine whether synthetic cathinones found in the so-called bath salts products behave as blockers or substrates. We also examined the structure-activity relationships for synthetic cathinone analogs related to the widely abused compound MDPV, a common constituent in "bath salts" possessing potent actions at the dopamine transporter.
Collapse
Affiliation(s)
- Ernesto Solis
- In Vivo Electrophysiology Unit, Behavioral Neuroscience Research Branch, National Institute on Drug Abuse - Intramural Research Program, National Institutes of Health, Triad Technology Center, 333 Cassell Drive, Suite 2200, Baltimore, MD, 21224, USA.
| |
Collapse
|
33
|
Zwanziger D, Schmidt M, Fischer J, Kleinau G, Braun D, Schweizer U, Moeller LC, Biebermann H, Fuehrer D. The long N-terminus of the human monocarboxylate transporter 8 is a target of ubiquitin-dependent proteasomal degradation which regulates protein expression and oligomerization capacity. Mol Cell Endocrinol 2016; 434:278-87. [PMID: 27222294 DOI: 10.1016/j.mce.2016.05.017] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/02/2015] [Revised: 04/19/2016] [Accepted: 05/20/2016] [Indexed: 11/24/2022]
Abstract
Monocarboxylate transporter 8 (MCT8) equilibrates thyroid hormones between the extra- and the intracellular sides. MCT8 exists either with a short or a long N-terminus, but potential functional differences between both variants are yet not known. We, therefore, generated MCT8 constructs which are different in N-terminal length: MCT8(1-613), MCT8(25-613), MCT8(49-613) and MCT8(75-613). The M75G substitution prevents translation of MCT8(75-613) and ensures expression of full-length MCT8 protein. The K56G substitution was made to prevent ubiquitinylation. Cell-surface expression, localization and proteasomal degradation were investigated using C-terminally GFP-tagged MCT8 constructs (HEK293 and MDCK1 cells) and oligomerization capacity was determined using N-terminally HA- and C-terminally FLAG-tagged MCT8 constructs (COS7 cells). MCT8(1-613)-GFP showed a lower protein expression than the shorter MCT8(75-613)-GFP protein. The proteasome inhibitor lactacystin increased MCT8(1-613)-GFP protein amount, suggesting proteasomal degradation of MCT8 with the long N-terminus. Ubiquitin conjugation of MCT8(1-613)-GFP was found by immuno-precipitation. A diminished ubiquitin conjugation caused by K56G substitution resulted in increased MCT8(1-613)-GFP protein expression. Sandwich ELISA was performed to investigate if the bands at higher molecular weight observed in Western blot analysis are due to MCT8 oligomerization, which was indeed shown. Our data imply a role of the long N-terminus of MCT8 as target of ubiquitin-dependent proteasomal degradation affecting MCT8 amount and subsequently oligomerization capacity.
Collapse
Affiliation(s)
- Denise Zwanziger
- University of Duisburg-Essen, Department of Endocrinology and Metabolism and Division of Laboratory Research, Hufelandstraße 55, 45147 Essen, Germany.
| | - Mathias Schmidt
- University of Duisburg-Essen, Department of Endocrinology and Metabolism and Division of Laboratory Research, Hufelandstraße 55, 45147 Essen, Germany.
| | - Jana Fischer
- Charitè-Berlin, Institute of Experimental Pediatric Endocrinology, Augustenburgerplatz 1, 13353 Berlin, Germany.
| | - Gunnar Kleinau
- Charitè-Berlin, Institute of Experimental Pediatric Endocrinology, Augustenburgerplatz 1, 13353 Berlin, Germany.
| | - Doreen Braun
- Rheinische Friedrich-Wilhelms Universität Bonn, Institut für Biochemie und Molekularbiologie, Nussallee 11, 53115 Bonn, Germany.
| | - Ulrich Schweizer
- Rheinische Friedrich-Wilhelms Universität Bonn, Institut für Biochemie und Molekularbiologie, Nussallee 11, 53115 Bonn, Germany.
| | - Lars Christian Moeller
- University of Duisburg-Essen, Department of Endocrinology and Metabolism and Division of Laboratory Research, Hufelandstraße 55, 45147 Essen, Germany.
| | - Heike Biebermann
- Charitè-Berlin, Institute of Experimental Pediatric Endocrinology, Augustenburgerplatz 1, 13353 Berlin, Germany.
| | - Dagmar Fuehrer
- University of Duisburg-Essen, Department of Endocrinology and Metabolism and Division of Laboratory Research, Hufelandstraße 55, 45147 Essen, Germany.
| |
Collapse
|
34
|
New psychoactive substances: an overview on recent publications on their toxicodynamics and toxicokinetics. Arch Toxicol 2016; 90:2421-44. [PMID: 27665567 DOI: 10.1007/s00204-016-1812-x] [Citation(s) in RCA: 50] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/09/2016] [Accepted: 08/04/2016] [Indexed: 02/07/2023]
Abstract
This review article covers English-written and PubMed-listed review articles and original studies published between January 2015 and April 2016 dealing with the toxicodynamics and toxicokinetics of new psychoactive substances. Compounds covered include stimulants and entactogens, synthetic cannabinoids, tryptamines, NBOMes, phencyclidine-like drugs, benzodiazepines, and opioids. First, an overview and discussion is provided on timely review articles followed by an overview and discussion on recent original studies. Both sections are then concluded by an opinion on these latest developments. This review shows that the NPS market is still highly dynamic and that the data published on their toxicodynamics and toxicokinetics can hardly keep pace with the appearance of new entities. However, data available are very helpful to understand and predict how NPS may behave in severe intoxication. The currently best-documented parameter is the in vitro metabolism of NPS, a prerequisite to allow detection of NPS in biological matrices in cases of acute intoxications or chronic consumption. However, additional data such as their chronic toxicity are still lacking.
Collapse
|
35
|
Solanki RR, Scholl JL, Watt MJ, Renner KJ, Forster GL. Amphetamine Withdrawal Differentially Increases the Expression of Organic Cation Transporter 3 and Serotonin Transporter in Limbic Brain Regions. J Exp Neurosci 2016; 10:93-100. [PMID: 27478387 PMCID: PMC4957605 DOI: 10.4137/jen.s40231] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/24/2016] [Revised: 06/27/2016] [Accepted: 06/28/2016] [Indexed: 12/26/2022] Open
Abstract
Amphetamine withdrawal increases anxiety and stress sensitivity related to blunted ventral hippocampus (vHipp) and enhances the central nucleus of the amygdala (CeA) serotonin responses. Extracellular serotonin levels are regulated by the serotonin transporter (SERT) and organic cation transporter 3 (OCT3), and vHipp OCT3 expression is enhanced during 24 hours of amphetamine withdrawal, while SERT expression is unaltered. Here, we tested whether OCT3 and SERT expression in the CeA is also affected during acute withdrawal to explain opposing regional alterations in limbic serotonergic neurotransmission and if respective changes continued with two weeks of withdrawal. We also determined whether changes in transporter expression were confined to these regions. Male rats received amphetamine or saline for two weeks followed by 24 hours or two weeks of withdrawal, with transporter expression measured using Western immunoblot. OCT3 and SERT expression increased in the CeA at both withdrawal timepoints. In the vHipp, OCT3 expression increased only at 24 hours of withdrawal, with an equivalent pattern seen in the dorsomedial hypothalamus. No changes were evident in any other regions sampled. These regionally specific changes in limbic OCT3 and SERT expression may partially contribute to the serotonergic imbalance and negative affect during amphetamine withdrawal.
Collapse
Affiliation(s)
- Rajeshwari R. Solanki
- Division of Basic Biomedical Sciences, Sanford School of Medicine, Center for Brain and Behavior Research, University of South Dakota, Vermillion, SD, USA
| | - Jamie L. Scholl
- Division of Basic Biomedical Sciences, Sanford School of Medicine, Center for Brain and Behavior Research, University of South Dakota, Vermillion, SD, USA
| | - Michael J. Watt
- Division of Basic Biomedical Sciences, Sanford School of Medicine, Center for Brain and Behavior Research, University of South Dakota, Vermillion, SD, USA
| | - Kenneth J. Renner
- Biology Department, Center for Brain and Behavior Research, University of South Dakota, Vermillion, SD, USA
| | - Gina L. Forster
- Division of Basic Biomedical Sciences, Sanford School of Medicine, Center for Brain and Behavior Research, University of South Dakota, Vermillion, SD, USA
| |
Collapse
|
36
|
Zestos AG, Mikelman SR, Kennedy RT, Gnegy ME. PKCβ Inhibitors Attenuate Amphetamine-Stimulated Dopamine Efflux. ACS Chem Neurosci 2016; 7:757-66. [PMID: 26996926 DOI: 10.1021/acschemneuro.6b00028] [Citation(s) in RCA: 31] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/16/2022] Open
Abstract
Amphetamine abuse afflicts over 13 million people, and there is currently no universally accepted treatment for amphetamine addiction. Amphetamine serves as a substrate for the dopamine transporter and reverses the transporter to cause an increase in extracellular dopamine. Activation of the beta subunit of protein kinase C (PKCβ) enhances extracellular dopamine in the presence of amphetamine by facilitating the reverse transport of dopamine and internalizing the D2 autoreceptor. We previously demonstrated that PKCβ inhibitors block amphetamine-stimulated dopamine efflux in synaptosomes from rat striatum in vitro. In this study, we utilized in vivo microdialysis in live, behaving rats to assess the effect of the PKCβ inhibitors, enzastaurin and ruboxistaurin, on amphetamine-stimulated locomotion and increases in monoamines and their metabolites. A 30 min perfusion of the nucleus accumbens core with 1 μM enzastaurin or 1 μM ruboxistaurin reduced efflux of dopamine and its metabolite 3-methoxytyramine induced by amphetamine by approximately 50%. The inhibitors also significantly reduced amphetamine-stimulated extracellular levels of norepinephrine. The stimulation of locomotor behavior by amphetamine, measured simultaneously with the analytes, was comparably reduced by the PKCβ inhibitors. Using a stable isotope label retrodialysis procedure, we determined that ruboxistaurin had no effect on basal levels of dopamine, norepinephrine, glutamate, or GABA. In addition, normal uptake function through the dopamine transporter was unaltered by the PKCβ inhibitors, as measured in rat synaptosomes. Our results support the utility of using PKCβ inhibitors to reduce the effects of amphetamine.
Collapse
Affiliation(s)
- Alexander G. Zestos
- Department
of Pharmacology, University of Michigan, 2301 MSRB III, 1150 W. Medical Center
Drive, Ann Arbor, Michigan 48109-5632, United States
- Department
of Chemistry, University of Michigan, 9300 North University Avenue, Ann Arbor, Michigan 48105, United States
| | - Sarah R. Mikelman
- Department
of Pharmacology, University of Michigan, 2301 MSRB III, 1150 W. Medical Center
Drive, Ann Arbor, Michigan 48109-5632, United States
| | - Robert T. Kennedy
- Department
of Pharmacology, University of Michigan, 2301 MSRB III, 1150 W. Medical Center
Drive, Ann Arbor, Michigan 48109-5632, United States
- Department
of Chemistry, University of Michigan, 9300 North University Avenue, Ann Arbor, Michigan 48105, United States
| | - Margaret E. Gnegy
- Department
of Pharmacology, University of Michigan, 2301 MSRB III, 1150 W. Medical Center
Drive, Ann Arbor, Michigan 48109-5632, United States
| |
Collapse
|
37
|
Tracer Flux Measurements to Study Outward Transport by Monoamine Neurotransmitter Transporters. NEUROMETHODS 2016. [DOI: 10.1007/978-1-4939-3765-3_2] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/11/2023]
|
38
|
Delgermurun D, Ito S, Ohta T, Yamaguchi S, Otsuguro KI. Endogenous 5-HT outflow from chicken aorta by 5-HT uptake inhibitors and amphetamine derivatives. J Vet Med Sci 2015; 78:71-6. [PMID: 26321443 PMCID: PMC4751119 DOI: 10.1292/jvms.15-0146] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/24/2022] Open
Abstract
Chemoreceptor cells aggregating in clusters in the chicken thoracic aorta
contain 5-hydroxytryptamine (5-HT) and have voltage-dependent ion channels and nicotinic
acetylcholine receptors, which are characteristics typically associated with neurons. The
aim of the present study was to investigate the effects of 5-HT uptake inhibitors,
fluvoxamine, fluoxetine and clomipramine (CLM), and amphetamine derivatives,
p-chloroamphetamine (PCA) and methamphetamine (MET), on endogenous 5-HT
outflow from the isolated chick thoracic aorta in vitro. 5-HT was
measured by using a HPLC system with electrochemical detection. The amphetamine
derivatives and 5-HT uptake inhibitors caused concentration-dependent increases in
endogenous 5-HT outflow. PCA was about ten times more effective in eliciting 5-HT outflow
than MET. The 5-HT uptake inhibitors examined had similar potency for 5-HT outflow. PCA
and CLM increased 5-HT outflow in a temperature-dependent manner. The outflow of 5-HT
induced by PCA or 5-HT uptake inhibitors was independent of extracellular Ca2+
concentration. The 5-HT outflow induced by CLM, but not that by PCA, was dependent on the
extracellular NaCl concentration. These results suggest that the 5-HT uptake system of
5-HT-containing chemoreceptor cells in the chicken thoracic aorta has characteristics
similar to those of 5-HT-containing neurons in the mammalian central nervous system
(CNS).
Collapse
Affiliation(s)
- Dugar Delgermurun
- Laboratory of Pharmacology, Department of Biomedical Sciences, Graduate School of Veterinary Medicine, Hokkaido University, Sapporo 060-0818, Japan
| | | | | | | | | |
Collapse
|
39
|
Sitte HH, Schütz GJ, Freissmuth M. Cooperativity between individual transporter protomers: new data fuelling old complexes. J Neurochem 2015; 133:163-6. [PMID: 25772534 DOI: 10.1111/jnc.13086] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/26/2015] [Accepted: 03/02/2015] [Indexed: 01/01/2023]
Abstract
Neurotransmitter transporters are arranged in an oligomeric quaternary structure as evidenced by crosslinking or fluorescence resonance energy transfer (FRET)-microscopy. In a study by Zhen and colleagues highlighted by this Editorial in the current issue of Journal of Neurochemistry, the combination of mutant and wild-type dopamine transporter (DAT) has been used to establish the cooperation between transporter protomers; the DAT mutant version has an altered affinity for the radiolabelled inhibitor [³H]CFT. Zhen and colleagues predict how saturation-binding curves ought to look, if the two binding sites (i.e. of the wild type and the mutant DAT) operated independently. The results are clear-cut: the experimental observations are inconsistent with curves obtained by mixing independent binding sites. Thus, by definition, the binding sites cooperate. Read the full article 'Dopamine transporter oligomerization: impact of combining protomers with differential cocaine analog binding affinities' on page 167.
Collapse
Affiliation(s)
- Harald H Sitte
- Center for Physiology and Pharmacology, Institute of Pharmacology, Medical University Vienna, Vienna, Austria
| | | | | |
Collapse
|
40
|
Han TK, Proctor WR, Costales CL, Cai H, Everett RS, Thakker DR. Four cation-selective transporters contribute to apical uptake and accumulation of metformin in Caco-2 cell monolayers. J Pharmacol Exp Ther 2015; 352:519-28. [PMID: 25563903 PMCID: PMC4352590 DOI: 10.1124/jpet.114.220350] [Citation(s) in RCA: 88] [Impact Index Per Article: 8.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/07/2014] [Accepted: 01/05/2015] [Indexed: 01/11/2023] Open
Abstract
Metformin is the frontline therapy for type II diabetes mellitus. The oral bioavailability of metformin is unexpectedly high, between 40 and 60%, given its hydrophilicity and positive charge at all physiologic pH values. Previous studies in Caco-2 cell monolayers, a cellular model of the human intestinal epithelium, showed that during absorptive transport metformin is taken up into the cells via transporters in the apical (AP) membrane; however, predominant transport to the basolateral (BL) side occurs via the paracellular route because intracellular metformin cannot egress across the BL membrane. Furthermore, these studies have suggested that the AP transporters can contribute to intestinal accumulation and absorption of metformin. Transporter-specific inhibitors as well as a novel approach involving a cocktail of transporter inhibitors with overlapping selectivity were used to identify the AP transporters that mediate metformin uptake in Caco-2 cell monolayers; furthermore, the relative contributions of these transporters in metformin AP uptake were also determined. The organic cation transporter 1, plasma membrane monoamine transporter (PMAT), serotonin reuptake transporter, and choline high-affinity transporter contributed to approximately 25%, 20%, 20%, and 15%, respectively, of the AP uptake of metformin. PMAT-knockdown Caco-2 cells were constructed to confirm the contribution of PMAT in metformin AP uptake because a PMAT-selective inhibitor is not available. The identification of four intestinal transporters that contribute to AP uptake and potentially intestinal absorption of metformin is a significant novel finding that can influence our understanding of metformin pharmacology and intestinal drug-drug interactions involving this highly prescribed drug.
Collapse
Affiliation(s)
- Tianxiang Kevin Han
- Division of Molecular Pharmaceutics (T.H., W.R.P., C.L.C.) and Division of Pharmacotherapy and Experimental Therapeutics (H.C., R.S.E., D.R.T.), UNC Eshelman School of Pharmacy, The University of North Carolina at Chapel Hill, Chapel Hill, North Carolina
| | - William R Proctor
- Division of Molecular Pharmaceutics (T.H., W.R.P., C.L.C.) and Division of Pharmacotherapy and Experimental Therapeutics (H.C., R.S.E., D.R.T.), UNC Eshelman School of Pharmacy, The University of North Carolina at Chapel Hill, Chapel Hill, North Carolina
| | - Chester L Costales
- Division of Molecular Pharmaceutics (T.H., W.R.P., C.L.C.) and Division of Pharmacotherapy and Experimental Therapeutics (H.C., R.S.E., D.R.T.), UNC Eshelman School of Pharmacy, The University of North Carolina at Chapel Hill, Chapel Hill, North Carolina
| | - Hao Cai
- Division of Molecular Pharmaceutics (T.H., W.R.P., C.L.C.) and Division of Pharmacotherapy and Experimental Therapeutics (H.C., R.S.E., D.R.T.), UNC Eshelman School of Pharmacy, The University of North Carolina at Chapel Hill, Chapel Hill, North Carolina
| | - Ruth S Everett
- Division of Molecular Pharmaceutics (T.H., W.R.P., C.L.C.) and Division of Pharmacotherapy and Experimental Therapeutics (H.C., R.S.E., D.R.T.), UNC Eshelman School of Pharmacy, The University of North Carolina at Chapel Hill, Chapel Hill, North Carolina
| | - Dhiren R Thakker
- Division of Molecular Pharmaceutics (T.H., W.R.P., C.L.C.) and Division of Pharmacotherapy and Experimental Therapeutics (H.C., R.S.E., D.R.T.), UNC Eshelman School of Pharmacy, The University of North Carolina at Chapel Hill, Chapel Hill, North Carolina
| |
Collapse
|
41
|
Fischer J, Kleinau G, Müller A, Kühnen P, Zwanziger D, Kinne A, Rehders M, Moeller LC, Führer D, Grüters A, Krude H, Brix K, Biebermann H. Modulation of monocarboxylate transporter 8 oligomerization by specific pathogenic mutations. J Mol Endocrinol 2015; 54:39-50. [PMID: 25527620 DOI: 10.1530/jme-14-0272] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/08/2022]
Abstract
The monocarboxylate transporter 8 (MCT8) is a member of the major facilitator superfamily (MFS). These membrane-spanning proteins facilitate translocation of a variety of substrates, MCT8 specifically transports iodothyronines. Mutations in MCT8 are the underlying cause of severe X-linked psychomotor retardation. At the molecular level, such mutations led to deficiencies in substrate translocation due to reduced cell-surface expression, impaired substrate binding, or decreased substrate translocation capabilities. However, the causal relationships between genotypes, molecular features of mutated MCT8, and patient characteristics have not yet been comprehensively deciphered. We investigated the relationship between pathogenic mutants of MCT8 and their capacity to form dimers (presumably oligomeric structures) as a potential regulatory parameter of the transport function of MCT8. Fourteen pathogenic variants of MCT8 were investigated in vitro with respect to their capacity to form oligomers. Particular mutations close to the substrate translocation channel (S194F, A224T, L434W, and R445C) were found to inhibit dimerization of MCT8. This finding is in contrast to those for other transporters or transmembrane proteins, in which substitutions predominantly at the outer-surface inhibit oligomerization. Moreover, specific mutations of MCT8 located in transmembrane helix 2 (del230F, V235M, and ins236V) increased the capacity of MCT8 variants to dimerize. We analyzed the localization of MCT8 dimers in a cellular context, demonstrating differences in MCT8 dimer formation and distribution. In summary, our results add a new link between the functions (substrate transport) and protein organization (dimerization) of MCT8, and might be of relevance for other members of the MFS. Finally, the findings are discussed in relationship to functional data combined with structural-mechanistical insights into MCT8.
Collapse
Affiliation(s)
- Jana Fischer
- Institut für Experimentelle Pädiatrische EndokrinologieCharité-Universitätsmedizin Berlin, Augustenburger Platz 1, 13353 Berlin, GermanyLeibniz-Institut für Molekulare PharmakologieBerlin, GermanyUniversitätsklinikum EssenKlinik für Endokrinologie und Stoffwechselerkrankungen, Essen, GermanyJacobs University BremenBremen, Germany
| | - Gunnar Kleinau
- Institut für Experimentelle Pädiatrische EndokrinologieCharité-Universitätsmedizin Berlin, Augustenburger Platz 1, 13353 Berlin, GermanyLeibniz-Institut für Molekulare PharmakologieBerlin, GermanyUniversitätsklinikum EssenKlinik für Endokrinologie und Stoffwechselerkrankungen, Essen, GermanyJacobs University BremenBremen, Germany
| | - Anne Müller
- Institut für Experimentelle Pädiatrische EndokrinologieCharité-Universitätsmedizin Berlin, Augustenburger Platz 1, 13353 Berlin, GermanyLeibniz-Institut für Molekulare PharmakologieBerlin, GermanyUniversitätsklinikum EssenKlinik für Endokrinologie und Stoffwechselerkrankungen, Essen, GermanyJacobs University BremenBremen, Germany
| | - Peter Kühnen
- Institut für Experimentelle Pädiatrische EndokrinologieCharité-Universitätsmedizin Berlin, Augustenburger Platz 1, 13353 Berlin, GermanyLeibniz-Institut für Molekulare PharmakologieBerlin, GermanyUniversitätsklinikum EssenKlinik für Endokrinologie und Stoffwechselerkrankungen, Essen, GermanyJacobs University BremenBremen, Germany
| | - Denise Zwanziger
- Institut für Experimentelle Pädiatrische EndokrinologieCharité-Universitätsmedizin Berlin, Augustenburger Platz 1, 13353 Berlin, GermanyLeibniz-Institut für Molekulare PharmakologieBerlin, GermanyUniversitätsklinikum EssenKlinik für Endokrinologie und Stoffwechselerkrankungen, Essen, GermanyJacobs University BremenBremen, Germany
| | - Anita Kinne
- Institut für Experimentelle Pädiatrische EndokrinologieCharité-Universitätsmedizin Berlin, Augustenburger Platz 1, 13353 Berlin, GermanyLeibniz-Institut für Molekulare PharmakologieBerlin, GermanyUniversitätsklinikum EssenKlinik für Endokrinologie und Stoffwechselerkrankungen, Essen, GermanyJacobs University BremenBremen, Germany
| | - Maren Rehders
- Institut für Experimentelle Pädiatrische EndokrinologieCharité-Universitätsmedizin Berlin, Augustenburger Platz 1, 13353 Berlin, GermanyLeibniz-Institut für Molekulare PharmakologieBerlin, GermanyUniversitätsklinikum EssenKlinik für Endokrinologie und Stoffwechselerkrankungen, Essen, GermanyJacobs University BremenBremen, Germany
| | - Lars C Moeller
- Institut für Experimentelle Pädiatrische EndokrinologieCharité-Universitätsmedizin Berlin, Augustenburger Platz 1, 13353 Berlin, GermanyLeibniz-Institut für Molekulare PharmakologieBerlin, GermanyUniversitätsklinikum EssenKlinik für Endokrinologie und Stoffwechselerkrankungen, Essen, GermanyJacobs University BremenBremen, Germany
| | - Dagmar Führer
- Institut für Experimentelle Pädiatrische EndokrinologieCharité-Universitätsmedizin Berlin, Augustenburger Platz 1, 13353 Berlin, GermanyLeibniz-Institut für Molekulare PharmakologieBerlin, GermanyUniversitätsklinikum EssenKlinik für Endokrinologie und Stoffwechselerkrankungen, Essen, GermanyJacobs University BremenBremen, Germany
| | - Annette Grüters
- Institut für Experimentelle Pädiatrische EndokrinologieCharité-Universitätsmedizin Berlin, Augustenburger Platz 1, 13353 Berlin, GermanyLeibniz-Institut für Molekulare PharmakologieBerlin, GermanyUniversitätsklinikum EssenKlinik für Endokrinologie und Stoffwechselerkrankungen, Essen, GermanyJacobs University BremenBremen, Germany
| | - Heiko Krude
- Institut für Experimentelle Pädiatrische EndokrinologieCharité-Universitätsmedizin Berlin, Augustenburger Platz 1, 13353 Berlin, GermanyLeibniz-Institut für Molekulare PharmakologieBerlin, GermanyUniversitätsklinikum EssenKlinik für Endokrinologie und Stoffwechselerkrankungen, Essen, GermanyJacobs University BremenBremen, Germany
| | - Klaudia Brix
- Institut für Experimentelle Pädiatrische EndokrinologieCharité-Universitätsmedizin Berlin, Augustenburger Platz 1, 13353 Berlin, GermanyLeibniz-Institut für Molekulare PharmakologieBerlin, GermanyUniversitätsklinikum EssenKlinik für Endokrinologie und Stoffwechselerkrankungen, Essen, GermanyJacobs University BremenBremen, Germany
| | - Heike Biebermann
- Institut für Experimentelle Pädiatrische EndokrinologieCharité-Universitätsmedizin Berlin, Augustenburger Platz 1, 13353 Berlin, GermanyLeibniz-Institut für Molekulare PharmakologieBerlin, GermanyUniversitätsklinikum EssenKlinik für Endokrinologie und Stoffwechselerkrankungen, Essen, GermanyJacobs University BremenBremen, Germany
| |
Collapse
|
42
|
Sitte HH, Freissmuth M. Amphetamines, new psychoactive drugs and the monoamine transporter cycle. Trends Pharmacol Sci 2014; 36:41-50. [PMID: 25542076 PMCID: PMC4502921 DOI: 10.1016/j.tips.2014.11.006] [Citation(s) in RCA: 192] [Impact Index Per Article: 17.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/29/2014] [Revised: 11/25/2014] [Accepted: 11/25/2014] [Indexed: 01/05/2023]
Abstract
In monoaminergic neurons, the vesicular transporters and the plasma membrane transporters operate in a relay. Amphetamine and its congeners target this relay to elicit their actions: most amphetamines are substrates, which pervert the relay to elicit efflux of monoamines into the synaptic cleft. However, some amphetamines act as transporter inhibitors. Both compound classes elicit profound psychostimulant effects, which render them liable to recreational abuse. Currently, a surge of new psychoactive substances occurs on a global scale. Chemists bypass drug bans by ingenuous structural variations, resulting in a rich pharmacology. A credible transport model must account for their distinct mode of action and link this to subtle differences in activity and undesired, potentially deleterious effects.
Collapse
Affiliation(s)
- Harald H Sitte
- Institute of Pharmacology, Center for Physiology and Pharmacology, Medical University Vienna, Waehringerstrasse 13A, 1090 Vienna, Austria; Center for Addiction Research and Science (AddRess), Medical University Vienna, Waehringerstrasse 13A, 1090 Vienna, Austria.
| | - Michael Freissmuth
- Institute of Pharmacology, Center for Physiology and Pharmacology, Medical University Vienna, Waehringerstrasse 13A, 1090 Vienna, Austria
| |
Collapse
|
43
|
Sandtner W, Schmid D, Schicker K, Gerstbrein K, Koenig X, Mayer FP, Boehm S, Freissmuth M, Sitte HH. A quantitative model of amphetamine action on the 5-HT transporter. Br J Pharmacol 2014; 171:1007-18. [PMID: 24251585 PMCID: PMC3925039 DOI: 10.1111/bph.12520] [Citation(s) in RCA: 29] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/04/2013] [Revised: 10/30/2013] [Accepted: 11/05/2013] [Indexed: 10/27/2022] Open
Abstract
BACKGROUND AND PURPOSE Amphetamines bind to the plasmalemmal transporters for the monoamines dopamine (DAT), noradrenaline (NET) and 5-HT (SERT); influx of amphetamine leads to efflux of substrates. Various models have been proposed to account for this amphetamine-induced reverse transport in mechanistic terms. A most notable example is the molecular stent hypothesis, which posits a special amphetamine-induced conformation that is not likely in alternative access models of transport. The current study was designed to evaluate the explanatory power of these models and the molecular stent hypothesis. EXPERIMENTAL APPROACH Xenopus laevis oocytes and HEK293 cells expressing human (h) SERT were voltage-clamped and exposed to 5-HT, p-chloroamphetamine (pCA) or methylenedioxyamphetamine (MDMA). KEY RESULTS In contrast to the currents induced by 5-HT, pCA-triggered currents through SERT decayed slowly in Xenopus laevis oocytes once the agonist was removed (consistent with the molecular stent hypothesis). However, when SERT was expressed in HEK293 cells, currents induced by 3 or 100 μM pCA decayed 10 or 100 times faster, respectively, after pCA removal. CONCLUSIONS AND IMPLICATIONS This discrepancy in decay rates is inconsistent with the molecular stent hypothesis. In contrast, a multistate version of the alternative access model accounts for all the observations and reproduces the kinetic parameters extracted from the electrophysiological recordings. A crucial feature that explains the action of amphetamines is their lipophilic nature, which allows for rapid diffusion through the membrane.
Collapse
Affiliation(s)
- Walter Sandtner
- Center of Physiology and Pharmacology, Medical University Vienna, Vienna, Austria
| | | | | | | | | | | | | | | | | |
Collapse
|
44
|
Sørensen L, Strømgaard K, Kristensen AS. Characterization of intracellular regions in the human serotonin transporter for phosphorylation sites. ACS Chem Biol 2014; 9:935-44. [PMID: 24450286 DOI: 10.1021/cb4007198] [Citation(s) in RCA: 20] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/28/2022]
Abstract
In the central nervous system, synaptic levels of the monoamine neurotransmitter serotonin are mainly controlled by the serotonin transporter (SERT), and drugs used in the treatment of various psychiatric diseases have SERT as primary target. SERT is a phosphoprotein that undergoes phosphorylation/dephosphorylation during transporter regulation by multiple pathways. In particular, activation and/or inhibition of kinases including PKC, PKG, p38MAPK, and CaMKII modulate SERT function and trafficking. The molecular mechanisms by which kinase activity is linked to SERT regulation are poorly understood, including the identity of specific phosphorylated residues. To elucidate SERT phosphorylation sites, we have generated peptides corresponding to the entire intracellular region of human SERT and performed in vitro phosphorylation assays with a panel of kinases suggested to be involved in SERT regulation or for which canonical phosphorylation sites are predicted. Peptide analysis by liquid chromatography-tandem mass spectrometry (LC-MS/MS) was used to identify and quantify site-specific phosphorylation. Five residues located in the N- and C-termini and in intracellular loop 1 and 2 were identified as phosphorylation sites; Ser149, Ser277, and Thr603 for PKC, Ser13 for CaMKII, and Thr616 for p38MAPK. Possible regulatory roles of these potential phosphoacceptors for SERT function and surface expression were investigated using phospho-mimicking and phosphodeficient mutations, coexpression of constitutively active kinases and pharmacological kinase induction in a heterologous expression system. Our results suggest that Ser277 is involved in an initial phase of PKC-mediated down-regulation of SERT. The five identified sites can guide future studies of direct links between SERT phosphorylation and regulatory processes.
Collapse
Affiliation(s)
- Lena Sørensen
- Department of Drug Design
and Pharmacology, University of Copenhagen, Universitetsparken 2, DK-2100 Copenhagen, Denmark
| | - Kristian Strømgaard
- Department of Drug Design
and Pharmacology, University of Copenhagen, Universitetsparken 2, DK-2100 Copenhagen, Denmark
| | - Anders S. Kristensen
- Department of Drug Design
and Pharmacology, University of Copenhagen, Universitetsparken 2, DK-2100 Copenhagen, Denmark
| |
Collapse
|
45
|
Torres B, Ruoho AE. N-terminus regulation of VMAT2 mediates methamphetamine-stimulated efflux. Neuroscience 2013; 259:194-202. [PMID: 24321511 DOI: 10.1016/j.neuroscience.2013.11.059] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/11/2013] [Revised: 10/28/2013] [Accepted: 11/28/2013] [Indexed: 10/25/2022]
Abstract
The 20 amino acid (AA) N-terminus of the vesicular monoamine transporter 2 (VMAT2) was examined as a regulator of VMAT2 function. Removal of the first 16 or 19 AAs of the N-terminus resulted in a molecule with reduced ability to sequester [(3)H]-5HT. A glutathione-S-transferase-construct of the N-terminus underwent phosphorylation in the presence of PKC at serines 15 and 18. These putative phosphorylation sites were examined for effects on function. Phospho-mimetic substitution of serines 15 and 18 with aspartate in the full-length VMAT2 resulted in reduced [(3)H]-5HT sequestration and reduced methamphetamine (METH)-stimulated efflux of preloaded [(3)H]-5HT. In contrast, mutation of serines 15 and 18 to alanines maintained intact net substrate sequestration but eliminated METH-stimulated efflux of pre-accumulated [(3)H]-5HT. In summary, these data suggest a model in which the VMAT2 N-terminus regulates monoamine sequestration.
Collapse
Affiliation(s)
- B Torres
- University of Wisconsin, Madison, Department of Neuroscience, 1300 University Avenue, Madison, WI 53706, USA
| | - A E Ruoho
- University of Wisconsin, Madison, Department of Neuroscience, 1300 University Avenue, Madison, WI 53706, USA.
| |
Collapse
|
46
|
Amphetamine actions at the serotonin transporter rely on the availability of phosphatidylinositol-4,5-bisphosphate. Proc Natl Acad Sci U S A 2013; 110:11642-7. [PMID: 23798435 DOI: 10.1073/pnas.1220552110] [Citation(s) in RCA: 71] [Impact Index Per Article: 5.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/26/2022] Open
Abstract
Nerve functions require phosphatidylinositol-4,5-bisphosphate (PIP2) that binds to ion channels, thereby controlling their gating. Channel properties are also attributed to serotonin transporters (SERTs); however, SERT regulation by PIP2 has not been reported. SERTs control neurotransmission by removing serotonin from the extracellular space. An increase in extracellular serotonin results from transporter-mediated efflux triggered by amphetamine-like psychostimulants. Herein, we altered the abundance of PIP2 by activating phospholipase-C (PLC), using a scavenging peptide, and inhibiting PIP2-synthesis. We tested the effects of the verified scarcity of PIP2 on amphetamine-triggered SERT functions in human cells. We observed an interaction between SERT and PIP2 in pull-down assays. On decreased PIP2 availability, amphetamine-evoked currents were markedly reduced compared with controls, as was amphetamine-induced efflux. Signaling downstream of PLC was excluded as a cause for these effects. A reduction of substrate efflux due to PLC activation was also found with recombinant noradrenaline transporters and in rat hippocampal slices. Transmitter uptake was not affected by PIP2 reduction. Moreover, SERT was revealed to have a positively charged binding site for PIP2. Mutation of the latter resulted in a loss of amphetamine-induced SERT-mediated efflux and currents, as well as a lack of PIP2-dependent effects. Substrate uptake and surface expression were comparable between mutant and WT SERTs. These findings demonstrate that PIP2 binding to monoamine transporters is a prerequisite for amphetamine actions without being a requirement for neurotransmitter uptake. These results open the way to target amphetamine-induced SERT-dependent actions independently of normal SERT function and thus to treat psychostimulant addiction.
Collapse
|
47
|
Seddik A, Holy M, Weissensteiner R, Zdrazil B, Sitte HH, Ecker GF. Probing the Selectivity of Monoamine Transporter Substrates by Means of Molecular Modeling. Mol Inform 2013; 32:409-413. [PMID: 23956802 PMCID: PMC3743209 DOI: 10.1002/minf.201300013] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/25/2013] [Accepted: 03/28/2013] [Indexed: 11/25/2022]
Affiliation(s)
- Amir Seddik
- University of Vienna, Department of Medicinal Chemistry, Pharmacoinformatics Research Group Vienna, Austria
| | | | | | | | | | | |
Collapse
|
48
|
Rosenauer R, Luf A, Holy M, Freissmuth M, Schmid R, Sitte HH. A combined approach using transporter-flux assays and mass spectrometry to examine psychostimulant street drugs of unknown content. ACS Chem Neurosci 2013; 4:182-90. [PMID: 23336057 PMCID: PMC3547486 DOI: 10.1021/cn3001763] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/09/2012] [Accepted: 12/03/2012] [Indexed: 11/09/2022] Open
Abstract
![]()
The illicit consumption of psychoactive compounds may
cause short and long-term health problems and addiction. This is also
true for amphetamines and cocaine, which target monoamine transporters.
In the recent past, an increasing number of new compounds with amphetamine-like
structure such as mephedrone or 3,4-methylenedioxypyrovalerone (MDPV)
entered the market of illicit drugs. Subtle structural changes circumvent
legal restrictions placed on the parent compound. These novel drugs
are effectively marketed “designer drugs” (also called
“research chemicals”) without any knowledge of the underlying
pharmacology, the potential harm or a registration of the manufacturing
process. Accordingly new entrants and their byproducts are identified
postmarketing by chemical analysis and their pharmacological properties
inferred by comparison to compounds of known structure. However, such
a heuristic approach fails, if the structures diverge substantially
from a known derivative. In addition, the understanding of structure–activity
relations is too rudimentary to predict detailed pharmacological activity.
Here, we tested a combined approach by examining the composition of
street drugs using mass spectrometry and by assessing the functional
activity of their constituents at the neuronal transporters for dopamine,
serotonin, and norepinephrine. We show that this approach is superior
to mere chemical analysis in recognizing novel and potentially harmful
street drugs.
Collapse
Affiliation(s)
- Rudolf Rosenauer
- Institute of Pharmacology, Center for Physiology
and Pharmacology, Medical University of Vienna, Waehringer Strasse 13a, A-1090 Vienna, Austria
| | - Anton Luf
- Clinical Department of Laboratory Medicine, Medical University of Vienna, Waehringer Guertel 18-20,
A-1090 Vienna, Austria
| | - Marion Holy
- Institute of Pharmacology, Center for Physiology
and Pharmacology, Medical University of Vienna, Waehringer Strasse 13a, A-1090 Vienna, Austria
| | - Michael Freissmuth
- Institute of Pharmacology, Center for Physiology
and Pharmacology, Medical University of Vienna, Waehringer Strasse 13a, A-1090 Vienna, Austria
| | - Rainer Schmid
- Clinical Department of Laboratory Medicine, Medical University of Vienna, Waehringer Guertel 18-20,
A-1090 Vienna, Austria
| | - Harald H. Sitte
- Institute of Pharmacology, Center for Physiology
and Pharmacology, Medical University of Vienna, Waehringer Strasse 13a, A-1090 Vienna, Austria
| |
Collapse
|
49
|
Sucic S, Koban F, El-Kasaby A, Kudlacek O, Stockner T, Sitte HH, Freissmuth M. Switching the clientele: a lysine residing in the C terminus of the serotonin transporter specifies its preference for the coat protein complex II component SEC24C. J Biol Chem 2013; 288:5330-41. [PMID: 23288844 PMCID: PMC3581386 DOI: 10.1074/jbc.m112.408237] [Citation(s) in RCA: 45] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/24/2022] Open
Abstract
The serotonin transporter (SERT) maintains serotonergic neurotransmission via rapid reuptake of serotonin from the synaptic cleft. SERT relies exclusively on the coat protein complex II component SEC24C for endoplasmic reticulum (ER) export. The closely related transporters for noradrenaline and dopamine depend on SEC24D. Here, we show that discrimination between SEC24C and SEC24D is specified by the residue at position +2 downstream from the ER export motif (607RI608 in SERT). Substituting Lys610 with tyrosine, the corresponding residue found in the noradrenaline and dopamine transporters, switched the SEC24 isoform preference: SERT-K610Y relied solely on SEC24D to reach the cell surface. This analysis was extended to other SLC6 (solute carrier 6) transporter family members: siRNA-dependent depletion of SEC24C, but not of SEC24D, reduced surface levels of the glycine transporter-1a, the betaine/GABA transporter and the GABA transporter-4. Experiments with dominant negative versions of SEC24C and SEC24D recapitulated these findings. We also verified that the presence of two ER export motifs (in concatemers of SERT and GABA transporter-1) supported recruitment of both SEC24C and SEC24D. To the best of our knowledge, this is the first report to document a change in SEC24 specificity by mutation of a single residue in the client protein. Our observations allowed for deducing a rule for SLC6 family members: a hydrophobic residue (Tyr or Val) in the +2 position specifies interaction with SEC24D, and a hydrophilic residue (Lys, Asn, or Gln) recruits SEC24C. Variations in SEC24C are linked to neuropsychiatric disorders. The present findings provide a mechanistic explanation. Variations in SEC24C may translate into distinct surface levels of neurotransmitter transporters.
Collapse
Affiliation(s)
- Sonja Sucic
- Institute of Pharmacology, Center of Physiology and Pharmacology, Medical University of Vienna, A-1090 Vienna, Austria
| | | | | | | | | | | | | |
Collapse
|
50
|
Barr JL, Scholl JL, Solanki RR, Watt MJ, Lowry CA, Renner KJ, Forster GL. Influence of chronic amphetamine treatment and acute withdrawal on serotonin synthesis and clearance mechanisms in the rat ventral hippocampus. Eur J Neurosci 2012; 37:479-90. [PMID: 23157166 DOI: 10.1111/ejn.12050] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/02/2012] [Revised: 10/01/2012] [Accepted: 10/02/2012] [Indexed: 01/11/2023]
Abstract
Amphetamine withdrawal in both humans and rats is associated with increased anxiety states, which are thought to contribute to drug relapse. Serotonin in the ventral hippocampus mediates affective behaviors, and reduced serotonin levels in this region are observed in rat models of high anxiety, including during withdrawal from chronic amphetamine. This goal of this study was to understand the mechanisms by which reduced ventral hippocampus serotonergic neurotransmission occurs during amphetamine withdrawal. Serotonin synthesis (assessed by accumulation of serotonin precursor as a measure of the capacity of in vivo tryptophan hydroxylase activity), expression of serotonergic transporters, and in vivo serotonergic clearance using in vivo microdialysis were assessed in the ventral hippocampus in adult male Sprague Dawley rats at 24 h withdrawal from chronic amphetamine. Overall, results showed that diminished extracellular serotonin at 24 h withdrawal from chronic amphetamine was not accompanied by a change in capacity for serotonin synthesis (in vivo tryptophan hydroxylase activity), or serotonin transporter expression or function in the ventral hippocampus, but instead was associated with increased expression and function of organic cation transporters (low-affinity, high-capacity serotonin transporters). These findings suggest that 24 h withdrawal from chronic amphetamine reduces the availability of extracellular serotonin in the ventral hippocampus by increasing organic cation transporter-mediated serotonin clearance, which may represent a future pharmacological target for reversing anxiety states during drug withdrawal.
Collapse
Affiliation(s)
- Jeffrey L Barr
- Neuroscience Group, Basic Biomedical Sciences, Sanford School of Medicine, University of South Dakota, Vermillion, SD, USA
| | | | | | | | | | | | | |
Collapse
|