1
|
Inagaki R, Kita S, Niwa N, Fukunaga K, Iwamoto T, Moriguchi S. Aberrant extracellular dopamine clearance in the prefrontal cortex exhibits ADHD-like behavior in NCX3 heterozygous mice. FEBS J 2025; 292:426-444. [PMID: 39624860 PMCID: PMC11734882 DOI: 10.1111/febs.17339] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/13/2024] [Revised: 06/24/2024] [Accepted: 11/19/2024] [Indexed: 01/16/2025]
Abstract
Attention-deficit/hyperactivity disorder (ADHD) is a neurodevelopmental disorder that involves dopaminergic dysfunction in the prefrontal cortex (PFC), manifesting hyperactivity, inattention, and cognitive deficits. However, the ADHD-associated candidate genes underlying dopaminergic neurotransmission alterations remain poorly defined. Here, we identified the abundant localization of sodium-calcium exchanger 3 (NCX3) levels in the dopaminergic neurons of the ventral tegmental area, a major source of dopaminergic innervation to the PFC. We confirmed that NCX3 knockdown in N27 cells caused aberrant dopamine influx through the strong interaction between calcium/calmodulin-dependent protein kinase II alpha and dopamine transporter. In addition, we assessed behavioral changes and underlying molecular properties in NCX3 heterozygous (NCX3+/-) mice. NCX3+/- mice exhibited hyperactivity, cognitive deficits, and social dysfunction which were alleviated by treating with methylphenidate. Furthermore, NCX3+/- mice displayed a persistent elevation of basal dopamine levels and decreased extracellular levels of dopamine triggered by social stimuli in the PFC of NCX3+/- mice. In agreement with the rise in extracellular dopamine levels in the PFC, NCX3+/- mice showed activation of dopamine D1 receptor signaling pathways in the PFC compared to wild-type mice. Thus, deficiency of NCX3 leads to impaired dopaminergic neurotransmission in the PFC, which likely accounts for the ADHD-like behavior in NCX3+/- mice.
Collapse
Affiliation(s)
- Ryo Inagaki
- Research Center for Pharmaceutical Development, Graduate School of Pharmaceutical SciencesTohoku UniversitySendaiJapan
| | - Satomi Kita
- Department of Pharmacology, Faculty of Pharmaceutical SciencesTokushima Bunri UniversityJapan
| | - Nozomu Niwa
- Research Center for Pharmaceutical Development, Graduate School of Pharmaceutical SciencesTohoku UniversitySendaiJapan
| | - Kohji Fukunaga
- Department of Pharmacology, Graduate School of Pharmaceutical SciencesTohoku UniversitySendaiJapan
| | - Takahiro Iwamoto
- Department of Pharmacology, Faculty of MedicineFukuoka UniversityJapan
| | - Shigeki Moriguchi
- Research Center for Pharmaceutical Development, Graduate School of Pharmaceutical SciencesTohoku UniversitySendaiJapan
| |
Collapse
|
2
|
Dehury B, Mishra S, Panda S, Singh MK, Simha NL, Pati S. Structural Dynamics of Neutral Amino Acid Transporter SLC6A19 in Simple and Complex Lipid Bilayers. J Cell Biochem 2025; 126:e30693. [PMID: 39749651 PMCID: PMC11696832 DOI: 10.1002/jcb.30693] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/27/2024] [Revised: 11/24/2024] [Accepted: 12/04/2024] [Indexed: 01/04/2025]
Abstract
B0AT1 (SLC6A19) is a major sodium-coupled neutral amino acid transporter that relies on angiotensin converting enzyme 2 (ACE2) or collectrin for membrane trafficking. Despite its significant role in disorders associated with amino acid metabolism, there is a deficit of comprehensive structure-function understanding of B0AT1 in lipid environment. Herein, we have employed molecular dynamics (MD) simulations to explore the architectural characteristics of B0AT1 in two distinct environments: a simplified POPC bilayer and a complex lipid system replicating the native membrane composition. Notably, our B0AT1 analysis in terms of structural stability and regions of maximum flexibility shows consistency in both the systems with enhanced structural features in the case of complex lipid system. Our findings suggest that diacylglycerol phospholipids significantly alter the pore radius, hydrophobic index, and surface charge distribution of B0AT1, thereby affecting the flexibility of transmembrane helices TM7, TM12, and loop connecting TM7-TM8, crucial for ACE2-B0AT1 interaction. Pro41, Ser190, Arg214, Arg240, Ser413, Pro414, Cys463, and Val582 are among the most prominent lipid binding residues that might influence B0AT1 functionality. We also perceive notable lipid mediated deviation in the degree of tilt and loss of helicity in TM1 and TM6 which might affect the substrate binding sites S1 and S2 in B0AT1. Considerably, destabilization in the structure of B0AT1 in lipid environment was evident upon mutation in TM domain, associated with Hartnup disorder through various structure-based protein stability tools. Our two-tiered approach allowed us to validate the use of POPC as a baseline for initial analyses of SLC transporters. Altogether, our all-atoms MD study provides a platform for future investigations into the structure-function mechanism of B0AT1 in realistic lipid mimetic bilayers and offers a framework for developing new therapeutic agents targeting this transporter.
Collapse
Affiliation(s)
- Budheswar Dehury
- Department of Bioinformatics, Manipal School of Life SciencesManipal Academy of Higher EducationManipalIndia
- Bioinformatics Division I Microbiology DivisionICMR‐Regional Medical Research CentreBhubaneswarOdishaIndia
| | - Sarbani Mishra
- Bioinformatics Division I Microbiology DivisionICMR‐Regional Medical Research CentreBhubaneswarOdishaIndia
| | - Sunita Panda
- Bioinformatics Division I Microbiology DivisionICMR‐Regional Medical Research CentreBhubaneswarOdishaIndia
| | | | - Nischal L. Simha
- Department of Bioinformatics, Manipal School of Life SciencesManipal Academy of Higher EducationManipalIndia
| | - Sanghamitra Pati
- Bioinformatics Division I Microbiology DivisionICMR‐Regional Medical Research CentreBhubaneswarOdishaIndia
| |
Collapse
|
3
|
Mayer FP, Stewart A, Varman DR, Moritz AE, Foster JD, Owens AW, Areal LB, Gowrishankar R, Velez M, Wickham K, Phelps H, Katamish R, Rabil M, Jayanthi LD, Vaughan RA, Daws LC, Blakely RD, Ramamoorthy S. Kappa Opioid Receptor Antagonism Restores Phosphorylation, Trafficking and Behavior induced by a Disease Associated Dopamine Transporter Variant. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2023.05.03.539310. [PMID: 37205452 PMCID: PMC10187322 DOI: 10.1101/2023.05.03.539310] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/21/2023]
Abstract
Aberrant dopamine (DA) signaling is implicated in schizophrenia, bipolar disorder (BPD), autism spectrum disorder (ASD), substance use disorder, and attention-deficit/hyperactivity disorder (ADHD). Treatment of these disorders remains inadequate, as exemplified by the therapeutic use of d-amphetamine and methylphenidate for the treatment of ADHD, agents with high abuse liability. In search for an improved and non-addictive therapeutic approach for the treatment of DA-linked disorders, we utilized a preclinical mouse model expressing the human DA transporter (DAT) coding variant DAT Val559, previously identified in individuals with ADHD, ASD, or BPD. DAT Val559, like several other disease-associated variants of DAT, exhibits anomalous DA efflux (ADE) that can be blocked by d-amphetamine and methylphenidate. Kappa opioid receptors (KORs) are expressed by DA neurons and modulate DA release and clearance, suggesting that targeting KORs might also provide an alternative approach to normalizing DA-signaling disrupted by perturbed DAT function. Here we demonstrate that KOR stimulation leads to enhanced surface trafficking and phosphorylation of Thr53 in wildtype DAT, effects achieved constitutively by the Val559 mutant. Moreover, these effects can be rescued by KOR antagonism of DAT Val559 in ex vivo preparations. Importantly, KOR antagonism also corrected in vivo DA release as well as sex-dependent behavioral abnormalities observed in DAT Val559 mice. Given their low abuse liability, our studies with a construct valid model of human DA associated disorders reinforce considerations of KOR antagonism as a pharmacological strategy to treat DA associated brain disorders.
Collapse
Affiliation(s)
- Felix P. Mayer
- Department of Biomedical Science, Charles E. Schmidt College of Medicine, Florida Atlantic University, Jupiter, FL, USA
- Stiles-Nicholson Brain Institute, Florida Atlantic University, Jupiter, FL, USA
| | - Adele Stewart
- Department of Biomedical Science, Charles E. Schmidt College of Medicine, Florida Atlantic University, Jupiter, FL, USA
- Stiles-Nicholson Brain Institute, Florida Atlantic University, Jupiter, FL, USA
| | - Durairaj Ragu Varman
- Department of Pharmacology and Toxicology, Virginia Commonwealth University, Richmond, VA, USA
| | - Amy E. Moritz
- Department of Biomedical Sciences, University of North Dakota School of Medicine and Health Sciences, Grand Forks, ND, USA
| | - James D. Foster
- Department of Biomedical Sciences, University of North Dakota School of Medicine and Health Sciences, Grand Forks, ND, USA
| | - Anthony W. Owens
- Department of Cellular and Integrative Physiology, University of Texas Health Science Center at San Antonio, TX, USA
| | - Lorena B. Areal
- Department of Biomedical Science, Charles E. Schmidt College of Medicine, Florida Atlantic University, Jupiter, FL, USA
| | - Raajaram Gowrishankar
- Department of Biomedical Science, Charles E. Schmidt College of Medicine, Florida Atlantic University, Jupiter, FL, USA
| | - Michelle Velez
- Department of Biomedical Science, Charles E. Schmidt College of Medicine, Florida Atlantic University, Jupiter, FL, USA
| | - Kyria Wickham
- Department of Biomedical Science, Charles E. Schmidt College of Medicine, Florida Atlantic University, Jupiter, FL, USA
| | - Hannah Phelps
- Department of Biomedical Science, Charles E. Schmidt College of Medicine, Florida Atlantic University, Jupiter, FL, USA
| | - Rania Katamish
- Department of Biomedical Science, Charles E. Schmidt College of Medicine, Florida Atlantic University, Jupiter, FL, USA
| | - Maximilian Rabil
- Department of Biomedical Science, Charles E. Schmidt College of Medicine, Florida Atlantic University, Jupiter, FL, USA
| | - Lankupalle D. Jayanthi
- Department of Pharmacology and Toxicology, Virginia Commonwealth University, Richmond, VA, USA
| | - Roxanne A. Vaughan
- Department of Biomedical Sciences, University of North Dakota School of Medicine and Health Sciences, Grand Forks, ND, USA
| | - Lynette C. Daws
- Department of Cellular and Integrative Physiology, University of Texas Health Science Center at San Antonio, TX, USA
- Department of Pharmacology, University of Texas Health Science Center, San Antonio, TX, USA
| | - Randy D. Blakely
- Department of Biomedical Science, Charles E. Schmidt College of Medicine, Florida Atlantic University, Jupiter, FL, USA
- Stiles-Nicholson Brain Institute, Florida Atlantic University, Jupiter, FL, USA
| | - Sammanda Ramamoorthy
- Department of Pharmacology and Toxicology, Virginia Commonwealth University, Richmond, VA, USA
| |
Collapse
|
4
|
Tu G, Fu T, Zheng G, Xu B, Gou R, Luo D, Wang P, Xue W. Computational Chemistry in Structure-Based Solute Carrier Transporter Drug Design: Recent Advances and Future Perspectives. J Chem Inf Model 2024; 64:1433-1455. [PMID: 38294194 DOI: 10.1021/acs.jcim.3c01736] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/01/2024]
Abstract
Solute carrier transporters (SLCs) are a class of important transmembrane proteins that are involved in the transportation of diverse solute ions and small molecules into cells. There are approximately 450 SLCs within the human body, and more than a quarter of them are emerging as attractive therapeutic targets for multiple complex diseases, e.g., depression, cancer, and diabetes. However, only 44 unique transporters (∼9.8% of the SLC superfamily) with 3D structures and specific binding sites have been reported. To design innovative and effective drugs targeting diverse SLCs, there are a number of obstacles that need to be overcome. However, computational chemistry, including physics-based molecular modeling and machine learning- and deep learning-based artificial intelligence (AI), provides an alternative and complementary way to the classical drug discovery approach. Here, we present a comprehensive overview on recent advances and existing challenges of the computational techniques in structure-based drug design of SLCs from three main aspects: (i) characterizing multiple conformations of the proteins during the functional process of transportation, (ii) identifying druggability sites especially the cryptic allosteric ones on the transporters for substrates and drugs binding, and (iii) discovering diverse small molecules or synthetic protein binders targeting the binding sites. This work is expected to provide guidelines for a deep understanding of the structure and function of the SLC superfamily to facilitate rational design of novel modulators of the transporters with the aid of state-of-the-art computational chemistry technologies including artificial intelligence.
Collapse
Affiliation(s)
- Gao Tu
- Chongqing Key Laboratory of Natural Product Synthesis and Drug Research, School of Pharmaceutical Sciences, Chongqing University, Chongqing 401331, China
| | - Tingting Fu
- College of Pharmaceutical Sciences, Zhejiang University, Hangzhou 310058, China
| | | | - Binbin Xu
- Chengdu Sintanovo Biotechnology Co., Ltd., Chengdu 610200, China
| | - Rongpei Gou
- Chongqing Key Laboratory of Natural Product Synthesis and Drug Research, School of Pharmaceutical Sciences, Chongqing University, Chongqing 401331, China
| | - Ding Luo
- Chongqing Key Laboratory of Natural Product Synthesis and Drug Research, School of Pharmaceutical Sciences, Chongqing University, Chongqing 401331, China
| | - Panpan Wang
- College of Chemistry and Pharmaceutical Engineering, Huanghuai University, Zhumadian 463000, China
| | - Weiwei Xue
- Chongqing Key Laboratory of Natural Product Synthesis and Drug Research, School of Pharmaceutical Sciences, Chongqing University, Chongqing 401331, China
| |
Collapse
|
5
|
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
|
6
|
Fu T, Zeng S, Zheng Q, Zhu F. The Important Role of Transporter Structures in Drug Disposition, Efficacy, and Toxicity. Drug Metab Dispos 2023; 51:1316-1323. [PMID: 37295948 DOI: 10.1124/dmd.123.001275] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/27/2023] [Revised: 05/27/2023] [Accepted: 06/02/2023] [Indexed: 06/12/2023] Open
Abstract
The ATP-binding cassette (ABC) and solute carrier (SLC) transporters are critical determinants of drug disposition, clinical efficacy, and toxicity as they specifically mediate the influx and efflux of various substrates and drugs. ABC transporters can modulate the pharmacokinetics of many drugs via mediating the translocation of drugs across biologic membranes. SLC transporters are important drug targets involved in the uptake of a broad range of compounds across the membrane. However, high-resolution experimental structures have been reported for a very limited number of transporters, which limits the study of their physiologic functions. In this review, we collected structural information on ABC and SLC transporters and described the application of computational methods in structure prediction. Taking P-glycoprotein (ABCB1) and serotonin transporter (SLC6A4) as examples, we assessed the pivotal role of structure in transport mechanisms, details of ligand-receptor interactions, drug selectivity, the molecular mechanisms of drug-drug interactions, and differences caused by genetic polymorphisms. The data collected contributes toward safer and more effective pharmacological treatments. SIGNIFICANCE STATEMENT: The experimental structure of ATP-binding cassette and solute carrier transporters was collected, and the application of computational methods in structure prediction was described. P-glycoprotein and serotonin transporter were used as examples to reveal the pivotal role of structure in transport mechanisms, drug selectivity, the molecular mechanisms of drug-drug interactions, and differences caused by genetic polymorphisms.
Collapse
Affiliation(s)
- Tingting Fu
- College of Pharmaceutical Sciences, The Second Affiliated Hospital, Zhejiang University School of Medicine, Zhejiang University, Hangzhou, China (F.Z.); School of Pharmaceutical Sciences, Jilin University, Changchun, China (T.F., Q.Z.); College of Pharmaceutical Sciences, Zhejiang University, Hangzhou, China (S.Z., F.Z.); and Innovation Institute for Artificial Intelligence in Medicine of Zhejiang University, Alibaba-Zhejiang University Joint Research Center of Future Digital Healthcare, Hangzhou, China (F.Z.)
| | - Su Zeng
- College of Pharmaceutical Sciences, The Second Affiliated Hospital, Zhejiang University School of Medicine, Zhejiang University, Hangzhou, China (F.Z.); School of Pharmaceutical Sciences, Jilin University, Changchun, China (T.F., Q.Z.); College of Pharmaceutical Sciences, Zhejiang University, Hangzhou, China (S.Z., F.Z.); and Innovation Institute for Artificial Intelligence in Medicine of Zhejiang University, Alibaba-Zhejiang University Joint Research Center of Future Digital Healthcare, Hangzhou, China (F.Z.)
| | - Qingchuan Zheng
- College of Pharmaceutical Sciences, The Second Affiliated Hospital, Zhejiang University School of Medicine, Zhejiang University, Hangzhou, China (F.Z.); School of Pharmaceutical Sciences, Jilin University, Changchun, China (T.F., Q.Z.); College of Pharmaceutical Sciences, Zhejiang University, Hangzhou, China (S.Z., F.Z.); and Innovation Institute for Artificial Intelligence in Medicine of Zhejiang University, Alibaba-Zhejiang University Joint Research Center of Future Digital Healthcare, Hangzhou, China (F.Z.)
| | - Feng Zhu
- College of Pharmaceutical Sciences, The Second Affiliated Hospital, Zhejiang University School of Medicine, Zhejiang University, Hangzhou, China (F.Z.); School of Pharmaceutical Sciences, Jilin University, Changchun, China (T.F., Q.Z.); College of Pharmaceutical Sciences, Zhejiang University, Hangzhou, China (S.Z., F.Z.); and Innovation Institute for Artificial Intelligence in Medicine of Zhejiang University, Alibaba-Zhejiang University Joint Research Center of Future Digital Healthcare, Hangzhou, China (F.Z.)
| |
Collapse
|
7
|
Zanfino G, Puzzo C, de Laurenzi V, Adriani W. Characterization of Behavioral Phenotypes in Heterozygous DAT Rat Based on Pedigree. Biomedicines 2023; 11:2565. [PMID: 37761006 PMCID: PMC10526166 DOI: 10.3390/biomedicines11092565] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/11/2023] [Revised: 09/07/2023] [Accepted: 09/11/2023] [Indexed: 09/29/2023] Open
Abstract
Dopamine is an essential neurotransmitter whose key roles include movement control, pleasure and reward, attentional and cognitive skills, and regulation of the sleep/wake cycle. Reuptake is carried out by the dopamine transporter (DAT; DAT1 SLC6A3 gene). In order to study the effects of hyper-dopaminergia syndrome, the gene was silenced in rats. DAT-KO rats show stereotypical behavior, hyperactivity, a deficit in working memory, and an altered circadian cycle. In addition to KO rats, heterozygous (DAT-HET) rats show relative hypofunction of DAT; exact phenotypic effects are still unknown and may depend on whether the sire or the dam was KO. Our goal was to elucidate the potential importance of the parental origin of the healthy or silenced allele and its impact across generations, along with the potential variations in maternal care. We thus generated specular lines to study the effects of (grand) parental roles in inheriting the wild or mutated allele. MAT-HETs are the progeny of a KO sire and a WT dam; by breeding MAT-HET males and KO females, we obtained subjects with a DAT -/- epigenotype, named QULL, to reflect additional epigenetic DAT modulation when embryos develop within a hyper-dopaminergic KO uterus. We aimed to verify if any behavioral anomaly was introduced by a QULL (instead of KO) rat acting as a direct father or indirect maternal grandfather (or both). We thus followed epigenotypes obtained after three generations and observed actual effects on impaired maternal care of the offspring (based on pedigree). In particular, offspring of MAT-HET-dam × QULL-sire breeding showed a compulsive and obsessive phenotype. Although the experimental groups were all heterozygous, the impact of having a sire of epigenotype QULL (who developed in the uterus of a KO grand-dam) has emerged clearly. Along the generations, the effects of the DAT epigenotype on the obsessive/compulsive phenotype do vary as a function of the uterine impact on either allele in one's genealogical line.
Collapse
Affiliation(s)
- Gioia Zanfino
- Center for Behavioral Sciences and Mental Health, Istituto Superiore di Sanità, 00161 Rome, Italy; (G.Z.); (C.P.)
| | - Concetto Puzzo
- Center for Behavioral Sciences and Mental Health, Istituto Superiore di Sanità, 00161 Rome, Italy; (G.Z.); (C.P.)
- Faculty of Psychology, International Telematic University Uninettuno, 00186 Rome, Italy
| | - Vincenzo de Laurenzi
- Center for Advanced Studies and Technology (CAST), “G. d’Annunzio” University of Chieti-Pescara, 66100 Chieti, Italy;
- Department of Innovative Technologies in Medicine and Dentistry, “G. d’Annunzio” University of Chieti-Pescara, 66100 Chieti, Italy
| | - Walter Adriani
- Center for Behavioral Sciences and Mental Health, Istituto Superiore di Sanità, 00161 Rome, Italy; (G.Z.); (C.P.)
- Faculty of Psychology, International Telematic University Uninettuno, 00186 Rome, Italy
| |
Collapse
|
8
|
Niello M, Sideromenos S, Gradisch R, O´Shea R, Schwazer J, Maier J, Kastner N, Sandtner W, Jäntsch K, Lupica CR, Hoffman AF, Lubec G, Loland CJ, Stockner T, Pollak DD, Baumann MH, Sitte HH. Persistent binding at dopamine transporters determines sustained psychostimulant effects. Proc Natl Acad Sci U S A 2023; 120:e2114204120. [PMID: 36730201 PMCID: PMC9963675 DOI: 10.1073/pnas.2114204120] [Citation(s) in RCA: 11] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/02/2021] [Accepted: 12/28/2022] [Indexed: 02/03/2023] Open
Abstract
Psychostimulants interacting with the dopamine transporter (DAT) can be used illicitly or for the treatment of specific neuropsychiatric disorders. However, they can also produce severe and persistent adverse events. Often, their pharmacological properties in vitro do not fully correlate to their pharmacological profile in vivo. Here, we investigated the pharmacological effects of enantiomers of pyrovalerone, α-pyrrolidinovalerophenone, and 3,4-methylenedioxypyrovalerone as compared to the traditional psychostimulants cocaine and methylphenidate, using a variety of in vitro, computational, and in vivo approaches. We found that in vitro drug-binding kinetics at DAT correlate with the time-course of in vivo psychostimulant action in mice. In particular, a slow dissociation (i.e., slow koff) of S-enantiomers of pyrovalerone analogs from DAT predicts their more persistent in vivo effects when compared to cocaine and methylphenidate. Overall, our findings highlight the critical importance of drug-binding kinetics at DAT for determining the in vivo profile of effects produced by psychostimulant drugs.
Collapse
Affiliation(s)
- Marco Niello
- Center for Physiology and Pharmacology, Institute of Pharmacology, Medical University of Vienna, 1090Vienna, Austria
| | - Spyridon Sideromenos
- Center for Physiology and Pharmacology, Department of Neurophysiology and Neuropharmacology, Medical University of Vienna, 1090Vienna, Austria
| | - Ralph Gradisch
- Center for Physiology and Pharmacology, Institute of Pharmacology, Medical University of Vienna, 1090Vienna, Austria
| | - Ronan O´Shea
- Electrophysiology Research Section, National Institute on Drug Abuse, NIH, Baltimore, MD21224
| | - Jakob Schwazer
- Center for Physiology and Pharmacology, Institute of Pharmacology, Medical University of Vienna, 1090Vienna, Austria
| | - Julian Maier
- Center for Physiology and Pharmacology, Institute of Pharmacology, Medical University of Vienna, 1090Vienna, Austria
| | - Nina Kastner
- Center for Physiology and Pharmacology, Institute of Pharmacology, Medical University of Vienna, 1090Vienna, Austria
| | - Walter Sandtner
- Center for Physiology and Pharmacology, Institute of Pharmacology, Medical University of Vienna, 1090Vienna, Austria
| | - Kathrin Jäntsch
- Center for Physiology and Pharmacology, Institute of Pharmacology, Medical University of Vienna, 1090Vienna, Austria
| | - Carl R. Lupica
- Electrophysiology Research Section, National Institute on Drug Abuse, NIH, Baltimore, MD21224
| | - Alexander F. Hoffman
- Electrophysiology Research Section, National Institute on Drug Abuse, NIH, Baltimore, MD21224
| | - Gert Lubec
- Department of Neuroproteomics, Paracelsus Medical University, 5020Salzburg, Austria
| | - Claus J. Loland
- Department of Neuroscience, Faculty of Health and Medical Sciences, University of Copenhagen, 2200Copenhagen, Denmark
| | - Thomas Stockner
- Center for Physiology and Pharmacology, Institute of Pharmacology, Medical University of Vienna, 1090Vienna, Austria
| | - Daniela D. Pollak
- Center for Physiology and Pharmacology, Department of Neurophysiology and Neuropharmacology, Medical University of Vienna, 1090Vienna, Austria
| | - Michael H. Baumann
- Designer Drug Research Unit, Intramural Research Program, National Institute on Drug Abuse, NIH, Baltimore, MD21224
| | - Harald H. Sitte
- Center for Physiology and Pharmacology, Institute of Pharmacology, Medical University of Vienna, 1090Vienna, Austria
- AddRess, Center for Addiction Research and Science, Medical University of Vienna, 1090Vienna, Austria
| |
Collapse
|
9
|
Shekar A, Mabry SJ, Cheng MH, Aguilar JI, Patel S, Zanella D, Saleeby DP, Zhu Y, Romanazzi T, Ulery-Reynolds P, Bahar I, Carter AM, Matthies HJG, Galli A. Syntaxin 1 Ser 14 phosphorylation is required for nonvesicular dopamine release. SCIENCE ADVANCES 2023; 9:eadd8417. [PMID: 36630507 PMCID: PMC9833662 DOI: 10.1126/sciadv.add8417] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/07/2022] [Accepted: 12/14/2022] [Indexed: 05/30/2023]
Abstract
Amphetamine (AMPH) is a psychostimulant that is commonly abused. The stimulant properties of AMPH are associated with its ability to increase dopamine (DA) neurotransmission. This increase is promoted by nonvesicular DA release mediated by reversal of DA transporter (DAT) function. Syntaxin 1 (Stx1) is a SNARE protein that is phosphorylated at Ser14 by casein kinase II. We show that Stx1 phosphorylation is critical for AMPH-induced nonvesicular DA release and, in Drosophila melanogaster, regulates the expression of AMPH-induced preference and sexual motivation. Our molecular dynamics simulations of the DAT/Stx1 complex demonstrate that phosphorylation of these proteins is pivotal for DAT to dwell in a DA releasing state. This state is characterized by the breakdown of two key salt bridges within the DAT intracellular gate, causing the opening and hydration of the DAT intracellular vestibule, allowing DA to bind from the cytosol, a mechanism that we hypothesize underlies nonvesicular DA release.
Collapse
Affiliation(s)
- Aparna Shekar
- Department of Surgery, University of Alabama at Birmingham, Birmingham, AL, USA
| | - Samuel J. Mabry
- Department of Surgery, University of Alabama at Birmingham, Birmingham, AL, USA
| | - Mary H. Cheng
- Department of Computational and Systems Biology, University of Pittsburgh School of Medicine, Pittsburgh, PA, USA
| | - Jenny I. Aguilar
- Department of Surgery, University of Alabama at Birmingham, Birmingham, AL, USA
| | - Shalin Patel
- Department of Surgery, University of Alabama at Birmingham, Birmingham, AL, USA
| | - Daniele Zanella
- Department of Surgery, University of Alabama at Birmingham, Birmingham, AL, USA
| | - David P. Saleeby
- Department of Surgery, University of Alabama at Birmingham, Birmingham, AL, USA
| | - Yanqi Zhu
- Department of Surgery, University of Alabama at Birmingham, Birmingham, AL, USA
| | - Tiziana Romanazzi
- Department of Biotechnology and Life Sciences, University of Insubria, Varese, Italy
| | | | - Ivet Bahar
- Department of Computational and Systems Biology, University of Pittsburgh School of Medicine, Pittsburgh, PA, USA
| | - Angela M. Carter
- Department of Surgery, University of Alabama at Birmingham, Birmingham, AL, USA
| | | | - Aurelio Galli
- Department of Surgery, University of Alabama at Birmingham, Birmingham, AL, USA
| |
Collapse
|
10
|
Kearney PJ, Bolden NC, Kahuno E, Conklin TL, Martin GE, Lubec G, Melikian HE. Presynaptic Gq-coupled receptors drive biphasic dopamine transporter trafficking that modulates dopamine clearance and motor function. J Biol Chem 2023; 299:102900. [PMID: 36640864 PMCID: PMC9943899 DOI: 10.1016/j.jbc.2023.102900] [Citation(s) in RCA: 8] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/09/2022] [Revised: 12/23/2022] [Accepted: 01/03/2023] [Indexed: 01/13/2023] Open
Abstract
Extracellular dopamine (DA) levels are constrained by the presynaptic DA transporter (DAT), a major psychostimulant target. Despite its necessity for DA neurotransmission, DAT regulation in situ is poorly understood, and it is unknown whether regulated DAT trafficking impacts dopaminergic signaling and/or behaviors. Leveraging chemogenetics and conditional gene silencing, we found that activating presynaptic Gq-coupled receptors, either hM3Dq or mGlu5, drove rapid biphasic DAT membrane trafficking in ex vivo striatal slices, with region-specific differences between ventral and dorsal striata. DAT insertion required D2 DA autoreceptors and intact retromer, whereas DAT retrieval required PKC activation and Rit2. Ex vivo voltammetric studies revealed that DAT trafficking impacts DA clearance. Furthermore, dopaminergic mGlu5 silencing elevated DAT surface expression and abolished motor learning, which was rescued by inhibiting DAT with a subthreshold CE-158 dose. We discovered that presynaptic DAT trafficking is complex, multimodal, and region specific, and for the first time, we identified cell autonomous mechanisms that govern presynaptic DAT tone. Importantly, the findings are consistent with a role for regulated DAT trafficking in DA clearance and motor function.
Collapse
Affiliation(s)
- Patrick J. Kearney
- Brudnick Neuropsychiatric Research Institute, Department of Neurobiology, UMASS Chan Medical School, Worcester, Massachusetts, USA,Morningside Graduate School of Biomedical Sciences, UMASS Chan Medical School, Worcester, Massachusetts, USA
| | - Nicholas C. Bolden
- Brudnick Neuropsychiatric Research Institute, Department of Neurobiology, UMASS Chan Medical School, Worcester, Massachusetts, USA,Morningside Graduate School of Biomedical Sciences, UMASS Chan Medical School, Worcester, Massachusetts, USA
| | - Elizabeth Kahuno
- Brudnick Neuropsychiatric Research Institute, Department of Neurobiology, UMASS Chan Medical School, Worcester, Massachusetts, USA
| | - Tucker L. Conklin
- Brudnick Neuropsychiatric Research Institute, Department of Neurobiology, UMASS Chan Medical School, Worcester, Massachusetts, USA
| | - Gilles E. Martin
- Brudnick Neuropsychiatric Research Institute, Department of Neurobiology, UMASS Chan Medical School, Worcester, Massachusetts, USA
| | - Gert Lubec
- Department of Neuroproteomics, Paracelsus Private Medical University, Salzburg, Austria
| | - Haley E. Melikian
- Brudnick Neuropsychiatric Research Institute, Department of Neurobiology, UMASS Chan Medical School, Worcester, Massachusetts, USA,For correspondence: Haley E. Melikian
| |
Collapse
|
11
|
Nepal B, Das S, Reith ME, Kortagere S. Overview of the structure and function of the dopamine transporter and its protein interactions. Front Physiol 2023; 14:1150355. [PMID: 36935752 PMCID: PMC10020207 DOI: 10.3389/fphys.2023.1150355] [Citation(s) in RCA: 11] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/24/2023] [Accepted: 02/21/2023] [Indexed: 03/06/2023] Open
Abstract
The dopamine transporter (DAT) plays an integral role in dopamine neurotransmission through the clearance of dopamine from the extracellular space. Dysregulation of DAT is central to the pathophysiology of numerous neuropsychiatric disorders and as such is an attractive therapeutic target. DAT belongs to the solute carrier family 6 (SLC6) class of Na+/Cl- dependent transporters that move various cargo into neurons against their concentration gradient. This review focuses on DAT (SCL6A3 protein) while extending the narrative to the closely related transporters for serotonin and norepinephrine where needed for comparison or functional relevance. Cloning and site-directed mutagenesis experiments provided early structural knowledge of DAT but our contemporary understanding was achieved through a combination of crystallization of the related bacterial transporter LeuT, homology modeling, and subsequently the crystallization of drosophila DAT. These seminal findings enabled a better understanding of the conformational states involved in the transport of substrate, subsequently aiding state-specific drug design. Post-translational modifications to DAT such as phosphorylation, palmitoylation, ubiquitination also influence the plasma membrane localization and kinetics. Substrates and drugs can interact with multiple sites within DAT including the primary S1 and S2 sites involved in dopamine binding and novel allosteric sites. Major research has centered around the question what determines the substrate and inhibitor selectivity of DAT in comparison to serotonin and norepinephrine transporters. DAT has been implicated in many neurological disorders and may play a role in the pathology of HIV and Parkinson's disease via direct physical interaction with HIV-1 Tat and α-synuclein proteins respectively.
Collapse
Affiliation(s)
- Binod Nepal
- Department of Microbiology and Immunology, Drexel University College of Medicine, Philadelphia, PA, United States
| | - Sanjay Das
- Department of Microbiology and Immunology, Drexel University College of Medicine, Philadelphia, PA, United States
| | - Maarten E. Reith
- Department of Psychiatry, New York University School of Medicine, New York City, NY, United States
| | - Sandhya Kortagere
- Department of Microbiology and Immunology, Drexel University College of Medicine, Philadelphia, PA, United States
- *Correspondence: Sandhya Kortagere,
| |
Collapse
|
12
|
Male DAT Val559 Mice Exhibit Compulsive Behavior under Devalued Reward Conditions Accompanied by Cellular and Pharmacological Changes. Cells 2022; 11:cells11244059. [PMID: 36552823 PMCID: PMC9777203 DOI: 10.3390/cells11244059] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/31/2022] [Revised: 12/05/2022] [Accepted: 12/12/2022] [Indexed: 12/23/2022] Open
Abstract
Identified across multiple psychiatric disorders, the dopamine (DA) transporter (DAT) Ala559Val substitution triggers non-vesicular, anomalous DA efflux (ADE), perturbing DA neurotransmission and behavior. We have shown that DAT Val559 mice display a waiting impulsivity and changes in cognitive performance associated with enhanced reward motivation. Here, utilizing a within-subject, lever-pressing paradigm designed to bias the formation of goal-directed or habitual behavior, we demonstrate that DAT Val559 mice modulate their nose poke behavior appropriately to match context, but demonstrate a perseverative checking behavior. Although DAT Val559 mice display no issues with the cognitive flexibility required to acquire and re-learn a visual pairwise discrimination task, devaluation of reward evoked habitual reward seeking in DAT Val559 mutants in operant tasks regardless of reinforcement schedule. The direct DA agonist apomorphine also elicits locomotor stereotypies in DAT Val559, but not WT mice. Our observation that dendritic spine density is increased in the dorsal medial striatum (DMS) of DAT Val559 mice speaks to an imbalance in striatal circuitry that might underlie the propensity of DAT Val559 mutants to exhibit compulsive behaviors when reward is devalued. Thus, DAT Val559 mice represent a model for dissection of how altered DA signaling perturbs circuits that normally balance habitual and goal-directed behaviors.
Collapse
|
13
|
Stewart A, Mayer FP, Gowrishankar R, Davis GL, Areal LB, Gresch PJ, Katamish RM, Peart R, Stilley SE, Spiess K, Rabil MJ, Diljohn FA, Wiggins AE, Vaughan RA, Hahn MK, Blakely RD. Behaviorally penetrant, anomalous dopamine efflux exposes sex and circuit dependent regulation of dopamine transporters. Mol Psychiatry 2022; 27:4869-4880. [PMID: 36117213 DOI: 10.1038/s41380-022-01773-7] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/31/2021] [Revised: 08/18/2022] [Accepted: 08/31/2022] [Indexed: 01/19/2023]
Abstract
Virtually all neuropsychiatric disorders display sex differences in prevalence, age of onset, and/or clinical symptomology. Although altered dopamine (DA) signaling is a feature of many of these disorders, sex-dependent mechanisms uniquely responsive to DA that drive sex-dependent behaviors remain unelucidated. Previously, we established that anomalous DA efflux (ADE) is a prominent feature of the DA transporter (DAT) variant Val559, a coding substitution identified in two male-biased disorders: attention-deficit/hyperactivity disorder and autism spectrum disorder. In vivo, Val559 ADE induces activation of nigrostriatal D2-type DA autoreceptors (D2ARs) that magnifies inappropriate, nonvesicular DA release by elevating phosphorylation and surface trafficking of ADE-prone DAT proteins. Here we demonstrate that DAT Val559 mice exhibit sex-dependent alterations in psychostimulant responses, social behavior, and cognitive performance. In a search for underlying mechanisms, we discovered that the ability of ADE to elicit D2AR regulation of DAT is both sex and circuit-dependent, with dorsal striatum D2AR/DAT coupling evident only in males, whereas D2AR/DAT coupling in the ventral striatum is exclusive to females. Moreover, systemic administration of the D2R antagonist sulpiride, which precludes ADE-driven DAT trafficking, can normalize DAT Val559 behavioral changes unique to each sex and without effects on the opposite sex or wildtype mice. Our studies support the sex- and circuit dependent capacity of D2ARs to regulate DAT as a critical determinant of the sex-biased effects of perturbed DA signaling in neurobehavioral disorders. Moreover, our work provides a cogent example of how a shared biological insult drives alternative physiological and behavioral trajectories as opposed to resilience.
Collapse
Affiliation(s)
- Adele Stewart
- Department of Biomedical Science, Florida Atlantic University, Jupiter, FL, USA.,Stiles-Nicholson Brain Institute, Florida Atlantic University, Jupiter, FL, USA
| | - Felix P Mayer
- Department of Biomedical Science, Florida Atlantic University, Jupiter, FL, USA
| | | | - Gwynne L Davis
- Department of Biomedical Science, Florida Atlantic University, Jupiter, FL, USA
| | - Lorena B Areal
- Department of Biomedical Science, Florida Atlantic University, Jupiter, FL, USA
| | - Paul J Gresch
- Department of Biomedical Science, Florida Atlantic University, Jupiter, FL, USA.,Stiles-Nicholson Brain Institute, Florida Atlantic University, Jupiter, FL, USA
| | - Rania M Katamish
- Department of Biomedical Science, Florida Atlantic University, Jupiter, FL, USA
| | - Rodeania Peart
- Wilkes Honors College, Florida Atlantic University, Jupiter, FL, USA
| | - Samantha E Stilley
- Department of Biomedical Science, Florida Atlantic University, Jupiter, FL, USA
| | - Keeley Spiess
- Department of Biomedical Science, Florida Atlantic University, Jupiter, FL, USA
| | - Maximilian J Rabil
- Department of Biomedical Science, Florida Atlantic University, Jupiter, FL, USA
| | | | - Angelica E Wiggins
- Department of Biomedical Science, Florida Atlantic University, Jupiter, FL, USA
| | - Roxanne A Vaughan
- Department of Biomedical Sciences, University of North Dakota School of Medicine and Health Sciences, Grand Forks, ND, USA
| | - Maureen K Hahn
- Department of Biomedical Science, Florida Atlantic University, Jupiter, FL, USA.,Stiles-Nicholson Brain Institute, Florida Atlantic University, Jupiter, FL, USA
| | - Randy D Blakely
- Department of Biomedical Science, Florida Atlantic University, Jupiter, FL, USA. .,Stiles-Nicholson Brain Institute, Florida Atlantic University, Jupiter, FL, USA.
| |
Collapse
|
14
|
Multimodal detection of dopamine by sniffer cells expressing genetically encoded fluorescent sensors. Commun Biol 2022; 5:578. [PMID: 35689020 PMCID: PMC9187629 DOI: 10.1038/s42003-022-03488-5] [Citation(s) in RCA: 15] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/28/2021] [Accepted: 05/13/2022] [Indexed: 12/21/2022] Open
Abstract
Dopamine supports locomotor control and higher brain functions such as motivation and learning. Consistently, dopaminergic dysfunction is involved in a spectrum of neurological and neuropsychiatric diseases. Detailed data on dopamine dynamics is needed to understand how dopamine signals translate into cellular and behavioral responses, and to uncover pathological disturbances in dopamine-related diseases. Genetically encoded fluorescent dopamine sensors have recently enabled unprecedented monitoring of dopamine dynamics in vivo. However, these sensors' utility for in vitro and ex vivo assays remains unexplored. Here, we present a blueprint for making dopamine sniffer cells for multimodal dopamine detection. We generated sniffer cell lines with inducible expression of seven different dopamine sensors and perform a head-to-head comparison of sensor properties to guide users in sensor selection. In proof-of-principle experiments, we apply the sniffer cells to record endogenous dopamine release from cultured neurons and striatal slices, and for determining tissue dopamine content. Furthermore, we use the sniffer cells to measure dopamine uptake and release via the dopamine transporter as a radiotracer free, high-throughput alternative to electrochemical- and radiotracer-based assays. Importantly, the sniffer cell framework can readily be applied to the growing list of genetically encoded fluorescent neurotransmitter sensors.
Collapse
|
15
|
Sutton C, Williams EQ, Homsi H, Beerepoot P, Nazari R, Han D, Ramsey AJ, Mash DC, Olson DE, Blough B, Salahpour A. Structure-Activity Relationships of Dopamine Transporter Pharmacological Chaperones. Front Cell Neurosci 2022; 16:832536. [PMID: 35614973 PMCID: PMC9124866 DOI: 10.3389/fncel.2022.832536] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/10/2021] [Accepted: 03/21/2022] [Indexed: 11/13/2022] Open
Abstract
Mutations in the dopamine transporter gene (SLC6A3) have been implicated in many human diseases. Among these is the infantile parkinsonism-dystonia known as Dopamine Transporter Deficiency Syndrome (DTDS). Afflicted individuals have minimal to no functional dopamine transporter protein. This is primarily due to retention of misfolded disease-causing dopamine transporter variants. This results in a variety of severe motor symptoms in patients and the disease ultimately leads to death in adolescence or young adulthood. Though no treatment is currently available, pharmacological chaperones targeting the dopamine transporter have been shown to rescue select DTDS disease-causing variants. Previous work has identified two DAT pharmacological chaperones with moderate potency and efficacy: bupropion and ibogaine. In this study, we carried out structure-activity relationships (SARs) for bupropion and ibogaine with the goal of identifying the chemical features required for pharmacological chaperone activity. Our results show that the isoquinuclidine substituent of ibogaine and its analogs is an important feature for pharmacological chaperone efficacy. For bupropion, the secondary amine group is essential for pharmacological chaperone activity. Lastly, we describe additional ibogaine and bupropion analogs with varying chemical modifications and variable pharmacological chaperone efficacies at the dopamine transporter. Our results contribute to the design and refinement of future dopamine transporter pharmacological chaperones with improved efficacies and potencies.
Collapse
Affiliation(s)
- Charles Sutton
- Department of Pharmacology and Toxicology, Faculty of Medicine, University of Toronto, Toronto, ON, Canada
| | - Erin Q. Williams
- Department of Pharmacology and Toxicology, Faculty of Medicine, University of Toronto, Toronto, ON, Canada
| | - Hoomam Homsi
- Department of Pharmacology and Toxicology, Faculty of Medicine, University of Toronto, Toronto, ON, Canada
| | - Pieter Beerepoot
- Department of Pharmacology and Toxicology, Faculty of Medicine, University of Toronto, Toronto, ON, Canada
| | - Reza Nazari
- Department of Pharmacology and Toxicology, Faculty of Medicine, University of Toronto, Toronto, ON, Canada
| | - Dong Han
- Department of Pharmacology and Toxicology, Faculty of Medicine, University of Toronto, Toronto, ON, Canada
| | - Amy J. Ramsey
- Department of Pharmacology and Toxicology, Faculty of Medicine, University of Toronto, Toronto, ON, Canada
| | - Deborah C. Mash
- Departments of Neurology and Molecular and Cellular Pharmacology, University of Miami Miller School of Medicine, Miami, FL, United States
| | - David E. Olson
- Department of Chemistry, College of Letters and Science, University of California, Davis, Davis, CA, United States
- Department of Biochemistry and Molecular Medicine, School of Medicine, University of California, Davis, Davis, CA, United States
- Center for Neuroscience, University of California, Davis, Davis, CA, United States
| | - Bruce Blough
- Center for Drug Discovery, RTI International, North Carolina, NC, United States
| | - Ali Salahpour
- Department of Pharmacology and Toxicology, Faculty of Medicine, University of Toronto, Toronto, ON, Canada
| |
Collapse
|
16
|
Reith MEA, Kortagere S, Wiers CE, Sun H, Kurian MA, Galli A, Volkow ND, Lin Z. The dopamine transporter gene SLC6A3: multidisease risks. Mol Psychiatry 2022; 27:1031-1046. [PMID: 34650206 PMCID: PMC9008071 DOI: 10.1038/s41380-021-01341-5] [Citation(s) in RCA: 39] [Impact Index Per Article: 13.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/17/2021] [Revised: 09/28/2021] [Accepted: 10/01/2021] [Indexed: 02/02/2023]
Abstract
The human dopamine transporter gene SLC6A3 has been consistently implicated in several neuropsychiatric diseases but the disease mechanism remains elusive. In this risk synthesis, we have concluded that SLC6A3 represents an increasingly recognized risk with a growing number of familial mutants associated with neuropsychiatric and neurological disorders. At least five loci were related to common and severe diseases including alcohol use disorder (high activity variant), attention-deficit/hyperactivity disorder (low activity variant), autism (familial proteins with mutated networking) and movement disorders (both regulatory variants and familial mutations). Association signals depended on genetic markers used as well as ethnicity examined. Strong haplotype selection and gene-wide epistases support multimarker assessment of functional variations and phenotype associations. Inclusion of its promoter region's functional markers such as DNPi (rs67175440) and 5'VNTR (rs70957367) may help delineate condensate-based risk action, testing a locus-pathway-phenotype hypothesis for one gene-multidisease etiology.
Collapse
Affiliation(s)
- Maarten E A Reith
- Department of Psychiatry, New York University School of Medicine, New York City, NY, 10016, USA
| | - Sandhya Kortagere
- Department of Microbiology and Immunology, Drexel University College of Medicine, Philadelphia, PA, 19129, USA
| | - Corinde E Wiers
- Laboratory of Neuroimaging, National Institute on Alcohol Abuse and Alcoholism, Bethesda, MD, 20817, USA
- Department of Psychiatry, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, 19104, USA
| | - Hui Sun
- Laboratory of Neuroimaging, National Institute on Alcohol Abuse and Alcoholism, Bethesda, MD, 20817, USA
| | - Manju A Kurian
- Molecular Neurosciences, Developmental Neurosciences, Zayed Centre for Research into Rare Diseases in Children, UCL Great Ormond Street Institute of Child Health, and Department of Neurology, Great Ormond Street Hospital, London, WC1N 1EH, UK
| | - Aurelio Galli
- Department of Surgery, University of Alabama at Birmingham, Birmingham, AL, 35294, USA
| | - Nora D Volkow
- Laboratory of Neuroimaging, National Institute on Alcohol Abuse and Alcoholism, Bethesda, MD, 20817, USA
- National Institute on Drug Abuse, Bethesda, MD, 20817, USA
| | - Zhicheng Lin
- Laboratory of Psychiatric Neurogenomics, McLean Hospital, and Department of Psychiatry, Harvard Medical School, Belmont, MA, 02478, USA.
| |
Collapse
|
17
|
Mackie PM, Gopinath A, Montas DM, Nielsen A, Smith A, Nolan RA, Runner K, Matt SM, McNamee J, Riklan JE, Adachi K, Doty A, Ramirez-Zamora A, Yan L, Gaskill PJ, Streit WJ, Okun MS, Khoshbouei H. Functional characterization of the biogenic amine transporters on human macrophages. JCI Insight 2022; 7:151892. [PMID: 35015729 PMCID: PMC8876465 DOI: 10.1172/jci.insight.151892] [Citation(s) in RCA: 17] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/01/2021] [Accepted: 01/05/2022] [Indexed: 11/17/2022] Open
Abstract
Monocyte-derived macrophages are key players in tissue homeostasis and diseases regulated by a variety of signaling molecules. Recent literature has highlighted the ability for biogenic amines to regulate macrophage functions, but the mechanisms governing biogenic amine signaling in and around immune cells remains nebulous. In the central nervous system (CNS), biogenic amine transporters are regarded as the master regulators of neurotransmitter signaling. While we and others have shown that macrophages express these transporters, relatively little is known of their function in these cells. To address these knowledge gaps, we investigated the function of norepinephrine (NET) and dopamine (DAT) transporters on human monocyte-derived macrophages. We found that both NET and DAT are present and can uptake substrate from the extracellular space at baseline. Not only was DAT expressed in cultured monocyte-derived macrophages (MDMs), but it was also detected in a subset of intestinal macrophages in situ. Surprisingly, we discovered a NET-independent, DAT-mediated immuno-modulatory mechanism in response to lipopolysaccharide (LPS). LPS induced reverse transport of dopamine through DAT, engaging an autocrine/paracrine signaling loop that regulated the macrophage response. Removing this signaling loop enhanced the pro-inflammatory response to LPS. Collectively, our data introduce a potential role for DAT in the regulation of innate immunity.
Collapse
Affiliation(s)
- Phillip M Mackie
- Department of Neuroscience, University of Florida, Gainesville, United States of America
| | - Adithya Gopinath
- Department of Neuroscience, McKnight Brain Insitute, University of Florida College of Medicine, Gainesville, United States of America
| | - Dominic M Montas
- Department of Neuroscience, University of Florida College of Medicine, Gainesville, United States of America
| | - Alyssa Nielsen
- Department of Neuroscience, University of Florida College of Medicine, Gainesville, United States of America
| | - Aidan Smith
- Department of Neuroscience, University of Florida College of Medicine, Gainesville, United States of America
| | - Rachel A Nolan
- Department of Pharmacology and Physiology, Drexel University College of Medicine, Philadelphia, United States of America
| | - Kaitlyn Runner
- Department of Pharmacology and Physiology, Drexel University College of Medicine, Philadelphia, United States of America
| | - Stephanie M Matt
- Department of Pharmacology and Physiology, Drexel University College of Medicine, Philadelphia, United States of America
| | - John McNamee
- Department of Neuroscience, University of Florida College of Medicine, Gainesville, United States of America
| | - Joshua E Riklan
- Department of Neuroscience, University of Florida College of Medicine, Gainesville, United States of America
| | - Kengo Adachi
- Neuronal Signal Transduction Group, Max Plank Florida Institute for Neuroscience, Jupiter, United States of America
| | - Andria Doty
- Flow Cytometry Core Facility, University of Florida College of Medicine, Gainesville, United States of America
| | - Adolfo Ramirez-Zamora
- Department of Neurology, University of Florida College of Medicine, Gainesville, United States of America
| | - Long Yan
- Neuronal Signal Transduction Group, Max Plank Florida Institute for Neuroscience, Jupiter, United States of America
| | - Peter J Gaskill
- Department of Pharmacology and Physiology, Drexel University College of Medicine, Philadelphia, United States of America
| | - Wolfgang J Streit
- Department of Neuroscience, University of Florida College of Medicine, Gainesville, United States of America
| | - Michael S Okun
- University of Florida College of Medicine, Gainesville, United States of America
| | - Habibeh Khoshbouei
- Department of Neuroscience, University of Florida College of Medicine, Gainesville, United States of America
| |
Collapse
|
18
|
Aguilar JI, Cheng MH, Font J, Schwartz AC, Ledwitch K, Duran A, Mabry SJ, Belovich AN, Zhu Y, Carter AM, Shi L, Kurian MA, Fenollar-Ferrer C, Meiler J, Ryan RM, Mchaourab HS, Bahar I, Matthies HJG, Galli A. Psychomotor impairments and therapeutic implications revealed by a mutation associated with infantile Parkinsonism-Dystonia. eLife 2021; 10:e68039. [PMID: 34002696 PMCID: PMC8131106 DOI: 10.7554/elife.68039] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/03/2021] [Accepted: 05/02/2021] [Indexed: 12/30/2022] Open
Abstract
Parkinson disease (PD) is a progressive, neurodegenerative disorder affecting over 6.1 million people worldwide. Although the cause of PD remains unclear, studies of highly penetrant mutations identified in early-onset familial parkinsonism have contributed to our understanding of the molecular mechanisms underlying disease pathology. Dopamine (DA) transporter (DAT) deficiency syndrome (DTDS) is a distinct type of infantile parkinsonism-dystonia that shares key clinical features with PD, including motor deficits (progressive bradykinesia, tremor, hypomimia) and altered DA neurotransmission. Here, we define structural, functional, and behavioral consequences of a Cys substitution at R445 in human DAT (hDAT R445C), identified in a patient with DTDS. We found that this R445 substitution disrupts a phylogenetically conserved intracellular (IC) network of interactions that compromise the hDAT IC gate. This is demonstrated by both Rosetta molecular modeling and fine-grained simulations using hDAT R445C, as well as EPR analysis and X-ray crystallography of the bacterial homolog leucine transporter. Notably, the disruption of this IC network of interactions supported a channel-like intermediate of hDAT and compromised hDAT function. We demonstrate that Drosophila melanogaster expressing hDAT R445C show impaired hDAT activity, which is associated with DA dysfunction in isolated brains and with abnormal behaviors monitored at high-speed time resolution. We show that hDAT R445C Drosophila exhibit motor deficits, lack of motor coordination (i.e. flight coordination) and phenotypic heterogeneity in these behaviors that is typically associated with DTDS and PD. These behaviors are linked with altered dopaminergic signaling stemming from loss of DA neurons and decreased DA availability. We rescued flight coordination with chloroquine, a lysosomal inhibitor that enhanced DAT expression in a heterologous expression system. Together, these studies shed some light on how a DTDS-linked DAT mutation underlies DA dysfunction and, possibly, clinical phenotypes shared by DTDS and PD.
Collapse
Affiliation(s)
- Jenny I Aguilar
- Department of Pharmacology, Vanderbilt UniversityNashvilleUnited States
- Department of Surgery, University of Alabama at BirminghamBirminghamUnited States
| | - Mary Hongying Cheng
- Department of Computational and Systems Biology, School of Medicine, University of PittsburghPittsburghUnited States
| | - Josep Font
- School of Medical Sciences, Faculty of Medicine and Health, University of SydneySydneyAustralia
| | - Alexandra C Schwartz
- Department of Molecular Physiology & Biophysics, Vanderbilt UniversityNashvilleUnited States
| | - Kaitlyn Ledwitch
- Center for Structural Biology, Vanderbilt UniversityNashvilleUnited States
- Department of Chemistry, Vanderbilt UniversityNashvilleUnited States
| | - Amanda Duran
- Center for Structural Biology, Vanderbilt UniversityNashvilleUnited States
- Department of Chemistry, Vanderbilt UniversityNashvilleUnited States
| | - Samuel J Mabry
- Department of Surgery, University of Alabama at BirminghamBirminghamUnited States
| | - Andrea N Belovich
- Department of Biomedical Sciences, Idaho College of Osteopathic MedicineMeridianUnited States
| | - Yanqi Zhu
- Department of Surgery, University of Alabama at BirminghamBirminghamUnited States
| | - Angela M Carter
- Department of Surgery, University of Alabama at BirminghamBirminghamUnited States
| | - Lei Shi
- Computational Chemistry and Molecular Biophysics Section, NIDA, NIHBaltimoreUnited States
| | - Manju A Kurian
- Molecular Neurosciences, Developmental Neurosciences, University College London (UCL)LondonUnited Kingdom
| | | | - Jens Meiler
- Center for Structural Biology, Vanderbilt UniversityNashvilleUnited States
- Department of Chemistry, Vanderbilt UniversityNashvilleUnited States
- Institute for Drug Discovery, Leipzig University Medical SchoolLeipzigGermany
| | - Renae Monique Ryan
- School of Medical Sciences, Faculty of Medicine and Health, University of SydneySydneyAustralia
| | - Hassane S Mchaourab
- Department of Molecular Physiology & Biophysics, Vanderbilt UniversityNashvilleUnited States
| | - Ivet Bahar
- Department of Computational and Systems Biology, School of Medicine, University of PittsburghPittsburghUnited States
| | - Heinrich JG Matthies
- Department of Surgery, University of Alabama at BirminghamBirminghamUnited States
| | - Aurelio Galli
- Department of Surgery, University of Alabama at BirminghamBirminghamUnited States
- Center for Inter-systemic Networks and Enteric Medical Advances, University of Alabama at BirminghamBirminghamUnited States
| |
Collapse
|
19
|
Dynamic control of the dopamine transporter in neurotransmission and homeostasis. NPJ Parkinsons Dis 2021; 7:22. [PMID: 33674612 PMCID: PMC7935902 DOI: 10.1038/s41531-021-00161-2] [Citation(s) in RCA: 69] [Impact Index Per Article: 17.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/18/2020] [Accepted: 01/08/2021] [Indexed: 01/31/2023] Open
Abstract
The dopamine transporter (DAT) transports extracellular dopamine into the intracellular space contributing to the regulation of dopamine neurotransmission. A reduction of DAT density is implicated in Parkinson's disease (PD) by neuroimaging; dopamine turnover is dopamine turnover is elevated in early symptomatic PD and in presymptomatic individuals with monogenic mutations causal for parkinsonism. As an integral plasma membrane protein, DAT surface expression is dynamically regulated through endocytic trafficking, enabling flexible control of dopamine signaling in time and space, which in turn critically modulates movement, motivation and learning behavior. Yet the cellular machinery and functional implications of DAT trafficking remain enigmatic. In this review we summarize mechanisms governing DAT trafficking under normal physiological conditions and discuss how PD-linked mutations may disturb DAT homeostasis. We highlight the complexity of DAT trafficking and reveal DAT dysregulation as a common theme in genetic models of parkinsonism.
Collapse
|
20
|
Schicker K, Bhat S, Farr C, Burtscher V, Horner A, Freissmuth M, Sandtner W. Descriptors of Secondary Active Transporter Function and How They Relate to Partial Reactions in the Transport Cycle. MEMBRANES 2021; 11:178. [PMID: 33802510 PMCID: PMC8001282 DOI: 10.3390/membranes11030178] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 01/28/2021] [Revised: 02/25/2021] [Accepted: 02/27/2021] [Indexed: 12/19/2022]
Abstract
Plasmalemmal solute carriers (SLCs) gauge and control solute abundance across cellular membranes. By virtue of this action, they play an important role in numerous physiological processes. Mutations in genes encoding the SLCs alter amino acid sequence that often leads to impaired protein function and onset of monogenic disorders. To understand how these altered proteins cause disease, it is necessary to undertake relevant functional assays. These experiments reveal descriptors of SLC function such as the maximal transport velocity (Vmax), the Michaelis constant for solute uptake (KM), potencies for inhibition of transporter function (IC50/EC50), and many more. In several instances, the mutated versions of different SLC transporters differ from their wild-type counterparts in the value of these descriptors. While determination of these experimental parameters can provide conjecture as to how the mutation gives rise to disease, they seldom provide any definitive insights on how a variant differ from the wild-type transporter in its operation. This is because the experimental determination of association between values of the descriptors and several partial reactions a transporter undergoes is casual, but not causal, at best. In the present study, we employ kinetic models that allow us to derive explicit mathematical terms and provide experimental descriptors as a function of the rate constants used to parameterize the kinetic model of the transport cycle. We show that it is possible to utilize these mathematical expressions to deduce, from experimental outcomes, how the mutation has impinged on partial reactions in the transport cycle.
Collapse
Affiliation(s)
- Klaus Schicker
- Center for Physiology and Pharmacology, Division of Neurophysiology and Neuropharmacology, Medical University of Vienna, 1090 Vienna, Austria;
| | - Shreyas Bhat
- Center of Physiology and Pharmacology, Institute of Pharmacology and the Gaston H. Glock Research Laboratories for Exploratory Drug Development, Medical University of Vienna, 1090 Vienna, Austria; (S.B.); (C.F.); (V.B.); (M.F.)
| | - Clemens Farr
- Center of Physiology and Pharmacology, Institute of Pharmacology and the Gaston H. Glock Research Laboratories for Exploratory Drug Development, Medical University of Vienna, 1090 Vienna, Austria; (S.B.); (C.F.); (V.B.); (M.F.)
| | - Verena Burtscher
- Center of Physiology and Pharmacology, Institute of Pharmacology and the Gaston H. Glock Research Laboratories for Exploratory Drug Development, Medical University of Vienna, 1090 Vienna, Austria; (S.B.); (C.F.); (V.B.); (M.F.)
| | - Andreas Horner
- Institute of Biophysics, Johannes Kepler University Linz, 4040 Linz, Austria;
| | - Michael Freissmuth
- Center of Physiology and Pharmacology, Institute of Pharmacology and the Gaston H. Glock Research Laboratories for Exploratory Drug Development, Medical University of Vienna, 1090 Vienna, Austria; (S.B.); (C.F.); (V.B.); (M.F.)
| | - Walter Sandtner
- Center of Physiology and Pharmacology, Institute of Pharmacology and the Gaston H. Glock Research Laboratories for Exploratory Drug Development, Medical University of Vienna, 1090 Vienna, Austria; (S.B.); (C.F.); (V.B.); (M.F.)
| |
Collapse
|
21
|
Abstract
Inhibitors of Na+/Cl- dependent high affinity transporters for norepinephrine (NE), serotonin (5-HT), and/or dopamine (DA) represent frequently used drugs for treatment of psychological disorders such as depression, anxiety, obsessive-compulsive disorder, attention deficit hyperactivity disorder, and addiction. These transporters remove NE, 5-HT, and/or DA after neuronal excitation from the interstitial space close to the synapses. Thereby they terminate transmission and modulate neuronal behavioral circuits. Therapeutic failure and undesired central nervous system side effects of these drugs have been partially assigned to neurotransmitter removal by low affinity transport. Cloning and functional characterization of the polyspecific organic cation transporters OCT1 (SLC22A1), OCT2 (SLC22A2), OCT3 (SLC22A3) and the plasma membrane monoamine transporter PMAT (SLC29A4) revealed that every single transporter mediates low affinity uptake of NE, 5-HT, and DA. Whereas the organic transporters are all located in the blood brain barrier, OCT2, OCT3, and PMAT are expressed in neurons or in neurons and astrocytes within brain areas that are involved in behavioral regulation. Areas of expression include the dorsal raphe, medullary motoric nuclei, hypothalamic nuclei, and/or the nucleus accumbens. Current knowledge of the transport of monoamine neurotransmitters by the organic cation transporters, their interactions with psychotropic drugs, and their locations in the brain is reported in detail. In addition, animal experiments including behavior tests in wildtype and knockout animals are reported in which the impact of OCT2, OCT3, and/or PMAT on regulation of salt intake, depression, mood control, locomotion, and/or stress effect on addiction is suggested.
Collapse
Affiliation(s)
- Hermann Koepsell
- Institute of Anatomy and Cell Biology, University Würzburg, Würzburg, Germany.
| |
Collapse
|
22
|
Bhat S, El-Kasaby A, Freissmuth M, Sucic S. Functional and Biochemical Consequences of Disease Variants in Neurotransmitter Transporters: A Special Emphasis on Folding and Trafficking Deficits. Pharmacol Ther 2020; 222:107785. [PMID: 33310157 PMCID: PMC7612411 DOI: 10.1016/j.pharmthera.2020.107785] [Citation(s) in RCA: 27] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/31/2020] [Accepted: 12/02/2020] [Indexed: 01/30/2023]
Abstract
Neurotransmitters, such as γ-aminobutyric acid, glutamate, acetyl choline, glycine and the monoamines, facilitate the crosstalk within the central nervous system. The designated neurotransmitter transporters (NTTs) both release and take up neurotransmitters to and from the synaptic cleft. NTT dysfunction can lead to severe pathophysiological consequences, e.g. epilepsy, intellectual disability, or Parkinson’s disease. Genetic point mutations in NTTs have recently been associated with the onset of various neurological disorders. Some of these mutations trigger folding defects in the NTT proteins. Correct folding is a prerequisite for the export of NTTs from the endoplasmic reticulum (ER) and the subsequent trafficking to their pertinent site of action, typically at the plasma membrane. Recent studies have uncovered some of the key features in the molecular machinery responsible for transporter protein folding, e.g., the role of heat shock proteins in fine-tuning the ER quality control mechanisms in cells. The therapeutic significance of understanding these events is apparent from the rising number of reports, which directly link different pathological conditions to NTT misfolding. For instance, folding-deficient variants of the human transporters for dopamine or GABA lead to infantile parkinsonism/dystonia and epilepsy, respectively. From a therapeutic point of view, some folding-deficient NTTs are amenable to functional rescue by small molecules, known as chemical and pharmacological chaperones.
Collapse
Affiliation(s)
- Shreyas Bhat
- Institute of Pharmacology and the Gaston H. Glock Research Laboratories for Exploratory Drug Development, Center of Physiology and Pharmacology, Medical University of Vienna, A-1090 Vienna, Austria
| | - Ali El-Kasaby
- Institute of Pharmacology and the Gaston H. Glock Research Laboratories for Exploratory Drug Development, Center of Physiology and Pharmacology, Medical University of Vienna, A-1090 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, A-1090 Vienna, Austria
| | - Sonja Sucic
- Institute of Pharmacology and the Gaston H. Glock Research Laboratories for Exploratory Drug Development, Center of Physiology and Pharmacology, Medical University of Vienna, A-1090 Vienna, Austria.
| |
Collapse
|
23
|
Chen R, Ferris MJ, Wang S. Dopamine D2 autoreceptor interactome: Targeting the receptor complex as a strategy for treatment of substance use disorder. Pharmacol Ther 2020; 213:107583. [PMID: 32473160 PMCID: PMC7434700 DOI: 10.1016/j.pharmthera.2020.107583] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/14/2019] [Accepted: 05/11/2020] [Indexed: 02/06/2023]
Abstract
Dopamine D2 autoreceptors (D2ARs), located in somatodendritic and axon terminal compartments of dopamine (DA) neurons, function to provide a negative feedback regulatory control on DA neuron firing, DA synthesis, reuptake and release. Dysregulation of D2AR-mediated DA signaling is implicated in vulnerability to substance use disorder (SUD). Due to the extreme low abundance of D2ARs compared to postsynaptic D2 receptors (D2PRs) and the lack of experimental tools to differentiate the signaling of D2ARs from D2PRs, the regulation of D2ARs by drugs of abuse is poorly understood. The recent availability of conditional D2AR knockout mice and newly developed virus-mediated gene delivery approaches have provided means to specifically study the function of D2ARs at the molecular, cellular and behavioral levels. There is a growing revelation of novel mechanisms and new proteins that mediate D2AR activity, suggesting that D2ARs act cooperatively with an array of membrane and intracellular proteins to tightly control DA transmission. This review highlights D2AR-interacting partners including transporters, G-protein-coupled receptors, ion channels, intracellular signaling modulators, and protein kinases. The complexity of the D2AR interaction network illustrates the functional divergence of D2ARs. Pharmacological targeting of multiple D2AR-interacting partners may be more effective to restore disrupted DA homeostasis by drugs of abuse.
Collapse
Affiliation(s)
- Rong Chen
- Dept. of Physiology & Pharmacology, Wake Forest School of Medicine, Winston Salem, NC 27157, United States of America; Center for the Neurobiology of Addiction Treatment, Wake Forest School of Medicine, Winston Salem, NC 27157, United States of America.
| | - Mark J Ferris
- Dept. of Physiology & Pharmacology, Wake Forest School of Medicine, Winston Salem, NC 27157, United States of America; Center for the Neurobiology of Addiction Treatment, Wake Forest School of Medicine, Winston Salem, NC 27157, United States of America
| | - Shiyu Wang
- Dept. of Physiology & Pharmacology, Wake Forest School of Medicine, Winston Salem, NC 27157, United States of America
| |
Collapse
|
24
|
Areal LB, Blakely RD. Neurobehavioral changes arising from early life dopamine signaling perturbations. Neurochem Int 2020; 137:104747. [PMID: 32325191 PMCID: PMC7261509 DOI: 10.1016/j.neuint.2020.104747] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/10/2020] [Revised: 04/11/2020] [Accepted: 04/14/2020] [Indexed: 12/11/2022]
Abstract
Dopamine (DA) signaling is critical to the modulation of multiple brain functions including locomotion, reinforcement, attention and cognition. The literature provides strong evidence that altered DA availability and actions can impact normal neurodevelopment, with both early and enduring consequences on anatomy, physiology and behavior. An appreciation for the developmental contributions of DA signaling to brain development is needed to guide efforts to preclude and remedy neurobehavioral disorders, such as attention-deficit/hyperactivity disorder, addiction, bipolar disorder, schizophrenia and autism spectrum disorder, each of which exhibits links to DA via genetic, cellular and/or pharmacological findings. In this review, we highlight research pursued in preclinical models that use genetic and pharmacological approaches to manipulate DA signaling at sensitive developmental stages, leading to changes at molecular, circuit and/or behavioral levels. We discuss how these alterations can be aligned with traits displayed by neuropsychiatric diseases. Lastly, we review human studies that evaluate contributions of developmental perturbations of DA systems to increased risk for neuropsychiatric disorders.
Collapse
Affiliation(s)
- Lorena B Areal
- Department of Biomedical Science, Florida Atlantic University, Jupiter, FL, 33458, USA
| | - Randy D Blakely
- Department of Biomedical Science, Florida Atlantic University, Jupiter, FL, 33458, USA; Brain Institute, Florida Atlantic University, Jupiter, FL, 33458, USA.
| |
Collapse
|
25
|
Fagan RR, Kearney PJ, Melikian HE. In Situ Regulated Dopamine Transporter Trafficking: There's No Place Like Home. Neurochem Res 2020; 45:1335-1343. [PMID: 32146647 DOI: 10.1007/s11064-020-03001-6] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/08/2019] [Revised: 02/24/2020] [Accepted: 02/26/2020] [Indexed: 12/13/2022]
Abstract
Dopamine (DA) is critical for motivation, reward, movement initiation, and learning. Mechanisms that control DA signaling have a profound impact on these important behaviors, and additionally play a role in DA-related neuropathologies. The presynaptic SLC6 DA transporter (DAT) limits extracellular DA levels by clearing released DA, and is potently inhibited by addictive and therapeutic psychostimulants. Decades of evidence support that the DAT is subject to acute regulation by a number of signaling pathways, and that endocytic trafficking strongly regulates DAT availability and function. DAT trafficking studies have been performed in a variety of model systems, including both in vitro and ex vivo preparations. In this review, we focus on the breadth of DAT trafficking studies, with specific attention to, and comparison of, how context may influence DAT's response to different stimuli. In particular, this overview highlights that stimulated DAT trafficking not only differs between in vitro and ex vivo environments, but also is influenced by both sex and anatomical subregions.
Collapse
Affiliation(s)
- Rita R Fagan
- Brudnick Neuropsychiatric Research Institute, Department of Neurobiology, University of Massachusetts Medical School, Worcester, MA, 01605, USA
| | - Patrick J Kearney
- Brudnick Neuropsychiatric Research Institute, Department of Neurobiology, University of Massachusetts Medical School, Worcester, MA, 01605, USA
| | - Haley E Melikian
- Brudnick Neuropsychiatric Research Institute, Department of Neurobiology, University of Massachusetts Medical School, Worcester, MA, 01605, USA.
| |
Collapse
|
26
|
Lebowitz JJ, Khoshbouei H. Heterogeneity of dopamine release sites in health and degeneration. Neurobiol Dis 2019; 134:104633. [PMID: 31698055 DOI: 10.1016/j.nbd.2019.104633] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/14/2019] [Revised: 09/12/2019] [Accepted: 10/02/2019] [Indexed: 02/06/2023] Open
Abstract
Despite comprising only ~ 0.001% of all neurons in the human brain, ventral midbrain dopamine neurons exert a profound influence on human behavior and cognition. As a neuromodulator, dopamine selectively inhibits or enhances synaptic signaling to coordinate neural output for action, attention, and affect. Humans invariably lose brain dopamine during aging, and this can be exacerbated in disease states such as Parkinson's Disease. Further, it is well established in multiple disease states that cell loss is selective for a subset of highly sensitive neurons within the nigrostriatal dopamine tract. Regional differences in dopamine tone are regulated pre-synaptically, with subcircuits of projecting dopamine neurons exhibiting distinct molecular and physiological signatures. Specifically, proteins at dopamine release sites that synthesize and package cytosolic dopamine, modulate its release and reuptake, and alter neuronal excitability show regional differences that provide linkages to the observed sensitivity to neurodegeneration. The aim of this review is to outline the major components of dopamine homeostasis at neurotransmitter release sites and describe the regional differences most relevant to understanding why some, but not all, dopamine neurons exhibit heightened vulnerability to neurodegeneration.
Collapse
Affiliation(s)
- Joseph J Lebowitz
- Department of Neuroscience, University of Florida College of Medicine, Gainesville, FL 32610, USA
| | - Habibeh Khoshbouei
- Department of Neuroscience, University of Florida College of Medicine, Gainesville, FL 32610, USA.
| |
Collapse
|
27
|
Simmler LD, Blakely RD. The SERT Met172 Mouse: An Engineered Model To Elucidate the Contributions of Serotonin Signaling to Cocaine Action. ACS Chem Neurosci 2019; 10:3053-3060. [PMID: 30817127 DOI: 10.1021/acschemneuro.9b00005] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022] Open
Abstract
Cocaine abuse and addiction remain highly prevalent and, unfortunately, poorly treated. It is well-known that essential aspects of cocaine's addictive actions involve the drug's ability to block the presynaptic dopamine (DA) transporter (DAT), thereby elevating extracellular levels of DA in brain circuits that subserve reward, reinforcement, and habit. Less well appreciated are the multiple DA-independent actions of cocaine, activities that we and others believe contribute key pieces to the puzzle of cocaine addiction, treatment, and relapse. In particular, a significant body of work points to altered serotonin (5-HT) signaling as one such component, not surprising given that, relative to DAT, cocaine acts as potently to block the 5-HT transporter (SERT) as to block DAT, and thereby elevates extracellular 5-HT levels throughout the brain when reward-eliciting DA elevations occur. To elucidate the contribution of SERT antagonism to the actions of cocaine, we engineered a mouse model that significantly reduces cocaine potency at SERT without disrupting the expression or function of SERT in vivo. In this short Perspective, we review the rationale for development of the SERT Met172 model, the studies that document the pharmacological impact of the Ile172Met substitution in vitro and in vivo, and our findings with the model that demonstrate serotonergic contributions to the genetic, physiological, and behavioral actions of cocaine.
Collapse
Affiliation(s)
- Linda D. Simmler
- Department of Basic Neurosciences, University of Geneva, Rue Michel-Servet 1, 1206 Geneva, Switzerland
| | - Randy D. Blakely
- Department of Biomedical Science, Charles E. Schmidt College of Medicine and Brain Institute, Florida Atlantic University, Jupiter, Florida 33458, United States
| |
Collapse
|
28
|
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
|
29
|
DiCarlo GE, Aguilar JI, Matthies HJ, Harrison FE, Bundschuh KE, West A, Hashemi P, Herborg F, Rickhag M, Chen H, Gether U, Wallace MT, Galli A. Autism-linked dopamine transporter mutation alters striatal dopamine neurotransmission and dopamine-dependent behaviors. J Clin Invest 2019; 129:3407-3419. [PMID: 31094705 DOI: 10.1172/jci127411] [Citation(s) in RCA: 98] [Impact Index Per Article: 16.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022] Open
Abstract
The precise regulation of synaptic dopamine (DA) content by the dopamine transporter (DAT) ensures the phasic nature of the DA signal, which underlies the ability of DA to encode reward prediction error, thereby driving motivation, attention, and behavioral learning. Disruptions to the DA system are implicated in a number of neuropsychiatric disorders, including attention deficit hyperactivity disorder (ADHD) and, more recently, Autism Spectrum Disorder (ASD). An ASD-associated de novo mutation in the SLC6A3 gene resulting in a threonine to methionine substitution at site 356 (DAT T356M) was recently identified and has been shown to drive persistent reverse transport of DA (i.e. anomalous DA efflux) in transfected cells and to drive hyperlocomotion in Drosophila melanogaster. A corresponding mutation in the leucine transporter, a DAT-homologous transporter, promotes an outward-facing transporter conformation upon substrate binding, a conformation possibly underlying anomalous dopamine efflux. Here we investigated in vivo the impact of this ASD-associated mutation on DA signaling and ASD-associated behaviors. We found that mice homozygous for this mutation display impaired striatal DA neurotransmission and altered DA-dependent behaviors that correspond with some of the behavioral phenotypes observed in ASD.
Collapse
Affiliation(s)
| | - Jenny I Aguilar
- Department of Pharmacology, Vanderbilt University, Nashville, Tennessee, USA.,Department of Surgery, University of Alabama at Birmingham, Birmingham, Alabama, USA
| | - Heinrich Jg Matthies
- Department of Surgery, University of Alabama at Birmingham, Birmingham, Alabama, USA
| | - Fiona E Harrison
- Vanderbilt University Brain Institute, Nashville, Tennessee, USA.,Department of Medicine, Vanderbilt University Medical Center, Nashville, Tennessee, USA
| | - Kyle E Bundschuh
- Vanderbilt University Brain Institute, Nashville, Tennessee, USA
| | - Alyssa West
- Department of Chemistry and Biochemistry, University of South Carolina, Columbia, South Carolina, USA
| | - Parastoo Hashemi
- Department of Chemistry and Biochemistry, University of South Carolina, Columbia, South Carolina, USA
| | - Freja Herborg
- Molecular Neuropharmacology and Genetics Laboratory, Department of Neuroscience, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark
| | - Mattias Rickhag
- Molecular Neuropharmacology and Genetics Laboratory, Department of Neuroscience, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark
| | - Hao Chen
- DRI Biosciences Corp., Frederick, Maryland, USA
| | - Ulrik Gether
- Molecular Neuropharmacology and Genetics Laboratory, Department of Neuroscience, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark
| | - Mark T Wallace
- Vanderbilt University Brain Institute, Nashville, Tennessee, USA.,Department of Hearing and Speech Sciences, Vanderbilt University, Nashville, Tennessee, USA
| | - Aurelio Galli
- Department of Surgery, University of Alabama at Birmingham, Birmingham, Alabama, USA
| |
Collapse
|
30
|
Identification and functional characterization of solute carrier family 6 genes in Ciona savignyi. Gene 2019; 705:142-148. [PMID: 31026570 DOI: 10.1016/j.gene.2019.04.056] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/22/2018] [Revised: 03/10/2019] [Accepted: 04/19/2019] [Indexed: 12/12/2022]
Abstract
The solute carrier 6 (SLC6) gene family, functioning as neurotransmitter transporters, plays the crucial roles in neurotransmission, cellular and organismal homeostasis. In this study, we found an expansion of SLC6 family gene in the genome of chordate invertebrate Ciona savignyi. A total of 40 candidate genes including 29 complete and 11 putative genes were identified as SLC6 family gene homologs. Phylogenetic analysis revealed that most of these Ciona SLC6 genes were highly conserved with the vertebrate ones, although gene duplication and gene losses did exist. Four genes were selected from SLC6 subfamilies to be further investigated for their functional characteristics on cell growth and migration through overexpression approach in cultured cell lines. The results showed both SLC6A7 and SLC6A17 from amino acid transporters AA1 and AA2 sub-families, respectively, significantly suppressed the cell proliferation and migration. While SLC6A1 and SLC6A4, which were classified into GABA and monoamine transporters, respectively, did not affect the cell proliferation and migration in HEK293T, HeLa, and MCF7 cells. The whole set of C. savignyi SLC6 genes identified in this study provides an important genomic resource for future biochemical, physiological, and phylogenetic studies on SLC6 gene family. Our experimental data demonstrated that Ciona amino acid transporters, such as SLC6A7 and SLC6A17, were essential for cell physiology and behaviors, indicating their crucially potential roles in the control of cell proliferation and migration during ascidian embryogenesis.
Collapse
|
31
|
Stewart A, Davis GL, Gresch PJ, Katamish RM, Peart R, Rabil MJ, Gowrishankar R, Carroll FI, Hahn MK, Blakely RD. Serotonin transporter inhibition and 5-HT 2C receptor activation drive loss of cocaine-induced locomotor activation in DAT Val559 mice. Neuropsychopharmacology 2019; 44:994-1006. [PMID: 30578419 PMCID: PMC6462012 DOI: 10.1038/s41386-018-0301-8] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/19/2018] [Revised: 11/19/2018] [Accepted: 12/15/2018] [Indexed: 11/18/2022]
Abstract
Dopamine (DA) signaling dysfunction is believed to contribute to multiple neuropsychiatric disorders including attention-deficit/hyperactivity disorder (ADHD). The rare DA transporter (DAT) coding substitution Ala559Val found in subjects with ADHD, bipolar disorder and autism, promotes anomalous DA efflux in vitro and, in DAT Val559 mice, leads to increased reactivity to imminent handling, waiting impulsivity, and enhanced motivation for reward. Here, we report that, in contrast to amphetamine and methylphenidate, which induce significant locomotor activation, cocaine administration to these mice elicits no locomotor effects, despite retention of conditioned place preference (CPP). Additionally, cocaine fails to elevate extracellular DA. Given that amphetamine and methylphenidate, unlike cocaine, lack high-affinity interactions with the serotonin (5-HT) transporter (SERT), we hypothesized that the lack of cocaine-induced hyperlocomotion in DAT Val559 mice arises from SERT blockade and augmented 5-HT signaling relative to cocaine actions on wildtype animals. Consistent with this idea, the SERT blocker fluoxetine abolished methylphenidate-induced locomotor activity in DAT Val559 mice, mimicking the effects seen with cocaine. Additionally, a cocaine analog (RTI-113) with greater selectivity for DAT over SERT retains locomotor activation in DAT Val559 mice. Furthermore, genetic elimination of high-affinity cocaine interactions at SERT in DAT Val559 mice, or specific inhibition of 5-HT2C receptors in these animals, restored cocaine-induced locomotion, but did not restore cocaine-induced elevations of extracellular DA. Our findings reveal a significant serotonergic plasticity arising in the DAT Val559 model that involves enhanced 5-HT2C signaling, acting independently of striatal DA release, capable of suppressing the activity of cocaine-sensitive motor circuits.
Collapse
Affiliation(s)
- Adele Stewart
- 0000 0004 0635 0263grid.255951.fDepartment of Biomedical Science, Florida Atlantic University, Jupiter, FL USA
| | - Gwynne L. Davis
- 0000 0004 0635 0263grid.255951.fDepartment of Biomedical Science, Florida Atlantic University, Jupiter, FL USA ,0000 0001 2264 7217grid.152326.1Neuroscience Graduate Program, Vanderbilt University, Nashville, TN USA
| | - Paul J. Gresch
- 0000 0004 0635 0263grid.255951.fDepartment of Biomedical Science, Florida Atlantic University, Jupiter, FL USA ,0000 0004 0635 0263grid.255951.fBrain Institute, Florida Atlantic University, Jupiter, FL USA
| | - Rania M. Katamish
- 0000 0004 0635 0263grid.255951.fDepartment of Biomedical Science, Florida Atlantic University, Jupiter, FL USA
| | - Rodeania Peart
- 0000 0004 0635 0263grid.255951.fWilkes Honors College, Florida Atlantic University, Jupiter, FL USA
| | - Maximilian J. Rabil
- 0000 0004 0635 0263grid.255951.fDepartment of Biomedical Science, Florida Atlantic University, Jupiter, FL USA
| | - Raajaram Gowrishankar
- 0000 0004 0635 0263grid.255951.fDepartment of Biomedical Science, Florida Atlantic University, Jupiter, FL USA ,0000 0001 2264 7217grid.152326.1Neuroscience Graduate Program, Vanderbilt University, Nashville, TN USA ,0000 0001 2264 7217grid.152326.1International Scholars Program, Vanderbilt University, Nashville, TN USA
| | - F. Ivy Carroll
- 0000000100301493grid.62562.35Research Triangle Institute, Research Triangle Park, NC USA
| | - Maureen K. Hahn
- 0000 0004 0635 0263grid.255951.fDepartment of Biomedical Science, Florida Atlantic University, Jupiter, FL USA ,0000 0004 0635 0263grid.255951.fBrain Institute, Florida Atlantic University, Jupiter, FL USA
| | - Randy D. Blakely
- 0000 0004 0635 0263grid.255951.fDepartment of Biomedical Science, Florida Atlantic University, Jupiter, FL USA ,0000 0004 0635 0263grid.255951.fBrain Institute, Florida Atlantic University, Jupiter, FL USA
| |
Collapse
|
32
|
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
|
33
|
Thal LB, Tomlinson ID, Quinlan MA, Kovtun O, Blakely RD, Rosenthal SJ. Single Quantum Dot Imaging Reveals PKCβ-Dependent Alterations in Membrane Diffusion and Clustering of an Attention-Deficit Hyperactivity Disorder/Autism/Bipolar Disorder-Associated Dopamine Transporter Variant. ACS Chem Neurosci 2019; 10:460-471. [PMID: 30153408 PMCID: PMC6411462 DOI: 10.1021/acschemneuro.8b00350] [Citation(s) in RCA: 22] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022] Open
Abstract
The dopamine transporter (DAT) is a transmembrane protein that terminates dopamine signaling in the brain by driving rapid dopamine reuptake into presynaptic nerve terminals. Several lines of evidence indicate that DAT dysfunction is linked to neuropsychiatric disorders such as attention-deficit/hyperactivity disorder (ADHD), bipolar disorder (BPD), and autism spectrum disorder (ASD). Indeed, individuals with these disorders have been found to express the rare, functional DAT coding variant Val559, which confers anomalous dopamine efflux (ADE) in vitro and in vivo. To elucidate the impact of the DAT Val559 variant on membrane diffusion dynamics, we implemented our antagonist-conjugated quantum dot (QD) labeling approach to monitor the lateral mobility of single particle-labeled transporters in transfected HEK-293 and SK-N-MC cells. Our results demonstrate significantly higher diffusion coefficients of DAT Val559 compared to those of DAT Ala559, effects likely determined by elevated N-terminal transporter phosphorylation. We also provide pharmacological evidence that PKCβ-mediated signaling supports enhanced DAT Val559 membrane diffusion rates. Additionally, our results are complimented with diffusion rates of phosphomimicked and phosphorylation-occluded DAT variants. Furthermore, we show DAT Val559 has a lower propensity for membrane clustering, which may be caused by a mutation-derived shift out of membrane microdomains leading to faster lateral membrane diffusion rates. These findings further demonstrate a functional impact of DAT Val559 and suggest that changes in transporter localization and lateral mobility may sustain ADE and contribute to alterations in dopamine signaling underlying multiple neuropsychiatric disorders.
Collapse
|
34
|
Mulvihill KG. Presynaptic regulation of dopamine release: Role of the DAT and VMAT2 transporters. Neurochem Int 2018; 122:94-105. [PMID: 30465801 DOI: 10.1016/j.neuint.2018.11.004] [Citation(s) in RCA: 42] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/22/2018] [Revised: 10/28/2018] [Accepted: 11/08/2018] [Indexed: 01/23/2023]
Abstract
The signaling dynamics of the neurotransmitter dopamine has been established to have an important role in a variety of behavioural processes including motor control, cognition, and emotional processing. Key regulators of transmitter release and the signaling dynamics of dopamine are the plasma membrane reuptake transporter (DAT) and the vesicular monoamine transporter (VMAT2). These proteins serve to remove dopamine molecules from the extracellular and cytosolic space, respectively and both determine the amount of transmitter released from synaptic vesicles. This review provides an overview of how these transporter proteins are involved in molecular regulation and function together to govern the dynamics of vesicular release with opposing effects on the quantal size and extracellular concentration of dopamine. These transporter proteins are both focal points of convergence for a variety of regulatory molecular cascades as well as targets for many pharmacological agents. The ratio between these transporters is argued to be useful as a molecular marker for delineating dopamine functional subsystems that may differ in transmitter release patterns.
Collapse
Affiliation(s)
- Kevin G Mulvihill
- Department of Psychology, Brock University, St. Catharines, ON, L2S 3A1, Canada.
| |
Collapse
|
35
|
Kovtun O, Tomlinson ID, Bailey DM, Thal LB, Ross EJ, Harris L, Frankland MP, Ferguson RS, Glaser Z, Greer J, Rosenthal SJ. Single Quantum Dot Tracking Illuminates Neuroscience at the Nanoscale. Chem Phys Lett 2018; 706:741-752. [PMID: 30270931 PMCID: PMC6157616 DOI: 10.1016/j.cplett.2018.06.019] [Citation(s) in RCA: 25] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/15/2022]
Abstract
The use of nanometer-sized semiconductor crystals, known as quantum dots, allows us to directly observe individual biomolecular transactions through a fluorescence microscope. Here, we review the evolution of single quantum dot tracking over the past two decades, highlight key biophysical discoveries facilitated by quantum dots, briefly discuss biochemical and optical implementation strategies for a single quantum dot tracking experiment, and report recent accomplishments of our group at the interface of molecular neuroscience and nanoscience.
Collapse
Affiliation(s)
- Oleg Kovtun
- Departments of Chemistry, Chemical Biology, Vanderbilt University
- Departments of Vanderbilt Institute of Nanoscale Science and Engineering
| | - Ian D. Tomlinson
- Departments of Chemistry, Chemical Biology, Vanderbilt University
- Departments of Vanderbilt Institute of Nanoscale Science and Engineering
| | - Danielle M. Bailey
- Departments of Chemistry, Chemical Biology, Vanderbilt University
- Departments of Pharmacology, Chemical Biology, Vanderbilt University
- Departments of Vanderbilt Institute of Nanoscale Science and Engineering
| | - Lucas B. Thal
- Departments of Chemistry, Chemical Biology, Vanderbilt University
- Departments of Vanderbilt Institute of Nanoscale Science and Engineering
- Departments of Vanderbilt Institute of Chemical Biology, Vanderbilt University, Nashville, TN
| | - Emily J. Ross
- Departments of Hudson Alpha Institute for Biotechnology, Huntsville, AL
| | - Lauren Harris
- Departments of Vanderbilt Institute of Chemical Biology, Vanderbilt University, Nashville, TN
| | | | | | - Zachary Glaser
- Departments of Chemistry, Chemical Biology, Vanderbilt University
| | - Jonathan Greer
- Departments of Chemistry, Chemical Biology, Vanderbilt University
| | - Sandra J. Rosenthal
- Departments of Chemistry, Chemical Biology, Vanderbilt University
- Departments of Pharmacology, Chemical Biology, Vanderbilt University
- Departments of Chemical and Biomolecular Engineering, Chemical Biology, Vanderbilt University
- Departments of Physics and Astronomy, Chemical Biology, Vanderbilt University
- Departments of Vanderbilt Institute of Nanoscale Science and Engineering
- Departments of Vanderbilt Institute of Chemical Biology, Vanderbilt University, Nashville, TN
| |
Collapse
|
36
|
Refai O, Blakely RD. Blockade and reversal of swimming-induced paralysis in C. elegans by the antipsychotic and D2-type dopamine receptor antagonist azaperone. Neurochem Int 2018; 123:59-68. [PMID: 29800604 DOI: 10.1016/j.neuint.2018.05.013] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/22/2018] [Revised: 05/21/2018] [Accepted: 05/21/2018] [Indexed: 10/16/2022]
Abstract
The catecholamine neurotransmitter dopamine (DA) exerts powerful modulatory control of physiology and behavior across phylogeny. Perturbations of DA signaling in humans are associated with multiple neurodegenerative and behavioral disorders, including Parkinson's disease, attention-deficit/hyperactivity disorder, addiction and schizophrenia. In the nematode C. elegans, DA signaling regulates mating behavior, learning, food seeking and locomotion. Previously, we demonstrated that loss of function mutations in the dat-1 gene that encodes the presynaptic DA transporter (DAT-1) results in a rapid cessation of movement when animals are placed in water, termed Swimming Induced Paralysis (Swip). Loss of function mutations in genes that support DA biosynthesis, DA vesicular packaging and DA action at the extrasynaptic D2-type DA receptor DOP-3 suppress Swip in dat-1 animals, consistent with paralysis as arising from excessive DA signaling. Although animals grown on the vesicular monoamine transporter antagonist reserpine diminish Swip, the drug must be applied chronically, can impact the signaling of multiple biogenic amines, and has been reported to have penetrant, off-target actions. Here, we demonstrate that the antipsychotic drug azaperone potently and rapidly suppresses Swip behavior in either dat-1 mutants, as well as in wildtype animals treated with the DAT-1 antagonist nisoxetine, with genetic experiments consistent with DOP-3 antagonism as the mechanism of Swip suppression. Reversal of Swip in previously paralyzed dat-1 animals by azaperone application demonstrates an otherwise functionally-intact swimming circuit in these mutants. Finally, whereas azaperone suppresses DA-dependent Swip, the drug fails to attenuate the DA-independent paralysis induced by βPEA, aldicarb or genetic disruption of γ-aminobutyric acid (GABA) signaling. We discuss our findings with respect to the use of azaperone as a potent and selective tool in the identification and analysis of presynaptic mechanisms that regulate DA signaling.
Collapse
Affiliation(s)
- Osama Refai
- Department of Biomedical Science, Charles E. Schmidt College of Medicine, Florida Atlantic University, FL, USA
| | - Randy D Blakely
- Department of Biomedical Science, Charles E. Schmidt College of Medicine, Florida Atlantic University, FL, USA; Brain Institute, Florida Atlantic University, Jupiter, FL, 33458, USA.
| |
Collapse
|
37
|
Region-Specific Regulation of Presynaptic Dopamine Homeostasis by D 2 Autoreceptors Shapes the In Vivo Impact of the Neuropsychiatric Disease-Associated DAT Variant Val559. J Neurosci 2018; 38:5302-5312. [PMID: 29739866 DOI: 10.1523/jneurosci.0055-18.2018] [Citation(s) in RCA: 25] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/09/2018] [Revised: 03/19/2018] [Accepted: 04/14/2018] [Indexed: 12/21/2022] Open
Abstract
Disruptions of dopamine (DA) signaling contribute to a broad spectrum of neuropsychiatric disorders, including attention-deficit hyperactivity disorder (ADHD), addiction, bipolar disorder, and schizophrenia. Despite evidence that risk for these disorders derives from heritable variation in DA-linked genes, a better understanding is needed of the molecular and circuit context through which gene variation drives distinct disease traits. Previously, we identified the DA transporter (DAT) variant Val559 in subjects with ADHD and established that the mutation supports anomalous DAT-mediated DA efflux (ADE). Here, we demonstrate that region-specific contributions of D2 autoreceptors (D2AR) to presynaptic DA homeostasis dictate the consequences of Val559 expression in adolescent male mice. We show that activation of D2ARs in the WT dorsal striatum (DS), but not ventral striatum (VS), increases DAT phosphorylation and surface trafficking. In contrast, the activity of tyrosine hydroxylase (TH) is D2AR-dependent in both regions. In the DS but not VS of Val559 mice, tonic activation of D2ARs drives a positive feedback loop that promotes surface expression of efflux-prone DATs, raising extracellular DA levels and overwhelming DAT-mediated DA clearance capacity. Whereas D2ARs that regulate DAT are tonically activated in the Val559 DS, D2ARs that regulate TH become desensitized, allowing maintenance of cytosolic DA needed to sustain ADE. Together with prior findings, our results argue for distinct D2AR pools that regulate DA synthesis versus DA release and inactivation and offer a clear example of how the penetrance of gene variation can be limited to a subset of expression sites based on differences in intersecting regulatory networks.SIGNIFICANCE STATEMENT Altered dopamine (DA) signaling has been linked to multiple neuropsychiatric disorders. In an effort to understand and model disease-associated DAergic disturbances, we previously screened the DA transporter (DAT) in subjects with attention-deficit hyperactivity disorder (ADHD) and identified multiple, functionally impactful, coding variants. One of these variants, Val559, supports anomalous DA efflux (ADE) and in transgenic mice leads to changes in locomotor patterns, psychostimulant sensitivity, and impulsivity. Here, we show that the penetrance of Val559 ADE is dictated by region-specific differences in how presynaptic D2-type autoreceptors (D2ARs) constrain DA signaling, biasing phenotypic effects to dorsal striatal projections. The Val559 model illustrates how the impact of genetic variation underlying neuropsychiatric disorders can be shaped by the differential engagement of synaptic regulatory mechanisms.
Collapse
|
38
|
Mackie P, Lebowitz J, Saadatpour L, Nickoloff E, Gaskill P, Khoshbouei H. The dopamine transporter: An unrecognized nexus for dysfunctional peripheral immunity and signaling in Parkinson's Disease. Brain Behav Immun 2018; 70:21-35. [PMID: 29551693 PMCID: PMC5953824 DOI: 10.1016/j.bbi.2018.03.020] [Citation(s) in RCA: 51] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/08/2017] [Revised: 03/13/2018] [Accepted: 03/14/2018] [Indexed: 02/06/2023] Open
Abstract
The second-most common neurodegenerative disease, Parkinson's Disease (PD) has three hallmarks: dysfunctional dopamine transmission due, at least in part, to dopamine neuron degeneration; intracellular inclusions of α-synuclein aggregates; and neuroinflammation. The origin and interplay of these features remains a puzzle, as does the underlying mechanism of PD pathogenesis and progression. When viewed in the context of neuroimmunology, dopamine also plays a role in regulating peripheral immune cells. Intriguingly, plasma dopamine levels are altered in PD, suggesting collateral dysregulation of peripheral dopamine transmission. The dopamine transporter (DAT), the main regulator of dopaminergic tone in the CNS, is known to exist in lymphocytes and monocytes/macrophages, but little is known about peripheral DAT biology or how DAT regulates the dopaminergic tone, much less how peripheral DAT alters immune function. Our review is guided by the hypothesis that dysfunctional peripheral dopamine signaling might be linked to the dysfunctional immune responses in PD and thereby suggests a potential bidirectional communication between central and peripheral dopamine systems. This review seeks to foster new perspectives concerning PD pathogenesis and progression.
Collapse
Affiliation(s)
- Phillip Mackie
- University of Florida College of Medicine, Department of Neuroscience, Gainesville, FL 32611, United States
| | - Joe Lebowitz
- University of Florida College of Medicine, Department of Neuroscience, Gainesville, FL 32611, United States
| | - Leila Saadatpour
- University of Florida College of Medicine, Department of Neuroscience, Gainesville, FL 32611, United States
| | - Emily Nickoloff
- Department of Pharmacology and Physiology, Drexel University College of Medicine, Philadelphia, PA 19102, United States
| | - Peter Gaskill
- Department of Pharmacology and Physiology, Drexel University College of Medicine, Philadelphia, PA 19102, United States
| | - Habibeh Khoshbouei
- University of Florida College of Medicine, Department of Neuroscience, Gainesville, FL 32611, United States.
| |
Collapse
|
39
|
Herborg F, Andreassen TF, Berlin F, Loland CJ, Gether U. Neuropsychiatric disease-associated genetic variants of the dopamine transporter display heterogeneous molecular phenotypes. J Biol Chem 2018; 293:7250-7262. [PMID: 29559554 DOI: 10.1074/jbc.ra118.001753] [Citation(s) in RCA: 37] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/05/2018] [Revised: 03/15/2018] [Indexed: 12/14/2022] Open
Abstract
Genetic factors are known to significantly contribute to the etiology of psychiatric diseases such as attention deficit hyperactivity disorder (ADHD) and autism spectrum and bipolar disorders, but the underlying molecular processes remain largely elusive. The dopamine transporter (DAT) has received continuous attention as a potential risk factor for psychiatric disease, as it is critical for dopamine homeostasis and serves as principal target for ADHD medications. Constrain metrics for the DAT-encoding gene, solute carrier family 6 member 3 (SLC6A3), indicate that missense mutations are under strong negative selection, pointing to pathophysiological outcomes when DAT function is compromised. Here, we systematically characterized six rare genetic variants of DAT (I312F, T356M, D421N, A559V, E602G, and R615C) identified in patients with neuropsychiatric disorders. We evaluated dopamine uptake and ligand interactions, along with ion coordination and electrophysiological properties, to elucidate functional phenotypes, and applied Zn2+ exposure and a substituted cysteine-accessibility approach to identify shared structural changes. Three variants (I312F, T356M, and D421N) exhibited impaired dopamine uptake associated with changes in ligand binding, ion coordination, and distinct conformational disturbances. Remarkably, we found that all three variants displayed gain-of-function electrophysiological phenotypes. I312F mediated an increased uncoupled anion conductance previously suggested to modulate neuronal excitability. T356M and D421N both mediated a cocaine-sensitive leakage of cations, which for T356M was potentiated by Zn2+, concurrent with partial functional rescue. Collectively, our findings support that gain of disruptive functions due to missense mutations in SLC6A3 may be key to understanding how dopaminergic dyshomeostasis arises in heterozygous carriers.
Collapse
Affiliation(s)
- Freja Herborg
- Molecular Neuropharmacology and Genetics Laboratory, Department of Neuroscience, Faculty of Health and Medical Sciences, Panum Institute-Maersk Tower 07.05, University of Copenhagen, DK-2200 Copenhagen, Denmark.
| | - Thorvald F Andreassen
- Molecular Neuropharmacology and Genetics Laboratory, Department of Neuroscience, Faculty of Health and Medical Sciences, Panum Institute-Maersk Tower 07.05, University of Copenhagen, DK-2200 Copenhagen, Denmark
| | - Frida Berlin
- Molecular Neuropharmacology and Genetics Laboratory, Department of Neuroscience, Faculty of Health and Medical Sciences, Panum Institute-Maersk Tower 07.05, University of Copenhagen, DK-2200 Copenhagen, Denmark
| | - Claus J Loland
- Molecular Neuropharmacology and Genetics Laboratory, Department of Neuroscience, Faculty of Health and Medical Sciences, Panum Institute-Maersk Tower 07.05, University of Copenhagen, DK-2200 Copenhagen, Denmark
| | - Ulrik Gether
- Molecular Neuropharmacology and Genetics Laboratory, Department of Neuroscience, Faculty of Health and Medical Sciences, Panum Institute-Maersk Tower 07.05, University of Copenhagen, DK-2200 Copenhagen, Denmark.
| |
Collapse
|
40
|
Abstract
Background Much of the structure-based mechanistic understandings of the function of SLC6A neurotransmitter transporters emerged from the study of their bacterial LeuT-fold homologs. It has become evident, however, that structural differences such as the long N- and C-termini of the eukaryotic neurotransmitter transporters are involved in an expanded set of functional properties to the eukaryotic transporters. These functional properties are not shared by the bacterial homologs, which lack the structural elements that appeared later in evolution. However, mechanistic insights into some of the measured functional properties of the eukaryotic transporters that have been suggested to involve these structural elements are sparse or merely descriptive. Results To learn how the structural elements added in evolution enable mechanisms of the eukaryotic transporters in ways not shared with their bacterial LeuT-like homologs, we focused on the human dopamine transporter (hDAT) as a prototype. We present the results of a study employing large-scale molecular dynamics simulations and comparative Markov state model analysis of experimentally determined properties of the wild-type and mutant hDAT constructs. These offer a quantitative outline of mechanisms in which a rich spectrum of interactions of the hDAT N-terminus and C-terminus contribute to the regulation of transporter function (e.g., by phosphorylation) and/or to entirely new phenotypes (e.g., reverse uptake (efflux)) that were added in evolution. Conclusions The findings are consistent with the proposal that the size of eukaryotic neurotransmitter transporter termini increased during evolution to enable more functions (e.g., efflux) not shared with the bacterial homologs. The mechanistic explanations for the experimental findings about the modulation of function in DAT, the serotonin transporter, and other eukaryotic transporters reveal separate roles for the distal and proximal segments of the much larger N-terminus in eukaryotic transporters compared to the bacterial ones. The involvement of the proximal and distal segments — such as the role of the proximal segment in sustaining transport in phosphatidylinositol 4,5-bisphosphate-depleted membranes and of the distal segment in modulating efflux — may represent an evolutionary adaptation required for the function of eukaryotic transporters expressed in various cell types of the same organism that differ in the lipid composition and protein complement of their membrane environment. Electronic supplementary material The online version of this article (10.1186/s12915-018-0495-6) contains supplementary material, which is available to authorized users.
Collapse
|
41
|
Dai H, Jackson CR, Davis GL, Blakely RD, McMahon DG. Is dopamine transporter-mediated dopaminergic signaling in the retina a noninvasive biomarker for attention-deficit/ hyperactivity disorder? A study in a novel dopamine transporter variant Val559 transgenic mouse model. J Neurodev Disord 2017; 9:38. [PMID: 29281965 PMCID: PMC5745861 DOI: 10.1186/s11689-017-9215-8] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/19/2017] [Accepted: 09/14/2017] [Indexed: 12/14/2022] Open
Abstract
Background Dopamine (DA) is a critical neuromodulator in the retina. Disruption of retinal DA synthesis and signaling significantly attenuates light-adapted, electroretinogram (ERG) responses, as well as contrast sensitivity and acuity. As these measures can be detected noninvasively, they may provide opportunities to detect disease processes linked to perturbed DA signaling. Recently, we identified a rare, functional DA transporter (DAT, SLC6A3) coding substitution, Ala559Val, in subjects with attention-deficit/hyperactivity disorder (ADHD), demonstrating that DAT Val559 imparts anomalous DA efflux (ADE) with attendant physiological, pharmacological, and behavioral phenotypes. To understand the broader impact of ADE on ADHD, noninvasive measures sensitive to DAT reversal are needed. Methods Here, we explored this question through ERG-based analysis of retinal light responses, as well as HPLC measurements of retinal DA in DAT Val559 mice. Results Male mice homozygous (HOM) for the DAT Val559 variant demonstrated increased, light-adapted ERG b-wave amplitudes compared to wild type (WT) and heterozygous (HET) mice, whereas dark-adapted responses were indistinguishable across genotypes. The elevated amplitude of the photopic light responses in HOM mice could be mimicked in WT mice by applying D1 and D4 DA receptor agonists and suppressed in HOM mice by introducing D4 antagonist, supporting elevated retinal DA signaling arising from ADE. Following the challenge with amphetamine, WT exhibited an increase in light-adapted response amplitudes, while HOM did not. Total retinal DA content was similar across genotypes. Interestingly, female DAT Val559 HOM animals revealed no significant difference in photopic ERG responses when compared with WT and HET littermates. Conclusions These data reveal that noninvasive, in vivo evaluation of retinal responses to light can reveal physiological signatures of ADE, suggesting a possible approach to the segregation of neurobehavioral disorders based on the DAT-dependent control of DA signaling.
Collapse
Affiliation(s)
- Heng Dai
- Department of Biological Sciences, Vanderbilt University, Box 35-1634 Station B, Nashville, TN, 37235-1634, USA
| | - Chad R Jackson
- Department of Biological Sciences, Vanderbilt University, Box 35-1634 Station B, Nashville, TN, 37235-1634, USA.,Present address: Defense Threat Reduction Agency, 8211 Terminal Road, Lorton, VA, 22079, USA
| | - Gwynne L Davis
- Department of Biomedical Sciences, Charles E. Schmidt College of Medicine and Brain Institute, Florida Atlantic University, Jupiter, FL, 33458, USA
| | - Randy D Blakely
- Department of Biomedical Sciences, Charles E. Schmidt College of Medicine and Brain Institute, Florida Atlantic University, Jupiter, FL, 33458, USA
| | - Douglas G McMahon
- Department of Biological Sciences, Vanderbilt University, Box 35-1634 Station B, Nashville, TN, 37235-1634, USA.
| |
Collapse
|
42
|
Efimova EV, Gainetdinov RR, Budygin EA, Sotnikova TD. Dopamine transporter mutant animals: a translational perspective. J Neurogenet 2017; 30:5-15. [PMID: 27276191 DOI: 10.3109/01677063.2016.1144751] [Citation(s) in RCA: 56] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022]
Abstract
The dopamine transporter (DAT) plays an important homeostatic role in the control of both the extracellular and intraneuronal concentrations of dopamine, thereby providing effective control over activity of dopaminergic transmission. Since brain dopamine is known to be involved in numerous neuropsychiatric disorders, investigations using mice with genetically altered DAT function and thus intensity of dopamine-mediated signaling have provided numerous insights into the pathology of these disorders and novel pathological mechanisms that could be targeted to provide new therapeutic approaches for these disorders. In this brief overview, we discuss recent investigations involving animals with genetically altered DAT function, particularly focusing on translational studies providing new insights into pathology and pharmacology of dopamine-related disorders. Perspective applications of these and newly developed models of DAT dysfunction are also discussed.
Collapse
Affiliation(s)
- Evgeniya V Efimova
- a Institute of Translational Biomedicine, St. Petersburg State University , St. Petersburg , Russia ;,b Skolkovo Institute of Science and Technology , Skolkovo , Moscow Region , Russia
| | - Raul R Gainetdinov
- a Institute of Translational Biomedicine, St. Petersburg State University , St. Petersburg , Russia ;,b Skolkovo Institute of Science and Technology , Skolkovo , Moscow Region , Russia
| | - Evgeny A Budygin
- a Institute of Translational Biomedicine, St. Petersburg State University , St. Petersburg , Russia ;,c Department of Neurobiology and Anatomy , Wake Forest School of Medicine , Winston-Salem , NC , USA
| | - Tatyana D Sotnikova
- a Institute of Translational Biomedicine, St. Petersburg State University , St. Petersburg , Russia
| |
Collapse
|
43
|
Garcia-Olivares J, Baust T, Harris S, Hamilton P, Galli A, Amara SG, Torres GE. Gβγ subunit activation promotes dopamine efflux through the dopamine transporter. Mol Psychiatry 2017; 22:1673-1679. [PMID: 28894302 PMCID: PMC5996372 DOI: 10.1038/mp.2017.176] [Citation(s) in RCA: 24] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/11/2016] [Revised: 07/05/2017] [Accepted: 07/28/2017] [Indexed: 01/30/2023]
Abstract
The dopamine transporter (DAT) is an important regulator of brain dopamine (DA) homeostasis, controlling the intensity and duration of DA signaling. DAT is the target for psychostimulants-like cocaine and amphetamine-and plays an important role in neuropsychiatric disorders, including attention-deficit hyperactivity disorder and drug addiction. Thus, a thorough understanding of the mechanisms that regulate DAT function is necessary for the development of clinical interventions to treat DA-related brain disorders. Previous studies have revealed a plethora of protein-protein interactions influencing DAT cellular localization and activity, suggesting that the fine-tuning of DA homeostasis involves multiple mechanisms. We recently reported that G-protein beta-gamma (Gβγ) subunits bind directly to DAT and decrease DA clearance. Here we show that Gβγ induces the release of DA through DAT. Specifically, a Gβγ-binding/activating peptide, mSIRK, increases DA efflux through DAT in heterologous cells and primary dopaminergic neurons in culture. Addition of the Gβγ inhibitor gallein or DAT inhibitors prevents this effect. Residues 582 to 596 in the DAT carboxy terminus were identified as the primary binding site of Gβγ. A TAT peptide containing the Gβγ-interacting domain of DAT blocked the ability of mSIRK to induce DA efflux, consistent with a direct interaction of Gβγ with the transporter. Finally, activation of a G-protein-coupled receptor, the muscarinic M5R, results in DAT-mediated DA efflux through a Gβγ-dependent mechanism. Collectively, our data show that Gβγ interacts with DAT to promote DA efflux. This novel mechanism may have important implications in the regulation of brain DA homeostasis.
Collapse
Affiliation(s)
- J Garcia-Olivares
- Laboratory of Cellular and Molecular Neurobiology, National Institute of Mental Health, Bethesda, MD, USA
| | - T Baust
- Department of Pharmacology and Therapeutics, University of Florida College of Medicine, Gainesville, FL, USA
| | - S Harris
- Department of Pharmacology and Therapeutics, University of Florida College of Medicine, Gainesville, FL, USA
| | - P Hamilton
- Department of Molecular Physiology and Biophysics, Vanderbilt University School of Medicine, Nashville, TN, USA
| | - A Galli
- Department of Molecular Physiology and Biophysics, Vanderbilt University School of Medicine, Nashville, TN, USA
| | - SG Amara
- Laboratory of Cellular and Molecular Neurobiology, National Institute of Mental Health, Bethesda, MD, USA
| | - GE Torres
- Department of Pharmacology and Therapeutics, University of Florida College of Medicine, Gainesville, FL, USA
| |
Collapse
|
44
|
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
|
45
|
Foster JD, Vaughan RA. Phosphorylation mechanisms in dopamine transporter regulation. J Chem Neuroanat 2017; 83-84:10-18. [PMID: 27836487 PMCID: PMC6705611 DOI: 10.1016/j.jchemneu.2016.10.004] [Citation(s) in RCA: 52] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/23/2016] [Revised: 10/26/2016] [Accepted: 10/29/2016] [Indexed: 12/17/2022]
Abstract
The dopamine transporter (DAT) is a plasma membrane phosphoprotein that actively translocates extracellular dopamine (DA) into presynaptic neurons. The transporter is the primary mechanism for control of DA levels and subsequent neurotransmission, and is the target for abused and therapeutic drugs that exert their effects by suppressing reuptake. The transport capacity of DAT is acutely regulated by signaling systems and drug exposure, providing neurons the ability to fine-tune DA clearance in response to specific conditions. Kinase pathways play major roles in these mechanisms, and this review summarizes the current status of DAT phosphorylation characteristics and the evidence linking transporter phosphorylation to control of reuptake and other functions. Greater understanding of these processes may aid in elucidation of their possible contributions to DA disease states and suggest specific phosphorylation sites as targets for therapeutic manipulation of reuptake.
Collapse
Affiliation(s)
- James D Foster
- 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.
| |
Collapse
|
46
|
Davis GL, Stewart A, Stanwood GD, Gowrishankar R, Hahn MK, Blakely RD. Functional coding variation in the presynaptic dopamine transporter associated with neuropsychiatric disorders drives enhanced motivation and context-dependent impulsivity in mice. Behav Brain Res 2017; 337:61-69. [PMID: 28964912 DOI: 10.1016/j.bbr.2017.09.043] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/11/2017] [Revised: 09/18/2017] [Accepted: 09/27/2017] [Indexed: 10/18/2022]
Abstract
Recent genetic analyses have provided evidence that clinical commonalities associated with different psychiatric diagnoses often have shared mechanistic underpinnings. The development of animal models expressing functional genetic variation attributed to multiple disorders offers a salient opportunity to capture molecular, circuit and behavioral alterations underlying this hypothesis. In keeping with studies suggesting dopaminergic contributions to attention-deficit hyperactivity disorder (ADHD), bipolar disorder (BPD) and autism spectrum disorder (ASD), subjects with these diagnoses have been found to express a rare, functional coding substitution in the dopamine (DA) transporter (DAT), Ala559Val. We developed DAT Val559 knock-in mice as a construct valid model of dopaminergic alterations that drive multiple clinical phenotypes, and here evaluate the impact of lifelong expression of the variant on impulsivity and motivation utilizing the 5- choice serial reaction time task (5-CSRTT) and Go/NoGo as well as tests of time estimation (peak interval analysis), reward salience (sucrose preference), and motivation (progressive ratio test). Our findings indicate that the DAT Val559 variant induces impulsivity behaviors that are dependent upon the reward context, with increased impulsive action observed when mice are required to delay responding for a reward, whereas mice are able to withhold responding if there is a probability of reward for a correct rejection. Utilizing peak interval and progressive ratio tests, we provide evidence that impulsivity is likely driven by an enhanced motivational phenotype that also may drive faster task acquisition in operant tasks. These data provide critical validation that DAT, and more generally, DA signaling perturbations can drive impulsivity that can manifest in specific contexts and not others, and may rely on motivational alterations, which may also drive increased maladaptive reward seeking.
Collapse
Affiliation(s)
- Gwynne L Davis
- Department of Pharmacology, Vanderbilt University, 2220 Pierce Ave, Nashville, TN 37232, United States; Department of Biomedical Science, Charles E. Schmidt College of Medicine, United States.
| | - Adele Stewart
- Department of Pharmacology, Vanderbilt University, 2220 Pierce Ave, Nashville, TN 37232, United States; Department of Biomedical Science, Charles E. Schmidt College of Medicine, United States; Brain Institute, Florida Atlantic University, 5353 Parkside Drive, Jupiter, FL 33458, United States.
| | - Gregg D Stanwood
- Biomedical Sciences, Florida State University College of Medicine, 1115 West Call Street, Tallahassee, FL, 32306, United States.
| | - Raajaram Gowrishankar
- Department of Pharmacology, Vanderbilt University, 2220 Pierce Ave, Nashville, TN 37232, United States; Department of Biomedical Science, Charles E. Schmidt College of Medicine, United States.
| | - Maureen K Hahn
- Department of Pharmacology, Vanderbilt University, 2220 Pierce Ave, Nashville, TN 37232, United States; Department of Biomedical Science, Charles E. Schmidt College of Medicine, United States; Brain Institute, Florida Atlantic University, 5353 Parkside Drive, Jupiter, FL 33458, United States.
| | - Randy D Blakely
- Department of Pharmacology, Vanderbilt University, 2220 Pierce Ave, Nashville, TN 37232, United States; Department of Biomedical Science, Charles E. Schmidt College of Medicine, United States; Brain Institute, Florida Atlantic University, 5353 Parkside Drive, Jupiter, FL 33458, United States.
| |
Collapse
|
47
|
Gaskill PJ, Miller DR, Gamble-George J, Yano H, Khoshbouei H. HIV, Tat and dopamine transmission. Neurobiol Dis 2017; 105:51-73. [PMID: 28457951 PMCID: PMC5541386 DOI: 10.1016/j.nbd.2017.04.015] [Citation(s) in RCA: 59] [Impact Index Per Article: 7.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/25/2017] [Revised: 04/04/2017] [Accepted: 04/16/2017] [Indexed: 01/02/2023] Open
Abstract
Human Immunodeficiency Virus (HIV) is a progressive infection that targets the immune system, affecting more than 37 million people around the world. While combinatorial antiretroviral therapy (cART) has lowered mortality rates and improved quality of life in infected individuals, the prevalence of HIV associated neurocognitive disorders is increasing and HIV associated cognitive decline remains prevalent. Recent research has suggested that HIV accessory proteins may be involved in this decline, and several studies have indicated that the HIV protein transactivator of transcription (Tat) can disrupt normal neuronal and glial function. Specifically, data indicate that Tat may directly impact dopaminergic neurotransmission, by modulating the function of the dopamine transporter and specifically damaging dopamine-rich regions of the CNS. HIV infection of the CNS has long been associated with dopaminergic dysfunction, but the mechanisms remain undefined. The specific effect(s) of Tat on dopaminergic neurotransmission may be, at least partially, a mechanism by which HIV infection directly or indirectly induces dopaminergic dysfunction. Therefore, precisely defining the specific effects of Tat on the dopaminergic system will help to elucidate the mechanisms by which HIV infection of the CNS induces neuropsychiatric, neurocognitive and neurological disorders that involve dopaminergic neurotransmission. Further, this will provide a discussion of the experiments needed to further these investigations, and may help to identify or develop new therapeutic approaches for the prevention or treatment of these disorders in HIV-infected individuals.
Collapse
Affiliation(s)
- Peter J Gaskill
- Department of Pharmacology and Physiology, Drexel University College of Medicine, Philadelphia, PA 19102, United States.
| | - Douglas R Miller
- Department of Neuroscience, University of Florida, Gainesville, FL 32611, United States
| | - Joyonna Gamble-George
- Department of Neuroscience, University of Florida, Gainesville, FL 32611, United States
| | - Hideaki Yano
- National Institute on Drug Abuse, National Institutes of Health, Baltimore, MD 21224, United States
| | - Habibeh Khoshbouei
- Department of Neuroscience, University of Florida, Gainesville, FL 32611, United States.
| |
Collapse
|
48
|
The Dopamine Transporter Recycles via a Retromer-Dependent Postendocytic Mechanism: Tracking Studies Using a Novel Fluorophore-Coupling Approach. J Neurosci 2017; 37:9438-9452. [PMID: 28847807 DOI: 10.1523/jneurosci.3885-16.2017] [Citation(s) in RCA: 49] [Impact Index Per Article: 6.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/20/2016] [Revised: 07/17/2017] [Accepted: 08/19/2017] [Indexed: 01/28/2023] Open
Abstract
Presynaptic reuptake, mediated by the dopamine (DA) transporter (DAT), terminates DAergic neurotransmission and constrains extracellular DA levels. Addictive and therapeutic psychostimulants inhibit DA reuptake and multiple DAT coding variants have been reported in patients with neuropsychiatric disorders. These findings underscore that DAT is critical for DA neurotransmission and homeostasis. DAT surface availability is regulated acutely by endocytic trafficking, and considerable effort has been directed toward understanding mechanisms that govern DAT's plasma membrane expression and postendocytic fate. Multiple studies have demonstrated DAT endocytic recycling and enhanced surface delivery in response to various stimuli. Paradoxically, imaging studies have not detected DAT targeting to classic recycling endosomes, suggesting that internalized DAT targets to either degradation or an undefined recycling compartment. Here, we leveraged PRIME (PRobe Incorporation Mediated by Enzyme) labeling to couple surface DAT directly to fluorophore, and tracked DAT's postendocytic itinerary in immortalized mesencephalic cells. Following internalization, DAT robustly targeted to retromer-positive endosomes, and DAT/retromer colocalization was observed in male mouse dopaminergic somatodendritic and terminal regions. Short hairpin RNA-mediated Vps35 knockdown revealed that DAT endocytic recycling requires intact retromer. DAT also targeted rab7-positive endosomes with slow, linear kinetics that were unaffected by either accelerating DAT internalization or binding a high-affinity cocaine analog. However, cocaine increased DAT exit from retromer-positive endosomes significantly. Finally, we found that the DAT carboxy-terminal PDZ-binding motif was required for DAT recycling and exit from retromer. These results define the DAT recycling mechanism and provide a unifying explanation for previous, seemingly disparate, DAT endocytic trafficking findings.SIGNIFICANCE STATEMENT The neuronal dopamine (DA) transporter (DAT) recaptures released DA and modulates DAergic neurotransmission, and a number of DAT coding variants have been reported in several DA-related disorders, including infantile parkinsonism, attention-deficit/hyperactivity disorder and autism spectrum disorder. DAT is also competitively inhibited by psychostimulants with high abuse potential. Therefore, mechanisms that acutely affect DAT availability will likely exert significant impact on both normal and pathological DAergic homeostasis. Here, we explore the cellular mechanisms that acutely control DAT surface expression. Our results reveal the intracellular mechanisms that mediate DAT endocytic recycling following constitutive and regulated internalization. In addition to shedding light on this critical process, these findings resolve conflict among multiple, seemingly disparate, previous reports on DAT's postendocytic fate.
Collapse
|
49
|
The Atypical MAP Kinase SWIP-13/ERK8 Regulates Dopamine Transporters through a Rho-Dependent Mechanism. J Neurosci 2017; 37:9288-9304. [PMID: 28842414 DOI: 10.1523/jneurosci.1582-17.2017] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/05/2017] [Revised: 07/31/2017] [Accepted: 08/12/2017] [Indexed: 12/26/2022] Open
Abstract
The neurotransmitter dopamine (DA) regulates multiple behaviors across phylogeny, with disrupted DA signaling in humans associated with addiction, attention-deficit/ hyperactivity disorder, schizophrenia, and Parkinson's disease. The DA transporter (DAT) imposes spatial and temporal limits on DA action, and provides for presynaptic DA recycling to replenish neurotransmitter pools. Molecular mechanisms that regulate DAT expression, trafficking, and function, particularly in vivo, remain poorly understood, though recent studies have implicated rho-linked pathways in psychostimulant action. To identify genes that dictate the ability of DAT to sustain normal levels of DA clearance, we pursued a forward genetic screen in Caenorhabditis elegans based on the phenotype swimming-induced paralysis (Swip), a paralytic behavior observed in hermaphrodite worms with loss-of-function dat-1 mutations. Here, we report the identity of swip-13, which encodes a highly conserved ortholog of the human atypical MAP kinase ERK8. We present evidence that SWIP-13 acts presynaptically to insure adequate levels of surface DAT expression and DA clearance. Moreover, we provide in vitro and in vivo evidence supporting a conserved pathway involving SWIP-13/ERK8 activation of Rho GTPases that dictates DAT surface expression and function.SIGNIFICANCE STATEMENT Signaling by the neurotransmitter dopamine (DA) is tightly regulated by the DA transporter (DAT), insuring efficient DA clearance after release. Molecular networks that regulate DAT are poorly understood, particularly in vivo Using a forward genetic screen in the nematode Caenorhabditis elegans, we implicate the atypical mitogen activated protein kinase, SWIP-13, in DAT regulation. Moreover, we provide in vitro and in vivo evidence that SWIP-13, as well as its human counterpart ERK8, regulate DAT surface availability via the activation of Rho proteins. Our findings implicate a novel pathway that regulates DA synaptic availability and that may contribute to risk for disorders linked to perturbed DA signaling. Targeting this pathway may be of value in the development of therapeutics in such disorders.
Collapse
|
50
|
Simmler LD, Liechti ME. Interactions of Cathinone NPS with Human Transporters and Receptors in Transfected Cells. Curr Top Behav Neurosci 2017; 32:49-72. [PMID: 27272068 DOI: 10.1007/7854_2016_20] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/22/2022]
Abstract
Pharmacological assays carried out in transfected cells have been very useful for describing the mechanism of action of cathinone new psychoactive substances (NPS). These in vitro characterizations provide fast and reliable information on psychoactive substances soon after they emerge for recreational use. Well-investigated comparator compounds, such as methamphetamine, 3,4-methylenedioxymethamphetamine, cocaine, and lysergic acid diethylamide, should always be included in the characterization to enhance the translation of the in vitro data into clinically useful information. We classified cathinone NPS according to their pharmacology at monoamine transporters and receptors. Cathinone NPS are monoamine uptake inhibitors and most induce transporter-mediated monoamine efflux with weak to no activity at pre- or postsynaptic receptors. Cathinones with a nitrogen-containing pyrrolidine ring emerged as NPS that are extremely potent transporter inhibitors but not monoamine releasers. Cathinones exhibit clinically relevant differences in relative potencies at serotonin vs. dopamine transporters. Additionally, cathinone NPS have more dopaminergic vs. serotonergic properties compared with their non-β-keto amphetamine analogs, suggesting more stimulant and reinforcing properties. In conclusion, in vitro pharmacological assays in heterologous expression systems help to predict the psychoactive and toxicological effects of NPS.
Collapse
Affiliation(s)
- Linda D Simmler
- Division of Clinical Pharmacology and Toxicology, Department of Biomedicine and Department of Clinical Research, University Hospital Basel, Basel, Switzerland
| | - Matthias E Liechti
- Division of Clinical Pharmacology and Toxicology, Department of Biomedicine and Department of Clinical Research, University Hospital Basel, Basel, Switzerland.
| |
Collapse
|