1
|
Nowak J, Aronin J, Beg F, O’Malley N, Ferrick M, Quattrin T, Pavlesen S, Hadjiargyrou M, Komatsu DE, Thanos PK. The Effects of Chronic Psychostimulant Administration on Bone Health: A Review. Biomedicines 2024; 12:1914. [PMID: 39200379 PMCID: PMC11351835 DOI: 10.3390/biomedicines12081914] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/27/2024] [Revised: 08/08/2024] [Accepted: 08/19/2024] [Indexed: 09/02/2024] Open
Abstract
(1) Background: Methylphenidate (MP) and amphetamine (AMP) are psychostimulants that are widely prescribed to treat Attention Deficit Hyperactivity Disorder (ADHD) and narcolepsy. In recent years, 6.1 million children received an ADHD diagnosis, and nearly 2/3 of these children were prescribed psychostimulants for treatment. The purpose of this review is to summarize the current literature on psychostimulant use and the resulting effects on bone homeostasis, biomechanical properties, and functional integrity. (2) Methods: Literature searches were conducted from Medline/PubMed electronic databases utilizing the search terms "methylphenidate" OR "amphetamine" OR "methylphenidate" AND "bone health" AND "bone remodeling" AND "osteoclast" AND "osteoblast" AND "dopamine" from 01/1985 to 04/2023. (3) Results: Of the 550 publications found, 44 met the inclusion criteria. Data from identified studies demonstrate that the use of MP and AMP results in decreases in specific bone properties and biomechanical integrity via downstream effects on osteoblasts and osteoclast-related genes. (4) Conclusions: The chronic use of psychostimulants negatively affects bone integrity and strength as a result of increased osteoclast activity. These data support the need to take this into consideration when planning the treatment type and duration for bone fractures.
Collapse
Affiliation(s)
- Jessica Nowak
- Behavioral Neuropharmacology and Neuroimaging Laboratory on Addictions (BNNLA), Clinical Research Institute on Addictions, Department of Pharmacology and Toxicology, Jacobs School of Medicine and Biomedical Sciences, University at Buffalo, Buffalo, NY 14203, USA
| | - Jacob Aronin
- Behavioral Neuropharmacology and Neuroimaging Laboratory on Addictions (BNNLA), Clinical Research Institute on Addictions, Department of Pharmacology and Toxicology, Jacobs School of Medicine and Biomedical Sciences, University at Buffalo, Buffalo, NY 14203, USA
| | - Faraaz Beg
- Behavioral Neuropharmacology and Neuroimaging Laboratory on Addictions (BNNLA), Clinical Research Institute on Addictions, Department of Pharmacology and Toxicology, Jacobs School of Medicine and Biomedical Sciences, University at Buffalo, Buffalo, NY 14203, USA
| | - Natasha O’Malley
- Department of Orthopaedics, University of Rochester Medical Center, Rochester, NY 14642, USA
| | - Michael Ferrick
- Department of Orthopaedics, Jacobs School of Medicine, University at Buffalo, Buffalo, NY 14203, USA
| | - Teresa Quattrin
- UBMD Pediatrics, JR Oishei Children’s Hospital, University at Buffalo, Buffalo, NY 14203, USA
| | - Sonja Pavlesen
- Clinical Research Center, UBMD Orthopaedics & Sports Medicine, 111 N Maplemere Rd., Suite 100, Buffalo, NY 14221, USA
| | - Michael Hadjiargyrou
- Department of Biological and Chemical Sciences, New York Institute of Technology, Westbury, NY 11568, USA;
| | - David E. Komatsu
- Department of Orthopaedics and Rehabilitation, Stony Brook University, Stony Brook, NY 11794, USA
| | - Panayotis K. Thanos
- Behavioral Neuropharmacology and Neuroimaging Laboratory on Addictions (BNNLA), Clinical Research Institute on Addictions, Department of Pharmacology and Toxicology, Jacobs School of Medicine and Biomedical Sciences, University at Buffalo, Buffalo, NY 14203, USA
| |
Collapse
|
2
|
Støier JF, Konomi-Pilkati A, Apuschkin M, Herborg F, Gether U. Amphetamine-induced reverse transport of dopamine does not require cytosolic Ca 2. J Biol Chem 2023; 299:105063. [PMID: 37468107 PMCID: PMC10448275 DOI: 10.1016/j.jbc.2023.105063] [Citation(s) in RCA: 8] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/25/2022] [Revised: 07/13/2023] [Accepted: 07/14/2023] [Indexed: 07/21/2023] Open
Abstract
Amphetamines (AMPHs) are substrates of the dopamine transporter (DAT) and reverse the direction of dopamine (DA) transport. This has been suggested to depend on activation of Ca2+-dependent pathways, but the mechanism underlying reverse transport via endogenously expressed DAT is still unclear. Here, to enable concurrent visualization by live imaging of extracellular DA dynamics and cytosolic Ca2+ levels, we employ the fluorescent Ca2+ sensor jRGECO1a expressed in cultured dopaminergic neurons together with the fluorescent DA sensor GRABDA1H expressed in cocultured "sniffer" cells. In the presence of the Na+-channel blocker tetrodotoxin to prevent exocytotic DA release, AMPH induced in the cultured neurons a profound dose-dependent efflux of DA that was blocked both by inhibition of DAT with cocaine and by inhibition of the vesicular monoamine transporter-2 with Ro-4-1284 or reserpine. However, the AMPH-induced DA efflux was not accompanied by an increase in cytosolic Ca2+ and was unaffected by blockade of voltage-gated calcium channels or chelation of cytosolic Ca2+. The independence of cytosolic Ca2+ was further supported by activation of N-methyl-D-aspartate-type ionotropic glutamate receptors leading to a marked increase in cytosolic Ca2+ without affecting AMPH-induced DA efflux. Curiously, AMPH elicited spontaneous Ca2+ spikes upon blockade of the D2 receptor, suggesting that AMPH can regulate intracellular Ca2+ in an autoreceptor-dependent manner regardless of the apparent independence of Ca2+ for AMPH-induced efflux. We conclude that AMPH-induced DA efflux in dopaminergic neurons does not require cytosolic Ca2+ but is strictly dependent on the concerted action of AMPH on both vesicular monoamine transporter-2 and DAT.
Collapse
Affiliation(s)
- Jonatan Fullerton Støier
- Molecular Neuropharmacology and Genetics Laboratory, Department of Neuroscience, Faculty of Health and Medical Sciences, Panum Institute - Maersk Tower 7.5, University of Copenhagen, Copenhagen, Denmark
| | - Ainoa Konomi-Pilkati
- Molecular Neuropharmacology and Genetics Laboratory, Department of Neuroscience, Faculty of Health and Medical Sciences, Panum Institute - Maersk Tower 7.5, University of Copenhagen, Copenhagen, Denmark
| | - Mia Apuschkin
- Molecular Neuropharmacology and Genetics Laboratory, Department of Neuroscience, Faculty of Health and Medical Sciences, Panum Institute - Maersk Tower 7.5, University of Copenhagen, Copenhagen, Denmark
| | - Freja Herborg
- Molecular Neuropharmacology and Genetics Laboratory, Department of Neuroscience, Faculty of Health and Medical Sciences, Panum Institute - Maersk Tower 7.5, University of Copenhagen, Copenhagen, Denmark
| | - Ulrik Gether
- Molecular Neuropharmacology and Genetics Laboratory, Department of Neuroscience, Faculty of Health and Medical Sciences, Panum Institute - Maersk Tower 7.5, University of Copenhagen, Copenhagen, Denmark.
| |
Collapse
|
3
|
Wang S, Neel AI, Adams KL, Sun H, Jones SR, Howlett AC, Chen R. Atorvastatin differentially regulates the interactions of cocaine and amphetamine with dopamine transporters. Neuropharmacology 2023; 225:109387. [PMID: 36567004 PMCID: PMC9872521 DOI: 10.1016/j.neuropharm.2022.109387] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/09/2022] [Revised: 12/12/2022] [Accepted: 12/18/2022] [Indexed: 12/24/2022]
Abstract
The function of the dopamine transporter (DAT) is regulated by membrane cholesterol content. A direct, acute removal of membrane cholesterol by methyl-β-cyclodextrin (MβCD) has been shown to reduce dopamine (DA) uptake and release mediated by the DAT. This is of particular interest because a few widely prescribed statins that lower peripheral cholesterol levels are blood-brain barrier (BBB) penetrants, and therefore could alter DAT function through brain cholesterol modulation. The goal of this study was to investigate the effects of prolonged atorvastatin treatment (24 h) on DAT function in neuroblastoma 2A cells stably expressing DAT. We found that atorvastatin treatment effectively lowered membrane cholesterol content in a concentration-dependent manner. Moreover, atorvastatin treatment markedly reduced DA uptake and abolished cocaine inhibition of DA uptake, independent of surface DAT levels. These deficits induced by atorvastatin treatment were reversed by cholesterol replenishment. However, atorvastatin treatment did not change amphetamine (AMPH)-induced DA efflux. This is in contrast to a small but significant reduction in DA efflux induced by acute depletion of membrane cholesterol using MβCD. This discrepancy may involve differential changes in membrane lipid composition resulting from chronic and acute cholesterol depletion. Our data suggest that the outward-facing conformation of DAT, which favors the binding of DAT blockers such as cocaine, is more sensitive to atorvastatin-induced cholesterol depletion than the inward-facing conformation, which favors the binding of DAT substrates such as AMPH. Our study on statin-DAT interactions may have clinical implications in our understanding of neurological side effects associated with chronic use of BBB penetrant statins.
Collapse
Affiliation(s)
- Shiyu Wang
- Department of Physiology & Pharmacology, Wake Forest School of Medicine, Winston Salem, NC, 27157, United States
| | - Anna I Neel
- Department of Physiology & Pharmacology, Wake Forest School of Medicine, Winston Salem, NC, 27157, United States
| | - Kristen L Adams
- Department of Physiology & Pharmacology, Wake Forest School of Medicine, Winston Salem, NC, 27157, United States
| | - Haiguo Sun
- Department of Physiology & Pharmacology, Wake Forest School of Medicine, Winston Salem, NC, 27157, United States
| | - Sara R Jones
- Department of Physiology & Pharmacology, Wake Forest School of Medicine, Winston Salem, NC, 27157, United States
| | - Allyn C Howlett
- Department of Physiology & Pharmacology, Wake Forest School of Medicine, Winston Salem, NC, 27157, United States
| | - Rong Chen
- Department of Physiology & Pharmacology, Wake Forest School of Medicine, Winston Salem, NC, 27157, United States.
| |
Collapse
|
4
|
Dutta CN, Christov-Moore L, Ombao H, Douglas PK. Neuroprotection in late life attention-deficit/hyperactivity disorder: A review of pharmacotherapy and phenotype across the lifespan. Front Hum Neurosci 2022; 16:938501. [PMID: 36226261 PMCID: PMC9548548 DOI: 10.3389/fnhum.2022.938501] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/07/2022] [Accepted: 08/16/2022] [Indexed: 11/13/2022] Open
Abstract
For decades, psychostimulants have been the gold standard pharmaceutical treatment for attention-deficit/hyperactivity disorder (ADHD). In the United States, an astounding 9% of all boys and 4% of girls will be prescribed stimulant drugs at some point during their childhood. Recent meta-analyses have revealed that individuals with ADHD have reduced brain volume loss later in life (>60 y.o.) compared to the normal aging brain, which suggests that either ADHD or its treatment may be neuroprotective. Crucially, these neuroprotective effects were significant in brain regions (e.g., hippocampus, amygdala) where severe volume loss is linked to cognitive impairment and Alzheimer's disease. Historically, the ADHD diagnosis and its pharmacotherapy came about nearly simultaneously, making it difficult to evaluate their effects in isolation. Certain evidence suggests that psychostimulants may normalize structural brain changes typically observed in the ADHD brain. If ADHD itself is neuroprotective, perhaps exercising the brain, then psychostimulants may not be recommended across the lifespan. Alternatively, if stimulant drugs are neuroprotective, then this class of medications may warrant further investigation for their therapeutic effects. Here, we take a bottom-up holistic approach to review the psychopharmacology of ADHD in the context of recent models of attention. We suggest that future studies are greatly needed to better appreciate the interactions amongst an ADHD diagnosis, stimulant treatment across the lifespan, and structure-function alterations in the aging brain.
Collapse
Affiliation(s)
- Cintya Nirvana Dutta
- Biostatistics Group, Computer, Electrical and Mathematical Sciences and Engineering Division, King Abdullah University of Science and Technology, Thuwal, Saudi Arabia
- School of Modeling, Simulation, and Training, and Computer Science, University of Central Florida, Orlando, FL, United States
| | - Leonardo Christov-Moore
- Brain and Creativity Institute, University of Southern California, Los Angeles, CA, United States
| | - Hernando Ombao
- Biostatistics Group, Computer, Electrical and Mathematical Sciences and Engineering Division, King Abdullah University of Science and Technology, Thuwal, Saudi Arabia
| | - Pamela K. Douglas
- School of Modeling, Simulation, and Training, and Computer Science, University of Central Florida, Orlando, FL, United States
- Department of Psychiatry and Biobehavioral Medicine, University of California, Los Angeles, Los Angeles, CA, United States
| |
Collapse
|
5
|
Wang S, Liu H, Roberts JB, Wiley AP, Marayati BF, Adams KL, Luessen DJ, Eldeeb K, Sun H, Zhang K, Chen R. Prolonged ethanol exposure modulates constitutive internalization and recycling of 5-HT1A receptors. J Neurochem 2022; 160:469-481. [PMID: 34928513 PMCID: PMC8828711 DOI: 10.1111/jnc.15564] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/04/2021] [Revised: 11/24/2021] [Accepted: 12/13/2021] [Indexed: 02/03/2023]
Abstract
Alcohol exposure alters the signaling of the serotoninergic system, which is involved in alcohol consumption, reward, and dependence. In particular, dysregulation of serotonin receptor type 1A (5-HT1AR) is associated with alcohol intake and withdrawal-induced anxiety-like behavior in rodents. However, how ethanol regulates 5-HT1AR activity and cell surface availability remains elusive. Using neuroblastoma 2a cells stably expressing human 5-HT1ARs tagged with hemagglutinin at the N-terminus, we found that prolonged ethanol exposure (18 h) reduced the basal surface levels of 5-HT1ARs in a concentration-dependent manner. This reduction is attributed to both enhanced receptor internalization and attenuated receptor recycling. Moreover, constitutive 5-HT1AR internalization in ethanol naïve cells was blocked by concanavalin A (ConA) but not nystatin, suggesting clathrin-dependent 5-HT1AR internalization. In contrast, constitutive 5-HT1AR internalization in ethanol-treated cells was blocked by nystatin but not by ConA, indicating that constitutive 5-HT1AR internalization switched from a clathrin- to a caveolin-dependent pathway. Dynasore, an inhibitor of dynamin, blocked 5-HT1AR internalization in both vehicle- and ethanol-treated cells. Furthermore, ethanol exposure enhanced the activity of dynamin I via dephosphorylation and reduced myosin Va levels, which may contribute to increased internalization and reduced recycling of 5-HT1ARs, respectively. Our findings suggest that prolonged ethanol exposure not only alters the endocytic trafficking of 5-HT1ARs but also the mechanism by which constitutive 5-HT1AR internalization occurs.
Collapse
Affiliation(s)
- Shiyu Wang
- Department of Physiology & Pharmacology, Wake Forest School of Medicine, Winston Salem, NC 27157
| | - Haoran Liu
- Department of Biology, Wake Forest University, Winston Salem, NC 27106
| | - Jonté B. Roberts
- Department of Physiology & Pharmacology, Wake Forest School of Medicine, Winston Salem, NC 27157
| | - Aidan P. Wiley
- Department of Physiology & Pharmacology, Wake Forest School of Medicine, Winston Salem, NC 27157
| | | | - Kristen L. Adams
- Department of Physiology & Pharmacology, Wake Forest School of Medicine, Winston Salem, NC 27157
| | - Deborah J. Luessen
- Department of Physiology & Pharmacology, Wake Forest School of Medicine, Winston Salem, NC 27157
| | - Khalil Eldeeb
- Department of Physiology & Pharmacology, Wake Forest School of Medicine, Winston Salem, NC 27157
- Campbell University School of Osteopathic Medicine, Lillington, NC 27546
| | - Haiguo Sun
- Department of Physiology & Pharmacology, Wake Forest School of Medicine, Winston Salem, NC 27157
| | - Ke Zhang
- Department of Biology, Wake Forest University, Winston Salem, NC 27106
- Center for Molecular Signaling, Wake Forest University, Winston Salem, NC 27106
| | - Rong Chen
- Department of Physiology & Pharmacology, Wake Forest School of Medicine, Winston Salem, NC 27157
- Center for Molecular Signaling, Wake Forest University, Winston Salem, NC 27106
| |
Collapse
|
6
|
Deng J, Kim K, Zheng X, Shang L, Zhan CG, Zheng F. Cocaine hydrolase blocks cocaine-induced dopamine transporter trafficking to the plasma membrane. Addict Biol 2022; 27:e13089. [PMID: 34363291 PMCID: PMC8720053 DOI: 10.1111/adb.13089] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/01/2021] [Revised: 07/21/2021] [Accepted: 07/27/2021] [Indexed: 01/03/2023]
Abstract
Cocaine blocks dopamine uptake via dopamine transporter (DAT) on plasma membrane of neuron cells and, as a result, produces the high and induces DAT trafficking to plasma membrane which contributes to the drug seeking or craving. In this study, we first examined the dose dependence of cocaine-induced DAT trafficking and hyperactivity in rats, demonstrating that cocaine at an intraperitoneal dose of 10 mg/kg or higher led to redistribution of most DAT to the plasma membrane while inducing significant hyperactivity in rats. However, administration of 5-mg/kg cocaine (ip) did not significantly induce DAT trafficking or hyperactivity in rats. So the threshold (intraperitoneal) dose of cocaine that can significantly induce DAT trafficking or hyperactivity should be between 5 and 10 mg/kg. These data suggest that when a cocaine dose is high enough to induce significant hyperactivity, it can also significantly induce DAT trafficking to the plasma membrane. Further, the threshold brain cocaine concentration required to induce significant hyperactivity and DAT trafficking was estimated to be ~2.0 ± 0.8 μg/g. Particularly, for treatment of cocaine abuse, previous studies demonstrated that an exogenous cocaine-metabolizing enzyme, for example, CocH3-Fc(M3), can effectively block cocaine-induced hyperactivity. However, it was unknown whether an enzyme could also effectively block cocaine-induced DAT trafficking to the plasma membrane. This study demonstrates, for the first time, that the enzyme is also capable of effectively blocking cocaine from reaching the brain even with a lethal dose of 60-mg/kg cocaine (ip) and, thus, powerfully preventing cocaine-induced physiological effects such as the hyperactivity and DAT trafficking.
Collapse
Affiliation(s)
- Jing Deng
- Molecular Modeling and Biopharmaceutical Center, College of Pharmacy, University of Kentucky, 789 South Limestone Street, Lexington, KY 40536,Department of Pharmaceutical Sciences, College of Pharmacy, University of Kentucky, 789 South Limestone Street, Lexington, KY 40536
| | - Kyungbo Kim
- Molecular Modeling and Biopharmaceutical Center, College of Pharmacy, University of Kentucky, 789 South Limestone Street, Lexington, KY 40536,Department of Pharmaceutical Sciences, College of Pharmacy, University of Kentucky, 789 South Limestone Street, Lexington, KY 40536
| | - Xirong Zheng
- Molecular Modeling and Biopharmaceutical Center, College of Pharmacy, University of Kentucky, 789 South Limestone Street, Lexington, KY 40536,Department of Pharmaceutical Sciences, College of Pharmacy, University of Kentucky, 789 South Limestone Street, Lexington, KY 40536
| | - Linyue Shang
- Molecular Modeling and Biopharmaceutical Center, College of Pharmacy, University of Kentucky, 789 South Limestone Street, Lexington, KY 40536,Department of Pharmaceutical Sciences, College of Pharmacy, University of Kentucky, 789 South Limestone Street, Lexington, KY 40536
| | - Chang-Guo Zhan
- Molecular Modeling and Biopharmaceutical Center, College of Pharmacy, University of Kentucky, 789 South Limestone Street, Lexington, KY 40536,Department of Pharmaceutical Sciences, College of Pharmacy, University of Kentucky, 789 South Limestone Street, Lexington, KY 40536
| | - Fang Zheng
- Molecular Modeling and Biopharmaceutical Center, College of Pharmacy, University of Kentucky, 789 South Limestone Street, Lexington, KY 40536,Department of Pharmaceutical Sciences, College of Pharmacy, University of Kentucky, 789 South Limestone Street, Lexington, KY 40536
| |
Collapse
|
7
|
Magee CP, Le BD, Siripathane YH, Wilkins DG, Hanson GR, Fleckenstein AE. Methcathinone decreases dopamine transporter function: Role of protein kinase C. J Neurochem 2021; 159:116-127. [PMID: 34320222 DOI: 10.1111/jnc.15483] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/02/2020] [Revised: 07/19/2021] [Accepted: 07/25/2021] [Indexed: 11/28/2022]
Abstract
Methcathinone (MCAT) is a psychostimulant of abuse that can cause both persistent striatal dopaminergic and serotonergic, as well as hippocampal serotonergic, deficits. Evidence suggests that the rapid effects of stimulants that are structurally and mechanistically similar to MCAT on monoamine transporter function may contribute to the abuse liability and/or persistent monoaminergic deficits caused by these agents. Thus, effects of MCAT on 1) striatal dopamine (DA) transporter (DAT); and 2) striatal and hippocampal serotonin transporter (SERT) function, as determined in tissues from adult male rats, were assessed. As reported previously, a single administration of MCAT rapidly (within 1 hr) decreases striatal [3 H]DA uptake. Similarly, incubation of rat synaptosomes with MCAT at 37℃ (but not 4˚C) decreased striatal [3 H]DA uptake. Incubation with MCAT likewise decreased [3 H]5HT but not vesicular [3 H]DA uptake. MCAT incubation in vitro was without effect on [3 H]DA uptake in striatal synaptosomes prepared from MCAT-treated rats. The decrease in [3 H]DA uptake caused by MCAT incubation: (a) reflected a decrease in Vmax , with minimal change in Km , and (b) was attenuated by co-incubation with the cell-permeable calcium chelator, N,N'-[1,2-ethanediylbis(oxy-2,1-phenylene)]bis[N-[2-[(acetyloxy)methoxy]-2-oxoethyl]-1,1'-bis[(acetyloxy)methyl] ester-glycine (BAPTA-AM), as well as the non-selective protein kinase-C (PKC) inhibitors bisindolylmaleimide-1 (BIM-1) and 2-[1-3(Aminopropyl)indol-3-yl]-3(1-methyl-1H-indol-3-yl)maleimide (or Bisindolylmaleimide VIII; Ro-31-7549). Taken together, these results suggest that in vitro MCAT incubation may model important aspects of MCAT administration in vivo, and that calcium and PKC contribute to the in vitro effects of MCAT on DAT.
Collapse
Affiliation(s)
- Charlotte P Magee
- Interdepartmental Program in Neuroscience, University of Utah, Salt Lake City, UT, USA.,School of Dentistry, University of Utah, Salt Lake City, UT, USA
| | - BaoMinh D Le
- School of Dentistry, University of Utah, Salt Lake City, UT, USA
| | | | - Diana G Wilkins
- Department of Pathology, University of Utah, Salt Lake City, UT, USA
| | - Glen R Hanson
- Interdepartmental Program in Neuroscience, University of Utah, Salt Lake City, UT, USA.,School of Dentistry, University of Utah, Salt Lake City, UT, USA
| | - Annette E Fleckenstein
- Interdepartmental Program in Neuroscience, University of Utah, Salt Lake City, UT, USA.,School of Dentistry, University of Utah, Salt Lake City, UT, USA
| |
Collapse
|
8
|
Ryan RM, Ingram SL, Scimemi A. Regulation of Glutamate, GABA and Dopamine Transporter Uptake, Surface Mobility and Expression. Front Cell Neurosci 2021; 15:670346. [PMID: 33927596 PMCID: PMC8076567 DOI: 10.3389/fncel.2021.670346] [Citation(s) in RCA: 21] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/21/2021] [Accepted: 03/15/2021] [Indexed: 01/31/2023] Open
Abstract
Neurotransmitter transporters limit spillover between synapses and maintain the extracellular neurotransmitter concentration at low yet physiologically meaningful levels. They also exert a key role in providing precursors for neurotransmitter biosynthesis. In many cases, neurons and astrocytes contain a large intracellular pool of transporters that can be redistributed and stabilized in the plasma membrane following activation of different signaling pathways. This means that the uptake capacity of the brain neuropil for different neurotransmitters can be dynamically regulated over the course of minutes, as an indirect consequence of changes in neuronal activity, blood flow, cell-to-cell interactions, etc. Here we discuss recent advances in the mechanisms that control the cell membrane trafficking and biophysical properties of transporters for the excitatory, inhibitory and modulatory neurotransmitters glutamate, GABA, and dopamine.
Collapse
Affiliation(s)
- Renae M. Ryan
- School of Medical Sciences, Faculty of Medicine and Health, University of Sydney, Sydney, NSW, Australia
| | - Susan L. Ingram
- Department of Neurological Surgery, Oregon Health & Science University, Portland, OR, United States
| | | |
Collapse
|
9
|
Jones SR, Fordahl SC. Bingeing on High-Fat Food Enhances Evoked Dopamine Release and Reduces Dopamine Uptake in the Nucleus Accumbens. Obesity (Silver Spring) 2021; 29:721-730. [PMID: 33660412 PMCID: PMC8048651 DOI: 10.1002/oby.23122] [Citation(s) in RCA: 13] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/06/2020] [Revised: 12/15/2020] [Accepted: 12/17/2020] [Indexed: 11/24/2022]
Abstract
OBJECTIVE Binge-eating disorder (BED) disrupts dopamine neuron function, in part by altering dopamine transporter (DAT) activity. This study characterized the effects of high-fat bingeing on presynaptic dopamine terminals and tested the hypothesis that acute low-dose amphetamine would restore DAT function. METHODS C57BL/6 mice were given limited access (LimA) to a high-fat diet (2 h/d, 3 d/wk) or standard chow (control). After 6 weeks, ex vivo fast-scan cyclic voltammetry was used to characterize dopamine-terminal adaptations in the nucleus accumbens. Prior to undergoing fast-scan cyclic voltammetry, some mice from each group were given amphetamine (0.5 mg/kg intraperitoneally). RESULTS Escalation of high fat intake, termed bingeing, occurred in the LimA group and coincided with increased phasic dopamine release, reduced dopamine uptake rates, and increased dopamine receptor 2 (D2 ) autoreceptor function. Acute amphetamine selectively reversed dopamine uptake changes in the LimA group and restored the potency of amphetamine to inhibit uptake. CONCLUSIONS High-fat bingeing enhanced dopaminergic signaling in the nucleus accumbens by promoting phasic dopamine release and reducing clearance. This study's data show that amphetamine was efficacious in restoring impaired DAT function caused by high-fat bingeing but did not reduce dopamine release to normal. These presynaptic changes should be considered if amphetamine-like dopamine releasers are used as treatments for BED.
Collapse
Affiliation(s)
- Sara R. Jones
- Department of Physiology and PharmacologySchool of MedicineWake Forest UniversityWinston‐SalemNorth CarolinaUSA
| | - Steve C. Fordahl
- Department of NutritionUNC GreensboroGreensboroNorth CarolinaUSA
| |
Collapse
|
10
|
Alonso IP, Pino JA, Kortagere S, Torres GE, España RA. Dopamine transporter function fluctuates across sleep/wake state: potential impact for addiction. Neuropsychopharmacology 2021; 46:699-708. [PMID: 33032296 PMCID: PMC8026992 DOI: 10.1038/s41386-020-00879-2] [Citation(s) in RCA: 17] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/26/2020] [Revised: 09/13/2020] [Accepted: 09/15/2020] [Indexed: 12/20/2022]
Abstract
The dopamine transporter (DAT) has been implicated in a variety of arousal-related processes including the regulation of motor activity, learning, motivated behavior, psychostimulant abuse, and, more recently, sleep/wake state. We previously demonstrated that DAT uptake regulates fluctuations in extracellular dopamine (DA) in the striatum across the light/dark cycle with DA levels at their highest during the dark phase and lowest during the light phase. Despite this evidence, whether fluctuations in DA uptake across the light/dark cycle are associated with changes in sleep/wake has not been tested. To address this, we employed a combination of sleep/wake recordings, fast scan cyclic voltammetry, and western blotting to examine whether sleep/wake state and/or light/dark phase impact DA terminal neurotransmission in male rats. Further, we assessed whether variations in plasma membrane DAT levels and/or phosphorylation of the threonine 53 site on the DAT accounts for fluctuations in DA neurotransmission. Given the extensive evidence indicating that psychostimulants increase DA through interactions with the DAT, we also examined to what degree the effects of cocaine at inhibiting the DAT vary across sleep/wake state. Results demonstrated a significant association between individual sleep/wake states and DA terminal neurotransmission, with higher DA uptake rate, increased phosphorylation of the DAT, and enhanced cocaine potency observed after periods of sleep. These findings suggest that sleep/wake state influences DA neurotransmission in a manner that is likely to impact a host of DA-dependent processes including a variety of neuropsychiatric disorders.
Collapse
Affiliation(s)
- I. P. Alonso
- grid.166341.70000 0001 2181 3113Department of Neurobiology and Anatomy, Drexel University College of Medicine, Philadelphia, PA 19129 USA
| | - J. A. Pino
- grid.440631.40000 0001 2228 7602Departamento de Medicina, Facultad de Medicina, Universidad de Atacama, 1532502 Copiapó, Chile
| | - S. Kortagere
- grid.166341.70000 0001 2181 3113Department of Microbiology and Immunology, Drexel University College of Medicine, Philadelphia, PA 19129 USA
| | - G. E. Torres
- grid.254250.40000 0001 2264 7145Department of Molecular, Cellular & Biomedical Sciences, CUNY School of Medicine at the City College of New York, New York, NY 10031 USA
| | - R. A. España
- grid.166341.70000 0001 2181 3113Department of Neurobiology and Anatomy, Drexel University College of Medicine, Philadelphia, PA 19129 USA
| |
Collapse
|
11
|
Ingram SM, Rana T, Manson AM, Yayah FM, Jackson EGB, Anderson C, Davids BO, Goodwin JS. Optogenetically-induced multimerization of the dopamine transporter increases uptake and trafficking to the plasma membrane. J Biol Chem 2021; 296:100787. [PMID: 34015332 PMCID: PMC8203837 DOI: 10.1016/j.jbc.2021.100787] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/22/2020] [Revised: 05/07/2021] [Accepted: 05/13/2021] [Indexed: 11/19/2022] Open
Abstract
The dopamine transporter (DAT) is essential for the reuptake of the released neurotransmitter dopamine (DA) in the brain. Psychostimulants, methamphetamine and cocaine, have been reported to induce the formation of DAT multimeric complexes in vivo and in vitro. The interpretation of DAT multimer function has been primarily in the context of compounds that induce structural and functional modifications of the DAT, complicating the understanding of the significance of DAT multimers. To examine multimerization in the absence of DAT ligands as well as in their presence, we developed a novel, optogenetic fusion chimera of cryptochrome 2 and DAT with an mCherry fluorescent reporter (Cry2-DAT). Using blue light to induce Cry2-DAT multimeric protein complex formation, we were able to simultaneously test the functional contributions of DAT multimerization in the absence or presence of substrates or inhibitors with high spatiotemporal precision. We found that blue light-stimulated Cry2-DAT multimers significantly increased IDT307 uptake and MFZ 9-18 binding in the absence of ligands as well as after methamphetamine and nomifensine treatment. Blue light-induced Cry2-DAT multimerization increased colocalization with recycling endosomal marker Rab11 and had decreased presence in Rab5-positive early endosomes and Rab7-positive late endosomes. Our data suggest that the increased uptake and binding results from induced and rapid trafficking of DAT multimers to the plasma membrane. Our data suggest that DAT multimers may function to help maintain DA homeostasis.
Collapse
Affiliation(s)
- Shalonda M Ingram
- Department of Biochemistry, Cancer Biology, Neuroscience, and Pharmacology, School of Medicine, Meharry Medical College, Nashville, Tennessee, USA
| | - Tanu Rana
- Department of Biochemistry, Cancer Biology, Neuroscience, and Pharmacology, School of Medicine, Meharry Medical College, Nashville, Tennessee, USA
| | - Ashley M Manson
- Department of Biochemistry, Cancer Biology, Neuroscience, and Pharmacology, School of Medicine, Meharry Medical College, Nashville, Tennessee, USA
| | - Faisal M Yayah
- Department of Biochemistry, Cancer Biology, Neuroscience, and Pharmacology, School of Medicine, Meharry Medical College, Nashville, Tennessee, USA
| | - Evan G B Jackson
- Department of Biochemistry, Cancer Biology, Neuroscience, and Pharmacology, School of Medicine, Meharry Medical College, Nashville, Tennessee, USA
| | - Christopher Anderson
- Department of Biochemistry, Cancer Biology, Neuroscience, and Pharmacology, School of Medicine, Meharry Medical College, Nashville, Tennessee, USA
| | - Benem-Orom Davids
- Department of Biochemistry, Cancer Biology, Neuroscience, and Pharmacology, School of Medicine, Meharry Medical College, Nashville, Tennessee, USA
| | - J Shawn Goodwin
- Department of Biochemistry, Cancer Biology, Neuroscience, and Pharmacology, School of Medicine, Meharry Medical College, Nashville, Tennessee, USA.
| |
Collapse
|
12
|
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
|
13
|
Ferdous N, Kudumala S, Sossi S, Carvelli L. Prolonged Amphetamine Treatments Cause Long-Term Decrease of Dopamine Uptake in Cultured Cells. Neurochem Res 2019; 45:1399-1409. [PMID: 31883055 PMCID: PMC7260268 DOI: 10.1007/s11064-019-02938-7] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/16/2019] [Revised: 12/12/2019] [Accepted: 12/18/2019] [Indexed: 11/24/2022]
Abstract
Amphetamine (AMPH) is a systemic stimulant used to treat a variety of diseases including Attention Deficit Hyperactive Disorder, narcolepsy and obesity. Previous data showed that by binding to catecholamine transporters, AMPH prevents the reuptake of the neurotransmitters dopamine (DA) and norepinephrine (NE). Because AMPH, either used therapeutically at final concentrations of 1–10 µM or abused as recreational drug (50–200 µM), is taken over long periods of time, we investigated the prolonged effects of this drug on the uptake of DA. We found that, in LLC-PK1 cells stably expressing the human DA transporter (hDAT), pretreatments with 1 or 50 µM AMPH caused significant reduction in DA uptake right after the 15-h pretreatment. Remarkably, after 50 but not 1 µM AMPH pretreatment, we observed a significant reduction in DA uptake also after one, two or three cell divisions. To test whether these long-term effects induced by AMPH where conserved in a model comparable to primordial neuronal cells and native neurons, we used the human neuroblastoma cell line SH-SY5Y cells, which were reported to endogenously express both hDAT and the NE transporter. Pretreatments with 50 µM AMPH caused a significant reduction of DA uptake both right after 15 h and 3 cell divisions followed by neuro-differentiation with retinoic acid (RA) for 5 days. Under these same conditions, AMPH did not change the intracellular concentrations of ATP, ROS and cell viability suggesting, therefore, that the reduction in DA uptake was not cause by AMPH-induced toxicity. Interestingly, while 1 µM AMPH did not cause long-term effects in the LLC-PK1 cells, in the SH-SY5Y cells, it decreased the DA uptake after one, two, but not three, cell divisions and 5-day RA differentiation. These data show that besides the well-known acute effects, AMPH can also produce long-term effects in vitro that are maintained during cell division and transmitted to the daughter cells.
Collapse
Affiliation(s)
- Nafisa Ferdous
- Department of Biomedical Science, University of North Dakota, Grand Forks, ND, USA
| | - Sirisha Kudumala
- Harriet L. Wilkes Honors College, Florida Atlantic University, FL, Jupiter, USA
| | - Serena Sossi
- Integrated Biology Program, Florida Atlantic University, FL, Boca Raton, USA
| | - Lucia Carvelli
- Brain Institute, Florida Atlantic University, Jupiter, FL, USA. .,Harriet L. Wilkes Honors College, Florida Atlantic University, FL, Jupiter, USA. .,Integrated Biology Program, Florida Atlantic University, FL, Boca Raton, USA.
| |
Collapse
|
14
|
Geresu B, Canseco-Alba A, Sanabria B, Lin Z, Liu QR, Onaivi ES, Engidawork E. Involvement of CB2 Receptors in the Neurobehavioral Effects of Catha Edulis (Vahl) Endl. (Khat) in Mice. Molecules 2019; 24:E3164. [PMID: 31480324 PMCID: PMC6749201 DOI: 10.3390/molecules24173164] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/19/2019] [Revised: 08/26/2019] [Accepted: 08/29/2019] [Indexed: 12/13/2022] Open
Abstract
There is behavioral evidence for the interaction between crude khat extract and the endocannabinoid system, whereby the endocannabinoid system alters khat extract-mediated behavioral effects through modulation of the monoaminergic system. The objective of this study was to investigate the role of the endocannabinoid system on the neurobehavioral effect of khat extract in mice following concomitant administration of khat extract and the CB2R agonist, JWH133. Locomotor activity test, immunohistochemistry, and reverse transcriptase polymerase chain reaction technique were utilized to assess locomotor activity, tyrosine hydroxylase immunoreactivity, and expression of dopamine transporter mRNA gene. The results show sub-acute administration of khat extract alone increased locomotor activity in mice and co-administration of the CB2R agonist, JWH133, reduced khat extract induced hyperlocomotor activity. The data revealed that cell type specific deletion of CB2Rs on dopaminergic neurons increased the hyperlocomotor behavior of khat extract. Furthermore, the results revealed that khat extract attenuated MPTP induced motor deficits, which is enhanced by JWH133. Khat extract also increased expression of tyrosine hydroxylase positive cells and expression of dopamine transporter mRNA gene in wild type mice. Nevertheless, JWH133 did not alter the effect of khat extract on tyrosine hydroxylase immunoreactivity and dopamine transporter mRNA expression when given together with khat extract. Taken together, the results suggest that the CB2Rs selectively interact with khat extract-mediated locomotor effects and could be utilized as therapeutic target in central nervous system movement disorders associated with dopamine dysregulation.
Collapse
MESH Headings
- 1-Methyl-4-phenyl-1,2,3,6-tetrahydropyridine
- Animals
- Behavior, Animal/drug effects
- Brain/drug effects
- Brain/physiology
- Cannabinoids/administration & dosage
- Cannabinoids/pharmacology
- Catha/chemistry
- Dopamine Plasma Membrane Transport Proteins/genetics
- Dopamine Plasma Membrane Transport Proteins/metabolism
- Dopaminergic Neurons/drug effects
- Dopaminergic Neurons/physiology
- Gene Deletion
- Gene Expression Regulation/drug effects
- Mice, Inbred C57BL
- Mice, Knockout
- Motor Activity/drug effects
- Plant Extracts/pharmacology
- RNA, Messenger/genetics
- RNA, Messenger/metabolism
- Receptor, Cannabinoid, CB2/agonists
- Receptor, Cannabinoid, CB2/metabolism
- Tyrosine 3-Monooxygenase/metabolism
Collapse
Affiliation(s)
- Berhanu Geresu
- Department of Pharmacology and Clinical Pharmacy, School of Pharmacy, Addis Ababa University, 1176 Addis Ababa, Ethiopia
| | - Ana Canseco-Alba
- Department of Biology, William Paterson University, Wayne, NJ 07470, USA
| | - Branden Sanabria
- Department of Biology, William Paterson University, Wayne, NJ 07470, USA
| | - Zhicheng Lin
- Department of Psychiatry, Harvard Medical School, Psychiatric Neurogenomics, Division of Alcohol and Drug Abuse, and Mailman Neuroscience Research Center, McLean Hospital, Belmont, MA 02478, USA
| | - Qing-Rong Liu
- Laboratory of Clinical Investigation, National Institute on Aging, National Institutes of Health, Baltimore, MD 21224, USA
| | - Emmanuel S Onaivi
- Department of Biology, William Paterson University, Wayne, NJ 07470, USA.
| | - Ephrem Engidawork
- Department of Pharmacology and Clinical Pharmacy, School of Pharmacy, Addis Ababa University, 1176 Addis Ababa, Ethiopia.
| |
Collapse
|
15
|
The effects of proteasome on baseline and methamphetamine-dependent dopamine transmission. Neurosci Biobehav Rev 2019; 102:308-317. [DOI: 10.1016/j.neubiorev.2019.05.008] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/13/2018] [Revised: 04/29/2019] [Accepted: 05/09/2019] [Indexed: 12/16/2022]
|
16
|
Yu Y, de Campos RPS, Hong S, Krastev DL, Sadanand S, Leung Y, Wheeler AR. A microfluidic platform for continuous monitoring of dopamine homeostasis in dopaminergic cells. MICROSYSTEMS & NANOENGINEERING 2019; 5:10. [PMID: 31057937 PMCID: PMC6409360 DOI: 10.1038/s41378-019-0049-2] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/27/2018] [Revised: 01/08/2019] [Accepted: 01/10/2019] [Indexed: 05/24/2023]
Abstract
Homeostasis of dopamine, a classical neurotransmitter, is a key indicator of neuronal health. Dysfunction in the regulation of dopamine is implicated in a long list of neurological disorders, including addiction, depression, and neurodegeneration. The existing methods used to evaluate dopamine homeostasis in vitro are inconvenient and do not allow for continuous non-destructive measurement. In response to this challenge, we introduce an integrated microfluidic system that combines dopaminergic cell culture and differentiation with electroanalytical measurements of extracellular dopamine in real-time at any point during an assay. We used the system to examine the behavior of differentiated SH-SY5Y cells upon exposure to four dopamine transporter ant/agonists (cocaine, ketamine, epigallocatechin gallate, and amphetamine) and study their pharmacokinetics. The IC50 values of cocaine, ketamine, and epigallocatechin gallate were determined to be (average ± standard deviation) 3.7 ± 1.1 µM, 51.4 ± 17.9 µM, and 2.6 ± 0.8 µM, respectively. Furthermore, we used the new system to study amphetamine-mediated dopamine release to probe the related phenomena of dopamine transporter-mediated reverse-transport and dopamine release from vesicles. We propose that this platform, which is the first platform to simultaneously evaluate uptake and release, could be useful to screen for drugs and other agents that target dopaminergic neurons and the function of the dopamine transporter. More broadly, this platform should be adaptable for any application that could benefit from high-temporal resolution electroanalysis combined with multi-day cell culture using small numbers of cells.
Collapse
Affiliation(s)
- Yue Yu
- Institute for Biomaterials and Biomedical Engineering, University of Toronto, 164 College St, Toronto, ON M5s 3G9 Canada
- Donnelly Centre for Cellular and Biomolecular Research, 160 College St., Toronto, ON M5S 3E1 Canada
| | - Richard P. S. de Campos
- Donnelly Centre for Cellular and Biomolecular Research, 160 College St., Toronto, ON M5S 3E1 Canada
- Department of Chemistry, University of Toronto, 80 St George St., Toronto, ON M5S 3H6 Canada
| | - Seolim Hong
- Department of Human Biology, University of Toronto, 300 Huron Street, Toronto, ON M5S 3J6 Canada
| | - Dimitar L. Krastev
- Donnelly Centre for Cellular and Biomolecular Research, 160 College St., Toronto, ON M5S 3E1 Canada
- Department of Human Biology, University of Toronto, 300 Huron Street, Toronto, ON M5S 3J6 Canada
| | - Siddharth Sadanand
- Donnelly Centre for Cellular and Biomolecular Research, 160 College St., Toronto, ON M5S 3E1 Canada
| | - Yen Leung
- Donnelly Centre for Cellular and Biomolecular Research, 160 College St., Toronto, ON M5S 3E1 Canada
| | - Aaron R. Wheeler
- Institute for Biomaterials and Biomedical Engineering, University of Toronto, 164 College St, Toronto, ON M5s 3G9 Canada
- Donnelly Centre for Cellular and Biomolecular Research, 160 College St., Toronto, ON M5S 3E1 Canada
- Department of Chemistry, University of Toronto, 80 St George St., Toronto, ON M5S 3H6 Canada
| |
Collapse
|
17
|
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
|
18
|
Amato D, Kruyer A, Samaha AN, Heinz A. Hypofunctional Dopamine Uptake and Antipsychotic Treatment-Resistant Schizophrenia. Front Psychiatry 2019; 10:314. [PMID: 31214054 PMCID: PMC6557273 DOI: 10.3389/fpsyt.2019.00314] [Citation(s) in RCA: 29] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/30/2019] [Accepted: 04/23/2019] [Indexed: 01/07/2023] Open
Abstract
Antipsychotic treatment resistance in schizophrenia remains a major issue in psychiatry. Nearly 30% of patients with schizophrenia do not respond to antipsychotic treatment, yet the underlying neurobiological causes are unknown. All effective antipsychotic medications are thought to achieve their efficacy by targeting the dopaminergic system. Here we review early literature describing the fundamental mechanisms of antipsychotic drug efficacy, highlighting mechanistic concepts that have persisted over time. We then reconsider the original framework for understanding antipsychotic efficacy in light of recent advances in our scientific understanding of the dopaminergic effects of antipsychotics. Based on these new insights, we describe a role for the dopamine transporter in the genesis of both antipsychotic therapeutic response and primary resistance. We believe that this discussion will help delineate the dopaminergic nature of antipsychotic treatment-resistant schizophrenia.
Collapse
Affiliation(s)
- Davide Amato
- Department of Neuroscience, Medical University of South Carolina, Charleston, SC, United States
| | - Anna Kruyer
- Department of Neuroscience, Medical University of South Carolina, Charleston, SC, United States
| | - Anne-Noël Samaha
- Department of Pharmacology and Physiology, Faculty of Medicine, Université de Montréal, Montreal, QC, Canada
| | - Andreas Heinz
- Department of Psychiatry, Charité University Medicine Berlin, Campus Charité Mitte, Berlin, Germany
| |
Collapse
|
19
|
Abstract
There is a plethora of amphetamine derivatives exerting stimulant, euphoric, anti-fatigue, and hallucinogenic effects; all structural properties allowing these effects are contained within the amphetamine structure. In the first part of this review, the interaction of amphetamine with the dopamine transporter (DAT), crucially involved in its behavioral effects, is covered, as well as the role of dopamine synthesis, the vesicular monoamine transporter VMAT2, and organic cation 3 transporter (OCT3). The second part deals with requirements in amphetamine's effect on the kinases PKC, CaMKII, and ERK, whereas the third part focuses on where we are in developing anti-amphetamine therapeutics. Thus, treatments are discussed that target DAT, VMAT2, PKC, CaMKII, and OCT3. As is generally true for the development of therapeutics for substance use disorder, there are multiple preclinically promising specific compounds against (meth)amphetamine, for which further development and clinical trials are badly needed.
Collapse
Affiliation(s)
- Maarten E A Reith
- Department of Psychiatry, New York University School of Medicine, New York, NY, USA.
| | - Margaret E Gnegy
- Department of Pharmacology, University of Michigan School of Medicine, Ann Arbor, MI, USA
| |
Collapse
|
20
|
Silwal AP, Lu HP. Mode-Selective Raman Imaging of Dopamine-Human Dopamine Transporter Interaction in Live Cells. ACS Chem Neurosci 2018; 9:3117-3127. [PMID: 30024721 DOI: 10.1021/acschemneuro.8b00301] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/30/2022] Open
Abstract
Dopamine (DA) is the catecholamine neurotransmitter which interacts with dopamine receptors (DARs) to generate dopaminergic signals in the nervous system. Dopamine transporter (DAT) interacts with DA to maintain DA's homeostasis in synaptic and perisynaptic space. DAT and DARs have great importance in the central nervous system (CNS) because they are associated with the targeted binding of drugs. Interactions of DA, its analogue with DARs, or DAT have been studied extensively to understand the mechanism of the dopaminergic signaling process and several neurodegenerative diseases, including schizophrenia, Parkinson's diseases, addiction, attention deficit hyperactivity disorder, and bipolar disorder. However, there is still a lack of a risk-free, label-free, and minimally invasive imaging approach to probe the interaction between DA and DAT or DARs. Here, we probed the DA, human dopamine transporter (hDAT), and DA-hDAT interactions in live cells using combined approach of two-photon excited (2PE) fluorescence imaging and mode-selective Raman measurement. We utilized the signature Raman peak at 1287 cm-1 to probe the location of DA and 807 and 1076 cm-1 to probe the DA-hDAT interaction in live cells. We found that the combined approach of mode-selective Raman imaging, 2PE fluorescence imaging, and computational methods is successful to probe and confirm the DA-hDAT interactions in living cells. The probing of the interactions of DARs or DAT with DA or other targeting drugs is crucial for the diagnosis and cure of several neurodegenerative diseases. Also, this analytical approach could be extended to probe other types of protein-ligand interactions.
Collapse
Affiliation(s)
- Achut P. Silwal
- Department of Chemistry and Center for Photochemical Sciences, Bowling Green State University, Ohio 43403, United States
| | - H. Peter Lu
- Department of Chemistry and Center for Photochemical Sciences, Bowling Green State University, Ohio 43403, United States
| |
Collapse
|
21
|
Silwal A, Lu HP. Raman Spectroscopic Analysis of Signaling Molecules-Dopamine Receptors Interactions in Living Cells. ACS OMEGA 2018; 3:14849-14857. [PMID: 30555993 PMCID: PMC6289496 DOI: 10.1021/acsomega.8b01727] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 07/20/2018] [Accepted: 10/16/2018] [Indexed: 06/09/2023]
Abstract
The selective interaction of signaling compounds including neurotransmitters and drugs with the dopamine receptors (DARs) is extremely important for the treatment of neurodegenerative diseases. Here, we report a method to probe the selective interactions of signaling compounds with D1 and D2 DARs in living cells using the combined approach of theoretical calculation and surface-enhanced Raman spectroscopy (SERS). When signaling compounds such as DA, amphetamine, methamphetamine, and methylenedioxypyrovalerone interact with D1 dopamine receptors (DRD1), the intracellular cyclic adenosine monophosphate (cAMP) level is increased. However, the intracellular level of cAMP is decreased when D2 dopamine receptors (DRD2) interact with the abovementioned signaling compounds. In our experiments, we have internalized the silica-coated silver nanoparticles (AgNP@SiO2) in living cells to adsorb biologically generated cAMP which was probed by using SERS. Besides adsorptions of cAMP, AgNP@SiO2 has a crucial role for the enhancement of Raman cross section of the samples. We observed the characteristic SERS peaks of cAMP when DRD1-overexpressed cells interact with the signaling compounds; these peaks were not observed for other cells including DRD2-overexpressed and DRD1-DRD2-coexpressed cells. Our experimental approach is successful to probe the intracellular cAMP and characterize the selectivity of signaling compounds to different types of DARs. Furthermore, our experimental approach is highly capable for in vivo studies because it can probe intracellular cAMP using a low input power of incident laser without significant cell damage. Our experimental results and density functional theory calculations showed that 780 and 1503 cm-1 are signature Raman peaks of cAMP. The SERS peak at 780 cm-1 is associated with C-O, C-C, and C-N stretching and symmetric and asymmetric bending of two O-H bonds of cAMP, whereas the SERS peak at 1503 cm-1 is contributed by the O9-H3 bending mode.
Collapse
|
22
|
Cellular and molecular responses to acute cocaine treatment in neuronal-like N2a cells: potential mechanism for its resistance in cell death. Cell Death Discov 2018; 4:13. [PMID: 30210816 PMCID: PMC6133924 DOI: 10.1038/s41420-018-0078-x] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/17/2018] [Revised: 06/11/2018] [Accepted: 06/22/2018] [Indexed: 02/07/2023] Open
Abstract
Cocaine is a highly abused drug that causes psychiatric and neurological problems. Its entry into neurons could alter cell-biochemistry and contribute in the manifestation of early pathological symptoms. We have previously shown the acute cocaine effects in rat C6 astroglia-like cells and found that these cells were highly sensitive to cocaine in terms of manifesting certain pathologies known to underlie psychological disorders. The present study was aimed to discern acute cocaine effects on the early onset of various changes in Neuro-2a (N2a) cells. Whole-cell patch-clamp recording of differentiated cells displayed the functional voltage-gated Na+ and K+ channels, which demonstrated the neuronal characteristics of the cells. Treatment of these cells with acute cocaine (1 h) at in vivo (nM to μM) and in vitro (mM) concentrations revealed that the cells remained almost 100% viable. Cocaine administration at 6.25 μM or 4 mM doses significantly reduced the inward currents but had no significant effect on outward currents, indicating the Na+ channel-blocking activity of cocaine. While no morphological change was observed at in vivo doses, treatment at in vitro doses altered the morphology, damaged the neurites, and induced cytoplasmic vacuoles; furthermore, general mitochondrial activity and membrane potential were significantly decreased. Mitochondrial dysfunction enabled the cells switch to anaerobic glycolysis, evidenced by dose-dependent increases in lactate and H2S, resulting unaltered ATP level in the cells. Further investigation on the mechanism of action unfolded that the cell's resistance to cocaine was through the activation of nuclear factor E2-related factor-2 (Nrf-2) gene and subsequent increase of antioxidants (glutathione [GSH], catalase and GSH peroxidase [GPx]). The data clearly indicate that the cells employed a detoxifying strategy against cocaine. On a broader perspective, we envision that extrapolating the knowledge of neuronal resistance to central nervous system (CNS) diseases could delay their onset or progression.
Collapse
|
23
|
Huang L, Liu M, Huang H, Wen Y, Zhang X, Wei Y. Recent Advances and Progress on Melanin-like Materials and Their Biomedical Applications. Biomacromolecules 2018; 19:1858-1868. [DOI: 10.1021/acs.biomac.8b00437] [Citation(s) in RCA: 174] [Impact Index Per Article: 24.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/20/2023]
Affiliation(s)
- Long Huang
- Department of Chemistry, Nanchang University, 999 Xuefu Avenue, Nanchang 330031, China
| | - Meiying Liu
- Department of Chemistry, Nanchang University, 999 Xuefu Avenue, Nanchang 330031, China
| | - Hongye Huang
- Department of Chemistry, Nanchang University, 999 Xuefu Avenue, Nanchang 330031, China
| | - Yuanqing Wen
- Department of Chemistry, Nanchang University, 999 Xuefu Avenue, Nanchang 330031, China
| | - Xiaoyong Zhang
- Department of Chemistry, Nanchang University, 999 Xuefu Avenue, Nanchang 330031, China
| | - Yen Wei
- Department of Chemistry and the Tsinghua Center for Frontier Polymer Research, Tsinghua University, Beijing 100084, P.R. China
- Department of Chemistry and Center for Nanotechnology, Chung-Yuan Christian University, Chung-Li 32023, Taiwan
| |
Collapse
|
24
|
Covarrubias-Pinto A, Acuña AI, Boncompain G, Papic E, Burgos PV, Perez F, Castro MA. Ascorbic acid increases SVCT2 localization at the plasma membrane by accelerating its trafficking from early secretory compartments and through the endocytic-recycling pathway. Free Radic Biol Med 2018; 120:181-191. [PMID: 29545069 DOI: 10.1016/j.freeradbiomed.2018.03.013] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/30/2018] [Revised: 03/06/2018] [Accepted: 03/08/2018] [Indexed: 11/30/2022]
Abstract
Ascorbic acid (Asc) is an antioxidant molecule essential for physiological functions. The concentration of extracellular Asc increases during synaptic transmission and renal reabsorption. These phenomena induce an increase of the Sodium-dependent-Vitamin-C-transporter 2 (SVCT2) at plasma membrane (PM) localization, as we previously demonstrated in neuronal and non-neuronal cells. Hence, the aim of this study was to evaluate intracellular SVCT2 trafficking kinetics in response to Asc. We observed two peaks of SVCT2 localization and function at the PM (at 5-10 min, "acute response", and 30-60 min, "post-acute response") when cells were incubated with Asc. We defined that the post-acute response was dependent on SVCT2 located in early secretory compartments, and its trafficking was abolished with Tunicamycin and Brefeldin A treatment. Moreover, using the RUSH system to retain and synchronize cargo secretion through the secretory pathway we demonstrated that the post-acute response increases SVCT2 trafficking kinetics from the ER to the PM suggesting the retention of SVCT2 at the early secretory pathway when Asc is absent. However, these observations do not explain the increased SVCT2 levels at the PM during the "acute" response, suggesting the involvement of a faster mechanism in close proximity with the PM. To investigate the possible role of endosomal compartments, we tested the effect of endocytosis inhibition. Expression of dominant-negative (DN) versions of the GTPase-dynamin II and clathrin-accessory protein AP180 showed a significant increase in SVCT2 levels at the PM. Moreover, expression of Rab11-DN, a GTPase implicated in cargo protein recycling from endosomes to the PM showed a similar outcome, strongly indicating that Asc impacts SVCT2 trafficking during the acute response. Therefore, our results revealed two mechanisms by which Asc modulates SVCT2 levels at the PM, one at the early secretory pathway and another at the endocytic compartments. We propose that these two mechanisms have key protective implications in the homeostasis of metabolically active and specialized tissues.
Collapse
Affiliation(s)
- A Covarrubias-Pinto
- Biochemistry and Microbiology Institute, Faculty of Sciences, Universidad Austral de Chile, Valdivia, Chile; Center for Interdisciplinary Studies of the Nervous System (CISNe), Universidad Austral de Chile, Valdivia, Chile
| | - A I Acuña
- Biochemistry and Microbiology Institute, Faculty of Sciences, Universidad Austral de Chile, Valdivia, Chile; Center for Interdisciplinary Studies of the Nervous System (CISNe), Universidad Austral de Chile, Valdivia, Chile
| | - G Boncompain
- Institut Curie, PSL Research University, CNRS UMR144, Paris, France
| | - E Papic
- Biochemistry and Microbiology Institute, Faculty of Sciences, Universidad Austral de Chile, Valdivia, Chile; Center for Interdisciplinary Studies of the Nervous System (CISNe), Universidad Austral de Chile, Valdivia, Chile
| | - P V Burgos
- Department of Physiology, Faculty of Medicine, Universidad Austral de Chile, Valdivia, Chile; Center for Interdisciplinary Studies of the Nervous System (CISNe), Universidad Austral de Chile, Valdivia, Chile; Center for Cell Biology and Biomedicine, School of Sciences and School of Medicine, Universidad San Sebastián, Santiago, Chile
| | - F Perez
- Institut Curie, PSL Research University, CNRS UMR144, Paris, France
| | - M A Castro
- Biochemistry and Microbiology Institute, Faculty of Sciences, Universidad Austral de Chile, Valdivia, Chile; Center for Interdisciplinary Studies of the Nervous System (CISNe), Universidad Austral de Chile, Valdivia, Chile; Research Initiative for Brain Rejuvenation (ReBrain), Chile.
| |
Collapse
|
25
|
Carpenter C, Zestos AG, Altshuler R, Sorenson RJ, Guptaroy B, Showalter HD, Kennedy RT, Jutkiewicz E, Gnegy ME. Direct and Systemic Administration of a CNS-Permeant Tamoxifen Analog Reduces Amphetamine-Induced Dopamine Release and Reinforcing Effects. Neuropsychopharmacology 2017; 42:1940-1949. [PMID: 28492278 PMCID: PMC5561350 DOI: 10.1038/npp.2017.95] [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: 03/08/2017] [Revised: 04/29/2017] [Accepted: 05/03/2017] [Indexed: 01/09/2023]
Abstract
Amphetamines (AMPHs) are globally abused. With no effective treatment for AMPH addiction to date, there is urgent need for the identification of druggable targets that mediate the reinforcing action of this stimulant class. AMPH-stimulated dopamine efflux is modulated by protein kinase C (PKC) activation. Inhibition of PKC reduces AMPH-stimulated dopamine efflux and locomotor activity. The only known CNS-permeant PKC inhibitor is the selective estrogen receptor modulator tamoxifen. In this study, we demonstrate that a tamoxifen analog, 6c, which more potently inhibits PKC than tamoxifen but lacks affinity for the estrogen receptor, reduces AMPH-stimulated increases in extracellular dopamine and reinforcement-related behavior. In rat striatal synaptosomes, 6c was almost fivefold more potent at inhibiting AMPH-stimulated dopamine efflux than [3H]dopamine uptake through the dopamine transporter (DAT). The compound did not compete with [3H]WIN 35,428 binding or affect surface DAT levels. Using microdialysis, direct accumbal administration of 1 μM 6c reduced dopamine overflow in freely moving rats. Using LC-MS, we demonstrate that 6c is CNS-permeant. Systemic treatment of rats with 6 mg/kg 6c either simultaneously or 18 h prior to systemic AMPH administration reduced both AMPH-stimulated dopamine overflow and AMPH-induced locomotor effects. Finally, 18 h pretreatment of rats with 6 mg/kg 6c s.c. reduces AMPH-self administration but not food self-administration. These results demonstrate the utility of tamoxifen analogs in reducing AMPH effects on dopamine and reinforcement-related behaviors and suggest a new avenue of development for therapeutics to reduce AMPH abuse.
Collapse
Affiliation(s)
- Colleen Carpenter
- Department of Pharmacology, University of Michigan, Ann Arbor, MI, USA
| | - Alexander G Zestos
- Department of Pharmacology, University of Michigan, Ann Arbor, MI, USA
- Department of Chemistry, University of Michigan, Ann Arbor, MI, USA
| | - Rachel Altshuler
- Department of Pharmacology, University of Michigan, Ann Arbor, MI, USA
| | - Roderick J Sorenson
- Department of Medicinal Chemistry, University of Michigan, Ann Arbor, MI, USA
- Vahlteich Medicinal Chemistry Core, University of Michigan, Ann Arbor, MI, USA
| | - Bipasha Guptaroy
- Department of Pharmacology, University of Michigan, Ann Arbor, MI, USA
| | - Hollis D Showalter
- Department of Medicinal Chemistry, University of Michigan, Ann Arbor, MI, USA
- Vahlteich Medicinal Chemistry Core, University of Michigan, Ann Arbor, MI, USA
| | - Robert T Kennedy
- Department of Pharmacology, University of Michigan, Ann Arbor, MI, USA
- Department of Chemistry, University of Michigan, Ann Arbor, MI, USA
| | - Emily Jutkiewicz
- Department of Pharmacology, University of Michigan, Ann Arbor, MI, USA
| | - Margaret E Gnegy
- Department of Pharmacology, University of Michigan, Ann Arbor, MI, USA
| |
Collapse
|
26
|
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
|
27
|
Silwal AP, Yadav R, Sprague JE, Lu HP. Raman Spectroscopic Signature Markers of Dopamine-Human Dopamine Transporter Interaction in Living Cells. ACS Chem Neurosci 2017; 8:1510-1518. [PMID: 28375605 DOI: 10.1021/acschemneuro.7b00048] [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] [Indexed: 12/11/2022] Open
Abstract
Dopamine (DA) controls many psychological and behavioral activities in the central nervous system (CNS) through interactions with the human dopamine transporter (hDAT) and dopamine receptors. The roles of DA in the function of the CNS are affected by the targeted binding of drugs to hDAT; thus, hDAT plays a critical role in neurophysiology and neuropathophysiology. An effective experimental method is necessary to study the DA-hDAT interaction and effects of variety of drugs like psychostimulants and antidepressants that are dependent on this interaction. In searching for obtaining and identifying the Raman spectral signatures, we have used surface enhanced Raman scattering (SERS) spectroscopy to record SERS spectra from DA, human embryonic kidney 293 cells (HEK293), hDAT-HEK293, DA-HEK293, and DA-hDAT-HEK293. We have demonstrated a specific 2D-distribution SERS spectral analytical approach to analyze DA-hDAT interaction. Our study shows that the Raman modes at 807, 839, 1076, 1090, 1538, and 1665 cm-1 are related to DA-hDAT interaction, where Raman shifts at 807 and 1076 cm-1 are the signature markers for the bound state of DA to probe DA-hDAT interaction. On the basis of density function theory (DFT) calculation, Raman shift of the bound state of DA at 807 cm-1 is related to combination of bending modes α(C3-O10-H21), α(C2-O11-H22), α(C7-C8-H18), α(C6-C4-H13), α(C7-C8-H19), and α(C7-C8-N9), and Raman shift at 1076 cm-1 is related to combination of bending modes α(H19-N9-C8), γ(N9-H19), γ(C8-H19), γ(N9-H20), γ(C8-H18), and α(C7-C8-H18). These findings demonstrate that protein-ligand interactions can be confirmed by probing change in Raman shift of ligand molecules, which could be crucial to understanding molecular interactions between neurotransmitters and their receptors or transporters.
Collapse
Affiliation(s)
- Achut P. Silwal
- Department of Chemistry
and Center for Photochemical Sciences, Bowling Green State University, Bowling
Green, Ohio 43403, United States
| | - Rajeev Yadav
- Department of Chemistry
and Center for Photochemical Sciences, Bowling Green State University, Bowling
Green, Ohio 43403, United States
| | - Jon E. Sprague
- The Ohio Attorney General’s Center for the Future of Forensic Science, Bowling Green, Ohio 43403, United States
| | - H. Peter Lu
- Department of Chemistry
and Center for Photochemical Sciences, Bowling Green State University, Bowling
Green, Ohio 43403, United States
| |
Collapse
|
28
|
Nora GJ, Harun R, Fine DF, Hutchison D, Grobart AC, Stezoski JP, Munoz MJ, Kochanek PM, Leak RK, Drabek T, Wagner AK. Ventricular fibrillation cardiac arrest produces a chronic striatal hyperdopaminergic state that is worsened by methylphenidate treatment. J Neurochem 2017; 142:305-322. [PMID: 28445595 DOI: 10.1111/jnc.14058] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/27/2016] [Revised: 04/19/2017] [Accepted: 04/20/2017] [Indexed: 01/21/2023]
Abstract
Cardiac arrest survival rates have improved with modern resuscitation techniques, but many survivors experience impairments associated with hypoxic-ischemic brain injury (HIBI). Currently, little is understood about chronic changes in striatal dopamine (DA) systems after HIBI. Given the common empiric clinical use of DA enhancing agents in neurorehabilitation, investigation evaluating dopaminergic alterations after cardiac arrest (CA) is necessary to optimize rehabilitation approaches. We hypothesized that striatal DA neurotransmission would be altered chronically after ventricular fibrillation cardiac arrest (VF-CA). Fast-scan cyclic voltammetry was used with median forebrain bundle (MFB) maximal electrical stimulations (60Hz, 10s) in rats to characterize presynaptic components of DA neurotransmission in the dorsal striatum (D-Str) and nucleus accumbens 14 days after a 5-min VF-CA when compared to Sham or Naïve. VF-CA increased D-Str-evoked overflow [DA], total [DA] released, and initial DA release rate versus controls, despite also increasing maximal velocity of DA reuptake (Vmax ). Methylphenidate (10 mg/kg), a DA transporter inhibitor, was administered to VF-CA and Shams after establishing a baseline, pre-drug 60 Hz, 5 s stimulation response. Methylphenidate increased initial evoked overflow [DA] more-so in VF-CA versus Sham and reduced D-Str Vmax in VF-CA but not Shams; these findings are consistent with upregulated striatal DA transporter in VF-CA versus Sham. Our work demonstrates that 5-min VF-CA increases electrically stimulated DA release with concomitant upregulation of DA reuptake 2 weeks after brief VF-CA insult. Future work should elucidate how CA insult duration, time after insult, and insult type influence striatal DA neurotransmission and related cognitive and motor functions.
Collapse
Affiliation(s)
- Gerald J Nora
- Department of Physical Medicine and Rehabilitation, University of Pittsburgh, Pittsburgh, Pennsylvania, USA
| | - Rashed Harun
- Department of Physical Medicine and Rehabilitation, University of Pittsburgh, Pittsburgh, Pennsylvania, USA
- Safar Center for Resuscitation Research, University of Pittsburgh, Pittsburgh, Pennsylvania, USA
- Center for Neuroscience, University of Pittsburgh, Pittsburgh, Pennsylvania, USA
| | - David F Fine
- Department of Physical Medicine and Rehabilitation, University of Pittsburgh, Pittsburgh, Pennsylvania, USA
- Safar Center for Resuscitation Research, University of Pittsburgh, Pittsburgh, Pennsylvania, USA
| | - Daniel Hutchison
- Mylan School of Pharmacy, Duquesne University, Pittsburgh, Pennsylvania, USA
| | - Adam C Grobart
- Department of Physical Medicine and Rehabilitation, University of Pittsburgh, Pittsburgh, Pennsylvania, USA
| | - Jason P Stezoski
- Safar Center for Resuscitation Research, University of Pittsburgh, Pittsburgh, Pennsylvania, USA
- Department of Critical Care Medicine, University of Pittsburgh, Pittsburgh, Pennsylvania, USA
| | - Miranda J Munoz
- Department of Physical Medicine and Rehabilitation, University of Pittsburgh, Pittsburgh, Pennsylvania, USA
| | - Patrick M Kochanek
- Safar Center for Resuscitation Research, University of Pittsburgh, Pittsburgh, Pennsylvania, USA
- Department of Critical Care Medicine, University of Pittsburgh, Pittsburgh, Pennsylvania, USA
| | - Rehana K Leak
- Mylan School of Pharmacy, Duquesne University, Pittsburgh, Pennsylvania, USA
| | - Tomas Drabek
- Safar Center for Resuscitation Research, University of Pittsburgh, Pittsburgh, Pennsylvania, USA
- Department of Anesthesiology, University of Pittsburgh, Pittsburgh, Pennsylvania, USA
| | - Amy K Wagner
- Department of Physical Medicine and Rehabilitation, University of Pittsburgh, Pittsburgh, Pennsylvania, USA
- Safar Center for Resuscitation Research, University of Pittsburgh, Pittsburgh, Pennsylvania, USA
- Center for Neuroscience, University of Pittsburgh, Pittsburgh, Pennsylvania, USA
- Department of Neuroscience, University of Pittsburgh, Pittsburgh, Pennsylvania, USA
| |
Collapse
|
29
|
Apostolou T, Moschopoulou G, Kolotourou E, Kintzios S. Assessment of in vitro dopamine-neuroblastoma cell interactions with a bioelectric biosensor: perspective for a novel in vitro functional assay for dopamine agonist/antagonist activity. Talanta 2017; 170:69-73. [PMID: 28501215 DOI: 10.1016/j.talanta.2017.03.098] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/08/2017] [Revised: 03/27/2017] [Accepted: 03/30/2017] [Indexed: 10/19/2022]
Abstract
Current receptor-binding assays for dopamine do not measure the in vitro whole cellular response against dopamine or potential agonist/antagonist molecules. We herewith report the development of a novel functional assay concept for studying the in vitro interaction of the neurotransmitter dopamine with neural cells bearing dopamine receptors. The concept is based on the ultra-rapid measurement of changes in the electric properties of cultured N2a mouse neuroblastoma cells (corresponding to cumulative changes of the cell membrane potential). A close relationship between cumulative cell membrane potential and dopamine concentration was observed. Membrane depolarization was observed at nanomolar dopamine concentrations, while hyperpolarization was associated with micromolar ones. Treatment with the dopamine D2-receptor antagonist eticlopride resulted to a concentration-dependent membrane depolarization. Treatment with sodium chloride caused considerable weakening of the dopamine-associated hyperpolarization effect. The observed bioelectric response to dopamine was highly inversely correlated with the pattern of dopamine release-uptake balance by N2a cells, as determined with cyclic voltammetry. The bioelectric approach was also used to evaluate the dopaminergic activity of chaste tree (Vitex agnus-castus) extracts. The novel assay concept offers promising perspectives for the development of advanced companion diagnostics system for the high throughput, fast functional characterization of neurotransmitter agonists and antagonists.
Collapse
Affiliation(s)
- Theofylaktos Apostolou
- Laboratory of Cell Technology, Faculty of Biotechnology, Agricultural University of Athens, Iera Odos 75, 11855 Athens, Greece
| | - Georgia Moschopoulou
- Laboratory of Cell Technology, Faculty of Biotechnology, Agricultural University of Athens, Iera Odos 75, 11855 Athens, Greece.
| | - Evdokia Kolotourou
- Laboratory of Cell Technology, Faculty of Biotechnology, Agricultural University of Athens, Iera Odos 75, 11855 Athens, Greece
| | - Spyridon Kintzios
- Laboratory of Cell Technology, Faculty of Biotechnology, Agricultural University of Athens, Iera Odos 75, 11855 Athens, Greece
| |
Collapse
|
30
|
Brodnik ZD, Ferris MJ, Jones SR, España RA. Reinforcing Doses of Intravenous Cocaine Produce Only Modest Dopamine Uptake Inhibition. ACS Chem Neurosci 2017; 8:281-289. [PMID: 27936579 DOI: 10.1021/acschemneuro.6b00304] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/14/2023] Open
Abstract
The reinforcing efficacy of cocaine is thought to stem from inhibition of the dopamine transporter (DAT) and subsequent increases in extracellular dopamine concentrations in the brain. In humans, this hypothesis has generally been supported by positron emission tomography imaging studies where the percent of DATs occupied by cocaine is used as a measure of cocaine activity in the brain. Interpretation of these studies, however, often relies on the assumption that measures of DAT occupancy directly correspond with functional DAT blockade. In the current studies, we used in vivo and in vitro fast scan cyclic voltammetry in mice to measure dopamine uptake inhibition following varying doses of cocaine as well as two high affinity DAT inhibitors. We then compared dopamine clearance rates following these drug treatments to dopamine clearance obtained from DAT knockout mice as a proxy for complete DAT blockade. We found that administration of abused doses of cocaine resulted in approximately 2% of maximal DAT blockade. Overall, our data indicate that abused doses of cocaine produce a relatively modest degree of DA uptake inhibition, and suggest that the relationship between DAT occupancy and functional blockade of the DAT is more complex than originally posited.
Collapse
Affiliation(s)
- Zachary D. Brodnik
- Department
of Neurobiology and Anatomy, Drexel University College of Medicine, Philadelphia, Pennsylvania 19129, United States
| | - Mark J. Ferris
- Department
of Physiology and Pharmacology, Wake Forest School of Medicine, Winston-Salem, North Carolina 27501, United States
| | - Sara R. Jones
- Department
of Physiology and Pharmacology, Wake Forest School of Medicine, Winston-Salem, North Carolina 27501, United States
| | - Rodrigo A. España
- Department
of Neurobiology and Anatomy, Drexel University College of Medicine, Philadelphia, Pennsylvania 19129, United States
| |
Collapse
|
31
|
Kim MS, Yu JH, Kim CH, Choi JY, Seo JH, Lee MY, Yi CH, Choi TH, Ryu YH, Lee JE, Lee BH, Kim H, Cho SR. Environmental enrichment enhances synaptic plasticity by internalization of striatal dopamine transporters. J Cereb Blood Flow Metab 2016; 36:2122-2133. [PMID: 26661218 PMCID: PMC5363660 DOI: 10.1177/0271678x15613525] [Citation(s) in RCA: 26] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/21/2015] [Revised: 09/20/2015] [Accepted: 09/21/2015] [Indexed: 12/20/2022]
Abstract
Environmental enrichment (EE) with a complex combination of physical, cognitive and social stimulations enhances synaptic plasticity and behavioral function. However, the mechanism remains to be elucidated in detail. We aimed to investigate dopamine-related synaptic plasticity underlying functional improvement after EE. For this, six-week-old CD-1 mice were randomly allocated to EE or standard conditions for two months. EE significantly enhanced behavioral functions such as rotarod and ladder walking tests. In a [18F]FPCIT positron emission tomography scan, binding values of striatal DAT were significantly decreased approximately 18% in the EE mice relative to the control mice. DAT inhibitor administrated to establish the relationship of the DAT down-regulation to the treatment effects also improved rotarod performances, suggesting that DAT inhibition recapitulated EE-mediated treatment benefits. Next, EE-induced internalization of DAT was confirmed using a surface biotinylation assay. In situ proximity ligation assay and immunoprecipitation demonstrated that EE significantly increased the phosphorylation of striatal DAT as well as the levels of DAT bound with protein kinase C (PKC). In conclusion, we suggest that EE enables phosphorylation of striatal DAT via a PKC-mediated pathway and causes DAT internalization. This is the first report to suggest an EE-mediated mechanism of synaptic plasticity by internalization of striatal DAT.
Collapse
Affiliation(s)
- Myung-Sun Kim
- Department and Research Institute of Rehabilitation Medicine, Yonsei University College of Medicine, Seoul, Korea.,Yonsei Stem Cell Center, Avison Biomedical Research Center, Seoul, Korea
| | - Ji Hea Yu
- Department and Research Institute of Rehabilitation Medicine, Yonsei University College of Medicine, Seoul, Korea.,Department of Medical Science, The Graduate School, Yonsei University, Seoul, Korea.,Brain Korea 21 PLUS Project for Medical Science, Yonsei University, Seoul, Korea
| | - Chul Hoon Kim
- Brain Korea 21 PLUS Project for Medical Science, Yonsei University, Seoul, Korea.,Department of Pharmacology, Yonsei University College of Medicine, Seoul, Korea.,Brain Research Institute, Yonsei University College of Medicine, Seoul, Korea
| | - Jae Yong Choi
- Department of Pharmacology, Yonsei University College of Medicine, Seoul, Korea.,Department of Nuclear Medicine, Yonsei University College of Medicine, Seoul, Korea
| | - Jung Hwa Seo
- Department and Research Institute of Rehabilitation Medicine, Yonsei University College of Medicine, Seoul, Korea.,Brain Korea 21 PLUS Project for Medical Science, Yonsei University, Seoul, Korea
| | - Min-Young Lee
- Department and Research Institute of Rehabilitation Medicine, Yonsei University College of Medicine, Seoul, Korea.,Yonsei Stem Cell Center, Avison Biomedical Research Center, Seoul, Korea
| | - Chi Hoon Yi
- Department of Molecular Imaging, Korea Institute of Radiological and Medical Sciences, Seoul, Korea
| | - Tae Hyun Choi
- Department of Molecular Imaging, Korea Institute of Radiological and Medical Sciences, Seoul, Korea
| | - Young Hoon Ryu
- Department of Nuclear Medicine, Yonsei University College of Medicine, Seoul, Korea
| | - Jong Eun Lee
- Brain Korea 21 PLUS Project for Medical Science, Yonsei University, Seoul, Korea.,Brain Research Institute, Yonsei University College of Medicine, Seoul, Korea.,Department of Anatomy, Yonsei University College of Medicine, Seoul, Korea
| | - Bae Hwan Lee
- Brain Korea 21 PLUS Project for Medical Science, Yonsei University, Seoul, Korea.,Brain Research Institute, Yonsei University College of Medicine, Seoul, Korea.,Department of Physiology, Yonsei University College of Medicine, Seoul, Korea
| | - Hyongbum Kim
- Department of Pharmacology, Yonsei University College of Medicine, Seoul, Korea
| | - Sung-Rae Cho
- Department and Research Institute of Rehabilitation Medicine, Yonsei University College of Medicine, Seoul, Korea .,Yonsei Stem Cell Center, Avison Biomedical Research Center, Seoul, Korea.,Brain Korea 21 PLUS Project for Medical Science, Yonsei University, Seoul, Korea.,Rehabilitation Institute of Neuromuscular Disease, Yonsei University College of Medicine, Seoul, Korea
| |
Collapse
|
32
|
Sulzer D, Cragg SJ, Rice ME. Striatal dopamine neurotransmission: regulation of release and uptake. ACTA ACUST UNITED AC 2016; 6:123-148. [PMID: 27141430 DOI: 10.1016/j.baga.2016.02.001] [Citation(s) in RCA: 262] [Impact Index Per Article: 29.1] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/18/2022]
Abstract
Dopamine (DA) transmission is governed by processes that regulate release from axonal boutons in the forebrain and the somatodendritic compartment in midbrain, and by clearance by the DA transporter, diffusion, and extracellular metabolism. We review how axonal DA release is regulated by neuronal activity and by autoreceptors and heteroreceptors, and address how quantal release events are regulated in size and frequency. In brain regions densely innervated by DA axons, DA clearance is due predominantly to uptake by the DA transporter, whereas in cortex, midbrain, and other regions with relatively sparse DA inputs, the norepinephrine transporter and diffusion are involved. We discuss the role of DA uptake in restricting the sphere of influence of DA and in temporal accumulation of extracellular DA levels upon successive action potentials. The tonic discharge activity of DA neurons may be translated into a tonic extracellular DA level, whereas their bursting activity can generate discrete extracellular DA transients.
Collapse
Affiliation(s)
- David Sulzer
- Depts of Psychiatry, Neurology, & Pharmacology, NY State Psychiatric Institute, Columbia University, New York, NY, USA
| | - Stephanie J Cragg
- Dept Physiology, Anatomy and Genetics, University of Oxford, Oxford, UK
| | - Margaret E Rice
- Depts of Neurosurgery & Neuroscience and Physiology, New York University School of Medicine, New York, NY, USA
| |
Collapse
|
33
|
Harun R, Hare KM, Brough EM, Munoz MJ, Grassi CM, Torres GE, Grace AA, Wagner AK. Fast-scan cyclic voltammetry demonstrates that L-DOPA produces dose-dependent, regionally selective bimodal effects on striatal dopamine kinetics in vivo. J Neurochem 2016; 136:1270-1283. [PMID: 26611352 PMCID: PMC4884169 DOI: 10.1111/jnc.13444] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/16/2015] [Revised: 11/17/2015] [Accepted: 11/18/2015] [Indexed: 11/29/2022]
Abstract
Parkinson's disease (PD) is a debilitating condition that is caused by a relatively specific degeneration of dopaminergic (DAergic) neurons of the substantia nigra pars compacta. L-DOPA was introduced as a viable treatment option for PD over 40 years ago and still remains the most common and effective therapy for PD. Though the effects of L-DOPA to augment striatal DA production are well known, little is actually known about how L-DOPA alters the kinetics of DA neurotransmission that contribute to its beneficial and adverse effects. In this study, we examined the effects of L-DOPA administration (50 mg/kg carbidopa + 0, 100, and 250 mg/kg L-DOPA) on regional electrically stimulated DA response kinetics using fast-scan cyclic voltammetry in anesthetized rats. We demonstrate that L-DOPA enhances DA release in both the dorsal striatum (D-STR) and nucleus accumbens (NAc), but surprisingly causes a delayed inhibition of release in the D-STR. In both regions, L-DOPA progressively attenuated reuptake kinetics, predominantly through a decrease in Vmax . These findings have important implications on understanding the pharmacodynamics of L-DOPA, which may be informative for understanding its therapeutic effects and also common side effects like L-DOPA-induced dyskinesias (LID). L-DOPA is commonly used to treat Parkinsonian symptoms, but little is known about how it affects presynaptic DA neurotransmission. Using in vivo fast-scan cyclic voltammetry, we show L-DOPA inhibits DA reuptake in a region-specific and dose-dependent manner, and L-DOPA has paradoxical effects on release. These findings may be important when considering mechanisms for L-DOPA's therapeutic benefits and adverse side-effects.
Collapse
Affiliation(s)
- Rashed Harun
- Center for Neuroscience, University of Pittsburgh, Pittsburgh, Pennsylvania, USA
- Department of Physical Medicine & Rehabilitation, University of Pittsburgh, School of Medicine, Pittsburgh, Pennsylvania, USA
- Safar Center for Resuscitation Research, Pittsburgh, Pennsylvania, USA
| | - Kristin M Hare
- Department of Physical Medicine & Rehabilitation, University of Pittsburgh, School of Medicine, Pittsburgh, Pennsylvania, USA
| | - Elizabeth M Brough
- Department of Physical Medicine & Rehabilitation, University of Pittsburgh, School of Medicine, Pittsburgh, Pennsylvania, USA
- Safar Center for Resuscitation Research, Pittsburgh, Pennsylvania, USA
| | - Miranda J Munoz
- Department of Physical Medicine & Rehabilitation, University of Pittsburgh, School of Medicine, Pittsburgh, Pennsylvania, USA
- Department of Biological Sciences, Carnegie Mellon University, Mellon College of Science, Pittsburgh, Pennsylvania, USA
| | - Christine M Grassi
- Department of Physical Medicine & Rehabilitation, University of Pittsburgh, School of Medicine, Pittsburgh, Pennsylvania, USA
| | - Gonzalo E Torres
- Center for Neuroscience, University of Pittsburgh, Pittsburgh, Pennsylvania, USA
- Department of Neurobiology, University of Pittsburgh, School of Medicine, Pittsburgh, Pennsylvania, USA
| | - Anthony A Grace
- Department of Neuroscience, University of Pittsburgh, Pittsburgh, Pennsylvania, USA
- Departments of Psychiatry and Psychology, University of Pittsburgh, Pittsburgh, Pennsylvania, USA
| | - Amy K Wagner
- Center for Neuroscience, University of Pittsburgh, Pittsburgh, Pennsylvania, USA
- Department of Physical Medicine & Rehabilitation, University of Pittsburgh, School of Medicine, Pittsburgh, Pennsylvania, USA
- Safar Center for Resuscitation Research, Pittsburgh, Pennsylvania, USA
- Department of Neuroscience, University of Pittsburgh, Pittsburgh, Pennsylvania, USA
| |
Collapse
|
34
|
Membrane potential shapes regulation of dopamine transporter trafficking at the plasma membrane. Nat Commun 2016; 7:10423. [PMID: 26804245 PMCID: PMC4737753 DOI: 10.1038/ncomms10423] [Citation(s) in RCA: 51] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/18/2015] [Accepted: 12/09/2015] [Indexed: 12/17/2022] Open
Abstract
The dopaminergic system is essential for cognitive processes, including reward, attention and motor control. In addition to DA release and availability of synaptic DA receptors, timing and magnitude of DA neurotransmission depend on extracellular DA-level regulation by the dopamine transporter (DAT), the membrane expression and trafficking of which are highly dynamic. Data presented here from real-time TIRF (TIRFM) and confocal microscopy coupled with surface biotinylation and electrophysiology suggest that changes in the membrane potential alone, a universal yet dynamic cellular property, rapidly alter trafficking of DAT to and from the surface membrane. Broadly, these findings suggest that cell-surface DAT levels are sensitive to membrane potential changes, which can rapidly drive DAT internalization from and insertion into the cell membrane, thus having an impact on the capacity for DAT to regulate extracellular DA levels. The dopaminergic system has important roles in a number of cognitive process. Here, the authors use detailed analysis of dopamine transporter trafficking to show its levels at the cell surface are sensitive to changes in membrane potential.
Collapse
|
35
|
Kang BM, Mun CW, Chun SI, Kim TH, Son DB, Kim HD. Noninvasive and repetitive measurement of cellular metabolite from human osteosarcoma cells (MG-63) using 3.0 tesla proton ( 1 H) MR spectroscopy. Magn Reson Med 2016; 76:1912-1918. [PMID: 26762686 DOI: 10.1002/mrm.26075] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/30/2015] [Revised: 10/18/2015] [Accepted: 11/06/2015] [Indexed: 01/14/2023]
Abstract
PURPOSE This study suggests a noninvasive and repetitive measurement method using 1 H magnetic resonance spectroscopy to monitor changes in cellular metabolites within a single sample. METHODS Longitudinal acquisition of cellular metabolites from three-dimensional cultured human osteosarcoma (MG-63) cells was conducted using 3.0 Tesla 1 H MRS for 2 weeks at three time points: days 1, 7, and 14. During the MR spectroscopy (MRS) scan, cell specimen temperatures were kept constant at 37°C by a lab-developed magnetic resonance compatible thermostatic device. A DNA assay and live/dead staining of the cell specimens were carried out at each time point to verify the MRS measurements. RESULTS Cell viability in the proposed device did not significantly differ from that of cells in a conventional incubator (P = 0.946). Cell proliferation and choline concentration increased during the first week, but remained constant during the second week. Lactate did not change during the first week, but increased during the second week. Likewise, cell viability remained constant until day 7, then decreased. CONCLUSION The proposed MRS technique results in a survivable environment for longitudinal studies of cells and provides a new way to measure metabolomic changes over time in single specimens of cells. Magn Reson Med 76:1912-1918, 2016. © 2016 International Society for Magnetic Resonance in Medicine.
Collapse
Affiliation(s)
- Bok-Man Kang
- Department of Biomedical Engineering/u-HARC, Inje University, Gyeongnam, Republic of Korea
| | - Chi-Woong Mun
- Department of Biomedical Engineering/u-HARC, Inje University, Gyeongnam, Republic of Korea.,Department of Health Science and Technology, Inje University, Gyeongnam, Republic of Korea
| | - Song-I Chun
- Department of Biomedical Engineering/u-HARC, Inje University, Gyeongnam, Republic of Korea
| | - Tae-Hyung Kim
- Department of Health Science and Technology, Inje University, Gyeongnam, Republic of Korea
| | - Doo-Beum Son
- Department of Radiolgoy, Haeundae Paik Hospital, Inje University, Haeundae-gu, Busan, Republic of Korea
| | - Hong-Dae Kim
- Department of Radiolgoy, Haeundae Paik Hospital, Inje University, Haeundae-gu, Busan, Republic of Korea
| |
Collapse
|
36
|
Abstract
UNLABELLED The dopamine (DA) transporter (DAT) controls dopaminergic neurotransmission by removing extracellular DA. Although DA reuptake is proposed to be regulated by DAT traffic to and from the cell surface, the membrane trafficking system involved in the endocytic cycling of DAT in the intact mammalian brain has not been characterized. Hence, we performed immunolabeling and quantitative analysis of the subcellular and regional distribution of DAT using the transgenic knock-in mouse expressing hemagglutinin (HA) epitope-tagged DAT (HA-DAT) and by using a combination of electron microscopy and a novel method for immunofluorescence labeling of HA-DAT in acute sagittal brain slices. Both approaches demonstrated that, in midbrain somatodendritic regions, HA-DAT was present in the plasma membrane, endoplasmic reticulum, and Golgi complex, with a small fraction in early and recycling endosomes and an even smaller fraction in late endosomes and lysosomes. In the striatum and in axonal tracts between the midbrain and striatum, HA-DAT was detected predominantly in the plasma membrane, and quantitative analysis revealed increased DAT density in striatal compared with midbrain plasma membranes. Endosomes were strikingly rare and lysosomes were absent in striatal axons, in which there was little intracellular HA-DAT. Acute administration of amphetamine in vivo (60 min) or to slices ex vivo (10-60 min) did not result in detectable changes in DAT distribution. Altogether, these data provide evidence for regional differences in DAT plasma membrane targeting and retention and suggest a surprisingly low level of endocytic trafficking of DAT in the striatum along with limited DAT endocytic activity in somatodendritic areas. SIGNIFICANCE STATEMENT The dopamine transporter (DAT) is the key regulator of the dopamine neurotransmission in the CNS. In the present study, we developed a new approach for studying DAT localization and dynamics in intact neurons in acute sagittal brain slices from the knock-in mouse expressing epitope-tagged DAT. For the first time, the fluorescence imaging analysis of DAT was combined with the immunogold labeling of DAT and quantitative electron microscopy. In contrast to numerous studies of DAT trafficking in heterologous expression systems and dissociated cultured neurons, studies in intact neurons revealed a surprisingly low amount of endocytic trafficking of DAT at steady state and after acute amphetamine treatment and suggested that non-vesicular transport could be the main mechanism establishing DAT distribution within the dopaminergic neuron.
Collapse
|
37
|
German CL, Baladi MG, McFadden LM, Hanson GR, Fleckenstein AE. Regulation of the Dopamine and Vesicular Monoamine Transporters: Pharmacological Targets and Implications for Disease. Pharmacol Rev 2015; 67:1005-24. [PMID: 26408528 PMCID: PMC4630566 DOI: 10.1124/pr.114.010397] [Citation(s) in RCA: 129] [Impact Index Per Article: 12.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022] Open
Abstract
Dopamine (DA) plays a well recognized role in a variety of physiologic functions such as movement, cognition, mood, and reward. Consequently, many human disorders are due, in part, to dysfunctional dopaminergic systems, including Parkinson's disease, attention deficit hyperactivity disorder, and substance abuse. Drugs that modify the DA system are clinically effective in treating symptoms of these diseases or are involved in their manifestation, implicating DA in their etiology. DA signaling and distribution are primarily modulated by the DA transporter (DAT) and by vesicular monoamine transporter (VMAT)-2, which transport DA into presynaptic terminals and synaptic vesicles, respectively. These transporters are regulated by complex processes such as phosphorylation, protein-protein interactions, and changes in intracellular localization. This review provides an overview of 1) the current understanding of DAT and VMAT2 neurobiology, including discussion of studies ranging from those conducted in vitro to those involving human subjects; 2) the role of these transporters in disease and how these transporters are affected by disease; and 3) and how selected drugs alter the function and expression of these transporters. Understanding the regulatory processes and the pathologic consequences of DAT and VMAT2 dysfunction underlies the evolution of therapeutic development for the treatment of DA-related disorders.
Collapse
Affiliation(s)
- Christopher L German
- School of Dentistry (C.L.G., M.G.B., G.R.H., A.E.F.) and Department of Pharmacology and Toxicology (L.M.M., G.R.H.), University of Utah, Salt Lake City, Utah
| | - Michelle G Baladi
- School of Dentistry (C.L.G., M.G.B., G.R.H., A.E.F.) and Department of Pharmacology and Toxicology (L.M.M., G.R.H.), University of Utah, Salt Lake City, Utah
| | - Lisa M McFadden
- School of Dentistry (C.L.G., M.G.B., G.R.H., A.E.F.) and Department of Pharmacology and Toxicology (L.M.M., G.R.H.), University of Utah, Salt Lake City, Utah
| | - Glen R Hanson
- School of Dentistry (C.L.G., M.G.B., G.R.H., A.E.F.) and Department of Pharmacology and Toxicology (L.M.M., G.R.H.), University of Utah, Salt Lake City, Utah
| | - Annette E Fleckenstein
- School of Dentistry (C.L.G., M.G.B., G.R.H., A.E.F.) and Department of Pharmacology and Toxicology (L.M.M., G.R.H.), University of Utah, Salt Lake City, Utah
| |
Collapse
|
38
|
Luderman KD, Chen R, Ferris MJ, Jones SR, Gnegy ME. Protein kinase C beta regulates the D₂-like dopamine autoreceptor. Neuropharmacology 2015; 89:335-41. [PMID: 25446677 DOI: 10.1016/j.neuropharm.2014.10.012] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/21/2014] [Revised: 10/03/2014] [Accepted: 10/07/2014] [Indexed: 01/15/2023]
Abstract
The focus of this study was the regulation of the D2-like dopamine autoreceptor (D2 autoreceptor) by protein kinase Cβ, a member of the protein kinase C (PKC) family. Together with the dopamine transporter, the D2 autoreceptor regulates the level of extracellular dopamine and thus dopaminergic signaling. PKC regulates neuronal signaling via several mechanisms, including desensitizing autoreceptors to increase the release of several different neurotransmitters. Here, using both PKCβ(-/-) mice and specific PKCβ inhibitors, we demonstrated that a lack of PKCβ activity enhanced the D2 autoreceptor-stimulated decrease in dopamine release following both chemical and electrical stimulations. Inhibition of PKCβ increased surface localization of D2R in mouse striatal synaptosomes, which could underlie the greater sensitivity to quinpirole following inhibition of PKCβ. PKCβ(-/-) mice displayed greater sensitivity to the quinpirole-induced suppression of locomotor activity, demonstrating that the regulation of the D2 autoreceptor by PKCβ is physiologically significant. Overall, we have found that PKCβ downregulates the D2 autoreceptor, providing an additional layer of regulation for dopaminergic signaling. We propose that in the absence of PKCβ activity, surface D2 autoreceptor localization and thus D2 autoreceptor signaling is increased, leading to less dopamine in the extracellular space and attenuated dopaminergic signaling.
Collapse
Affiliation(s)
- Kathryn D Luderman
- Department of Pharmacology, University of Michigan, 1150 West Medical Center Drive, Ann Arbor, MI, 48109-5632, USA.
| | | | | | | | | |
Collapse
|
39
|
Tang QY, Kolanos R, De Felice LJ, Glennon RA. Structural analysis of dopamine- and amphetamine-induced depolarization currents in the human dopamine transporter. ACS Chem Neurosci 2015; 6:551-8. [PMID: 25594379 DOI: 10.1021/cn500282f] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/03/2023] Open
Abstract
Amphetamine (AMPH) induces depolarizing currents through the human dopamine transporter (hDAT). Recently we discovered that the S(+) enantiomer of AMPH induces a current through hDAT that persists long after its removal from the external milieu. The persistent current is less prominent for R(-)AMPH and essentially absent for dopamine (DA)-induced currents. Related agents such as methamphetamine also exhibit persistent currents, which are present in both frog oocyte and mammalian HEK expression systems. Here, we study hDAT-expressing Xenopus laevis oocytes voltage-clamped and exposed from outside to DA, S(+)AMPH, R(-)AMPH, and related synthesized compounds, including stereoisomers. The goal of the study was to determine how structural transitioning from dopamine to amphetamine influences hDAT potency and action. At saturating concentrations, S(+)AMPH or R(-)AMPH induce a sharply rising depolarizing current from -60 mV that is comparable in amplitude to DA-induced currents. The magnitude and duration of the currents and the presence or absence of persistent currents depend on the concentration, duration of exposure, and chemical structure and enantiomeric versions of the agents.
Collapse
Affiliation(s)
- Qiong-Yao Tang
- Department
of Physiology and Biophysics, Virginia Commonwealth University School of Medicine, Richmond, Virginia 23298, United States
- Jiangsu
Province Key Laboratory of Anesthesiology, Xuzhou Medical College, Xuzhou 221004, People’s Republic of China
| | - Renata Kolanos
- Department
of Medicinal Chemistry, Virginia Commonwealth University School of Pharmacy, Richmond, Virginia 23298, United States
| | - Louis J De Felice
- Department
of Physiology and Biophysics, Virginia Commonwealth University School of Medicine, Richmond, Virginia 23298, United States
| | - Richard A Glennon
- Department
of Medicinal Chemistry, Virginia Commonwealth University School of Pharmacy, Richmond, Virginia 23298, United States
| |
Collapse
|
40
|
Kovtun O, Sakrikar D, Tomlinson ID, Chang JC, Arzeta-Ferrer X, Blakely RD, Rosenthal SJ. Single-quantum-dot tracking reveals altered membrane dynamics of an attention-deficit/hyperactivity-disorder-derived dopamine transporter coding variant. ACS Chem Neurosci 2015; 6:526-34. [PMID: 25747272 PMCID: PMC5530757 DOI: 10.1021/cn500202c] [Citation(s) in RCA: 29] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/14/2023] Open
Abstract
The presynaptic, cocaine- and amphetamine-sensitive dopamine (DA) transporter (DAT, SLC6A3) controls the intensity and duration of synaptic dopamine signals by rapid clearance of DA back into presynaptic nerve terminals. Abnormalities in DAT-mediated DA clearance have been linked to a variety of neuropsychiatric disorders, including addiction, autism, and attention deficit/hyperactivity disorder (ADHD). Membrane trafficking of DAT appears to be an important, albeit incompletely understood, post-translational regulatory mechanism; its dysregulation has been recently proposed as a potential risk determinant of these disorders. In this study, we demonstrate a link between an ADHD-associated DAT mutation (Arg615Cys, R615C) and variation on DAT transporter cell surface dynamics, a combination only previously studied with ensemble biochemical and optical approaches that featured limited spatiotemporal resolution. Here, we utilize high-affinity, DAT-specific antagonist-conjugated quantum dot (QD) probes to establish the dynamic mobility of wild-type and mutant DATs at the plasma membrane of living cells. Single DAT-QD complex trajectory analysis revealed that the DAT 615C variant exhibited increased membrane mobility relative to DAT 615R, with diffusion rates comparable to those observed after lipid raft disruption. This phenomenon was accompanied by a loss of transporter mobilization triggered by amphetamine, a common component of ADHD medications. Together, our data provides the first dynamic imaging of single DAT proteins, providing new insights into the relationship between surface dynamics and trafficking of both wild-type and disease-associated transporters. Our approach should be generalizable to future studies that explore the possibilities of perturbed surface DAT dynamics that may arise as a consequence of genetic alterations, regulatory changes, and drug use that contribute to the etiology or treatment of neuropsychiatric disorders.
Collapse
Affiliation(s)
- Oleg Kovtun
- Department of Chemistry, Vanderbilt University, Nashville, Tennessee 37235, United States
- McCoy & McCoy Laboratories, Inc, Madisonville, Kentucky 42431, United States
| | - Dhananjay Sakrikar
- Department of Pharmacology, Vanderbilt University, Nashville, Tennessee 37235, United States
| | - Ian D. Tomlinson
- Department of Chemistry, Vanderbilt University, Nashville, Tennessee 37235, United States
| | - Jerry C. Chang
- Laboratory of Molecular and Cellular Neuroscience, The Rockefeller University, New York, New York 10065, United States
| | - Xochitl Arzeta-Ferrer
- Department of Chemistry, Vanderbilt University, Nashville, Tennessee 37235, United States
| | - Randy D. Blakely
- Department of Pharmacology, Vanderbilt University, Nashville, Tennessee 37235, United States
- Department of Psychiatry, Vanderbilt University, Nashville, Tennessee 37235, United States
- Silvio O. Conte Center for Neuroscience Research, Vanderbilt University, Nashville, Tennessee 37235, United States
| | - Sandra J. Rosenthal
- Department of Chemistry, Vanderbilt University, Nashville, Tennessee 37235, United States
- Department of Pharmacology, Vanderbilt University, Nashville, Tennessee 37235, United States
- Department of Chemical and Biomolecular Engineering, Vanderbilt University, Nashville, Tennessee 37235, United States
- Department of Physics and Astronomy, Vanderbilt University, Nashville, Tennessee 37235, United States
- Vanderbilt Institute of Nanoscale Science and Engineering, Vanderbilt University, Nashville, Tennessee 37235, United States
- Materials Science and Technology Division, Oak Ridge National Laboratory, Oak Ridge, Tennessee 37831, United States
| |
Collapse
|
41
|
Siciliano CA, Calipari ES, Ferris MJ, Jones SR. Adaptations of presynaptic dopamine terminals induced by psychostimulant self-administration. ACS Chem Neurosci 2015; 6:27-36. [PMID: 25491345 PMCID: PMC4304501 DOI: 10.1021/cn5002705] [Citation(s) in RCA: 42] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/24/2014] [Revised: 12/04/2014] [Indexed: 12/27/2022] Open
Abstract
A great deal of research has focused on investigating neurobiological alterations induced by chronic psychostimulant use in an effort to describe, understand, and treat the pathology of psychostimulant addiction. It has been known for several decades that dopamine neurotransmission in the nucleus accumbens is integrally involved in the selection and execution of motivated and goal-directed behaviors, and that psychostimulants act on this system to exert many of their effects. As such, a large body of work has focused on defining the consequences of psychostimulant use on dopamine signaling in the striatum as it relates to addictive behaviors. Here, we review presynaptic dopamine terminal alterations observed following self-administration of cocaine and amphetamine, as well as possible mechanisms by which these alterations occur and their impact on the progression of addiction.
Collapse
Affiliation(s)
- Cody A. Siciliano
- Department
of Physiology and Pharmacology, Wake Forest
School of Medicine, Winston-Salem, North Carolina 27157, United States
| | - Erin S. Calipari
- Fishberg
Department of Neuroscience, Icahn School
of Medicine at Mount Sinai, New
York, New York 10029, United States
| | - Mark J. Ferris
- Department
of Physiology and Pharmacology, Wake Forest
School of Medicine, Winston-Salem, North Carolina 27157, United States
| | - Sara R. Jones
- Department
of Physiology and Pharmacology, Wake Forest
School of Medicine, Winston-Salem, North Carolina 27157, United States
| |
Collapse
|
42
|
Prince CD, Rau AR, Yorgason JT, España RA. Hypocretin/Orexin regulation of dopamine signaling and cocaine self-administration is mediated predominantly by hypocretin receptor 1. ACS Chem Neurosci 2015; 6:138-46. [PMID: 25496218 PMCID: PMC4304483 DOI: 10.1021/cn500246j] [Citation(s) in RCA: 66] [Impact Index Per Article: 6.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/11/2023] Open
Abstract
![]()
Extensive
evidence suggests that the hypocretins/orexins influence
cocaine reinforcement and dopamine signaling via actions at hypocretin
receptor 1. By comparison, the involvement of hypocretin receptor
2 in reward and reinforcement processes has received relatively little
attention. Thus, although there is some evidence that hypocretin receptor
2 regulates intake of some drugs of abuse, it is currently unclear
to what extent hypocretin receptor 2 participates in the regulation
of dopamine signaling or cocaine self-administration, particularly
under high effort conditions. To address this, we examined the effects
of hypocretin receptor 1, and/or hypocretin receptor 2 blockade on
dopamine signaling and cocaine reinforcement. We used in vivo fast
scan cyclic voltammetry to test the effects of hypocretin antagonists
on dopamine signaling in the nucleus accumbens core and a progressive
ratio schedule to examine the effects of these antagonists on cocaine
self-administration. Results demonstrate that blockade of either hypocretin
receptor 1 or both hypocretin receptor 1 and 2 significantly reduces
the effects of cocaine on dopamine signaling and decreases the motivation
to take cocaine. In contrast, blockade of hypocretin receptor 2 alone
had no significant effects on dopamine signaling or self-administration.
These findings suggest a differential involvement of the two hypocretin
receptors, with hypocretin receptor 1 appearing to be more involved
than hypocretin receptor 2 in the regulation of dopamine signaling
and cocaine self-administration. When considered with the existing
literature, these data support the hypothesis that hypocretins exert
a permissive influence on dopamine signaling and motivated behavior
via preferential actions on hypocretin receptor 1.
Collapse
Affiliation(s)
- Courtney D. Prince
- Department
of Neurobiology and Anatomy, Drexel University College of Medicine, Philadelphia, Pennsylvania 19129, United States
| | - Andrew R. Rau
- Department
of Physiology and Pharmacology, Wake Forest School of Medicine, Winston-Salem, North Carolina 27157, United States
| | - Jordan T. Yorgason
- Vollum
Institute, Oregon Health and Science University, Portland, Oregon 97239, United States
| | - Rodrigo A. España
- Department
of Neurobiology and Anatomy, Drexel University College of Medicine, Philadelphia, Pennsylvania 19129, United States
| |
Collapse
|
43
|
Neurobiological model of stimulated dopamine neurotransmission to interpret fast-scan cyclic voltammetry data. Brain Res 2014; 1599:67-84. [PMID: 25527399 DOI: 10.1016/j.brainres.2014.12.020] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/15/2014] [Revised: 12/05/2014] [Accepted: 12/06/2014] [Indexed: 11/22/2022]
Abstract
Fast-scan cyclic voltammetry (FSCV) is an electrochemical method that can assess real-time in vivo dopamine (DA) concentration changes to study the kinetics of DA neurotransmission. Electrical stimulation of dopaminergic (DAergic) pathways can elicit FSCV DA responses that largely reflect a balance of DA release and reuptake. Interpretation of these evoked DA responses requires a framework to discern the contribution of DA release and reuptake. The current, widely implemented interpretive framework for doing so is the Michaelis-Menten (M-M) model, which is grounded on two assumptions- (1) DA release rate is constant during stimulation, and (2) DA reuptake occurs through dopamine transporters (DAT) in a manner consistent with M-M enzyme kinetics. Though the M-M model can simulate evoked DA responses that rise convexly, response types that predominate in the ventral striatum, the M-M model cannot simulate dorsal striatal responses that rise concavely. Based on current neurotransmission principles and experimental FSCV data, we developed a novel, quantitative, neurobiological framework to interpret DA responses that assumes DA release decreases exponentially during stimulation and continues post-stimulation at a diminishing rate. Our model also incorporates dynamic M-M kinetics to describe DA reuptake as a process of decreasing reuptake efficiency. We demonstrate that this quantitative, neurobiological model is an extension of the traditional M-M model that can simulate heterogeneous regional DA responses following manipulation of stimulation duration, frequency, and DA pharmacology. The proposed model can advance our interpretive framework for future in vivo FSCV studies examining regional DA kinetics and their alteration by disease and DA pharmacology.
Collapse
|
44
|
Simonson B, Morani AS, Ewald AWM, Walker L, Kumar N, Simpson D, Miller JH, Prisinzano TE, Kivell BM. Pharmacology and anti-addiction effects of the novel κ opioid receptor agonist Mesyl Sal B, a potent and long-acting analogue of salvinorin A. Br J Pharmacol 2014; 172:515-31. [PMID: 24641310 DOI: 10.1111/bph.12692] [Citation(s) in RCA: 43] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/20/2013] [Revised: 03/09/2014] [Accepted: 03/11/2014] [Indexed: 12/15/2022] Open
Abstract
BACKGROUND AND PURPOSE Acute activation of κ opioid (KOP) receptors results in anticocaine-like effects, but adverse effects, such as dysphoria, aversion, sedation and depression, limit their clinical development. Salvinorin A, isolated from the plant Salvia divinorum, and its semi-synthetic analogues have been shown to have potent KOP receptor agonist activity and may induce a unique response with similar anticocaine addiction effects as the classic KOP receptor agonists, but with a different side effect profile. EXPERIMENTAL APPROACH We evaluated the duration of effects of Mesyl Sal B in vivo utilizing antinociception assays and screened for cocaine-prime induced cocaine-seeking behaviour in self-administering rats to predict anti-addiction effects. Cellular transporter uptake assays and in vitro voltammetry were used to assess modulation of dopamine transporter (DAT) function and to investigate transporter trafficking and kinase signalling pathways modulated by KOP receptor agonists. KEY RESULTS Mesyl Sal B had a longer duration of action than SalA, had anti-addiction properties and increased DAT function in vitro in a KOP receptor-dependent and Pertussis toxin-sensitive manner. These effects on DAT function required ERK1/2 activation. We identified differences between Mesyl Sal B and SalA, with Mesyl Sal B increasing the Vmax of dopamine uptake without altering cell-surface expression of DAT. CONCLUSIONS AND IMPLICATIONS SalA analogues, such as Mesyl Sal B, have potential for development as anticocaine agents. Further tests are warranted to elucidate the mechanisms by which the novel salvinorin-based neoclerodane diterpene KOP receptor ligands produce both anti-addiction and adverse side effects. LINKED ARTICLES This article is part of a themed section on Opioids: New Pathways to Functional Selectivity. To view the other articles in this section visit http://dx.doi.org/10.1111/bph.2015.172.issue-2.
Collapse
Affiliation(s)
- B Simonson
- School of Biological Science, Centre for Biodiscovery, Victoria University of Wellington, Wellington, New Zealand
| | | | | | | | | | | | | | | | | |
Collapse
|
45
|
Single and binge methamphetamine administrations have different effects on the levels of dopamine D2 autoreceptor and dopamine transporter in rat striatum. Int J Mol Sci 2014; 15:5884-906. [PMID: 24717411 PMCID: PMC4013602 DOI: 10.3390/ijms15045884] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/02/2013] [Revised: 03/15/2014] [Accepted: 03/25/2014] [Indexed: 01/17/2023] Open
Abstract
Methamphetamine (METH) is a central nervous system psychostimulant with a high potential for abuse. At high doses, METH causes a selective degeneration of dopaminergic terminals in the striatum. Dopamine D2 receptor antagonists and dopamine transporter (DAT) inhibitors protect against neurotoxicity of the drug by decreasing intracellular dopamine content and, consequently, dopamine autoxidation and production of reactive oxygen species. In vitro, amphetamines regulate D2 receptor and DAT functions via regulation of their intracellular trafficking. No data exists on axonal transport of both proteins and there is limited data on their interactions in vivo. The aim of the present investigation was to examine synaptosomal levels of presynaptic D2 autoreceptor and DAT after two different regimens of METH and to determine whether METH affects the D2 autoreceptor-DAT interaction in the rat striatum. We found that, as compared to saline controls, administration of single high-dose METH decreased D2 autoreceptor immunoreactivity and increased DAT immunoreactivity in rat striatal synaptosomes whereas binge high-dose METH increased immunoreactivity of D2 autoreceptor and had no effect on DAT immunoreactivity. Single METH had no effect on D2 autoreceptor-DAT interaction whereas binge METH increased the interaction between the two proteins in the striatum. Our results suggest that METH can affect axonal transport of both the D2 autoreceptor and DAT in an interaction-dependent and -independent manner.
Collapse
|
46
|
Protein phosphatases but not reactive oxygen species play functional role in acute amphetamine-mediated dopamine release. Cell Biochem Biophys 2014; 66:831-41. [PMID: 23625176 DOI: 10.1007/s12013-013-9608-6] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/26/2022]
Abstract
Drug abuse-induced neurodegeneration can be triggered by elevated production of reactive oxygen species (ROS). Involvement of oxidative stress in acute amphetamine (AMPH)-mediated dopamine (DA) release, however, has not been completely understood yet. In order to elucidate the dopaminergic response of PC12 cells to a single dose of 10 μM AMPH, ROS production was measured as related to the extracellular DA level. Due to the spontaneous oxidation of peroxide-sensitive fluorophore 2',7'-dichlorofluorescin diacetate (DCFH-DA) to 2',7'-dichlorofluorescein (DCF), the increase in fluorescence could not be unambiguously attributed to AMPH-triggered ROS production. Based on Amplex Red fluorescence, no ROS production was detected after acute AMPH application. Our data strongly suggest that ROS development was not the main triggering factor for immediate DA release after acute AMPH treatment. On the other hand, AMPH-induced elevation of DA levels in rat brain striatal slices was quenched by the water soluble antioxidant, N-acetylcysteine (NAC) at 10 mM. In this study, we also investigated the contribution of protein phosphatases to the AMPH-induced rat brain striatal dopaminergic response. The experimental protocol, double AMPH challenge was applied for screening the effect of NAC and cantharidin on AMPH-mediated DA release. Here we show that AMPH-mediated DA release increased nearly twofold in striatal rat brain slices pretreated for 30 min with 1000 μM cantharidin, a selective PP1 and PP2A inhibitor. These findings prove the lack of ROS inhibitory action on protein phosphatase activity in acute AMPH-mediated DA efflux.
Collapse
|
47
|
Huang EYK, Tsui PF, Kuo TT, Tsai JJ, Chou YC, Ma HI, Chiang YH, Chen YH. Amantadine ameliorates dopamine-releasing deficits and behavioral deficits in rats after fluid percussion injury. PLoS One 2014; 9:e86354. [PMID: 24497943 PMCID: PMC3907421 DOI: 10.1371/journal.pone.0086354] [Citation(s) in RCA: 20] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/05/2013] [Accepted: 12/08/2013] [Indexed: 12/22/2022] Open
Abstract
Aims To investigate the role of dopamine in cognitive and motor learning skill deficits after a traumatic brain injury (TBI), we investigated dopamine release and behavioral changes at a series of time points after fluid percussion injury, and explored the potential of amantadine hydrochloride as a chronic treatment to provide behavioral recovery. Materials and Methods In this study, we sequentially investigated dopamine release at the striatum and behavioral changes at 1, 2, 4, 6, and 8 weeks after fluid percussion injury. Rats subjected to 6-Pa cerebral cortical fluid percussion injury were treated by using subcutaneous infusion pumps filled with either saline (sham group) or amantadine hydrochloride, with a releasing rate of 3.6mg/kg/hour for 8 weeks. The dopamine-releasing conditions and metabolism were analyzed sequentially by fast scan cyclic voltammetry (FSCV) and high-pressure liquid chromatography (HPLC). Novel object recognition (NOR) and fixed-speed rotarod (FSRR) behavioral tests were used to determine treatment effects on cognitive and motor deficits after injury. Results Sequential dopamine-release deficits were revealed in 6-Pa-fluid-percussion cerebral cortical injured animals. The reuptake rate (tau value) of dopamine in injured animals was prolonged, but the tau value became close to the value for the control group after amantadine therapy. Cognitive and motor learning impairments were shown evidenced by the NOR and FSRR behavioral tests after injury. Chronic amantadine therapy reversed dopamine-release deficits, and behavioral impairment after fluid percussion injuries were ameliorated in the rats treated by using amantadine-pumping infusion. Conclusion Chronic treatment with amantadine hydrochloride can ameliorate dopamine-release deficits as well as cognitive and motor deficits caused by cerebral fluid-percussion injury.
Collapse
Affiliation(s)
| | - Pi-Fen Tsui
- Department of Pharmacology, National Defense Medical Center, Taipei, Taiwan
| | - Tung-Tai Kuo
- Graduate Institute of Computer and Communication Engineering, National Taipei University of Technology, Taipei, Taiwan
| | - Jing-Jr. Tsai
- Department of Neurological Surgery, Tri-Service General Hospital, National Defense Medical Center, Taipei, Taiwan
| | - Yu-Ching Chou
- School of Public Health, National Defense Medical Center, Taipei, Taiwan
| | - Hsin-I Ma
- Department of Neurological Surgery, Tri-Service General Hospital, National Defense Medical Center, Taipei, Taiwan
| | - Yung-Hsiao Chiang
- Department of Neurosurgery, Taipei Medical University Hospital, the PhD Program for Neural Regenerative Medicine, Graduate Institute of Neural Regenerative Medicine, Taipei Medical University, Taipei, Taiwan
| | - Yuan-Hao Chen
- Department of Neurological Surgery, Tri-Service General Hospital, National Defense Medical Center, Taipei, Taiwan
- * E-mail:
| |
Collapse
|
48
|
Kivell BM, Ewald AWM, Prisinzano TE. Salvinorin A analogs and other κ-opioid receptor compounds as treatments for cocaine abuse. ADVANCES IN PHARMACOLOGY (SAN DIEGO, CALIF.) 2014; 69:481-511. [PMID: 24484985 DOI: 10.1016/b978-0-12-420118-7.00012-3] [Citation(s) in RCA: 39] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
Abstract
Acute activation of kappa-opioid receptors produces anti-addictive effects by regulating dopamine levels in the brain. Unfortunately, classic kappa-opioid agonists have undesired side effects such as sedation, aversion, and depression, which restrict their clinical use. Salvinorin A (Sal A), a novel kappa-opioid receptor agonist extracted from the plant Salvia divinorum, has been identified as a potential therapy for drug abuse and addiction. Here, we review the preclinical effects of Sal A in comparison with traditional kappa-opioid agonists and several new analogs. Sal A retains the anti-addictive properties of traditional kappa-opioid receptor agonists with several improvements including reduced side effects. However, the rapid metabolism of Sal A makes it undesirable for clinical development. In an effort to improve the pharmacokinetics and tolerability of this compound, kappa-opioid receptor agonists based on the structure of Sal A have been synthesized. While work in this field is still in progress, several analogs with improved pharmacokinetic profiles have been shown to have anti-addictive effects. While in its infancy, it is clear that these compounds hold promise for the future development of anti-addictive therapeutics.
Collapse
Affiliation(s)
- Bronwyn M Kivell
- School of Biological Sciences, Centre for Biodiscovery, Victoria University of Wellington, Wellington, New Zealand
| | - Amy W M Ewald
- School of Biological Sciences, Centre for Biodiscovery, Victoria University of Wellington, Wellington, New Zealand
| | - Thomas E Prisinzano
- Department of Medicinal Chemistry, University of Kansas, Lawrence, Kansas, USA.
| |
Collapse
|
49
|
Monoamine transporter inhibitors and substrates as treatments for stimulant abuse. ADVANCES IN PHARMACOLOGY (SAN DIEGO, CALIF.) 2014; 69:129-76. [PMID: 24484977 DOI: 10.1016/b978-0-12-420118-7.00004-4] [Citation(s) in RCA: 46] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/06/2023]
Abstract
The acute and chronic effects of abused psychostimulants on monoamine transporters and associated neurobiology have encouraged development of candidate medications that target these transporters. Monoamine transporters, in general, and dopamine transporters, in particular, are critical molecular targets that mediate abuse-related effects of psychostimulants such as cocaine and amphetamine. Moreover, chronic administration of psychostimulants can cause enduring changes in neurobiology reflected in dysregulation of monoamine neurochemistry and behavior. The current review will evaluate evidence for the efficacy of monoamine transporter inhibitors and substrates to reduce abuse-related effects of stimulants in preclinical assays of stimulant self-administration, drug discrimination, and reinstatement. In considering deployment of monoamine transport inhibitors and substrates as agonist-type medications to treat stimulant abuse, the safety and abuse liability of the medications are an obvious concern, and this will also be addressed. Future directions in drug discovery should identify novel medications that retain efficacy to decrease stimulant use but possess lower abuse liability and evaluate the degree to which efficacious medications can attenuate or reverse neurobiological effects of chronic stimulant use.
Collapse
|
50
|
Oaks AW, Sidhu A. Parallel mechanisms for direct and indirect membrane protein trafficking by synucleins. Commun Integr Biol 2013; 6:e26794. [PMID: 24563712 PMCID: PMC3917945 DOI: 10.4161/cib.26794] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/10/2013] [Accepted: 10/11/2013] [Indexed: 12/11/2022] Open
Abstract
More than 2 decades of work have yet to conclusively determine the physiological role of the synuclein proteins, even though these abundant brain constituents are participants in a broad array of cellular processes. Among proposed physiological roles is a functional interaction between the synuclein proteins and monoamine transporters contributing to transporter trafficking through direct protein–protein interactions. Recent work shows that an antagonistic effect of the synuclein proteins on the secretory functions of the endoplasmic reticulum and the Golgi apparatus appears to simultaneously influence trafficking of the dopamine transporter and other membrane proteins. Here, we highlight these new findings in view of the broader literature identifying the role of synucleins in protein trafficking and suggest emerging themes for ongoing and future work in the field of synuclein biology.
Collapse
Affiliation(s)
- Adam W Oaks
- Laboratory of Molecular Neurochemistry; Department of Biochemistry and Molecular & Cellular Biology; Georgetown University Medical Center; Washington, DC USA
| | - Anita Sidhu
- Laboratory of Molecular Neurochemistry; Department of Biochemistry and Molecular & Cellular Biology; Georgetown University Medical Center; Washington, DC USA
| |
Collapse
|