1
|
Camacho-Hernandez GA, Jahan K, Newman AH. Illuminating the monoamine transporters: Fluorescently labelled ligands to study dopamine, serotonin and norepinephrine transporters. Basic Clin Pharmacol Toxicol 2023; 133:473-484. [PMID: 36527444 PMCID: PMC11309735 DOI: 10.1111/bcpt.13827] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/11/2022] [Revised: 12/14/2022] [Accepted: 12/15/2022] [Indexed: 12/23/2022]
Abstract
Fluorescence microscopy has revolutionized the visualization of physiological processes in live-cell systems. With the recent innovations in super resolution microscopy, these events can be examined with high precision and accuracy. The development of fluorescently labelled small molecules has provided a significant advance in understanding the physiological relevance of targeted proteins that can now be visualized at the cellular level. One set of physiologically important target proteins are the monoamine transporters (MATs) that play an instrumental role in maintaining monoamine signalling homeostasis. Understanding the mechanisms underlying their regulation and dysregulation is fundamental to treating several neuropsychiatric conditions such as attention deficit hyperactivity disorder (ADHD), anxiety, depression and substance use disorders. Herein, we describe the rationale behind the small molecule design of fluorescently labelled ligands (FLL) either as MAT substrates or inhibitors as well as their applications to advance our understanding of this class of transporters in health and disease.
Collapse
Affiliation(s)
- Gisela Andrea Camacho-Hernandez
- Medicinal Chemistry Section, Molecular Targets and Medications Discovery Branch, National Institutes on Drug Abuse - Intramural Research Program, Baltimore, Maryland, USA
| | - Khorshada Jahan
- Medicinal Chemistry Section, Molecular Targets and Medications Discovery Branch, National Institutes on Drug Abuse - Intramural Research Program, Baltimore, Maryland, USA
| | - Amy Hauck Newman
- Medicinal Chemistry Section, Molecular Targets and Medications Discovery Branch, National Institutes on Drug Abuse - Intramural Research Program, Baltimore, Maryland, USA
| |
Collapse
|
2
|
Small C, Cheng MH, Belay SS, Bulloch SL, Zimmerman B, Sorkin A, Block ER. The Alkylamine Stimulant 1,3-Dimethylamylamine Exhibits Substrate-Like Regulation of Dopamine Transporter Function and Localization. J Pharmacol Exp Ther 2023; 386:266-273. [PMID: 37348963 PMCID: PMC10353075 DOI: 10.1124/jpet.122.001573] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/30/2022] [Revised: 02/16/2023] [Accepted: 05/04/2023] [Indexed: 06/24/2023] Open
Abstract
The alkylamine stimulant 1,3-dimethylamylamine (DMAA) is used nonmedically as an appetite suppressant and exercise performance enhancer despite adverse cardiovascular effects that have limited its legal status. There is scant research describing the mechanism of action of DMAA, making it difficult to gauge risks or therapeutic potential. An important molecular target of structurally related phenethylamines, such as amphetamine, for regulating mood, cognition, movement, and the development of substance use disorder is the dopamine transporter, which limits the range and magnitude of dopamine signaling via reuptake from the extracellular space. The present studies were therefore initiated to characterize the effects of DMAA on dopamine transporter function. Specifically, we tested the hypothesis that DMAA exhibits substrate-like effects on dopamine transporter function and trafficking. In transport assays in human embryonic kidney cells, DMAA inhibited dopamine uptake by the human dopamine transporter in a competitive manner. Docking analysis and molecular dynamics simulations supported these findings, revealing that DMAA binds to the S1 substrate binding site and induces a conformational change from outward-facing open to outward-facing closed states, similar to the known substrates. Further supporting substrate-like effects of DMAA, the drug stimulated dopamine transporter endocytosis in a heterologous expression system via cocaine- and protein kinase A-sensitive mechanisms, mirroring findings with amphetamine. Together, these data indicate that DMAA elicits neurologic effects by binding to and regulating function of the dopamine transporter. Furthermore, pharmacologic distinctions from amphetamine reveal structural determinants for regulating transporter conformation and add mechanistic insight for the regulation of dopamine transporter endocytosis. SIGNIFICANCE STATEMENT: The alkylamine stimulant 1,3-dimethylamylamine (DMAA) is used as an appetite suppressant and athletic performance enhancer and is structurally similar to amphetamine, but there is scant research describing its mechanism of action. Characterizing the effects of DMAA on dopamine transporter function supports evaluation of potential risks and therapeutic potential while also revealing mechanistic details of dynamic transporter-substrate interactions.
Collapse
Affiliation(s)
- Cassandra Small
- Science Department, Chatham University, Pittsburgh, Pennsylvania (C.S., S.S.B., S.L.B., B.Z., E.R.B.) and Departments of Computational and Systems Biology (M.H.C.) and Cell Biology (A.S.), School of Medicine, University of Pittsburgh, Pittsburgh, Pennsylvania
| | - Mary Hongying Cheng
- Science Department, Chatham University, Pittsburgh, Pennsylvania (C.S., S.S.B., S.L.B., B.Z., E.R.B.) and Departments of Computational and Systems Biology (M.H.C.) and Cell Biology (A.S.), School of Medicine, University of Pittsburgh, Pittsburgh, Pennsylvania
| | - Saron S Belay
- Science Department, Chatham University, Pittsburgh, Pennsylvania (C.S., S.S.B., S.L.B., B.Z., E.R.B.) and Departments of Computational and Systems Biology (M.H.C.) and Cell Biology (A.S.), School of Medicine, University of Pittsburgh, Pittsburgh, Pennsylvania
| | - Sarah L Bulloch
- Science Department, Chatham University, Pittsburgh, Pennsylvania (C.S., S.S.B., S.L.B., B.Z., E.R.B.) and Departments of Computational and Systems Biology (M.H.C.) and Cell Biology (A.S.), School of Medicine, University of Pittsburgh, Pittsburgh, Pennsylvania
| | - Brooke Zimmerman
- Science Department, Chatham University, Pittsburgh, Pennsylvania (C.S., S.S.B., S.L.B., B.Z., E.R.B.) and Departments of Computational and Systems Biology (M.H.C.) and Cell Biology (A.S.), School of Medicine, University of Pittsburgh, Pittsburgh, Pennsylvania
| | - Alexander Sorkin
- Science Department, Chatham University, Pittsburgh, Pennsylvania (C.S., S.S.B., S.L.B., B.Z., E.R.B.) and Departments of Computational and Systems Biology (M.H.C.) and Cell Biology (A.S.), School of Medicine, University of Pittsburgh, Pittsburgh, Pennsylvania
| | - Ethan R Block
- Science Department, Chatham University, Pittsburgh, Pennsylvania (C.S., S.S.B., S.L.B., B.Z., E.R.B.) and Departments of Computational and Systems Biology (M.H.C.) and Cell Biology (A.S.), School of Medicine, University of Pittsburgh, Pittsburgh, Pennsylvania
| |
Collapse
|
3
|
Freissmuth M. Regulated Rapid Round Trips: Endocytotic Cycling of the Dopamine Transporter Shapes Motor Learning. J Biol Chem 2023; 299:104618. [PMID: 36935007 PMCID: PMC10124912 DOI: 10.1016/j.jbc.2023.104618] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 03/15/2023] [Indexed: 03/19/2023] Open
Abstract
The level of dopamine transporters (DATs) in the neuronal plasma membrane shapes learning and motor coordination in mice. Mechanisms underlying the regulated internalization of DAT and its return to the cell surface have been intensively studied in heterologous cells and in neuronal cell bodies. However, whether this cycling also happens in synaptic boutons, or axon terminals, thought to be the major functional site for DAT expression, was an open question that Kearney and colleagues recently addressed in the JBC. They showed that DAT cycling in the presynaptic specialization of dopaminergic neurons is subject to control by a cell-autonomous loop comprising dopamine autoreceptors and metabotropic glutamate receptors. These results should inform future studies in neural development and motor learning.
Collapse
Affiliation(s)
- Michael Freissmuth
- Institute of Pharmacology and the Gaston H. Glock Research Laboratories for Exploratory Drug Development, Center of Physiology and Pharmacology, Medical University of Vienna, Austria.
| |
Collapse
|
4
|
Kearney PJ, Bolden NC, Kahuno E, Conklin TL, Martin GE, Lubec G, Melikian HE. Presynaptic Gq-coupled receptors drive biphasic dopamine transporter trafficking that modulates dopamine clearance and motor function. J Biol Chem 2023; 299:102900. [PMID: 36640864 PMCID: PMC9943899 DOI: 10.1016/j.jbc.2023.102900] [Citation(s) in RCA: 8] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/09/2022] [Revised: 12/23/2022] [Accepted: 01/03/2023] [Indexed: 01/13/2023] Open
Abstract
Extracellular dopamine (DA) levels are constrained by the presynaptic DA transporter (DAT), a major psychostimulant target. Despite its necessity for DA neurotransmission, DAT regulation in situ is poorly understood, and it is unknown whether regulated DAT trafficking impacts dopaminergic signaling and/or behaviors. Leveraging chemogenetics and conditional gene silencing, we found that activating presynaptic Gq-coupled receptors, either hM3Dq or mGlu5, drove rapid biphasic DAT membrane trafficking in ex vivo striatal slices, with region-specific differences between ventral and dorsal striata. DAT insertion required D2 DA autoreceptors and intact retromer, whereas DAT retrieval required PKC activation and Rit2. Ex vivo voltammetric studies revealed that DAT trafficking impacts DA clearance. Furthermore, dopaminergic mGlu5 silencing elevated DAT surface expression and abolished motor learning, which was rescued by inhibiting DAT with a subthreshold CE-158 dose. We discovered that presynaptic DAT trafficking is complex, multimodal, and region specific, and for the first time, we identified cell autonomous mechanisms that govern presynaptic DAT tone. Importantly, the findings are consistent with a role for regulated DAT trafficking in DA clearance and motor function.
Collapse
Affiliation(s)
- Patrick J. Kearney
- Brudnick Neuropsychiatric Research Institute, Department of Neurobiology, UMASS Chan Medical School, Worcester, Massachusetts, USA,Morningside Graduate School of Biomedical Sciences, UMASS Chan Medical School, Worcester, Massachusetts, USA
| | - Nicholas C. Bolden
- Brudnick Neuropsychiatric Research Institute, Department of Neurobiology, UMASS Chan Medical School, Worcester, Massachusetts, USA,Morningside Graduate School of Biomedical Sciences, UMASS Chan Medical School, Worcester, Massachusetts, USA
| | - Elizabeth Kahuno
- Brudnick Neuropsychiatric Research Institute, Department of Neurobiology, UMASS Chan Medical School, Worcester, Massachusetts, USA
| | - Tucker L. Conklin
- Brudnick Neuropsychiatric Research Institute, Department of Neurobiology, UMASS Chan Medical School, Worcester, Massachusetts, USA
| | - Gilles E. Martin
- Brudnick Neuropsychiatric Research Institute, Department of Neurobiology, UMASS Chan Medical School, Worcester, Massachusetts, USA
| | - Gert Lubec
- Department of Neuroproteomics, Paracelsus Private Medical University, Salzburg, Austria
| | - Haley E. Melikian
- Brudnick Neuropsychiatric Research Institute, Department of Neurobiology, UMASS Chan Medical School, Worcester, Massachusetts, USA,For correspondence: Haley E. Melikian
| |
Collapse
|
5
|
Wu HF, Yu W, Saito-Diaz K, Huang CW, Carey J, Lefcort F, Hart GW, Liu HX, Zeltner N. Norepinephrine transporter defects lead to sympathetic hyperactivity in Familial Dysautonomia models. Nat Commun 2022; 13:7032. [PMID: 36396637 PMCID: PMC9671909 DOI: 10.1038/s41467-022-34811-7] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/03/2021] [Accepted: 11/08/2022] [Indexed: 11/19/2022] Open
Abstract
Familial dysautonomia (FD), a rare neurodevelopmental and neurodegenerative disorder affects the sympathetic and sensory nervous system. Although almost all patients harbor a mutation in ELP1, it remains unresolved exactly how function of sympathetic neurons (symNs) is affected; knowledge critical for understanding debilitating disease hallmarks, including cardiovascular instability or dysautonomic crises, that result from dysregulated sympathetic activity. Here, we employ the human pluripotent stem cell (hPSC) system to understand symN disease mechanisms and test candidate drugs. FD symNs are intrinsically hyperactive in vitro, in cardiomyocyte co-cultures, and in animal models. We report reduced norepinephrine transporter expression, decreased intracellular norepinephrine (NE), decreased NE re-uptake, and excessive extracellular NE in FD symNs. SymN hyperactivity is not a direct ELP1 mutation result, but may connect to NET via RAB proteins. We found that candidate drugs lowered hyperactivity independent of ELP1 modulation. Our findings may have implications for other symN disorders and may allow future drug testing and discovery.
Collapse
Affiliation(s)
- Hsueh-Fu Wu
- Center for Molecular Medicine, University of Georgia, Athens, GA, USA
- Department of Biochemistry and Molecular Biology, University of Georgia, Athens, GA, USA
| | - Wenxin Yu
- Regenerative Bioscience Center, Department of Animal and Dairy Science, College of Agricultural and Environmental Sciences, University of Georgia, Athens, GA, USA
| | - Kenyi Saito-Diaz
- Center for Molecular Medicine, University of Georgia, Athens, GA, USA
| | - Chia-Wei Huang
- Department of Biochemistry and Molecular Biology, University of Georgia, Athens, GA, USA
- Complex Carbohydrate Research Center, University of Georgia, Athens, GA, USA
| | - Joseph Carey
- Department of Microbiology and Cell Biology, Montana State University, Bozeman, MT, USA
| | - Frances Lefcort
- Department of Microbiology and Cell Biology, Montana State University, Bozeman, MT, USA
| | - Gerald W Hart
- Department of Biochemistry and Molecular Biology, University of Georgia, Athens, GA, USA
- Complex Carbohydrate Research Center, University of Georgia, Athens, GA, USA
| | - Hong-Xiang Liu
- Regenerative Bioscience Center, Department of Animal and Dairy Science, College of Agricultural and Environmental Sciences, University of Georgia, Athens, GA, USA
| | - Nadja Zeltner
- Center for Molecular Medicine, University of Georgia, Athens, GA, USA.
- Department of Biochemistry and Molecular Biology, University of Georgia, Athens, GA, USA.
- Department of Cellular Biology, University of Georgia, Athens, GA, USA.
| |
Collapse
|
6
|
Yin L, Zhou J, Li T, Wang X, Xue W, Zhang J, Lin L, Wang N, Kang X, Zhou Y, Liu H, Li Y. Inhibition of the dopamine transporter promotes lysosome biogenesis and ameliorates Alzheimer's disease-like symptoms in mice. Alzheimers Dement 2022; 19:1343-1357. [PMID: 36130073 DOI: 10.1002/alz.12776] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/15/2022] [Revised: 07/11/2022] [Accepted: 07/22/2022] [Indexed: 12/11/2022]
Abstract
INTRODUCTION Lysosomes are degradative organelles that maintain cellular homeostasis and protein quality control. Transcription factor EB (TFEB)-mediated lysosome biogenesis enhances lysosome-dependent degradation and alleviates neurodegenerative diseases, but the mechanisms underlying TFEB regulation and modification are still poorly understood. METHODS By screening novel small-molecule compounds, we identified a group of lysosome-enhancing compounds (LYECs) that promote TFEB activation and lysosome biogenesis. RESULTS One of these compounds, LH2-051, significantly inhibited the function of the dopamine transporter (DAT) and subsequently promoted lysosome biogenesis. We uncovered cyclin-dependent kinase 9 (CDK9) as a novel regulator of DAT-mediated lysosome biogenesis and identified six novel CDK9-phosphorylated sites on TFEB. We observed that signal transduction by the DAT-CDK9-TFEB axis occurs on lysosomes. Finally, we found that LH2-051 enhanced the degradation of amyloid beta plaques and improved the memory of amyloid precursor protein (APP)/Presenilin 1 (PS1) mice. DISCUSSION We identified the DAT-CDK9-TFEB signaling axis as a novel regulator of lysosome biogenesis. Our study sheds light on the mechanisms of protein quality control under pathophysiological conditions.
Collapse
Affiliation(s)
- Limin Yin
- Department of Pharmacology, State Key Laboratory of Medical Neurobiology and MOE Frontiers Center for Brain Science, Key Laboratory of Metabolism and Molecular Medicine, Ministry of Education, School of Basic Medical Science, Fudan University, Shanghai, China
| | - Jianhui Zhou
- State Key Laboratory of Drug Research, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai, China.,School of Chinese Materia Medica, Nanjing University of Chinese Medicine, Nanjing, China
| | - Tianyou Li
- Department of Pharmacology, State Key Laboratory of Medical Neurobiology and MOE Frontiers Center for Brain Science, Key Laboratory of Metabolism and Molecular Medicine, Ministry of Education, School of Basic Medical Science, Fudan University, Shanghai, China
| | - Xinghua Wang
- School of Chinese Materia Medica, Nanjing University of Chinese Medicine, Nanjing, China
| | - Wenlong Xue
- Department of Pharmacology, State Key Laboratory of Medical Neurobiology and MOE Frontiers Center for Brain Science, Key Laboratory of Metabolism and Molecular Medicine, Ministry of Education, School of Basic Medical Science, Fudan University, Shanghai, China
| | - Jie Zhang
- Department of Pharmacology, State Key Laboratory of Medical Neurobiology and MOE Frontiers Center for Brain Science, Key Laboratory of Metabolism and Molecular Medicine, Ministry of Education, School of Basic Medical Science, Fudan University, Shanghai, China
| | - Lingxi Lin
- Department of Pharmacology, State Key Laboratory of Medical Neurobiology and MOE Frontiers Center for Brain Science, Key Laboratory of Metabolism and Molecular Medicine, Ministry of Education, School of Basic Medical Science, Fudan University, Shanghai, China
| | - Ning Wang
- Department of Pharmacology, State Key Laboratory of Medical Neurobiology and MOE Frontiers Center for Brain Science, Key Laboratory of Metabolism and Molecular Medicine, Ministry of Education, School of Basic Medical Science, Fudan University, Shanghai, China
| | - Xinyi Kang
- Department of Pharmacology, State Key Laboratory of Medical Neurobiology and MOE Frontiers Center for Brain Science, Key Laboratory of Metabolism and Molecular Medicine, Ministry of Education, School of Basic Medical Science, Fudan University, Shanghai, China
| | - Yu Zhou
- State Key Laboratory of Drug Research, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai, China.,School of Chinese Materia Medica, Nanjing University of Chinese Medicine, Nanjing, China.,School of Pharmacy, University of Chinese Academy of Sciences, Beijing, China
| | - Hong Liu
- State Key Laboratory of Drug Research, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai, China.,School of Chinese Materia Medica, Nanjing University of Chinese Medicine, Nanjing, China.,School of Pharmacy, University of Chinese Academy of Sciences, Beijing, China
| | - Yang Li
- Department of Pharmacology, State Key Laboratory of Medical Neurobiology and MOE Frontiers Center for Brain Science, Key Laboratory of Metabolism and Molecular Medicine, Ministry of Education, School of Basic Medical Science, Fudan University, Shanghai, China
| |
Collapse
|
7
|
Szegö EM, Van den Haute C, Höfs L, Baekelandt V, Van der Perren A, Falkenburger BH. Rab7 reduces α-synuclein toxicity in rats and primary neurons. Exp Neurol 2021; 347:113900. [PMID: 34695425 DOI: 10.1016/j.expneurol.2021.113900] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/12/2021] [Revised: 09/19/2021] [Accepted: 10/18/2021] [Indexed: 11/04/2022]
Abstract
During the pathogenesis of Parkinson's disease (PD), aggregation of alpha-synuclein (αSyn) induces a vicious cycle of cellular impairments that lead to neurodegeneration. Consequently, removing toxic αSyn aggregates constitutes a plausible strategy against PD. In this work, we tested whether stimulating the autolysosomal degradation of αSyn aggregates through the Ras-related in brain 7 (Rab7) pathway can reverse αSyn-induced cellular impairment and prevent neurodegeneration in vivo. The disease-related A53T mutant of αSyn was expressed in primary neurons and in dopaminergic neurons of the rat brain simultaneously with wild type (WT) Rab7 or the T22N mutant as negative control. The cellular integrity was quantified by morphological and biochemical analyses. In primary neurons, WT Rab7 rescued the αSyn-induced loss of neurons and neurites. Furthermore, Rab7 decreased the amount of reactive oxygen species and the amount of Triton X-100 insoluble αSyn. In rat brain, WT Rab7 reduced αSyn-induced loss of dopaminergic axon terminals in the striatum and the loss of dopaminergic dendrites in the substantia nigra pars reticulata. Further, WT Rab7 lowered αSyn pathology as quantified by phosphorylated αSyn staining. Finally, WT Rab7 attenuated αSyn-induced DNA damage in primary neurons and rat brain. In brief, Rab7 reduced αSyn-induced pathology, ameliorated αSyn-induced neuronal degeneration, oxidative stress and DNA damage. These findings indicate that Rab7 is able to disrupt the vicious cycle of cellular impairment, αSyn pathology and neurodegeneration present in PD. Stimulation of Rab7 and the autolysosomal degradation pathway could therefore constitute a beneficial strategy for PD.
Collapse
Affiliation(s)
- Eva M Szegö
- Department of Neurology, TU Dresden, Dresden, Germany.
| | - Chris Van den Haute
- Laboratory for Neurobiology and Gene Therapy, Department of Neurosciences, Leuven Brain Institute, KU Leuven, Leuven, Belgium; Leuven Viral Vector Core, KU Leuven, Leuven, Belgium.
| | - Lennart Höfs
- Department of Neurology, TU Dresden, Dresden, Germany.
| | - Veerle Baekelandt
- Laboratory for Neurobiology and Gene Therapy, Department of Neurosciences, Leuven Brain Institute, KU Leuven, Leuven, Belgium; Leuven Viral Vector Core, KU Leuven, Leuven, Belgium.
| | - Anke Van der Perren
- Laboratory for Neurobiology and Gene Therapy, Department of Neurosciences, Leuven Brain Institute, KU Leuven, Leuven, Belgium.
| | - Björn H Falkenburger
- Department of Neurology, TU Dresden, Dresden, Germany; Deutsches Zentrum für Neurodegenerative Erkrankungen, Dresden, Germany; Department of Neurology, RWTH Aachen University, Aachen, Germany; JARA-BRAIN Institute Molecular Neuroscience and Neuroimaging, Forschungszentrum Jülich GmbH and RWTH Aachen University, Aachen, Germany.
| |
Collapse
|
8
|
The presynaptic glycine transporter GlyT2 is regulated by the Hedgehog pathway in vitro and in vivo. Commun Biol 2021; 4:1197. [PMID: 34663888 PMCID: PMC8523746 DOI: 10.1038/s42003-021-02718-6] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/19/2020] [Accepted: 09/22/2021] [Indexed: 01/20/2023] Open
Abstract
The identity of a glycinergic synapse is maintained presynaptically by the activity of a surface glycine transporter, GlyT2, which recaptures glycine back to presynaptic terminals to preserve vesicular glycine content. GlyT2 loss-of-function mutations cause Hyperekplexia, a rare neurological disease in which loss of glycinergic neurotransmission causes generalized stiffness and strong motor alterations. However, the molecular underpinnings controlling GlyT2 activity remain poorly understood. In this work, we identify the Hedgehog pathway as a robust controller of GlyT2 expression and transport activity. Modulating the activation state of the Hedgehog pathway in vitro in rodent primary spinal cord neurons or in vivo in zebrafish embryos induced a selective control in GlyT2 expression, regulating GlyT2 transport activity. Our results indicate that activation of Hedgehog reduces GlyT2 expression by increasing its ubiquitination and degradation. This work describes a new molecular link between the Hedgehog signaling pathway and presynaptic glycine availability. By modulating the activation state of the Hedgehog pathway, de la Rocha-Muñoz et al demonstrate that Hedgehog signaling controls the expression and transport activity of the neuronal glycine transporter GlyT2. This work begins to reveal a potential link between the Hedgehog signaling pathway and presynaptic glycine availability.
Collapse
|
9
|
Fernández G, Krapacher F, Ferreras S, Quassollo G, Mari MM, Pisano MV, Montemerlo A, Rubianes MD, Bregonzio C, Arias C, Paglini MG. Lack of Cdk5 activity is involved on Dopamine Transporter expression and function: Evidences from an animal model of Attention-Deficit Hyperactivity Disorder. Exp Neurol 2021; 346:113866. [PMID: 34537209 DOI: 10.1016/j.expneurol.2021.113866] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/18/2021] [Revised: 09/08/2021] [Accepted: 09/14/2021] [Indexed: 11/29/2022]
Abstract
Attention deficit/Hyperactivity disorder (ADHD) is one of the most diagnosed psychiatric disorders nowadays. The core symptoms of the condition include hyperactivity, impulsiveness and inattention. The main pharmacological treatment consists of psychostimulant drugs affecting Dopamine Transporter (DAT) function. We have previously shown that genetically modified mice lacking p35 protein (p35KO), which have reduced Cdk5 activity, present key hallmarks resembling those described in animal models useful for studying ADHD. The p35KO mouse displays spontaneous hyperactivity and shows a calming effect of methylphenidate or amphetamine treatment. Interestingly, dopaminergic neurotransmission is altered in these mice as they have an increased Dopamine (DA) content together with a low DA turnover. This led us to hypothesize that the lack of Cdk5 activity affects DAT expression and/or function in this animal model. In this study, we performed biochemical assays, cell-based approaches, quantitative fluorescence analysis and functional studies that allowed us to demonstrate that p35KO mice exhibit decreased DA uptake and reduced cell surface DAT expression levels in the striatum (STR). These findings are supported by in vitro observations in which the inhibition of Cdk5 activity in N2a cells induced a significant increase in constitutive DAT endocytosis with a concomitant increase in DAT localization to recycling endosomes. Taken together, these data provide evidences regarding the role of Cdk5/p35 in DAT expression and function, thus contributing to the knowledge of DA neurotransmission physiology and also providing therapeutic options for the treatment of DA pathologies such as ADHD.
Collapse
Affiliation(s)
- Guillermo Fernández
- Laboratory of Neurophysiology, Instituto de Investigación Médica Mercedes y Martín Ferreyra, INIMEC-CONICET, Universidad Nacional de Córdoba, Córdoba, Argentina
| | - Favio Krapacher
- Laboratory of Neurophysiology, Instituto de Investigación Médica Mercedes y Martín Ferreyra, INIMEC-CONICET, Universidad Nacional de Córdoba, Córdoba, Argentina
| | - Soledad Ferreras
- Laboratory of Neurophysiology, Instituto de Investigación Médica Mercedes y Martín Ferreyra, INIMEC-CONICET, Universidad Nacional de Córdoba, Córdoba, Argentina
| | - Gonzalo Quassollo
- Laboratory of Neurophysiology, Instituto de Investigación Médica Mercedes y Martín Ferreyra, INIMEC-CONICET, Universidad Nacional de Córdoba, Córdoba, Argentina
| | - Macarena Mariel Mari
- Laboratory of Neurophysiology, Instituto de Investigación Médica Mercedes y Martín Ferreyra, INIMEC-CONICET, Universidad Nacional de Córdoba, Córdoba, Argentina
| | - María Victoria Pisano
- Laboratory of Neurophysiology, Instituto de Investigación Médica Mercedes y Martín Ferreyra, INIMEC-CONICET, Universidad Nacional de Córdoba, Córdoba, Argentina
| | - Antonella Montemerlo
- Instituto de Investigaciones en Fisicoquímica de Córdoba, INFIQC-CONICET, Facultad de Ciencias Químicas, Universidad Nacional de Córdoba, Córdoba, Argentina
| | - María Dolores Rubianes
- Instituto de Investigaciones en Fisicoquímica de Córdoba, INFIQC-CONICET, Facultad de Ciencias Químicas, Universidad Nacional de Córdoba, Córdoba, Argentina
| | - Claudia Bregonzio
- Instituto de Farmacología Experimental Córdoba, IFEC-CONICET, Departamento de Farmacología, Facultad de Ciencias Químicas, Universidad Nacional de Córdoba, Córdoba, Argentina
| | - Carlos Arias
- Laboratory of Neurophysiology, Instituto de Investigación Médica Mercedes y Martín Ferreyra, INIMEC-CONICET, Universidad Nacional de Córdoba, Córdoba, Argentina; Instituto de Investigaciones Psicológicas, IIPSI-CONICET, Facultad de Psicología, Universidad Nacional de Córdoba, Córdoba, Argentina
| | - María Gabriela Paglini
- Laboratory of Neurophysiology, Instituto de Investigación Médica Mercedes y Martín Ferreyra, INIMEC-CONICET, Universidad Nacional de Córdoba, Córdoba, Argentina; Instituto de Virología "Dr. J. M. Vanella", Facultad de Ciencias Médicas, Universidad Nacional de Córdoba, Córdoba, Argentina.
| |
Collapse
|
10
|
Dynamic control of the dopamine transporter in neurotransmission and homeostasis. NPJ Parkinsons Dis 2021; 7:22. [PMID: 33674612 PMCID: PMC7935902 DOI: 10.1038/s41531-021-00161-2] [Citation(s) in RCA: 59] [Impact Index Per Article: 19.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/18/2020] [Accepted: 01/08/2021] [Indexed: 01/31/2023] Open
Abstract
The dopamine transporter (DAT) transports extracellular dopamine into the intracellular space contributing to the regulation of dopamine neurotransmission. A reduction of DAT density is implicated in Parkinson's disease (PD) by neuroimaging; dopamine turnover is dopamine turnover is elevated in early symptomatic PD and in presymptomatic individuals with monogenic mutations causal for parkinsonism. As an integral plasma membrane protein, DAT surface expression is dynamically regulated through endocytic trafficking, enabling flexible control of dopamine signaling in time and space, which in turn critically modulates movement, motivation and learning behavior. Yet the cellular machinery and functional implications of DAT trafficking remain enigmatic. In this review we summarize mechanisms governing DAT trafficking under normal physiological conditions and discuss how PD-linked mutations may disturb DAT homeostasis. We highlight the complexity of DAT trafficking and reveal DAT dysregulation as a common theme in genetic models of parkinsonism.
Collapse
|
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: 0] [Impact Index Per Article: 0] [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
|
Gaggi G, Di Credico A, Izzicupo P, Alviano F, Di Mauro M, Di Baldassarre A, Ghinassi B. Human Mesenchymal Stromal Cells Unveil an Unexpected Differentiation Potential toward the Dopaminergic Neuronal Lineage. Int J Mol Sci 2020; 21:ijms21186589. [PMID: 32916865 PMCID: PMC7555006 DOI: 10.3390/ijms21186589] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/08/2020] [Revised: 08/29/2020] [Accepted: 09/08/2020] [Indexed: 12/12/2022] Open
Abstract
Degeneration of dopaminergic neurons represents the cause of many neurodegenerative diseases, with increasing incidence worldwide. The replacement of dead cells with new healthy ones may represent an appealing therapeutic approach to these pathologies, but currently, only pluripotent stem cells can generate dopaminergic neurons with high efficiency. However, with the use of these cells arises safety and/or ethical issues. Human mesenchymal stromal cells (hFM-MSCs) are perinatal stem cells that can be easily isolated from the amniochorionic membrane after delivery. Generally considered multipotent, their real differentiative potential is not completely elucidated. The aim of this study was to analyze their stemness characteristics and to evaluate whether they may overcome their mesenchymal fate, generating dopaminergic neurons. We demonstrated that hFM-MSCs expressed embryonal genes OCT4, NANOG, SOX2, KLF4, OVOL1, and ESG1, suggesting they have some features of pluripotency. Moreover, hFM-MSCs that underwent a dopaminergic differentiation protocol gradually increased the transcription of dopaminergic markers LMX1b, NURR1, PITX3, and DAT. We finally obtained a homogeneous population of cells resembling the morphology of primary midbrain dopaminergic neurons that expressed the functional dopaminergic markers TH, DAT, and Nurr1. In conclusion, our results suggested that hFM-MSCs retain the expression of pluripotency genes and are able to differentiate not only into mesodermal cells, but also into neuroectodermal dopaminergic neuron-like cells.
Collapse
Affiliation(s)
- Giulia Gaggi
- Human Anatomy and Cell Differentiation Lab, Department of Medicine and Aging Sciences, University “G. D’Annunzio” of Chieti-Pescara, 66100 Chieti, Italy; (G.G.); (A.D.C.); (P.I.); (B.G.)
| | - Andrea Di Credico
- Human Anatomy and Cell Differentiation Lab, Department of Medicine and Aging Sciences, University “G. D’Annunzio” of Chieti-Pescara, 66100 Chieti, Italy; (G.G.); (A.D.C.); (P.I.); (B.G.)
| | - Pascal Izzicupo
- Human Anatomy and Cell Differentiation Lab, Department of Medicine and Aging Sciences, University “G. D’Annunzio” of Chieti-Pescara, 66100 Chieti, Italy; (G.G.); (A.D.C.); (P.I.); (B.G.)
| | - Francesco Alviano
- Department of Experimental Diagnostic and Speciality Medicine, Unit of Histology, Embriology and Applied Biology, University of Bologna, 40126 Bologna, Italy;
| | - Michele Di Mauro
- Cardio-Thoracic Surgery Unit, Heart and Vascular Centre, Maastricht University Medical Centre (MUMC), Cardiovascular Research Institute Maastricht (CARIM), 6202 Maastricht, The Netherlands;
| | - Angela Di Baldassarre
- Human Anatomy and Cell Differentiation Lab, Department of Medicine and Aging Sciences, University “G. D’Annunzio” of Chieti-Pescara, 66100 Chieti, Italy; (G.G.); (A.D.C.); (P.I.); (B.G.)
- Correspondence:
| | - Barbara Ghinassi
- Human Anatomy and Cell Differentiation Lab, Department of Medicine and Aging Sciences, University “G. D’Annunzio” of Chieti-Pescara, 66100 Chieti, Italy; (G.G.); (A.D.C.); (P.I.); (B.G.)
| |
Collapse
|
13
|
Jeon M, Lin G, Stephen ZR, Vechey JE, Singh M, Revia R, Newman AH, Martinez D, Zhang M. Cocaine analogue conjugated magnetic nanoparticles for labeling and imaging dopaminergic neurons. Biomater Sci 2020; 8:4166-4175. [PMID: 32515443 DOI: 10.1039/d0bm00546k] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2022]
Abstract
Molecular imaging of the dopamine transporter (DAT) with Positron Emission Tomography (PET) or Single Photon Emission Computed Tomography (SPECT) has been widely used in studies of neurological and psychiatric disorders. Nevertheless, there is a great interest in expanding molecular imaging to include magnetic resonance technology, because of the superior spatial resolution this technology may provide. Here we present a magnetic nanoparticle (NP) that specifically targets dopaminergic neurons and allows DAT imaging with magnetic resonance imaging (MRI). The nanoparticle (namely, NP-DN) is composed of an iron oxide core and a polyethylene glycol (PEG) coating to which a DAT specific dopaminergic neurolabeler (DN) is conjugated. NP-DN displayed long-term stability with favorable hydrodynamic size and surface charge suitable for in vivo application. In vitro studies showed NP-DN was non-toxic, displayed specificity towards DAT-expressing neurons, and demonstrated a 3-fold increase in DAT labeling over non-targeted NP. Our study shows NP-DN provides excellent contrast enhancement for MRI and demonstrates great potential for neuroimaging.
Collapse
Affiliation(s)
- Mike Jeon
- Department of Material Sciences and Engineering, University of Washington, Seattle, WA 98195, USA.
| | | | | | | | | | | | | | | | | |
Collapse
|
14
|
Fagan RR, Kearney PJ, Sweeney CG, Luethi D, Schoot Uiterkamp FE, Schicker K, Alejandro BS, O'Connor LC, Sitte HH, Melikian HE. Dopamine transporter trafficking and Rit2 GTPase: Mechanism of action and in vivo impact. J Biol Chem 2020; 295:5229-5244. [PMID: 32132171 PMCID: PMC7170531 DOI: 10.1074/jbc.ra120.012628] [Citation(s) in RCA: 20] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/22/2020] [Revised: 02/20/2020] [Indexed: 12/20/2022] Open
Abstract
Following its evoked release, dopamine (DA) signaling is rapidly terminated by presynaptic reuptake, mediated by the cocaine-sensitive DA transporter (DAT). DAT surface availability is dynamically regulated by endocytic trafficking, and direct protein kinase C (PKC) activation acutely diminishes DAT surface expression by accelerating DAT internalization. Previous cell line studies demonstrated that PKC-stimulated DAT endocytosis requires both Ack1 inactivation, which releases a DAT-specific endocytic brake, and the neuronal GTPase, Rit2, which binds DAT. However, it is unknown whether Rit2 is required for PKC-stimulated DAT endocytosis in DAergic terminals or whether there are region- and/or sex-dependent differences in PKC-stimulated DAT trafficking. Moreover, the mechanisms by which Rit2 controls PKC-stimulated DAT endocytosis are unknown. Here, we directly examined these important questions. Ex vivo studies revealed that PKC activation acutely decreased DAT surface expression selectively in ventral, but not dorsal, striatum. AAV-mediated, conditional Rit2 knockdown in DAergic neurons impacted baseline DAT surface:intracellular distribution in DAergic terminals from female ventral, but not dorsal, striatum. Further, Rit2 was required for PKC-stimulated DAT internalization in both male and female ventral striatum. FRET and surface pulldown studies in cell lines revealed that PKC activation drives DAT-Rit2 surface dissociation and that the DAT N terminus is required for both PKC-mediated DAT-Rit2 dissociation and DAT internalization. Finally, we found that Rit2 and Ack1 independently converge on DAT to facilitate PKC-stimulated DAT endocytosis. Together, our data provide greater insight into mechanisms that mediate PKC-regulated DAT internalization and reveal unexpected region-specific differences in PKC-stimulated DAT trafficking in bona fide DAergic terminals.
Collapse
Affiliation(s)
- Rita R Fagan
- Brudnick Neuropsychiatric Research Institute, Department of Neurobiology, University of Massachusetts Medical School, Worcester, Massachusetts 01605
| | - Patrick J Kearney
- Brudnick Neuropsychiatric Research Institute, Department of Neurobiology, University of Massachusetts Medical School, Worcester, Massachusetts 01605
| | - Carolyn G Sweeney
- Brudnick Neuropsychiatric Research Institute, Department of Neurobiology, University of Massachusetts Medical School, Worcester, Massachusetts 01605
| | - Dino Luethi
- Institute of Pharmacology, Center for Physiology and Pharmacology, Medical University Vienna, Vienna A-1090, Austria
| | - Florianne E Schoot Uiterkamp
- Institute of Pharmacology, Center for Physiology and Pharmacology, Medical University Vienna, Vienna A-1090, Austria
| | - Klaus Schicker
- Institute of Pharmacology, Center for Physiology and Pharmacology, Medical University Vienna, Vienna A-1090, Austria
| | - Brian S Alejandro
- Brudnick Neuropsychiatric Research Institute, Department of Neurobiology, University of Massachusetts Medical School, Worcester, Massachusetts 01605
| | - Lauren C O'Connor
- Brudnick Neuropsychiatric Research Institute, Department of Neurobiology, University of Massachusetts Medical School, Worcester, Massachusetts 01605
| | - Harald H Sitte
- Institute of Pharmacology, Center for Physiology and Pharmacology, Medical University Vienna, Vienna A-1090, Austria
| | - Haley E Melikian
- Brudnick Neuropsychiatric Research Institute, Department of Neurobiology, University of Massachusetts Medical School, Worcester, Massachusetts 01605.
| |
Collapse
|
15
|
Fagan RR, Kearney PJ, Melikian HE. In Situ Regulated Dopamine Transporter Trafficking: There's No Place Like Home. Neurochem Res 2020; 45:1335-1343. [PMID: 32146647 DOI: 10.1007/s11064-020-03001-6] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/08/2019] [Revised: 02/24/2020] [Accepted: 02/26/2020] [Indexed: 12/13/2022]
Abstract
Dopamine (DA) is critical for motivation, reward, movement initiation, and learning. Mechanisms that control DA signaling have a profound impact on these important behaviors, and additionally play a role in DA-related neuropathologies. The presynaptic SLC6 DA transporter (DAT) limits extracellular DA levels by clearing released DA, and is potently inhibited by addictive and therapeutic psychostimulants. Decades of evidence support that the DAT is subject to acute regulation by a number of signaling pathways, and that endocytic trafficking strongly regulates DAT availability and function. DAT trafficking studies have been performed in a variety of model systems, including both in vitro and ex vivo preparations. In this review, we focus on the breadth of DAT trafficking studies, with specific attention to, and comparison of, how context may influence DAT's response to different stimuli. In particular, this overview highlights that stimulated DAT trafficking not only differs between in vitro and ex vivo environments, but also is influenced by both sex and anatomical subregions.
Collapse
Affiliation(s)
- Rita R Fagan
- Brudnick Neuropsychiatric Research Institute, Department of Neurobiology, University of Massachusetts Medical School, Worcester, MA, 01605, USA
| | - Patrick J Kearney
- Brudnick Neuropsychiatric Research Institute, Department of Neurobiology, University of Massachusetts Medical School, Worcester, MA, 01605, USA
| | - Haley E Melikian
- Brudnick Neuropsychiatric Research Institute, Department of Neurobiology, University of Massachusetts Medical School, Worcester, MA, 01605, USA.
| |
Collapse
|
16
|
Qi X, Wang S, Zhang L, Liu L, Wen Y, Ma M, Cheng S, Li P, Cheng B, Du Y, Liang X, Zhao Y, Ding M, Zhang F. An integrative analysis of transcriptome-wide association study and mRNA expression profile identified candidate genes for attention-deficit/hyperactivity disorder. Psychiatry Res 2019; 282:112639. [PMID: 31685286 DOI: 10.1016/j.psychres.2019.112639] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/18/2019] [Revised: 10/18/2019] [Accepted: 10/24/2019] [Indexed: 02/01/2023]
Abstract
BACKGROUND Attention-deficit/hyperactivity disorder (ADHD) is a common neurodevelopmental disorder, but the genetic mechanism of ADHD remains elusive now. METHODS Tissue specific transcriptome-wide association study (TWAS) of ADHD was performed by FUSION utilizing a genome-wide association study (GWAS) dataset of ADHD (including 20,183 ADHD cases and 35,191 healthy controls) and gene expression reference from brain and blood. Furthermore, the genes identified by TWAS were compared with the differently expressed genes detected by mRNA expression profiles of ADHD rat model and autism spectrum disorders (ASD) patients. Functional enrichment and annotation analysis of the identified genes were performed by DAVID and FUMAGWAS tool. RESULTS For brain tissue, TWAS identified 148 genes with P value < 0.05, such as TDO2 (PTWAS=4.01×10-2), CHD1L (PTWAS=9.64×10-3) and KIAA0319L (PTWAS=4.05×10-4). Further 11 common genes were examined in the mRNA expression datasets, such as ACSM5 (PTWAS=3.62×10-2, PmRNA=0.005), CCDC24 (PTWAS=1.49×10-2, PmRNA=2.35×10-3) and MVP (PTWAS=5.55×10-3, PmRNA=5.40×10-3). Pathway enrichment analysis of the genes identified by TWAS detected 3 pathways for ADHD, including Other glycan degradation (P value=0.021), Viral myocarditis (P value=0.034) and Endocytosis (P value=0.041). CONCLUSIONS Through integrating GWAS and mRNA expression data, we identified a group of ADHD-associated genes and pathways, providing novel clues for understanding the genetic mechanism of ADHD.
Collapse
Affiliation(s)
- Xin Qi
- School of Public Health, Health Science Center, Xi'an Jiaotong University, Xi'an, P. R. China
| | - Sen Wang
- School of Public Health, Health Science Center, Xi'an Jiaotong University, Xi'an, P. R. China
| | - Lu Zhang
- School of Public Health, Health Science Center, Xi'an Jiaotong University, Xi'an, P. R. China
| | - Li Liu
- School of Public Health, Health Science Center, Xi'an Jiaotong University, Xi'an, P. R. China
| | - Yan Wen
- School of Public Health, Health Science Center, Xi'an Jiaotong University, Xi'an, P. R. China
| | - Mei Ma
- School of Public Health, Health Science Center, Xi'an Jiaotong University, Xi'an, P. R. China
| | - Shiqiang Cheng
- School of Public Health, Health Science Center, Xi'an Jiaotong University, Xi'an, P. R. China
| | - Ping Li
- School of Public Health, Health Science Center, Xi'an Jiaotong University, Xi'an, P. R. China
| | - Bolun Cheng
- School of Public Health, Health Science Center, Xi'an Jiaotong University, Xi'an, P. R. China
| | - Yanan Du
- School of Public Health, Health Science Center, Xi'an Jiaotong University, Xi'an, P. R. China
| | - Xiao Liang
- School of Public Health, Health Science Center, Xi'an Jiaotong University, Xi'an, P. R. China
| | - Yan Zhao
- School of Public Health, Health Science Center, Xi'an Jiaotong University, Xi'an, P. R. China
| | - Miao Ding
- School of Public Health, Health Science Center, Xi'an Jiaotong University, Xi'an, P. R. China
| | - Feng Zhang
- School of Public Health, Health Science Center, Xi'an Jiaotong University, Xi'an, P. R. China.
| |
Collapse
|
17
|
Kovtun O, Tomlinson ID, Bailey DM, Thal LB, Ross EJ, Harris L, Frankland MP, Ferguson RS, Glaser Z, Greer J, Rosenthal SJ. Single Quantum Dot Tracking Illuminates Neuroscience at the Nanoscale. Chem Phys Lett 2018; 706:741-752. [PMID: 30270931 PMCID: PMC6157616 DOI: 10.1016/j.cplett.2018.06.019] [Citation(s) in RCA: 25] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/15/2022]
Abstract
The use of nanometer-sized semiconductor crystals, known as quantum dots, allows us to directly observe individual biomolecular transactions through a fluorescence microscope. Here, we review the evolution of single quantum dot tracking over the past two decades, highlight key biophysical discoveries facilitated by quantum dots, briefly discuss biochemical and optical implementation strategies for a single quantum dot tracking experiment, and report recent accomplishments of our group at the interface of molecular neuroscience and nanoscience.
Collapse
Affiliation(s)
- Oleg Kovtun
- Departments of Chemistry, Chemical Biology, Vanderbilt University
- Departments of Vanderbilt Institute of Nanoscale Science and Engineering
| | - Ian D. Tomlinson
- Departments of Chemistry, Chemical Biology, Vanderbilt University
- Departments of Vanderbilt Institute of Nanoscale Science and Engineering
| | - Danielle M. Bailey
- Departments of Chemistry, Chemical Biology, Vanderbilt University
- Departments of Pharmacology, Chemical Biology, Vanderbilt University
- Departments of Vanderbilt Institute of Nanoscale Science and Engineering
| | - Lucas B. Thal
- Departments of Chemistry, Chemical Biology, Vanderbilt University
- Departments of Vanderbilt Institute of Nanoscale Science and Engineering
- Departments of Vanderbilt Institute of Chemical Biology, Vanderbilt University, Nashville, TN
| | - Emily J. Ross
- Departments of Hudson Alpha Institute for Biotechnology, Huntsville, AL
| | - Lauren Harris
- Departments of Vanderbilt Institute of Chemical Biology, Vanderbilt University, Nashville, TN
| | | | | | - Zachary Glaser
- Departments of Chemistry, Chemical Biology, Vanderbilt University
| | - Jonathan Greer
- Departments of Chemistry, Chemical Biology, Vanderbilt University
| | - Sandra J. Rosenthal
- Departments of Chemistry, Chemical Biology, Vanderbilt University
- Departments of Pharmacology, Chemical Biology, Vanderbilt University
- Departments of Chemical and Biomolecular Engineering, Chemical Biology, Vanderbilt University
- Departments of Physics and Astronomy, Chemical Biology, Vanderbilt University
- Departments of Vanderbilt Institute of Nanoscale Science and Engineering
- Departments of Vanderbilt Institute of Chemical Biology, Vanderbilt University, Nashville, TN
| |
Collapse
|
18
|
Sambo DO, Lin M, Owens A, Lebowitz JJ, Richardson B, Jagnarine DA, Shetty M, Rodriquez M, Alonge T, Ali M, Katz J, Yan L, Febo M, Henry LK, Bruijnzeel AW, Daws L, Khoshbouei H. The sigma-1 receptor modulates methamphetamine dysregulation of dopamine neurotransmission. Nat Commun 2017; 8:2228. [PMID: 29263318 PMCID: PMC5738444 DOI: 10.1038/s41467-017-02087-x] [Citation(s) in RCA: 79] [Impact Index Per Article: 11.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/09/2017] [Accepted: 11/05/2017] [Indexed: 12/16/2022] Open
Abstract
Dopamine neurotransmission is highly dysregulated by the psychostimulant methamphetamine, a substrate for the dopamine transporter (DAT). Through interactions with DAT, methamphetamine increases extracellular dopamine levels in the brain, leading to its rewarding and addictive properties. Methamphetamine also interacts with the sigma-1 receptor (σ1R), an inter-organelle signaling modulator. Using complementary strategies, we identified a novel mechanism for σ1R regulation of dopamine neurotransmission in response to methamphetamine. We found that σ1R activation prevents methamphetamine-induced, DAT-mediated increases in firing activity of dopamine neurons. In vitro and in vivo amperometric measurements revealed that σ1R activation decreases methamphetamine-stimulated dopamine efflux without affecting basal dopamine neurotransmission. Consistent with these findings, σ1R activation decreases methamphetamine-induced locomotion, motivated behavior, and enhancement of brain reward function. Notably, we revealed that the σ1R interacts with DAT at or near the plasma membrane and decreases methamphetamine-induced Ca2+ signaling, providing potential mechanisms. Broadly, these data provide evidence for σ1R regulation of dopamine neurotransmission and support the σ1R as a putative target for the treatment of methamphetamine addiction.
Collapse
Affiliation(s)
- Danielle O Sambo
- Department of Neuroscience, University of Florida, Gainesville, FL, 32611, USA
| | - Min Lin
- Department of Neuroscience, University of Florida, Gainesville, FL, 32611, USA
| | - Anthony Owens
- Department of Cellular & Integrative Physiology, University of Texas Health Science Center at San Antonio, San Antonio, TX, 78229, USA
| | - Joseph J Lebowitz
- Department of Neuroscience, University of Florida, Gainesville, FL, 32611, USA
| | - Ben Richardson
- Department of Neuroscience, University of Florida, Gainesville, FL, 32611, USA
| | - Darin A Jagnarine
- Department of Psychiatry, University of Florida, Gainesville, FL, 32611, USA
| | - Madhur Shetty
- Department of Biomedical Sciences, University of North Dakota, Grand Forks, ND, 58203, USA
| | - Meghan Rodriquez
- Department of Biomedical Sciences, University of North Dakota, Grand Forks, ND, 58203, USA
| | - Taiwo Alonge
- Department of Neuroscience, University of Florida, Gainesville, FL, 32611, USA
| | - Mishaal Ali
- Department of Neuroscience, University of Florida, Gainesville, FL, 32611, USA
| | - Jonathan Katz
- Psychobiology Section, Intramural Research Program, National Institute on Drug Abuse, National Institutes of Health, Baltimore, MD, 21224, USA
| | - Long Yan
- Max Plank Institute for Neuroscience Jupiter, Jupiter, FL, 33458, USA
| | - Marcelo Febo
- Department of Psychiatry, University of Florida, Gainesville, FL, 32611, USA
| | - L Keith Henry
- Department of Biomedical Sciences, University of North Dakota, Grand Forks, ND, 58203, USA
| | | | - Lynette Daws
- Department of Cellular & Integrative Physiology, University of Texas Health Science Center at San Antonio, San Antonio, TX, 78229, USA
| | - Habibeh Khoshbouei
- Department of Neuroscience, University of Florida, Gainesville, FL, 32611, USA.
| |
Collapse
|
19
|
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: 47] [Impact Index Per Article: 6.7] [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
|
20
|
Yuan J, Liu XD, Han M, Lv RB, Wang YK, Zhang GM, Li Y. Comparison of striatal dopamine transporter levels in chronic heroin-dependent and methamphetamine-dependent subjects. Addict Biol 2017; 22:229-234. [PMID: 26040446 DOI: 10.1111/adb.12271] [Citation(s) in RCA: 21] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/29/2022]
Abstract
To compare the effects of heroin and methamphetamine (METH) addiction on dopamine transporters (DATs) in the same dose and duration, we assessed DAT levels in the striatum by 99m Tc-TRODAT-1 single-photon emission computed tomography (SPECT) brain images in people with heroin and METH dependence. We recruited 21 healthy human controls, 23 heroin-dependent subjects and 25 METH abusers. The heroin- and METH-dependent subjects exhibited negative urine toxicology after undergoing physiological detoxification. All subjects underwent SPECT brain imaging, and specific tracer uptake ratios (SURs) were assessed bilaterally in the regions of interest. A significant SUR reduction in heroin-dependent subjects and METH-dependent subjects compared with healthy controls was found in the left striatum, right striatum, left caudate nucleus, right caudate nucleus, left putamen and right putamen. There were no significant differences in the heroin group and METH group for the left striatum, right striatum, left caudate nucleus, right caudate nucleus, left putamen and right putamen. The scores of craving, HAMA (Hamilton Anxiety Rating Scale), in heroin abusers were lower than in the METH abusers. Our results show that people with heroin and METH dependence who are currently abstinent had lower DAT levels in the striatum than healthy controls. There were no differences in striatal DAT in heroin and METH users. These results suggest that chronic heroin and METH abuse appears to produce similar effects in striatal DAT in humans. METH users may have more serious craving and anxiety symptoms than heroin users with prolonged abstinence.
Collapse
Affiliation(s)
- Jie Yuan
- Department of Nuclear Medicine; Fudan University; China
| | - Xing Dang Liu
- Department of Nuclear Medicine; Fudan University; China
| | - Mei Han
- Department of Huashan Hospital; Fudan University; China
- Key Laboratory of Radiopharmaceuticals, Ministry of Education, College of Chemistry; Beijing Normal University; China
| | - Rong Bin Lv
- Department of Nuclear Medicine; Fudan University; China
| | - Yuan Kai Wang
- Department of Nuclear Medicine; Fudan University; China
| | | | - Yu Li
- Drug Rehabilitation Centre; China
| |
Collapse
|
21
|
Foster SR, Bräuner-Osborne H. Investigating Internalization and Intracellular Trafficking of GPCRs: New Techniques and Real-Time Experimental Approaches. Handb Exp Pharmacol 2017; 245:41-61. [PMID: 29018878 DOI: 10.1007/164_2017_57] [Citation(s) in RCA: 23] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/23/2022]
Abstract
The ability to regulate the interaction between cells and their extracellular environment is essential for the maintenance of appropriate physiological function. For G protein-coupled receptors (GPCRs), this regulation occurs through multiple mechanisms that provide spatial and temporal control for signal transduction. One of the major mechanisms for GPCR regulation involves their endocytic trafficking, which serves to internalize the receptors from the plasma membrane and thereby attenuate G protein-dependent signaling. However, there is accumulating evidence to suggest that GPCRs can signal independently of G proteins, as well as from intracellular compartments including endosomes. It is in this context that receptor internalization and intracellular trafficking have attracted renewed interest within the GPCR field. In this chapter, we will review the current understanding and methodologies that have been used to investigate internalization and intracellular signaling of GPCRs, with a particular focus on emerging real-time techniques. These recent developments have improved our understanding of the complexities of GPCR internalization and intracellular signaling and suggest that the broader biological relevance and potential therapeutic implications of these processes remain to be explored.
Collapse
Affiliation(s)
- Simon R Foster
- Department of Drug Design and Pharmacology, Faculty of Health and Medical Sciences, University of Copenhagen, Universitetsparken 2, 2100, Copenhagen, Denmark
| | - Hans Bräuner-Osborne
- Department of Drug Design and Pharmacology, Faculty of Health and Medical Sciences, University of Copenhagen, Universitetsparken 2, 2100, Copenhagen, Denmark.
| |
Collapse
|
22
|
Liebick M, Schläger C, Oppermann M. Analysis of Chemokine Receptor Trafficking by Site-Specific Biotinylation. PLoS One 2016; 11:e0157502. [PMID: 27310579 PMCID: PMC4911081 DOI: 10.1371/journal.pone.0157502] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/04/2016] [Accepted: 05/31/2016] [Indexed: 12/17/2022] Open
Abstract
Chemokine receptors undergo internalization and desensitization in response to ligand activation. Internalized receptors are either preferentially directed towards recycling pathways (e.g. CCR5) or sorted for proteasomal degradation (e.g. CXCR4). Here we describe a method for the analysis of receptor internalization and recycling based on specific Bir A-mediated biotinylation of an acceptor peptide coupled to the receptor, which allows a more detailed analysis of receptor trafficking compared to classical antibody-based detection methods. Studies on constitutive internalization of the chemokine receptors CXCR4 (12.1% ± 0.99% receptor internalization/h) and CCR5 (13.7% ± 0.68%/h) reveals modulation of these processes by inverse (TAK779; 10.9% ± 0.95%/h) or partial agonists (Met-CCL5; 15.6% ± 0.5%/h). These results suggest an actively driven internalization process. We also demonstrate the advantages of specific biotinylation compared to classical antibody detection during agonist-induced receptor internalization, which may be used for immunofluorescence analysis as well. Site-specific biotinylation may be applicable to studies on trafficking of transmembrane proteins, in general.
Collapse
MESH Headings
- Amides/pharmacology
- Animals
- Antibodies, Monoclonal/biosynthesis
- Antibodies, Monoclonal/chemistry
- Antibodies, Monoclonal/isolation & purification
- Basophils/cytology
- Basophils/drug effects
- Basophils/metabolism
- Biotin/chemistry
- Biotin/metabolism
- Biotinylation
- CCR5 Receptor Antagonists/pharmacology
- Carbon-Nitrogen Ligases/genetics
- Carbon-Nitrogen Ligases/metabolism
- Cell Line, Tumor
- Chemokine CCL5/pharmacology
- Escherichia coli Proteins/genetics
- Escherichia coli Proteins/metabolism
- Gene Expression
- Genetic Vectors/chemistry
- Genetic Vectors/metabolism
- Mice
- Protein Transport/drug effects
- Quaternary Ammonium Compounds/pharmacology
- Rats
- Receptors, CXCR4/antagonists & inhibitors
- Receptors, CXCR4/genetics
- Receptors, CXCR4/metabolism
- Receptors, CXCR5/antagonists & inhibitors
- Receptors, CXCR5/genetics
- Receptors, CXCR5/metabolism
- Recombinant Fusion Proteins/genetics
- Recombinant Fusion Proteins/metabolism
- Repressor Proteins/genetics
- Repressor Proteins/metabolism
- Transfection
Collapse
Affiliation(s)
- Marcel Liebick
- Department of Cellular and Molecular Immunology, University of Göttingen, Göttingen, Niedersachsen, Germany
| | - Christian Schläger
- Department of Cellular and Molecular Immunology, University of Göttingen, Göttingen, Niedersachsen, Germany
| | - Martin Oppermann
- Department of Cellular and Molecular Immunology, University of Göttingen, Göttingen, Niedersachsen, Germany
| |
Collapse
|
23
|
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 2016; 67:1005-24. [PMID: 26408528 DOI: 10.1124/pr.114.010397] [Citation(s) in RCA: 120] [Impact Index Per Article: 15.0] [Reference Citation Analysis] [Abstract] [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
|
24
|
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: 45] [Impact Index Per Article: 5.6] [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
|
25
|
Vuorenpää A, Jørgensen TN, Newman AH, Madsen KL, Scheinin M, Gether U. Differential Internalization Rates and Postendocytic Sorting of the Norepinephrine and Dopamine Transporters Are Controlled by Structural Elements in the N Termini. J Biol Chem 2016; 291:5634-5651. [PMID: 26786096 DOI: 10.1074/jbc.m115.702050] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/03/2015] [Indexed: 11/06/2022] Open
Abstract
The norepinephrine transporter (NET) mediates reuptake of synaptically released norepinephrine in central and peripheral noradrenergic neurons. The molecular processes governing availability of NET in the plasma membrane are poorly understood. Here we use the fluorescent cocaine analogue JHC 1-64, as well as several other approaches, to investigate the trafficking itinerary of NET in live noradrenergic neurons. Confocal imaging revealed extensive constitutive internalization of JHC 1-64-labeled NET in the neuronal somata, proximal extensions and presynaptic boutons. Phorbol 12-myristate 13-acetate increased intracellular accumulation of JHC 1-64-labeled NET and caused a parallel reduction in uptake capacity. Internalized NET strongly colocalized with the "long loop" recycling marker Rab11, whereas less overlap was seen with the "short loop" recycling marker Rab4 and the late endosomal marker Rab7. Moreover, mitigating Rab11 function by overexpression of dominant negative Rab11 impaired NET function. Sorting of NET to the Rab11 recycling compartment was further supported by confocal imaging and reversible biotinylation experiments in transfected differentiated CATH.a cells. In contrast to NET, the dopamine transporter displayed markedly less constitutive internalization and limited sorting to the Rab11 recycling compartment in the differentiated CATH.a cells. Exchange of domains between the two homologous transporters revealed that this difference was determined by non-conserved structural elements in the intracellular N terminus. We conclude that NET displays a distinct trafficking itinerary characterized by continuous shuffling between the plasma membrane and the Rab11 recycling compartment and that the functional integrity of the Rab11 compartment is critical for maintaining proper presynaptic NET function.
Collapse
Affiliation(s)
- Anne Vuorenpää
- From the Molecular Neuropharmacology and Genetics Laboratory, Department of Neuroscience and Pharmacology, Faculty of Health and Medical Sciences, Panum Institute 18.6, University of Copenhagen, DK-2200 Copenhagen, Denmark,; the Department of Pharmacology, Drug Development, and Therapeutics, University of Turku, Turku FI-20014, Finland,; the Unit of Clinical Pharmacology, Turku University Hospital, Turku FI-20520, Finland, and
| | - Trine N Jørgensen
- From the Molecular Neuropharmacology and Genetics Laboratory, Department of Neuroscience and Pharmacology, Faculty of Health and Medical Sciences, Panum Institute 18.6, University of Copenhagen, DK-2200 Copenhagen, Denmark
| | - Amy H Newman
- the Medicinal Chemistry Section, National Institute on Drug Abuse, Intramural Research Program, National Institutes of Health, Baltimore, Maryland 21224
| | - Kenneth L Madsen
- From the Molecular Neuropharmacology and Genetics Laboratory, Department of Neuroscience and Pharmacology, Faculty of Health and Medical Sciences, Panum Institute 18.6, University of Copenhagen, DK-2200 Copenhagen, Denmark
| | - Mika Scheinin
- the Department of Pharmacology, Drug Development, and Therapeutics, University of Turku, Turku FI-20014, Finland,; the Unit of Clinical Pharmacology, Turku University Hospital, Turku FI-20520, Finland, and
| | - Ulrik Gether
- From the Molecular Neuropharmacology and Genetics Laboratory, Department of Neuroscience and Pharmacology, Faculty of Health and Medical Sciences, Panum Institute 18.6, University of Copenhagen, DK-2200 Copenhagen, Denmark,.
| |
Collapse
|
26
|
Long-term controlled GDNF over-expression reduces dopamine transporter activity without affecting tyrosine hydroxylase expression in the rat mesostriatal system. Neurobiol Dis 2016; 88:44-54. [PMID: 26777664 DOI: 10.1016/j.nbd.2016.01.002] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/09/2015] [Revised: 12/07/2015] [Accepted: 01/07/2016] [Indexed: 01/14/2023] Open
Abstract
The dopamine (DA) transporter (DAT) is a plasma membrane glycoprotein expressed in dopaminergic (DA-) cells that takes back DA into presynaptic neurons after its release. DAT dysfunction has been involved in different neuro-psychiatric disorders including Parkinson's disease (PD). On the other hand, numerous studies support that the glial cell line-derived neurotrophic factor (GDNF) has a protective effect on DA-cells. However, studies in rodents show that prolonged GDNF over-expression may cause a tyrosine hydroxylase (TH, the limiting enzyme in DA synthesis) decline. The evidence of TH down-regulation suggests that another player in DA handling, DAT, may also be regulated by prolonged GDNF over-expression, and the possibility that this effect is induced at GDNF expression levels lower than those inducing TH down-regulation. This issue was investigated here using intrastriatal injections of a tetracycline-inducible adeno-associated viral vector expressing human GDNF cDNA (AAV-tetON-GDNF) in rats, and doxycycline (DOX; 0.01, 0.03, 0.5 and 3mg/ml) in the drinking water during 5weeks. We found that 3mg/ml DOX promotes an increase in striatal GDNF expression of 12× basal GDNF levels and both DA uptake decrease and TH down-regulation in its native and Ser40 phosphorylated forms. However, 0.5mg/ml DOX promotes a GDNF expression increase of 3× basal GDNF levels with DA uptake decrease but not TH down-regulation. The use of western-blot under non-reducing conditions, co-immunoprecipitation and in situ proximity ligation assay revealed that the DA uptake decrease is associated with the formation of DAT dimers and an increase in DAT-α-synuclein interactions, without changes in total DAT levels or its compartmental distribution. In conclusion, at appropriate GDNF transduction levels, DA uptake is regulated through DAT protein-protein interactions without interfering with DA synthesis.
Collapse
|
27
|
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
|
28
|
Kuver A, Smith SS. Flumazenil decreases surface expression of α4β2δ GABAA receptors by increasing the rate of receptor internalization. Brain Res Bull 2015; 120:131-43. [PMID: 26592470 DOI: 10.1016/j.brainresbull.2015.11.015] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/19/2015] [Revised: 11/15/2015] [Accepted: 11/18/2015] [Indexed: 11/25/2022]
Abstract
Increases in expression of α4βδ GABAA receptors (GABARs), triggered by fluctuations in the neurosteroid THP (3α-OH-5α[β]-pregnan-20-one), are associated with changes in mood and cognition. We tested whether α4βδ trafficking and surface expression would be altered by in vitro exposure to flumazenil, a benzodiazepine ligand which reduces α4βδ expression in vivo. We first determined that flumazenil (100 nM-100 μM, IC50=∼1 μM) acted as a negative modulator, reducing GABA (10 μM)-gated current in the presence of 100 nM THP (to increase receptor efficacy), assessed with whole cell patch clamp recordings of recombinant α4β2δ expressed in HEK-293 cells. Surface expression of recombinant α4β2δ receptors was detected using a 3XFLAG reporter at the C-terminus of α4 (α4F) using confocal immunocytochemical techniques following 48 h exposure of cells to GABA (10 μM)+THP (100 nM). Flumazenil (10 μM) decreased surface expression of α4F by ∼60%, while increasing its intracellular accumulation, after 48 h. Reduced surface expression of α4β2δ after flumazenil treatment was confirmed by decreases in the current responses to 100 nM of the GABA agonist gaboxadol. Flumazenil-induced decreases in surface expression of α4β2δ were prevented by the dynamin blocker, dynasore, and by leupeptin, which blocks lysosomal enzymes, suggesting that flumazenil is acting to increase endocytosis and lysosomal degradation of the receptor. Flumazenil increased the rate of receptor removal from the cell surface by 2-fold, assessed using botulinum toxin B to block insertion of new receptors. These findings may suggest new therapeutic strategies for regulation of α4β2δ expression using flumazenil.
Collapse
Affiliation(s)
- Aarti Kuver
- Department of Physiology and Pharmacology, SUNY Downstate Medical Center, 450 Clarkson Ave, Brooklyn, NY 11203, USA
| | - Sheryl S Smith
- Department of Physiology and Pharmacology, SUNY Downstate Medical Center, 450 Clarkson Ave, Brooklyn, NY 11203, USA.
| |
Collapse
|
29
|
Fang XQ, Qiao H, Groveman BR, Feng S, Pflueger M, Xin WK, Ali MK, Lin SX, Xu J, Duclot F, Kabbaj M, Wang W, Ding XS, Santiago-Sim T, Jiang XH, Salter MW, Yu XM. Regulated internalization of NMDA receptors drives PKD1-mediated suppression of the activity of residual cell-surface NMDA receptors. Mol Brain 2015; 8:75. [PMID: 26584860 PMCID: PMC4653853 DOI: 10.1186/s13041-015-0167-1] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/06/2015] [Accepted: 11/13/2015] [Indexed: 12/13/2022] Open
Abstract
Background Constitutive and regulated internalization of cell surface proteins has been extensively investigated. The regulated internalization has been characterized as a principal mechanism for removing cell-surface receptors from the plasma membrane, and signaling to downstream targets of receptors. However, so far it is still not known whether the functional properties of remaining (non-internalized) receptor/channels may be regulated by internalization of the same class of receptor/channels. The N-methyl-D-aspartate receptor (NMDAR) is a principal subtype of glutamate-gated ion channel and plays key roles in neuronal plasticity and memory functions. NMDARs are well-known to undergo two types of regulated internalization – homologous and heterologous, which can be induced by high NMDA/glycine and DHPG, respectively. In the present work, we investigated effects of regulated NMDAR internalization on the activity of residual cell-surface NMDARs and neuronal functions. Results In electrophysiological experiments we discovered that the regulated internalization of NMDARs not only reduced the number of cell surface NMDARs but also caused an inhibition of the activity of remaining (non-internalized) surface NMDARs. In biochemical experiments we identified that this functional inhibition of remaining surface NMDARs was mediated by increased serine phosphorylation of surface NMDARs, resulting from the activation of protein kinase D1 (PKD1). Knockdown of PKD1 did not affect NMDAR internalization but prevented the phosphorylation and inhibition of remaining surface NMDARs and NMDAR-mediated synaptic functions. Conclusion These data demonstrate a novel concept that regulated internalization of cell surface NMDARs not only reduces the number of NMDARs on the cell surface but also causes an inhibition of the activity of remaining surface NMDARs through intracellular signaling pathway(s). Furthermore, modulating the activity of remaining surface receptors may be an effective approach for treating receptor internalization-induced changes in neuronal functions of the CNS. Electronic supplementary material The online version of this article (doi:10.1186/s13041-015-0167-1) contains supplementary material, which is available to authorized users.
Collapse
Affiliation(s)
- Xiao-Qian Fang
- Department of Biomedical Sciences, Florida State University, Tallahassee, FL, 32306, USA.
| | - Haifa Qiao
- Department of Biomedical Sciences, Florida State University, Tallahassee, FL, 32306, USA.
| | - Bradley R Groveman
- Department of Biomedical Sciences, Florida State University, Tallahassee, FL, 32306, USA.
| | - Shuang Feng
- Department of Biomedical Sciences, Florida State University, Tallahassee, FL, 32306, USA.
| | - Melissa Pflueger
- Department of Biomedical Sciences, Florida State University, Tallahassee, FL, 32306, USA.
| | - Wen-Kuan Xin
- Faculty of Dentistry, University of Toronto, Toronto, M5G 1G6, Canada.
| | - Mohammad K Ali
- Department of Biomedical Sciences, Florida State University, Tallahassee, FL, 32306, USA.
| | - Shuang-Xiu Lin
- Department of Biomedical Sciences, Florida State University, Tallahassee, FL, 32306, USA.
| | - Jindong Xu
- Faculty of Dentistry, University of Toronto, Toronto, M5G 1G6, Canada.
| | - Florian Duclot
- Department of Biomedical Sciences, Florida State University, Tallahassee, FL, 32306, USA.
| | - Mohamed Kabbaj
- Department of Biomedical Sciences, Florida State University, Tallahassee, FL, 32306, USA.
| | - Wei Wang
- Department of Neurology, 1st Affiliated Hospital of Nanjing Medical University, Nanjing, 210029, PR China.
| | - Xin-Sheng Ding
- Department of Neurology, 1st Affiliated Hospital of Nanjing Medical University, Nanjing, 210029, PR China. .,BenQ Neurological Institute, Nanjing Medical University, Nanjing, 210021, PR China.
| | - Teresa Santiago-Sim
- Department of Neurosurgery, The University of Texas Medical School at Houston Vivian L. Smith, Houston, TX, 77030, USA.
| | - Xing-Hong Jiang
- Department of Physiology and Neurobiology, Medical College of Soochow University, Suzhou, 215123, China.
| | - Michael W Salter
- Program in Neuroscience and Mental Health, Hospital for Sick Children, University of Toronto, Toronto, M5G 1X8, Canada.
| | - Xian-Min Yu
- Department of Biomedical Sciences, Florida State University, Tallahassee, FL, 32306, USA. .,Faculty of Dentistry, University of Toronto, Toronto, M5G 1G6, Canada. .,BenQ Neurological Institute, Nanjing Medical University, Nanjing, 210021, PR China. .,Department of Physiology and Neurobiology, Medical College of Soochow University, Suzhou, 215123, China.
| |
Collapse
|
30
|
Barrera SP, Castrejon-Tellez V, Trinidad M, Robles-Escajeda E, Vargas-Medrano J, Varela-Ramirez A, Miranda M. PKC-Dependent GlyT1 Ubiquitination Occurs Independent of Phosphorylation: Inespecificity in Lysine Selection for Ubiquitination. PLoS One 2015; 10:e0138897. [PMID: 26418248 PMCID: PMC4587969 DOI: 10.1371/journal.pone.0138897] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/29/2014] [Accepted: 09/06/2015] [Indexed: 01/08/2023] Open
Abstract
Neurotransmitter transporter ubiquitination is emerging as the main mechanism for endocytosis and sorting of cargo into lysosomes. In this study, we demonstrate PKC-dependent ubiquitination of three different isoforms of the glycine transporter 1 (GlyT1). Incubation of cells expressing transporter with the PKC activator phorbol ester induced a dramatic, time-dependent increase in GlyT1 ubiquitination, followed by accumulation of GlyT1 in EEA1 positive early endosomes. This occurred via a mechanism that was abolished by inhibition of PKC. GlyT1 endocytosis was confirmed in both retinal sections and primary cultures of mouse amacrine neurons. Replacement of only all lysines in the N-and C-termini to arginines prevented ubiquitination and endocytosis, displaying redundancy in the mechanism of ubiquitination. Interestingly, a 40–50% reduction in glycine uptake was detected in phorbol-ester stimulated cells expressing the WT-GlyT1, whereas no significant change was for the mutant protein, demonstrating that endocytosis participates in the reduction of uptake. Consistent with previous findings for the dopamine transporter DAT, ubiquitination of GlyT1 tails functions as sorting signal to deliver transporter into the lysosome and removal of ubiquitination sites dramatically attenuated the rate of GlyT1 degradation. Finally, we showed for the first time that PKC-dependent GlyT1 phosphorylation was not affected by removal of ubiquitination sites, suggesting separate PKC-dependent signaling events for these posttranslational modifications.
Collapse
Affiliation(s)
- Susana P. Barrera
- Department of Biological Sciences and Border Biomedical Research Center, University of Texas at El Paso, El Paso, TX, 79968, United States of America
| | - Vicente Castrejon-Tellez
- Department of Biological Sciences and Border Biomedical Research Center, University of Texas at El Paso, El Paso, TX, 79968, United States of America
| | - Margarita Trinidad
- Department of Biological Sciences and Border Biomedical Research Center, University of Texas at El Paso, El Paso, TX, 79968, United States of America
| | - Elisa Robles-Escajeda
- Department of Biological Sciences and Border Biomedical Research Center, University of Texas at El Paso, El Paso, TX, 79968, United States of America
| | - Javier Vargas-Medrano
- Department of Biological Sciences and Border Biomedical Research Center, University of Texas at El Paso, El Paso, TX, 79968, United States of America
| | - Armando Varela-Ramirez
- Department of Biological Sciences and Border Biomedical Research Center, University of Texas at El Paso, El Paso, TX, 79968, United States of America
| | - Manuel Miranda
- Department of Biological Sciences and Border Biomedical Research Center, University of Texas at El Paso, El Paso, TX, 79968, United States of America
- * E-mail:
| |
Collapse
|
31
|
Levoye A, Zwier JM, Jaracz-Ros A, Klipfel L, Cottet M, Maurel D, Bdioui S, Balabanian K, Prézeau L, Trinquet E, Durroux T, Bachelerie F. A Broad G Protein-Coupled Receptor Internalization Assay that Combines SNAP-Tag Labeling, Diffusion-Enhanced Resonance Energy Transfer, and a Highly Emissive Terbium Cryptate. Front Endocrinol (Lausanne) 2015; 6:167. [PMID: 26617570 PMCID: PMC4638144 DOI: 10.3389/fendo.2015.00167] [Citation(s) in RCA: 49] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/28/2015] [Accepted: 10/19/2015] [Indexed: 12/15/2022] Open
Abstract
Although G protein-coupled receptor (GPCR) internalization has long been considered as a major aspect of the desensitization process that tunes ligand responsiveness, internalization is also involved in receptor resensitization and signaling, as well as the ligand scavenging function of some atypical receptors. Internalization thus contributes to the diversity of GPCR-dependent signaling, and its dynamics and quantification in living cells has generated considerable interest. We developed a robust and sensitive assay to follow and quantify ligand-induced and constitutive-induced GPCR internalization but also receptor recycling in living cells. This assay is based on diffusion-enhanced resonance energy transfer (DERET) between cell surface GPCRs labeled with a luminescent terbium cryptate donor and a fluorescein acceptor present in the culture medium. GPCR internalization results in a quantifiable reduction of energy transfer. This method yields a high signal-to-noise ratio due to time-resolved measurements. For various GPCRs belonging to different classes, we demonstrated that constitutive and ligand-induced internalization could be monitored as a function of time and ligand concentration, thus allowing accurate quantitative determination of kinetics of receptor internalization but also half-maximal effective or inhibitory concentrations of compounds. In addition to its selectivity and sensitivity, we provided evidence that DERET-based internalization assay is particularly suitable for characterizing biased ligands. Furthermore, the determination of a Z'-factor value of 0.45 indicates the quality and suitability of DERET-based internalization assay for high-throughput screening (HTS) of compounds that may modulate GPCRs internalization.
Collapse
Affiliation(s)
- Angélique Levoye
- INSERM U1148, Laboratory of Vascular Translational Science, Université Paris 13, Sorbonne Paris Cité, Paris, France
- *Correspondence: Angélique Levoye,
| | | | - Agnieszka Jaracz-Ros
- INSERM UMR996, Inflammation, Chemokines and Immunopathology, Université Paris-Sud, Université Paris-Saclay, Clamart, France
| | - Laurence Klipfel
- INSERM UMR996, Inflammation, Chemokines and Immunopathology, Université Paris-Sud, Université Paris-Saclay, Clamart, France
| | - Martin Cottet
- CNRS UMR 5203, INSERM U1191, Institut de Génomique Fonctionnelle, Université Montpellier 1 & 2, Montpellier, France
| | - Damien Maurel
- CNRS UMR 5203, INSERM U1191, Institut de Génomique Fonctionnelle, Université Montpellier 1 & 2, Montpellier, France
| | | | - Karl Balabanian
- INSERM UMR996, Inflammation, Chemokines and Immunopathology, Université Paris-Sud, Université Paris-Saclay, Clamart, France
| | - Laurent Prézeau
- CNRS UMR 5203, INSERM U1191, Institut de Génomique Fonctionnelle, Université Montpellier 1 & 2, Montpellier, France
| | | | - Thierry Durroux
- CNRS UMR 5203, INSERM U1191, Institut de Génomique Fonctionnelle, Université Montpellier 1 & 2, Montpellier, France
| | - Françoise Bachelerie
- INSERM UMR996, Inflammation, Chemokines and Immunopathology, Université Paris-Sud, Université Paris-Saclay, Clamart, France
| |
Collapse
|
32
|
Smooth muscle 22α facilitates angiotensin II-induced signaling and vascular contraction. J Mol Med (Berl) 2014; 93:547-58. [PMID: 25515236 DOI: 10.1007/s00109-014-1240-4] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/06/2014] [Revised: 11/23/2014] [Accepted: 11/25/2014] [Indexed: 01/29/2023]
Abstract
UNLABELLED Smooth muscle 22α (SM22α) is involved in stress fiber formation and enhances contractility in vascular smooth muscle cells (VSMCs). In many cases, SM22α acts as an adapter protein to assemble signaling complexes and regulate signaling, but whether SM22α regulates contractile signaling induced by angiotensin II (AngII) remains unclear. To address this issue, we established a hypertension model of Sm22α(-/-) mice, and demonstrated that hypertension induced by AngII was attenuated in Sm22α(-/-) mice. A decreased vasoconstriction was observed in aortic rings from Sm22α(-/-) mice. Furthermore, loss of SM22α resulted in a reduced contractile response to AngII in VSMCs in vitro. The phosphorylation of extracellular signal-regulated kinase 1/2 (ERK1/2) induced by AngII was impaired following depletion of SM22α, in parallel with a reduced contractility. The decay of ERK1/2 activity was associated with increased expression of mitogen-activated protein kinase phosphatase 3 (MKP3). Inhibition of MKP3 activity rescued ERK1/2 activity. SM22α depletion caused an enhanced interaction of MKP3 with ERK1/2, and a reduced ubiquitination and degradation of MKP3. Knockdown of SM22α extended the half-life of MKP3. In conclusion, SM22α promotes AngII-induced contraction by maintenance of ERK1/2 signaling cascades through facilitating ubiquitination and degradation of MKP3. KEY MESSAGE The vasoconstriction is attenuated in aortic rings from Sm22α(-/-) mice. MKP3 mediates dephosphorylation of ERK1/2 in AngII-induced VSMC contraction. SM22α inhibits the interaction of ERK1/2 with MKP3. SM22α promotes ubiquitination and degradation of MKP3. SM22α facilitates AngII-induced contraction by maintenance of ERK1/2 signaling.
Collapse
|
33
|
Rahbek-Clemmensen T, Bay T, Eriksen J, Gether U, Jørgensen TN. The serotonin transporter undergoes constitutive internalization and is primarily sorted to late endosomes and lysosomal degradation. J Biol Chem 2014; 289:23004-23019. [PMID: 24973209 DOI: 10.1074/jbc.m113.495754] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/06/2022] Open
Abstract
The serotonin transporter (SERT) plays a critical role in regulating serotonin signaling by mediating reuptake of serotonin from the extracellular space. The molecular and cellular mechanisms controlling SERT levels in the membrane remain poorly understood. To study trafficking of the surface resident SERT, two functional epitope-tagged variants were generated. Fusion of a FLAG-tagged one-transmembrane segment protein Tac to the SERT N terminus generated a transporter with an extracellular epitope suited for trafficking studies (TacSERT). Likewise, a construct with an extracellular antibody epitope was generated by introducing an HA (hemagglutinin) tag in the extracellular loop 2 of SERT (HA-SERT). By using TacSERT and HA-SERT in antibody-based internalization assays, we show that SERT undergoes constitutive internalization in a dynamin-dependent manner. Confocal images of constitutively internalized SERT demonstrated that SERT primarily co-localized with the late endosomal/lysosomal marker Rab7, whereas little co-localization was observed with the Rab11, a marker of the "long loop" recycling pathway. This sorting pattern was distinct from that of a prototypical recycling membrane protein, the β2-adrenergic receptor. Furthermore, internalized SERT co-localized with the lysosomal marker LysoTracker and not with transferrin. The sorting pattern was further confirmed by visualizing internalization of SERT using the fluorescent cocaine analog JHC1-64 and by reversible and pulse-chase biotinylation assays showing evidence for lysosomal degradation of the internalized transporter. Finally, we found that SERT internalized in response to stimulation with 12-myristate 13-acetate co-localized primarily with Rab7- and LysoTracker-positive compartments. We conclude that SERT is constitutively internalized and that the internalized transporter is sorted mainly to degradation.
Collapse
Affiliation(s)
- Troels Rahbek-Clemmensen
- Molecular Neuropharmacology Laboratory, Department of Neuroscience and Pharmacology, Faculty of Health Sciences, Panum Institute, and University of Copenhagen, DK-2200 Copenhagen, Denmark; Lundbeck Foundation Center for Biomembranes in Nanomedicine, University of Copenhagen, DK-2200 Copenhagen, Denmark
| | - Tina Bay
- Molecular Neuropharmacology Laboratory, Department of Neuroscience and Pharmacology, Faculty of Health Sciences, Panum Institute, and University of Copenhagen, DK-2200 Copenhagen, Denmark
| | - Jacob Eriksen
- Molecular Neuropharmacology Laboratory, Department of Neuroscience and Pharmacology, Faculty of Health Sciences, Panum Institute, and University of Copenhagen, DK-2200 Copenhagen, Denmark; Lundbeck Foundation Center for Biomembranes in Nanomedicine, University of Copenhagen, DK-2200 Copenhagen, Denmark
| | - Ulrik Gether
- Molecular Neuropharmacology Laboratory, Department of Neuroscience and Pharmacology, Faculty of Health Sciences, Panum Institute, and University of Copenhagen, DK-2200 Copenhagen, Denmark; Lundbeck Foundation Center for Biomembranes in Nanomedicine, University of Copenhagen, DK-2200 Copenhagen, Denmark.
| | - Trine Nygaard Jørgensen
- Molecular Neuropharmacology Laboratory, Department of Neuroscience and Pharmacology, Faculty of Health Sciences, Panum Institute, and University of Copenhagen, DK-2200 Copenhagen, Denmark
| |
Collapse
|
34
|
Heal DJ, Gosden J, Smith SL. Dopamine reuptake transporter (DAT) "inverse agonism"--a novel hypothesis to explain the enigmatic pharmacology of cocaine. Neuropharmacology 2014; 87:19-40. [PMID: 24953830 DOI: 10.1016/j.neuropharm.2014.06.012] [Citation(s) in RCA: 34] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/19/2014] [Revised: 05/07/2014] [Accepted: 06/09/2014] [Indexed: 12/20/2022]
Abstract
The long held view is cocaine's pharmacological effects are mediated by monoamine reuptake inhibition. However, drugs with rapid brain penetration like sibutramine, bupropion, mazindol and tesofensine, which are equal to or more potent than cocaine as dopamine reuptake inhibitors, produce no discernable subjective effects such as drug "highs" or euphoria in drug-experienced human volunteers. Moreover they are dysphoric and aversive when given at high doses. In vivo experiments in animals demonstrate that cocaine's monoaminergic pharmacology is profoundly different from that of other prescribed monoamine reuptake inhibitors, with the exception of methylphenidate. These findings led us to conclude that the highly unusual stimulant profile of cocaine and related compounds, eg methylphenidate, is not mediated by monoamine reuptake inhibition alone. We describe the experimental findings which suggest cocaine serves as a negative allosteric modulator to alter the function of the dopamine reuptake transporter (DAT) and reverse its direction of transport. This results in a firing-dependent, retro-transport of dopamine into the synaptic cleft. The proposed mechanism of cocaine is, therefore, different from other small molecule negative allostereric modulators of the monoamine reuptake transporters, eg SoRI-6238, which merely reduce the rate of inward transport. Because the physiological role of DAT is to remove dopamine from the synapse and the action of cocaine is the opposite of this, we have postulated that cocaine's effect is analogous to an inverse agonist. If this hypothesis is validated then cocaine is the prototypical compound that exemplifies a new class of monoaminergic drugs; DAT "inverse agonists". This article is part of the Special Issue entitled 'CNS Stimulants'.
Collapse
Affiliation(s)
- David J Heal
- RenaSci Limited, BioCity, Pennyfoot Street, Nottingham NG1 1GF, UK.
| | - Jane Gosden
- RenaSci Limited, BioCity, Pennyfoot Street, Nottingham NG1 1GF, UK
| | - Sharon L Smith
- RenaSci Limited, BioCity, Pennyfoot Street, Nottingham NG1 1GF, UK.
| |
Collapse
|
35
|
Kumar V, Rahbek-Clemmensen T, Billesbølle CB, Jorgensen TN, Gether U, Newman AH. Novel and high affinity fluorescent ligands for the serotonin transporter based on (s)-citalopram. ACS Med Chem Lett 2014; 5:696-9. [PMID: 24944746 DOI: 10.1021/ml5000806] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/21/2014] [Accepted: 03/21/2014] [Indexed: 01/06/2023] Open
Abstract
Novel rhodamine-labeled ligands, based on (S)-citalopram, were synthesized and evaluated for uptake inhibition at the human serotonin, dopamine, and norepinephrine transporters (hSERT, hDAT, and hNET, respectively) and for binding at SERT, in transiently transfected COS7 cells. Compound 14 demonstrated high affinity binding and selectivity for SERT (K i = 3 nM). Visualization of SERT, using confocal laser scanning microscopy, validated compound 14 as a novel tool for studying SERT expression and distribution in living cells.
Collapse
Affiliation(s)
- Vivek Kumar
- Medicinal Chemistry Section, Molecular
Targets and Medications Discovery Branch, National Institute on Drug
Abuse-Intramural Research Program, National Institutes of Health, 333 Cassell Drive, Baltimore, Maryland 21224, United States
| | - Troels Rahbek-Clemmensen
- Molecular Neuropharmacology Laboratory and Center for
Pharmacogenomics, Department of Neuroscience and Pharmacology, The
Faculty of Health and Medical Sciences, University of Copenhagen, DK-2200 Copenhagen N, Denmark
| | - Christian B. Billesbølle
- Molecular Neuropharmacology Laboratory and Center for
Pharmacogenomics, Department of Neuroscience and Pharmacology, The
Faculty of Health and Medical Sciences, University of Copenhagen, DK-2200 Copenhagen N, Denmark
| | - Trine Nygaard Jorgensen
- Molecular Neuropharmacology Laboratory and Center for
Pharmacogenomics, Department of Neuroscience and Pharmacology, The
Faculty of Health and Medical Sciences, University of Copenhagen, DK-2200 Copenhagen N, Denmark
| | - Ulrik Gether
- Molecular Neuropharmacology Laboratory and Center for
Pharmacogenomics, Department of Neuroscience and Pharmacology, The
Faculty of Health and Medical Sciences, University of Copenhagen, DK-2200 Copenhagen N, Denmark
| | - Amy Hauck Newman
- Medicinal Chemistry Section, Molecular
Targets and Medications Discovery Branch, National Institute on Drug
Abuse-Intramural Research Program, National Institutes of Health, 333 Cassell Drive, Baltimore, Maryland 21224, United States
| |
Collapse
|
36
|
Endosome-to-Plasma Membrane Recycling of VEGFR2 Receptor Tyrosine Kinase Regulates Endothelial Function and Blood Vessel Formation. Cells 2014; 3:363-85. [PMID: 24785348 PMCID: PMC4092869 DOI: 10.3390/cells3020363] [Citation(s) in RCA: 47] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/02/2013] [Revised: 03/04/2014] [Accepted: 03/17/2014] [Indexed: 12/13/2022] Open
Abstract
Rab GTPases are implicated in endosome-to-plasma membrane recycling, but how such membrane traffic regulators control vascular endothelial growth factor receptor 2 (VEGFR2/KDR) dynamics and function are not well understood. Here, we evaluated two different recycling Rab GTPases, Rab4a and Rab11a, in regulating endothelial VEGFR2 trafficking and signalling with implications for endothelial cell migration, proliferation and angiogenesis. In primary endothelial cells, VEGFR2 displays co-localisation with Rab4a, but not Rab11a GTPase, on early endosomes. Expression of a guanosine diphosphate (GDP)-bound Rab4a S22N mutant caused increased VEGFR2 accumulation in endosomes. TfR and VEGFR2 exhibited differences in endosome-to-plasma membrane recycling in the presence of chloroquine. Depletion of Rab4a, but not Rab11a, levels stimulated VEGF-A-dependent intracellular signalling. However, depletion of either Rab4a or Rab11a levels inhibited VEGF-A-stimulated endothelial cell migration. Interestingly, depletion of Rab4a levels stimulated VEGF-A-regulated endothelial cell proliferation. Rab4a and Rab11a were also both required for endothelial tubulogenesis. Evaluation of a transgenic zebrafish model showed that both Rab4 and Rab11a are functionally required for blood vessel formation and animal viability. Rab-dependent endosome-to-plasma membrane recycling of VEGFR2 is important for intracellular signalling, cell migration and proliferation during angiogenesis.
Collapse
|
37
|
Kantamneni S, Gonzàlez-Gonzàlez IM, Luo J, Cimarosti H, Jacobs SC, Jaafari N, Henley JM. Differential regulation of GABAB receptor trafficking by different modes of N-methyl-D-aspartate (NMDA) receptor signaling. J Biol Chem 2014; 289:6681-6694. [PMID: 24425870 PMCID: PMC3945329 DOI: 10.1074/jbc.m113.487348] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/01/2013] [Revised: 12/24/2013] [Indexed: 12/31/2022] Open
Abstract
Inhibitory GABAB receptors (GABABRs) can down-regulate most excitatory synapses in the CNS by reducing postsynaptic excitability. Functional GABABRs are heterodimers of GABAB1 and GABAB2 subunits and here we show that the trafficking and surface expression of GABABRs is differentially regulated by synaptic or pathophysiological activation of NMDA receptors (NMDARs). Activation of synaptic NMDARs using a chemLTP protocol increases GABABR recycling and surface expression. In contrast, excitotoxic global activation of synaptic and extrasynaptic NMDARs by bath application of NMDA causes the loss of surface GABABRs. Intriguingly, exposing neurons to extreme metabolic stress using oxygen/glucose deprivation (OGD) increases GABAB1 but decreases GABAB2 surface expression. The increase in surface GABAB1 involves enhanced recycling and is blocked by the NMDAR antagonist AP5. The decrease in surface GABAB2 is also blocked by AP5 and by inhibiting degradation pathways. These results indicate that NMDAR activity is critical in GABABR trafficking and function and that the individual subunits can be separately controlled to regulate neuronal responsiveness and survival.
Collapse
Affiliation(s)
- Sriharsha Kantamneni
- School of Biochemistry, Medical Sciences Building, University of Bristol, Bristol BS8 1TD, United Kingdom.
| | | | - Jia Luo
- School of Biochemistry, Medical Sciences Building, University of Bristol, Bristol BS8 1TD, United Kingdom
| | - Helena Cimarosti
- School of Biochemistry, Medical Sciences Building, University of Bristol, Bristol BS8 1TD, United Kingdom
| | - Susan C Jacobs
- School of Biochemistry, Medical Sciences Building, University of Bristol, Bristol BS8 1TD, United Kingdom
| | - Nadia Jaafari
- School of Biochemistry, Medical Sciences Building, University of Bristol, Bristol BS8 1TD, United Kingdom
| | - Jeremy M Henley
- School of Biochemistry, Medical Sciences Building, University of Bristol, Bristol BS8 1TD, United Kingdom.
| |
Collapse
|
38
|
Kucheryavykh LY, Rolón-Reyes K, Kucheryavykh YV, Skatchkov S, Eaton MJ, Sanabria P, Wessinger WD, Inyushin M. Glioblastoma development in mouse brain: general reduction of OCTs and mislocalization of OCT3 transporter and subsequent uptake of ASP + substrate to the nuclei. ACTA ACUST UNITED AC 2014; 3:3-9. [PMID: 25165637 DOI: 10.1166/jnsne.2014.1091] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/01/2023]
Abstract
Organic cation transporters (OCTs) were first found and then isolated from cultured glioma cells. When glioma cells are implanted into brain the fate of OCTs varies with time after implantation and transporter type. Here we report that OCT1, OCT2 and OCT3 immunofluorescence is significantly reduced over time in implanted GL261 glioma cells, during tumor development in the brain. By day 21 after glioma implantation, OCT1, OCT2 and OCT3 immunofluorescence was reduced more than 10-fold in the cytoplasm of glioma cells, while OCT3 immunofluorescence became concentrated in the nucleus. The well-known fluorescent substrate for OCT transporters, 4-(4-(dimethylamino)-styryl)-N-methylpyridinium iodide (ASP+), previously shown to accumulate in glioma-cell cytoplasm in in vivo slices, began to accumulate in the nucleus of these cells, but not in cytoplasm, after 21 days post-implantation. Considering this mislocalization phenomenon, and other literature on similar nuclear mislocalization of different transporters, receptors and channels in glioma cells, we suggest that it is one of the "omens" preceding the motility and aggressivity changes in glioma behavior.
Collapse
Affiliation(s)
| | | | | | - Serguei Skatchkov
- Dept. of Biochemistry and Physiology, Universidad Central del Caribe, Bayamon, PR 00960, USA
| | - Misty J Eaton
- Dept. of Biochemistry, Universidad Central del Caribe, Bayamon, PR 00960, USA
| | - Priscila Sanabria
- Dept. of Physiology, Universidad Central del Caribe, Bayamon, PR 00960, USA
| | - William D Wessinger
- Dept. of Pharmacology and Toxicology, University of Arkansas for Medical Sciences, Little Rock, AR 72205, USA
| | - Mikhail Inyushin
- Dept. of Physiology, Universidad Central del Caribe, Bayamon, PR 00960, USA
| |
Collapse
|
39
|
Abstract
Urea and urea transporters (UT) are critical to the production of concentrated urine and hence in maintaining body fluid balance. The UT-A1 urea transporter is the major and most important UT isoform in the kidney. Native UT-A1, expressed in the terminal inner medullary collecting duct (IMCD) epithelial cells, is a glycosylated protein with two glycoforms of 117 and 97 kDa. Vasopressin is the major hormone in vivo that rapidly increases urea permeability in the IMCD through increases in phosphorylation and apical plasma-membrane accumulation of UT-A1. The cell signaling pathway for vasopressin-mediated UT-A1 phosphorylation and activity involves two cAMP-dependent signaling pathways: protein kinase A (PKA) and exchange protein activated by cAMP (Epac). In this chapter, we will discuss UT-A1 regulation by phosphorylation, ubiquitination, and glycosylation.
Collapse
Affiliation(s)
- Guangping Chen
- Department of Physiology, and Renal Division Department of Medicine, Emory University School of Medicine, Whitehead Research Building Room 605N, 615 Michael Street, Atlanta, GA, 30322, USA,
| |
Collapse
|
40
|
Dopamine transporter endocytic trafficking in striatal dopaminergic neurons: differential dependence on dynamin and the actin cytoskeleton. J Neurosci 2013; 33:17836-46. [PMID: 24198373 DOI: 10.1523/jneurosci.3284-13.2013] [Citation(s) in RCA: 58] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022] Open
Abstract
Dopaminergic signaling profoundly impacts rewarding behaviors, movement, and executive function. The presynaptic dopamine (DA) transporter (DAT) recaptures released DA, thereby limiting synaptic DA availability and maintaining dopaminergic tone. DAT constitutively internalizes and PKC activation rapidly accelerates DAT endocytosis, resulting in DAT surface loss. Longstanding evidence supports PKC-stimulated DAT trafficking in heterologous expression studies. However, PKC-stimulated DAT internalization is not readily observed in cultured dopaminergic neurons. Moreover, conflicting reports implicate both classic and nonclassic endocytic mechanisms mediating DAT trafficking. Prior DAT trafficking studies relied primarily upon chronic gene disruption and dominant-negative protein expression, or were performed in cell lines and cultured neurons, yielding results difficult to translate to adult dopaminergic neurons. Here, we use newly described dynamin inhibitors to test whether constitutive and PKC-stimulated DAT internalization are dynamin-dependent in adult dopaminergic neurons. Ex vivo biotinylation studies in mouse striatal slices demonstrate that acute PKC activation drives native DAT surface loss, and that surface DAT surprisingly partitions between endocytic-willing and endocytic-resistant populations. Acute dynamin inhibition reveals that constitutive DAT internalization is dynamin-independent, whereas PKC-stimulated DAT internalization is dynamin-dependent. Moreover, total internal reflection fluorescence microscopy experiments demonstrate that constitutive DAT internalization occurs equivalently from lipid raft and nonraft microdomains, whereas PKC-stimulated DAT internalization arises exclusively from lipid rafts. Finally, DAT endocytic recycling relies on a dynamin-dependent mechanism that acts in concert with the actin cytoskeleton. These studies are the first comprehensive investigation of native DAT trafficking in ex vivo adult neurons, and reveal that DAT surface dynamics are governed by complex multimodal mechanisms.
Collapse
|
41
|
Su H, Chen M, Sands JM, Chen G. Activation of the cAMP/PKA pathway induces UT-A1 urea transporter monoubiquitination and targets it for lysosomal degradation. Am J Physiol Renal Physiol 2013; 305:F1775-82. [PMID: 24133116 PMCID: PMC3882448 DOI: 10.1152/ajprenal.00393.2013] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/08/2013] [Accepted: 10/16/2013] [Indexed: 11/22/2022] Open
Abstract
Regulation of urea transporter UT-A1 in the kidney is important for the urinary concentrating mechanism. We previously reported that activation of the cAMP/PKA pathway by forskolin (FSK) leads to UT-A1 ubiquitination, endocytosis, and degradation. In this study, we discovered that FSK-induced UT-A1 ubiquitination is monoubiquitination as judged by immunoblotting with specific ubiquitin antibodies to the different linkages of the ubiquitin chain. UT-A1 monoubiquitination induced by FSK was processed mainly on the cell plasma membrane. Monoubiquitination facilitates UT-A1 endocytosis, and internalized UT-A1 is accumulated in the early endosome. Inhibition of ubiquitination by E1 ubiquitin-activating enzyme inhibitor PYR-41 significantly reduced FSK-induced UT-A1 endocytosis and degradation. Interestingly, FSK-stimulated UT-A1 degradation occurs through a lysosomal protein degradation system. We further found that the PKA phosphorylation sites of UT-A1 at Ser486 and Ser499 are required for FSK-induced UT-A1 monoubiquitination. The physiological significance was confirmed using rat kidney inner medullary collecting duct suspensions, which showed that vasopressin treatment promotes UT-A1 ubiquitination. We conclude that unlike under basal conditions in which UT-A1 is subject to polyubiquitination and proteasome-mediated protein degradation, activation of UT-A1 by FSK induces UT-A1 monoubiquitination and protein lysosomal degradation.
Collapse
Affiliation(s)
- Hua Su
- Dept. of Physiology, Emory Univ. School of Medicine, Whitehead Research Bldg. Rm. 615, 615 Michael St., Atlanta, GA 30322.
| | | | | | | |
Collapse
|
42
|
Rickhag M, Hansen FH, Sørensen G, Strandfelt KN, Andresen B, Gotfryd K, Madsen KL, Vestergaard-Klewe I, Ammendrup-Johnsen I, Eriksen J, Newman AH, Füchtbauer EM, Gomeza J, Woldbye DPD, Wörtwein G, Gether U. A C-terminal PDZ domain-binding sequence is required for striatal distribution of the dopamine transporter. Nat Commun 2013; 4:1580. [PMID: 23481388 PMCID: PMC3646413 DOI: 10.1038/ncomms2568] [Citation(s) in RCA: 38] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/06/2012] [Accepted: 01/31/2013] [Indexed: 01/27/2023] Open
Abstract
The dopamine transporter (DAT) mediates reuptake of dopamine from the synaptic cleft. The cellular mechanisms controlling DAT levels in striatal nerve terminals remain poorly understood. DAT contains a C-terminal PDZ (PSD-95/Discs-large/ZO-1) domain binding sequence believed to bind synaptic scaffolding proteins, but its functional significance is uncertain. Here we demonstrate that two different DAT knock-in mice with disrupted PDZ-binding motifs (DAT-AAA and DAT+Ala) are characterized by dramatic loss of DAT expression in the striatum, causing hyperlocomotion and attenuated response to amphetamine. In cultured dopaminergic neurons and striatal slices from DAT-AAA mice, we find markedly reduced DAT surface levels and evidence for enhanced constitutive internalization. In DAT-AAA neurons, but not in wild type neurons, surface levels are rescued in part by expression of a dominant-negative dynamin mutation (K44A). Our findings suggest that PDZ domain interactions are critical for synaptic distribution of DAT in vivo and thereby for proper maintenance of dopamine homeostasis.
Collapse
Affiliation(s)
- Mattias Rickhag
- Molecular Neuropharmacology Laboratory, Lundbeck Foundation Center for Biomembranes in Nanomedicine, Department of Neuroscience and Pharmacology, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
43
|
Hong WC, Amara SG. Differential targeting of the dopamine transporter to recycling or degradative pathways during amphetamine- or PKC-regulated endocytosis in dopamine neurons. FASEB J 2013; 27:2995-3007. [PMID: 23612789 DOI: 10.1096/fj.12-218727] [Citation(s) in RCA: 50] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/11/2022]
Abstract
The dopamine transporter (DAT) clears the extracellular dopamine released during neurotransmission and is a major target for both therapeutic and addictive psychostimulant amphetamines. Amphetamine exposure or activation of protein kinase C (PKC) by the phorbol ester PMA has been shown to down-regulate cell surface DAT. However, in dopamine neurons, the trafficking itinerary and fate of internalized DAT has not been elucidated. By monitoring surface-labeled DAT in transfected dopamine neurons from embryonic rat mesencephalic cultures, we find distinct sorting and fates of internalized DAT after amphetamine or PMA treatment. Although both drugs promote DAT internalization above constitutive endocytosis in dopamine neurons, PMA induces ubiquitination of DAT and leads to accumulation of DAT on LAMP1-positive endosomes. In contrast, after amphetamine exposure DAT is sorted to recycling endosomes positive for Rab11 and the transferrin receptor. Furthermore, quantitative assessment of DAT recycling using an antibody-feeding assay reveals that significantly less DAT returns to the surface of dopamine neurons after internalization by PMA, compared with vehicle or amphetamine treatment. These results demonstrate that, in neurons, the DAT is sorted differentially to recycling and degradative pathways after psychostimulant exposure or PKC activation, which may allow for either the transient or sustained inhibition of DAT during dopamine neurotransmission.
Collapse
Affiliation(s)
- Weimin C Hong
- Department of Neurobiology, University of Pittsburgh, Pittsburgh, Pennsylvania, USA
| | | |
Collapse
|
44
|
de Juan-Sanz J, Núñez E, López-Corcuera B, Aragón C. Constitutive endocytosis and turnover of the neuronal glycine transporter GlyT2 is dependent on ubiquitination of a C-terminal lysine cluster. PLoS One 2013; 8:e58863. [PMID: 23484054 PMCID: PMC3590132 DOI: 10.1371/journal.pone.0058863] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/31/2012] [Accepted: 02/07/2013] [Indexed: 12/13/2022] Open
Abstract
Inhibitory glycinergic neurotransmission is terminated by sodium and chloride-dependent plasma membrane glycine transporters (GlyTs). The mainly glial glycine transporter GlyT1 is primarily responsible for the completion of inhibitory neurotransmission and the neuronal glycine transporter GlyT2 mediates the reuptake of the neurotransmitter that is used to refill synaptic vesicles in the terminal, a fundamental role in the physiology and pathology of glycinergic neurotransmission. Indeed, inhibitory glycinergic neurotransmission is modulated by the exocytosis and endocytosis of GlyT2. We previously reported that constitutive and Protein Kinase C (PKC)-regulated endocytosis of GlyT2 is mediated by clathrin and that PKC accelerates GlyT2 endocytosis by increasing its ubiquitination. However, the role of ubiquitination in the constitutive endocytosis and turnover of this protein remains unexplored. Here, we show that ubiquitination of a C-terminus four lysine cluster of GlyT2 is required for constitutive endocytosis, sorting into the slow recycling pathway and turnover of the transporter. Ubiquitination negatively modulates the turnover of GlyT2, such that increased ubiquitination driven by PKC activation accelerates transporter degradation rate shortening its half-life while decreased ubiquitination increases transporter stability. Finally, ubiquitination of GlyT2 in neurons is highly responsive to the free pool of ubiquitin, suggesting that the deubiquitinating enzyme (DUB) ubiquitin C-terminal hydrolase-L1 (UCHL1), as the major regulator of neuronal ubiquitin homeostasis, indirectly modulates the turnover of GlyT2. Our results contribute to the elucidation of the mechanisms underlying the dynamic trafficking of this important neuronal protein which has pathological relevance since mutations in the GlyT2 gene (SLC6A5) are the second most common cause of human hyperekplexia.
Collapse
Affiliation(s)
- Jaime de Juan-Sanz
- Centro de Biología Molecular ‘‘Severo Ochoa’’, Universidad Autónoma de Madrid, Consejo Superior de Investigaciones Científicas, Madrid, Spain
- Centro de Investigación Biomédica en Red de Enfermedades Raras (CIBERER), ISCIII, Madrid, Spain
- IdiPAZ-Hospital Universitario La Paz, Madrid, Spain
| | - Enrique Núñez
- Centro de Biología Molecular ‘‘Severo Ochoa’’, Universidad Autónoma de Madrid, Consejo Superior de Investigaciones Científicas, Madrid, Spain
- Centro de Investigación Biomédica en Red de Enfermedades Raras (CIBERER), ISCIII, Madrid, Spain
- IdiPAZ-Hospital Universitario La Paz, Madrid, Spain
| | - Beatriz López-Corcuera
- Centro de Biología Molecular ‘‘Severo Ochoa’’, Universidad Autónoma de Madrid, Consejo Superior de Investigaciones Científicas, Madrid, Spain
- Centro de Investigación Biomédica en Red de Enfermedades Raras (CIBERER), ISCIII, Madrid, Spain
- IdiPAZ-Hospital Universitario La Paz, Madrid, Spain
| | - Carmen Aragón
- Centro de Biología Molecular ‘‘Severo Ochoa’’, Universidad Autónoma de Madrid, Consejo Superior de Investigaciones Científicas, Madrid, Spain
- Centro de Investigación Biomédica en Red de Enfermedades Raras (CIBERER), ISCIII, Madrid, Spain
- IdiPAZ-Hospital Universitario La Paz, Madrid, Spain
- * E-mail:
| |
Collapse
|
45
|
Zhang P, Jørgensen TN, Loland CJ, Newman AH. A rhodamine-labeled citalopram analogue as a high-affinity fluorescent probe for the serotonin transporter. Bioorg Med Chem Lett 2012; 23:323-6. [PMID: 23168018 DOI: 10.1016/j.bmcl.2012.10.089] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/07/2012] [Revised: 10/15/2012] [Accepted: 10/19/2012] [Indexed: 11/27/2022]
Abstract
A novel fluorescent ligand was synthesized as a high-affinity, high specificity probe for visualizing the serotonin transporter (SERT). The rhodamine fluorophore was extended from an aniline substitution on the 5-position of the dihydroisobenzofuran ring of citalopram (2, 1-(3-(dimethylamino)propyl)-1-(4-fluorophenyl)-1,3-dihydroisobenzofuran-5-carbonitrile), using an ethylamino linker. The resulting rhodamine-labeled ligand 8 inhibited [(3)H]5-HT uptake in COS-7 cells (K(i)=225 nM) with similar potency to the tropane-based JHC 1-064 (1), but with higher specificity towards the SERT relative to the transporters for dopamine and norepinephrine. Visualization of the SERT with compound 8 was demonstrated by confocal microscopy in HEK293 cells stably expressing EGFP-SERT.
Collapse
Affiliation(s)
- Peng Zhang
- Molecular Targets and Medications Discovery Branch, Medicinal Chemistry Section, National Institute on Drug Abuse-Intramural Research Program, National Institutes of Health, 333 Cassell Drive, Baltimore, MD 21224, USA
| | | | | | | |
Collapse
|
46
|
Su H, Carter CB, Laur O, Sands JM, Chen G. Forskolin stimulation promotes urea transporter UT-A1 ubiquitination, endocytosis, and degradation in MDCK cells. Am J Physiol Renal Physiol 2012; 303:F1325-32. [PMID: 22914781 PMCID: PMC3518190 DOI: 10.1152/ajprenal.00248.2012] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/01/2012] [Accepted: 08/21/2012] [Indexed: 11/22/2022] Open
Abstract
The adenylyl cyclase stimulator forskolin (FSK) stimulates UT-A1 phosphorylation, membrane trafficking, and urea transport activity. Here, we found that FSK stimulation induces UT-A1 ubiquitination in UT-A1 Madin-Darby canine kidney (MDCK) cells. This suggests that phosphorylation by FSK also triggers the protein degradation machinery for UT-A1. UT-A1-MDCK cells were treated with 100 μg/ml cycloheximide to inhibit protein synthesis, with or without 10 μM FSK. Total UT-A1 protein abundance was significantly reduced after FSK treatment, concomitantly ubiquitinated UT-A1 was increased. We then specifically investigated the effect of FSK on UT-A1 expressed on the cell plasma membrane. FSK treatment accelerated UT-A1 removal from the cell plasma membrane by increasing UT-A1 endocytosis as judged by biotinylation/MesNa treatment and confocal microscopy. We further found that inhibition of the clathrin-mediated endocytic pathway, but not the caveolin-mediated endocytic pathway, significantly blocks FSK-stimulated UT-A1 endocytosis. The PKA inhibitor H89 and the proteasome inhibitors MG132 and lactacystin reduced FSK-induced membrane UT-A1 reduction. Our study shows that FSK activates the UT-A1 urea transporter and the activation/phosphorylation subsequently triggers the downregulation of UT-A1, which represents an important mechanism for the cell to return to the basal conditions after vasopressin stimulation.
Collapse
Affiliation(s)
- Hua Su
- Department of Medicine, Renal Division, Emory University School of Medicine, Atlanta, GA 30322, USA
| | | | | | | | | |
Collapse
|
47
|
Keith DJ, Wolfrum K, Eshleman AJ, Janowsky A. Melittin initiates dopamine transporter internalization and recycling in transfected HEK-293 cells. Eur J Pharmacol 2012; 690:13-21. [PMID: 22683840 DOI: 10.1016/j.ejphar.2012.05.020] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/09/2012] [Revised: 05/02/2012] [Accepted: 05/15/2012] [Indexed: 12/26/2022]
Abstract
The dopamine transporter removes the neurotransmitter from the synapse, regulating dopamine availability. The transporter can be internalized and its function is blocked by cocaine and other ligands. Melittin inhibits dopamine transporter function and causes internalization of the recombinant transporter in stably transfected HEK-293 cells, but the specific pathways for internalization and disposition of the transporter are unknown. Here we report that melittin treatment increased both transporter internalization and colocalization with clathrin, effects that were blocked by pretreatment with cocaine. Density gradient centrifugation revealed that melittin treatment caused the dopamine transporter to associate with a density fraction containing the early endosome marker Rab 5A. Confocal microscopy revealed that melittin treatment also increased transporter colocalization with Rab 5A and decreased colocalization with the late endosome marker Rab 7 and the recycling endosome marker Rab 11. Following 60 min of melittin treatment, the transporter was trafficked back to the membrane. By comparison, phorbol ester treatment increased transporter colocalization with early endosome antigen 1 and Rab 7 in a time-dependent manner. Cocaine treatment alone does not affect transporter trafficking in these cells. Results indicate multiple dopamine transporter internalization and recycling pathways that depend on transporter-ligand interactions and post-translational modifications.
Collapse
Affiliation(s)
- Dove J Keith
- Graduate Program in Neuroscience, VA Medical Center, Portland, OR 97239, USA
| | | | | | | |
Collapse
|
48
|
Martínez-Villarreal J, García Tardón N, Ibáñez I, Giménez C, Zafra F. Cell surface turnover of the glutamate transporter GLT-1 is mediated by ubiquitination/deubiquitination. Glia 2012; 60:1356-65. [PMID: 22593014 DOI: 10.1002/glia.22354] [Citation(s) in RCA: 47] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/30/2012] [Accepted: 04/23/2012] [Indexed: 11/11/2022]
Abstract
The main glutamate transporter in the brain, GLT-1, mediates glutamatergic neurotransmission in both physiological and pathological conditions. GLT-1 activity is controlled by both constitutive and regulated trafficking, and although recent evidence indicates that the turnover of this protein in the plasma membrane is accelerated by protein kinase C via an ubiquitin-dependent process, the mechanisms driving the constitutive trafficking of GLT-1 remain unexplored. Here, we used a heterologous system and primary astrocytes to investigate the turnover of GLT-1 and the role of ubiquitin attachment in this process. We show that GLT-1 is endocytosed constitutively in a clathrin-dependent manner, recycling the transporter into endosomes containing EEA1 and Rab4, a marker of rapidly recycling endosomes, and not Rab11 or Rab7, markers of the slow recycling and late endosomal compartments, respectively. We also show that this process is dependent on ubiquitination, because the inhibitor of the ubiquitin-activating enzyme E1, 4[4-(5-nitro-furan-2-ylmethylene)-3,5-dioxo-pyrazolidin-1-yl]-benzoic acid ethyl ester, promotes the retention of GLT-1 at the plasma membrane. Moreover, site-directed mutagenesis demonstrated the involvement of lysines 517 and 526 of GLT-1 in the constitutive internalization of the transporter. The translocation of GLT-1 from the recycling endosomes to the plasma membrane was blocked by LDN-57444, a specific inhibitor to the deubiquitinating enzyme (DUB) ubiquitin C-terminal hydrolase-L1, but not by an inhibitor of the related DUB ubiquitin C-terminal hydrolase-L3, supporting the existence of specific ubiquitination/deubiquitination cycles that ensure the correct concentrations of GLT-1 at the cell surface.
Collapse
Affiliation(s)
- Jaime Martínez-Villarreal
- Centro de Biología Molecular Severo Ochoa, Facultad de Ciencias, Consejo Superior de Investigaciones Científicas, Universidad Autónoma de Madrid, Madrid, Spain
| | | | | | | | | |
Collapse
|
49
|
Madsen KL, Thorsen TS, Rahbek-Clemmensen T, Eriksen J, Gether U. Protein interacting with C kinase 1 (PICK1) reduces reinsertion rates of interaction partners sorted to Rab11-dependent slow recycling pathway. J Biol Chem 2012; 287:12293-308. [PMID: 22303009 DOI: 10.1074/jbc.m111.294702] [Citation(s) in RCA: 35] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022] Open
Abstract
The scaffolding protein PICK1 (protein interacting with C kinase 1) contains an N-terminal PSD-95/Discs large/ZO-1 (PDZ) domain and a central lipid-binding Bin/amphiphysin/Rvs (BAR) domain. PICK1 is thought to regulate trafficking of its PDZ binding partners but different and even opposing functions have been suggested. Here, we apply ELISA-based assays and confocal microscopy in HEK293 cells with inducible PICK1 expression to assess in an isolated system the ability of PICK1 to regulate trafficking of natural and engineered PDZ binding partners. The dopamine transporter (DAT), which primarily sorts to degradation upon internalization, did not form perinuclear clusters with PICK1, and PICK1 did not affect DAT internalization/recycling. However, transfer of the PICK1-binding DAT C terminus to the β(2)-adrenergic receptor, which sorts to recycling upon internalization, led to formation of PICK1 co-clusters in Rab11-positive compartments. Furthermore, PICK1 inhibited Rab11-mediated recycling of the receptor in a BAR and PDZ domain-dependent manner. In contrast, transfer of the DAT C terminus to the δ-opioid receptor, which sorts to degradation, did not result in PICK1 co-clusters or any change in internalization/recycling. Further support for a role of PICK1 determined by its PDZ cargo was obtained for the PICK1 interaction partner prolactin-releasing peptide receptor (GPR10). GPR10 co-localized with Rab11 and clustered with PICK1 upon constitutive internalization but co-localized with the late endosomal marker Rab7 and did not cluster with PICK1 upon agonist-induced internalization. Our data suggest a selective role of PICK1 in clustering and reducing the recycling rates of PDZ domain binding partners sorted to the Rab11-dependent recycling pathway.
Collapse
Affiliation(s)
- Kenneth L Madsen
- Molecular Neuropharmacology Laboratory and Lundbeck Foundation Center for Biomembranes in Nanomedicine, Department of Neuroscience and Pharmacology, University of Copenhagen, DK-2200 Copenhagen, Denmark
| | | | | | | | | |
Collapse
|
50
|
Rao A, Richards TL, Simmons D, Zahniser NR, Sorkin A. Epitope-tagged dopamine transporter knock-in mice reveal rapid endocytic trafficking and filopodia targeting of the transporter in dopaminergic axons. FASEB J 2012; 26:1921-33. [PMID: 22267337 DOI: 10.1096/fj.11-196113] [Citation(s) in RCA: 34] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
Abstract
The plasma membrane dopamine (DA) transporter (DAT) is essential for reuptake of extracellular DA. DAT function in heterologous cells is regulated by subcellular targeting, endocytosis, and intracellular trafficking, but the mechanisms regulating neuronal DAT remain poorly understood. Hence, we generated a knock-in mouse expressing a hemagglutinin (HA)-epitope-tagged DAT to study endogenous transporter trafficking. Introduction of the HA tag into the second extracellular loop of mouse DAT did not perturb its expression level, distribution pattern, or substrate uptake kinetics. Live-cell fluorescence microscopy imaging using fluorescently labeled HA-specific antibody and a quantitative HA-antibody endocytosis assay demonstrated that in axons HA-DAT was primarily located in the plasma membrane and internalized mostly in growth cones and varicosities, where synaptic vesicle markers were also concentrated. Formation of varicosities was frequently preceded or accompanied by highly dynamic filopodia-like membrane protrusions. Remarkably, HA-DAT often concentrated at the tips of these filopodia. This pool of HA-DATs exhibited low lateral membrane mobility. Thus, DAT-containing filopodia may be involved in synaptogenesis in developing DA neurons. Treatment of neurons with amphetamine increased mobility of filopodial HA-DAT and accelerated HA-DAT endocytosis in axons, suggesting that chronic amphetamine may interfere with DA synapse development. Interestingly, phorbol esters did not accelerate endocytosis of axonal DAT.
Collapse
Affiliation(s)
- Anjali Rao
- Department of Cell Biology and Physiology, University of Pittsburgh School of Medicine, S368 Biomedical Science Tower South, 3500 Terrace St., Pittsburgh, PA 15261, USA
| | | | | | | | | |
Collapse
|