1
|
Bagalkot T, Sorkin A. Amphetamine Induces Sex-Dependent Loss of the Striatal Dopamine Transporter in Sensitized Mice. eNeuro 2024; 11:ENEURO.0491-23.2023. [PMID: 38164591 PMCID: PMC10849026 DOI: 10.1523/eneuro.0491-23.2023] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/23/2023] [Revised: 12/10/2023] [Accepted: 12/12/2023] [Indexed: 01/03/2024] Open
Abstract
Dopamine transporter (DAT) controls dopamine signaling in the brain through the reuptake of synaptically released dopamine. DAT is a target of abused psychostimulants such as amphetamine (Amph). Acute Amph administration induces transient DAT endocytosis, which, among other Amph effects on dopaminergic neurons, elevates extracellular dopamine. However, the effects of repeated Amph abuse, leading to behavioral sensitization and drug addiction, on DAT are unknown. Hence, we developed a 14 d Amph-sensitization protocol in knock-in mice expressing HA-epitope-tagged DAT (HA-DAT) and investigated the effects of Amph challenge on sensitized HA-DAT animals. The Amph challenge resulted in the highest locomotor activity on Day 14 in both sexes, which was sustained for 1 h in male but not female mice. Strikingly, significant (by 30-60%) loss of the HA-DAT protein in the striatum was caused by the Amph challenge of sensitized males but not females. Amph also reduced V max of dopamine transport in the striatal synaptosomes of males without changing K m values. Consistently, immunofluorescence microscopy revealed a significant increase of HA-DAT colocalization with the endosomal protein VPS35 only in Amph-challenged males. Amph-induced loss of striatal HA-DAT in sensitized mice was blocked by chloroquine, vacuolin-1, and inhibitor of Rho-associated kinases ROCK1/2, indicative of the involvement of endocytic trafficking in the DAT protein loss. Interestingly, an apparent degradation of HA-DAT protein was observed in the nucleus accumbens and not in the dorsal striatum. We propose that Amph challenge in sensitized mice triggers Rho-mediated endocytosis and post-endocytic trafficking of DAT in a brain-region-specific and sex-dependent manner.
Collapse
Affiliation(s)
- Tarique Bagalkot
- Department of Cell Biology, University of Pittsburgh School of Medicine, Pittsburgh 15261, Pennsylvania
| | - Alexander Sorkin
- Department of Cell Biology, University of Pittsburgh School of Medicine, Pittsburgh 15261, Pennsylvania
| |
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
|
Bagalkot T, Sorkin A. Endocytic down-regulation of the striatal dopamine transporter by amphetamine in sensitized mice in sex-dependent manner. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2023:2023.05.17.541165. [PMID: 37293021 PMCID: PMC10245703 DOI: 10.1101/2023.05.17.541165] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/10/2023]
Abstract
Dopamine transporter (DAT) controls dopamine signaling in the brain through the reuptake of synaptically released dopamine. DAT is a target of abused psychostimulants such as amphetamine (Amph). Acute Amph is proposed to cause transient DAT endocytosis which among other Amph effects on dopaminergic neurons elevates extracellular dopamine. However, the effects of repeated Amph abuse, leading to behavioral sensitization and drug addiction, on DAT traffic are unknown. Hence, we developed a 14-day Amph-sensitization protocol in knock-in mice expressing HA-epitope tagged DAT (HA-DAT) and investigated effects of Amph challenge on HA-DAT in sensitized animals. Amph challenge resulted in the highest locomotor activity on day 14 in both sexes, which was however sustained for 1 hour in male but not female mice. Strikingly, significant (by 30-60%) reduction in the amount of the HA-DAT protein in striatum was observed in response to Amph challenge of sensitized males but not females. Amph reduced Vmax of dopamine transport in striatal synaptosomes of males without changing Km values. Consistently, immunofluorescence microscopy revealed a significant increase of HA-DAT co-localization with the endosomal protein VPS35 only in males. Amph-induced HA-DAT down-regulation in the striatum of sensitized mice was blocked by chloroquine, vacuolin-1 (inhibitor of PIKfive kinase), and inhibitor of Rho-associated kinases (ROCK1/2), indicative of the involvement of endocytic trafficking in DAT down-regulation. Interestingly, HA-DAT protein down-regulation was observed in nucleus accumbens and not in dorsal striatum. We propose that Amph challenge in sensitized mice leads to ROCK-dependent endocytosis and post-endocytic traffic of DAT in a brain-region-specific and sex-dependent manner.
Collapse
|
4
|
Saenz J, Yao O, Khezerlou E, Aggarwal M, Zhou X, Barker DJ, DiCicco-Bloom E, Pan PY. Cocaine-regulated trafficking of dopamine transporters in cultured neurons revealed by a pH sensitive reporter. iScience 2023; 26:105782. [PMID: 36594015 PMCID: PMC9804146 DOI: 10.1016/j.isci.2022.105782] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/18/2022] [Revised: 11/07/2022] [Accepted: 12/07/2022] [Indexed: 12/14/2022] Open
Abstract
Cocaine acts by inhibiting plasma membrane dopamine transporter (DAT) function and altering its surface expression. The precise manner and mechanism by which cocaine regulates DAT trafficking, especially at neuronal processes, are poorly understood. In this study, we engineered and validated the use of DAT-pHluorin for studying DAT localization and its dynamic trafficking at neuronal processes of cultured mouse midbrain neurons. We demonstrate that unlike neuronal soma and dendrites, which contain a majority of the DATs in weakly acidic intracellular compartments, axonal DATs at both shafts and boutons are primarily (75%) localized to the plasma membrane, whereas large varicosities contain abundant intracellular DAT within acidic intracellular structures. We also demonstrate that cocaine exposure leads to a Synaptojanin1-sensitive DAT internalization process followed by membrane reinsertion that lasts for days. Thus, our study reveals the previously unknown dynamics and molecular regulation for cocaine-regulated DAT trafficking in neuronal processes.
Collapse
Affiliation(s)
- Jacqueline Saenz
- Rutgers University Robert Wood Johnson Medical School, Department of Neuroscience and Cell Biology, 675 Hoes Lane West, Piscataway, NJ 08854, USA
- Rutgers Graduate School of Biomedical Sciences, Molecular Biosciences Graduate Program, Piscataway, NJ 08854, USA
| | - Oscar Yao
- Rutgers University Robert Wood Johnson Medical School, Department of Neuroscience and Cell Biology, 675 Hoes Lane West, Piscataway, NJ 08854, USA
| | - Elnaz Khezerlou
- Rutgers University Robert Wood Johnson Medical School, Department of Neuroscience and Cell Biology, 675 Hoes Lane West, Piscataway, NJ 08854, USA
| | - Meha Aggarwal
- Rutgers University Robert Wood Johnson Medical School, Department of Neuroscience and Cell Biology, 675 Hoes Lane West, Piscataway, NJ 08854, USA
| | - Xiaofeng Zhou
- Rutgers University Robert Wood Johnson Medical School, Department of Neuroscience and Cell Biology, 683 Hoes Lane West, Piscataway, NJ 08854, USA
| | - David J. Barker
- Rutgers, The State University of New Jersey, Department of Psychology, 152 Frelinghuysen Road, Piscataway, NJ 08854, USA
| | - Emanuel DiCicco-Bloom
- Rutgers University Robert Wood Johnson Medical School, Department of Neuroscience and Cell Biology, 683 Hoes Lane West, Piscataway, NJ 08854, USA
| | - Ping-Yue Pan
- Rutgers University Robert Wood Johnson Medical School, Department of Neuroscience and Cell Biology, 675 Hoes Lane West, Piscataway, NJ 08854, USA
| |
Collapse
|
5
|
Piniella D, Martínez-Blanco E, Bartolomé-Martín D, Sanz-Martos AB, Zafra F. Identification by proximity labeling of novel lipidic and proteinaceous potential partners of the dopamine transporter. Cell Mol Life Sci 2021; 78:7733-7756. [PMID: 34709416 PMCID: PMC8629785 DOI: 10.1007/s00018-021-03998-1] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/28/2021] [Revised: 10/07/2021] [Accepted: 10/17/2021] [Indexed: 12/05/2022]
Abstract
Dopamine (DA) transporters (DATs) are regulated by trafficking and modulatory processes that probably rely on stable and transient interactions with neighboring proteins and lipids. Using proximity-dependent biotin identification (BioID), we found novel potential partners for DAT, including several membrane proteins, such as the transmembrane chaperone 4F2hc, the proteolipid M6a and a potential membrane receptor for progesterone (PGRMC2). We also detected two cytoplasmic proteins: a component of the Cullin1-dependent ubiquitination machinery termed F-box/LRR-repeat protein 2 (FBXL2), and the enzyme inositol 5-phosphatase 2 (SHIP2). Immunoprecipitation (IP) and immunofluorescence studies confirmed either a physical association or a close spatial proximity between these proteins and DAT. M6a, SHIP2 and the Cullin1 system were shown to increase DAT activity in coexpression experiments, suggesting a functional role for their association. Deeper analysis revealed that M6a, which is enriched in neuronal protrusions (filopodia or dendritic spines), colocalized with DAT in these structures. In addition, the product of SHIP2 enzymatic activity (phosphatidylinositol 3,4-bisphosphate [PI(3,4)P2]) was tightly associated with DAT, as shown by co-IP and by colocalization of mCherry-DAT with a specific biosensor for this phospholipid. PI(3,4)P2 strongly stimulated transport activity in electrophysiological recordings, and conversely, inhibition of SHIP2 reduced DA uptake in several experimental systems including striatal synaptosomes and the dopaminergic cell line SH-SY5Y. In summary, here we report several potential new partners for DAT and a novel regulatory lipid, which may represent new pharmacological targets for DAT, a pivotal protein in dopaminergic function of the brain.
Collapse
Affiliation(s)
- Dolores Piniella
- Centro de Biología Molecular Severo Ochoa and Departamento de Biología Molecular, Facultad de Ciencias, Consejo Superior de Investigaciones Científicas and Universidad Autónoma de Madrid, C / Nicolás Cabrera 1, 28049, Madrid, Spain
- IdiPAZ, Instituto de Salud Carlos III, Madrid, Spain
| | - Elena Martínez-Blanco
- Centro de Biología Molecular Severo Ochoa and Departamento de Biología Molecular, Facultad de Ciencias, Consejo Superior de Investigaciones Científicas and Universidad Autónoma de Madrid, C / Nicolás Cabrera 1, 28049, Madrid, Spain
- IdiPAZ, Instituto de Salud Carlos III, Madrid, Spain
| | - David Bartolomé-Martín
- Centro de Biología Molecular Severo Ochoa and Departamento de Biología Molecular, Facultad de Ciencias, Consejo Superior de Investigaciones Científicas and Universidad Autónoma de Madrid, C / Nicolás Cabrera 1, 28049, Madrid, Spain
- IdiPAZ, Instituto de Salud Carlos III, Madrid, Spain
- Departamento de Bioquímica, Microbiología, Biología Celular y Genética, Universidad de La Laguna, Tenerife, Spain
| | - Ana B Sanz-Martos
- Departamento de Ciencias Farmacéuticas y de la Salud, Facultad de Farmacia, Universidad CEU-San Pablo, CEU Universities, 28925, Madrid, Spain
| | - Francisco Zafra
- Centro de Biología Molecular Severo Ochoa and Departamento de Biología Molecular, Facultad de Ciencias, Consejo Superior de Investigaciones Científicas and Universidad Autónoma de Madrid, C / Nicolás Cabrera 1, 28049, Madrid, Spain.
- IdiPAZ, Instituto de Salud Carlos III, Madrid, Spain.
| |
Collapse
|
6
|
Tomlinson ID, Kovtun O, Torres R, Bellocchio LG, Josephs T, Rosenthal SJ. A Novel Biotinylated Homotryptamine Derivative for Quantum Dot Imaging of Serotonin Transporter in Live Cells. Front Cell Neurosci 2021; 15:667044. [PMID: 34867196 PMCID: PMC8637195 DOI: 10.3389/fncel.2021.667044] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/11/2021] [Accepted: 10/19/2021] [Indexed: 11/21/2022] Open
Abstract
The serotonin transporter (SERT) is the primary target for selective serotonin reuptake inhibitor (SSRI) antidepressants that are thought to exert their therapeutic effects by increasing the synaptic concentration of serotonin. Consequently, probes that can be utilized to study cellular trafficking of SERT are valuable research tools. We have developed a novel ligand (IDT785) that is composed of a SERT antagonist (a tetrahydro pyridyl indole derivative) conjugated to a biotinylated poly ethylene glycol (PEG) via a phenethyl linker. This compound was determined to be biologically active and inhibited SERT-mediated reuptake of IDT307 with the half-maximal inhibitory concentration of 7.2 ± 0.3 μM. We demonstrated that IDT785 enabled quantum dot (QD) labeling of membrane SERT in transfected HEK-293 cultures that could be blocked using the high affinity serotonin reuptake inhibitor paroxetine. Molecular docking studies suggested that IDT785 might be binding to the extracellular vestibule binding site rather than the orthosteric substrate binding site, which could be attributable to the hydrophilicity of the PEG chain and the increased loss of degrees of freedom that would be required to penetrate into the orthosteric binding site. Using IDT785, we were able to study the membrane localization and membrane dynamics of YFP-SERT heterologously expressed in HEK-293 cells and demonstrated that SERT expression was enriched in the membrane edge and in thin cellular protrusions.
Collapse
Affiliation(s)
- Ian D. Tomlinson
- Department of Chemistry, Vanderbilt University, Nashville, TN, United States
| | - Oleg Kovtun
- Department of Chemistry, Vanderbilt University, Nashville, TN, United States
| | - Ruben Torres
- Department of Chemistry, Vanderbilt University, Nashville, TN, United States
- Vanderbilt Institute of Chemical Biology, Vanderbilt University, Nashville, TN, United States
| | | | - Travis Josephs
- Neuroscience Program, Vanderbilt University, Nashville, TN, United States
| | - Sandra J. Rosenthal
- Department of Chemistry, Vanderbilt University, Nashville, TN, United States
- Vanderbilt Institute of Chemical Biology, Vanderbilt University, Nashville, TN, United States
- Department of Pharmacology, Vanderbilt University, Nashville, TN, United States
- Department of Chemical and Biomolecular Engineering, Vanderbilt University, Nashville, TN, United States
- Department of Physics and Astronomy, Vanderbilt University, Nashville, TN, United States
- Vanderbilt Institute of Nanoscale Science and Engineering, Vanderbilt University, Nashville, TN, United States
| |
Collapse
|
7
|
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
|
8
|
Sorkina T, Cheng MH, Bagalkot TR, Wallace C, Watkins SC, Bahar I, Sorkin A. Direct coupling of oligomerization and oligomerization-driven endocytosis of the dopamine transporter to its conformational mechanics and activity. J Biol Chem 2021; 296:100430. [PMID: 33610553 PMCID: PMC8010718 DOI: 10.1016/j.jbc.2021.100430] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/10/2020] [Revised: 02/08/2021] [Accepted: 02/16/2021] [Indexed: 12/15/2022] Open
Abstract
Dopamine transporter (DAT) mediates the reuptake of synaptically released dopamine, and thus controls the duration and intensity of dopamine neurotransmission. Mammalian DAT has been observed to form oligomers, although the mechanisms of oligomerization and its role in DAT activity and trafficking remain largely unknown. We discovered a series of small molecule compounds that stabilize trimers and induce high-order oligomers of DAT and concomitantly promote its clathrin-independent endocytosis. Using a combination of chemical cross-linking, fluorescence resonance energy transfer microscopy, antibody-uptake endocytosis assay, live-cell lattice light sheet microscopy, ligand binding and substrate transport kinetics analyses, and molecular modeling and simulations, we investigated molecular basis of DAT oligomerization and endocytosis induced by these compounds. Our study showed that small molecule–induced DAT oligomerization and endocytosis are favored by the inward-facing DAT conformation and involve interactions of four hydrophobic residues at the interface between transmembrane (TM) helices TM4 and TM9. Surprisingly, a corresponding quadruple DAT mutant displays altered dopamine transport kinetics and increased cocaine-analog binding. The latter is shown to originate from an increased preference for outward-facing conformation and inward-to-outward transition. Taken together, our results demonstrate a direct coupling between conformational dynamics of DAT, functional activity of the transporter, and its oligomerization leading to endocytosis. The high specificity of such coupling for DAT makes the TM4-9 hub a new target for pharmacological modulation of DAT activity and subcellular localization.
Collapse
Affiliation(s)
- Tatiana Sorkina
- Department of Cell Biology, School of Medicine, University of Pittsburgh, Pittsburgh, Pennsylvania, USA
| | - Mary Hongying Cheng
- Department of Computational and Systems Biology, School of Medicine, University of Pittsburgh, Pittsburgh, Pennsylvania, USA
| | - Tarique R Bagalkot
- Department of Cell Biology, School of Medicine, University of Pittsburgh, Pittsburgh, Pennsylvania, USA
| | - Callen Wallace
- Department of Cell Biology, School of Medicine, University of Pittsburgh, Pittsburgh, Pennsylvania, USA
| | - Simon C Watkins
- Department of Cell Biology, School of Medicine, University of Pittsburgh, Pittsburgh, Pennsylvania, USA
| | - Ivet Bahar
- Department of Computational and Systems Biology, School of Medicine, University of Pittsburgh, Pittsburgh, Pennsylvania, USA
| | - Alexander Sorkin
- Department of Cell Biology, School of Medicine, University of Pittsburgh, Pittsburgh, Pennsylvania, USA.
| |
Collapse
|
9
|
Bagalkot TR, Block ER, Bucchin K, Balcita-Pedicino JJ, Calderon M, Sesack SR, Sorkin A. Dopamine Transporter Localization in Medial Forebrain Bundle Axons Indicates Its Long-Range Transport Primarily by Membrane Diffusion with a Limited Contribution of Vesicular Traffic on Retromer-Positive Compartments. J Neurosci 2021; 41:234-250. [PMID: 33234607 PMCID: PMC7810657 DOI: 10.1523/jneurosci.0744-20.2020] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/31/2020] [Revised: 11/05/2020] [Accepted: 11/13/2020] [Indexed: 12/18/2022] Open
Abstract
Dopamine transporter (DAT) controls dopamine neurotransmission by clearing synaptically released dopamine. However, trafficking itineraries of DAT, which determine its cell-surface concentration near synapses, are poorly characterized. It is especially unknown how DAT is transported between spatially distant midbrain somatodendritic and striatal axonal compartments. To examine this "long-range" trafficking, the localization and membrane diffusion of HA-epitope tagged DAT in the medial forebrain bundle (MFB) of a knock-in mouse (both sexes) were analyzed using confocal, super-resolution and EM in intact brain and acute brain slices. HA-DAT was abundant in the plasma membrane of MFB axons, similar to the striatum, although the intracellular fraction of HA-DAT in MFB was more substantial. Intracellular HA-DAT colocalized with VPS35, a subunit of the retromer complex mediating recycling from endosomes, in a subset of axons. Late endosomes, lysosomes, and endoplasmic reticulum were abundant in the soma but minimally present in MFB axons, suggesting that biosynthesis and lysosomal degradation of DAT are confined to soma. Together, the data suggest that membrane diffusion is the main mode of long-range DAT transport through MFB, although the contribution of vesicular traffic can be significant in a population of MFB axons. Based on HA-DAT diffusion rates, plasma membrane DAT in MFB axons turns over with a halftime of ∼20 d, which explains the extremely slow turnover of DAT protein in the brain. Unexpectedly, the mean diameter of DAT-labeled MFB axons was observed to be twice larger than reported for striatum. The implications of this finding for dopamine neuron physiology are discussed.SIGNIFICANCE STATEMENT The dopamine transporter (DAT) is a key regulator of dopamine neurotransmission and a target of abused psychostimulants. In the present study, we examined, for the first time, mechanisms of the long-range traffic of DAT in intact brain and acute brain slices from the knock-in mouse expressing epitope-tagged DAT. Using a combination of confocal, super-resolution and EM, we defined DAT localization and its membrane diffusion parameters in medial forebrain bundle axonal tracts connecting midbrain somatodendritic and striatal axonal compartments of dopaminergic neurons. In contrast to the widely accepted model of long-range axonal transport, our studies suggest that DAT traffics between midbrain and striatum, mainly by lateral diffusion in the plasma membrane with only a limited contribution of vesicular transport in recycling endosomes.
Collapse
Affiliation(s)
- Tarique R Bagalkot
- Department of Cell Biology, University of Pittsburgh School of Medicine, Pittsburgh, Pennsylvania 15261
| | - Ethan R Block
- Department of Cell Biology, University of Pittsburgh School of Medicine, Pittsburgh, Pennsylvania 15261
- Chatham University, Pittsburgh, Pennsylvania 15232
| | - Kristen Bucchin
- Department of Neuroscience, Dietrich School of Arts and Sciences, University of Pittsburgh, Pittsburgh, Pennsylvania 15213
| | - Judith Joyce Balcita-Pedicino
- Department of Neuroscience, Dietrich School of Arts and Sciences, University of Pittsburgh, Pittsburgh, Pennsylvania 15213
| | - Michael Calderon
- Department of Cell Biology, University of Pittsburgh School of Medicine, Pittsburgh, Pennsylvania 15261
| | - Susan R Sesack
- Department of Neuroscience, Dietrich School of Arts and Sciences, University of Pittsburgh, Pittsburgh, Pennsylvania 15213
| | - Alexander Sorkin
- Department of Cell Biology, University of Pittsburgh School of Medicine, Pittsburgh, Pennsylvania 15261
| |
Collapse
|
10
|
Baena V, Owen CM, Uliasz TF, Lowther KM, Yee SP, Terasaki M, Egbert JR, Jaffe LA. Cellular Heterogeneity of the Luteinizing Hormone Receptor and Its Significance for Cyclic GMP Signaling in Mouse Preovulatory Follicles. Endocrinology 2020; 161:5834711. [PMID: 32384146 PMCID: PMC7574965 DOI: 10.1210/endocr/bqaa074] [Citation(s) in RCA: 19] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/07/2020] [Accepted: 05/02/2020] [Indexed: 12/14/2022]
Abstract
Meiotic arrest and resumption in mammalian oocytes are regulated by 2 opposing signaling proteins in the cells of the surrounding follicle: the guanylyl cyclase natriuretic peptide receptor 2 (NPR2), and the luteinizing hormone receptor (LHR). NPR2 maintains a meiosis-inhibitory level of cyclic guanosine 5'-monophosphate (cGMP) until LHR signaling causes dephosphorylation of NPR2, reducing NPR2 activity, lowering cGMP to a level that releases meiotic arrest. However, the signaling pathway between LHR activation and NPR2 dephosphorylation remains incompletely understood, due in part to imprecise information about the cellular localization of these 2 proteins. To investigate their localization, we generated mouse lines in which hemagglutinin epitope tags were added to the endogenous LHR and NPR2 proteins, and used immunofluorescence and immunogold microscopy to localize these proteins with high resolution. The results showed that the LHR protein is absent from the cumulus cells and inner mural granulosa cells, and is present in only 13% to 48% of the outer mural granulosa cells. In contrast, NPR2 is present throughout the follicle, and is more concentrated in the cumulus cells. Less than 20% of the NPR2 is in the same cells that express the LHR. These results suggest that to account for the LH-induced inactivation of NPR2, LHR-expressing cells send a signal that inactivates NPR2 in neighboring cells that do not express the LHR. An inhibitor of gap junction permeability attenuates the LH-induced cGMP decrease in the outer mural granulosa cells, consistent with this mechanism contributing to how NPR2 is inactivated in cells that do not express the LHR.
Collapse
Affiliation(s)
- Valentina Baena
- Department of Cell Biology, University of Connecticut Health Center, Farmington, Connecticut
| | - Corie M Owen
- Department of Cell Biology, University of Connecticut Health Center, Farmington, Connecticut
| | - Tracy F Uliasz
- Department of Cell Biology, University of Connecticut Health Center, Farmington, Connecticut
| | - Katie M Lowther
- Department of Cell Biology, University of Connecticut Health Center, Farmington, Connecticut
| | - Siu-Pok Yee
- Department of Cell Biology, University of Connecticut Health Center, Farmington, Connecticut
| | - Mark Terasaki
- Department of Cell Biology, University of Connecticut Health Center, Farmington, Connecticut
| | - Jeremy R Egbert
- Department of Cell Biology, University of Connecticut Health Center, Farmington, Connecticut
| | - Laurinda A Jaffe
- Department of Cell Biology, University of Connecticut Health Center, Farmington, Connecticut
- Correspondence: Laurinda A. Jaffe, Department of Cell Biology, University of Connecticut Health Center, Farmington, CT 06030 USA. E-mail:
| |
Collapse
|
11
|
Zhang V, Kucharski R, Landers C, Richards SN, Bröer S, Martin RE, Maleszka R. Characterization of a Dopamine Transporter and Its Splice Variant Reveals Novel Features of Dopaminergic Regulation in the Honey Bee. Front Physiol 2019; 10:1375. [PMID: 31736791 PMCID: PMC6838227 DOI: 10.3389/fphys.2019.01375] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/22/2019] [Accepted: 10/17/2019] [Indexed: 11/25/2022] Open
Abstract
Dopamine is an important neuromodulator involved in reward-processing, movement control, motivational responses, and other aspects of behavior in most animals. In honey bees (Apis mellifera), the dopaminergic system has been implicated in an elaborate pheromonal communication network between individuals and in the differentiation of females into reproductive (queen) and sterile (worker) castes. Here we have identified and characterized a honey bee dopamine transporter (AmDAT) and a splice variant lacking exon 3 (AmDATΔex3). Both transcripts are present in the adult brain and antennae as well as at lower levels within larvae and ovaries. When expressed separately in the Xenopus oocyte system, AmDAT localizes to the oocyte surface whereas the splice variant is retained at an internal membrane. Oocytes expressing AmDAT exhibit a 12-fold increase in the uptake of [3H]dopamine relative to non-injected oocytes, whereas the AmDATΔex3-expressing oocytes show no change in [3H]dopamine transport. Electrophysiological measurements of AmDAT activity revealed it to be a high-affinity, low-capacity transporter of dopamine. The transporter also recognizes noradrenaline as a major substrate and tyramine as a minor substrate, but does not transport octopamine, L-Dopa, or serotonin. Dopamine transport via AmDAT is inhibited by cocaine in a reversible manner, but is unaffected by octopamine. Co-expression of AmDAT and AmDATΔex3 in oocytes results in a substantial reduction in AmDAT-mediated transport, which was also detected as a significant decrease in the level of AmDAT protein. This down-regulatory effect is not attributable to competition with AmDATΔex3 for ER ribosomes, nor to a general inhibition of the oocyte's translational machinery. In vivo, the expression of both transcripts shows a high level of inter-individual variability. Gene-focused, ultra-deep amplicon sequencing detected methylation of the amdat locus at ten 5'-C-phosphate-G-3' dinucleotides (CpGs), but only in 5-10% of all reads in whole brains or antennae. These observations, together with the localization of the amdat transcript to a few clusters of dopaminergic neurons, imply that amdat methylation is positively linked to its transcription. Our findings suggest that multiple cellular mechanisms, including gene splicing and epigenomic communication systems, may be adopted to increase the potential of a conserved gene to contribute to lineage-specific behavioral outcomes.
Collapse
Affiliation(s)
- Vicky Zhang
- Research School of Biology, The Australian National University, Canberra, ACT, Australia
| | - Robert Kucharski
- Research School of Biology, The Australian National University, Canberra, ACT, Australia
- Faculty of Science and Technology, University of Canberra, Bruce, ACT, Australia
| | - Courtney Landers
- Research School of Biology, The Australian National University, Canberra, ACT, Australia
| | - Sashika N. Richards
- Research School of Biology, The Australian National University, Canberra, ACT, Australia
| | - Stefan Bröer
- Research School of Biology, The Australian National University, Canberra, ACT, Australia
| | - Rowena E. Martin
- Research School of Biology, The Australian National University, Canberra, ACT, Australia
| | - Ryszard Maleszka
- Research School of Biology, The Australian National University, Canberra, ACT, Australia
| |
Collapse
|
12
|
Thal LB, Tomlinson ID, Quinlan MA, Kovtun O, Blakely RD, Rosenthal SJ. Single Quantum Dot Imaging Reveals PKCβ-Dependent Alterations in Membrane Diffusion and Clustering of an Attention-Deficit Hyperactivity Disorder/Autism/Bipolar Disorder-Associated Dopamine Transporter Variant. ACS Chem Neurosci 2019; 10:460-471. [PMID: 30153408 PMCID: PMC6411462 DOI: 10.1021/acschemneuro.8b00350] [Citation(s) in RCA: 21] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022] Open
Abstract
The dopamine transporter (DAT) is a transmembrane protein that terminates dopamine signaling in the brain by driving rapid dopamine reuptake into presynaptic nerve terminals. Several lines of evidence indicate that DAT dysfunction is linked to neuropsychiatric disorders such as attention-deficit/hyperactivity disorder (ADHD), bipolar disorder (BPD), and autism spectrum disorder (ASD). Indeed, individuals with these disorders have been found to express the rare, functional DAT coding variant Val559, which confers anomalous dopamine efflux (ADE) in vitro and in vivo. To elucidate the impact of the DAT Val559 variant on membrane diffusion dynamics, we implemented our antagonist-conjugated quantum dot (QD) labeling approach to monitor the lateral mobility of single particle-labeled transporters in transfected HEK-293 and SK-N-MC cells. Our results demonstrate significantly higher diffusion coefficients of DAT Val559 compared to those of DAT Ala559, effects likely determined by elevated N-terminal transporter phosphorylation. We also provide pharmacological evidence that PKCβ-mediated signaling supports enhanced DAT Val559 membrane diffusion rates. Additionally, our results are complimented with diffusion rates of phosphomimicked and phosphorylation-occluded DAT variants. Furthermore, we show DAT Val559 has a lower propensity for membrane clustering, which may be caused by a mutation-derived shift out of membrane microdomains leading to faster lateral membrane diffusion rates. These findings further demonstrate a functional impact of DAT Val559 and suggest that changes in transporter localization and lateral mobility may sustain ADE and contribute to alterations in dopamine signaling underlying multiple neuropsychiatric disorders.
Collapse
|
13
|
Jaykumar AB, Caceres PS, Ortiz PA. Single-molecule labeling for studying trafficking of renal transporters. Am J Physiol Renal Physiol 2018; 315:F1243-F1249. [PMID: 30043625 DOI: 10.1152/ajprenal.00082.2017] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022] Open
Abstract
The ability to detect and track single molecules presents the advantage of visualizing the complex behavior of transmembrane proteins with a time and space resolution that would otherwise be lost with traditional labeling and biochemical techniques. Development of new imaging probes has provided a robust method to study their trafficking and surface dynamics. This mini-review focuses on the current technology available for single-molecule labeling of transmembrane proteins, their advantages, and limitations. We also discuss the application of these techniques to the study of renal transporter trafficking in light of recent research.
Collapse
Affiliation(s)
- Ankita Bachhawat Jaykumar
- Hypertension and Vascular Research Division, Department of Internal Medicine, Henry Ford Hospital , Detroit, Michigan.,Department of Physiology, Wayne State University School of Medicine , Detroit, Michigan
| | - Paulo S Caceres
- Hypertension and Vascular Research Division, Department of Internal Medicine, Henry Ford Hospital , Detroit, Michigan
| | - Pablo A Ortiz
- Hypertension and Vascular Research Division, Department of Internal Medicine, Henry Ford Hospital , Detroit, Michigan.,Department of Physiology, Wayne State University School of Medicine , Detroit, Michigan
| |
Collapse
|
14
|
Sorkina T, Ma S, Larsen MB, Watkins SC, Sorkin A. Small molecule induced oligomerization, clustering and clathrin-independent endocytosis of the dopamine transporter. eLife 2018; 7:32293. [PMID: 29630493 PMCID: PMC5896956 DOI: 10.7554/elife.32293] [Citation(s) in RCA: 27] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/25/2017] [Accepted: 03/22/2018] [Indexed: 12/14/2022] Open
Abstract
Clathrin-independent endocytosis (CIE) mediates internalization of many transmembrane proteins but the mechanisms of cargo recruitment during CIE are poorly understood. We found that the cell-permeable furopyrimidine AIM-100 promotes dramatic oligomerization, clustering and CIE of human and mouse dopamine transporters (DAT), but not of their close homologues, norepinephrine and serotonin transporters. All effects of AIM-100 on DAT and the occupancy of substrate binding sites in the transporter were mutually exclusive, suggesting that AIM-100 may act by binding to DAT. Surprisingly, AIM-100-induced DAT endocytosis was independent of dynamin, cholesterol-rich microdomains and actin cytoskeleton, implying that a novel endocytic mechanism is involved. AIM-100 stimulated trafficking of internalized DAT was also unusual: DAT accumulated in early endosomes without significant recycling or degradation. We propose that AIM-100 augments DAT oligomerization through an allosteric mechanism associated with the DAT conformational state, and that oligomerization-triggered clustering leads to a coat-independent endocytosis and subsequent endosomal retention of DAT.
Collapse
Affiliation(s)
- Tatiana Sorkina
- Department of Cell Biology, University of Pittsburgh School of Medicine, Pittsburgh, United States
| | - Shiqi Ma
- Department of Cell Biology, University of Pittsburgh School of Medicine, Pittsburgh, United States
| | - Mads Breum Larsen
- Department of Cell Biology, University of Pittsburgh School of Medicine, Pittsburgh, United States
| | - Simon C Watkins
- Department of Cell Biology, University of Pittsburgh School of Medicine, Pittsburgh, United States
| | - Alexander Sorkin
- Department of Cell Biology, University of Pittsburgh School of Medicine, Pittsburgh, United States
| |
Collapse
|
15
|
Heterogeneities in Axonal Structure and Transporter Distribution Lower Dopamine Reuptake Efficiency. eNeuro 2018; 5:eN-NWR-0298-17. [PMID: 29430519 PMCID: PMC5804147 DOI: 10.1523/eneuro.0298-17.2017] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/16/2017] [Revised: 11/28/2017] [Accepted: 12/07/2017] [Indexed: 12/13/2022] Open
Abstract
Efficient clearance of dopamine (DA) from the synapse is key to regulating dopaminergic signaling. This role is fulfilled by DA transporters (DATs). Recent advances in the structural characterization of DAT from Drosophila (dDAT) and in high-resolution imaging of DA neurons and the distribution of DATs in living cells now permit us to gain a mechanistic understanding of DA reuptake events in silico. Using electron microscopy images and immunofluorescence of transgenic knock-in mouse brains that express hemagglutinin-tagged DAT in DA neurons, we reconstructed a realistic environment for MCell simulations of DA reuptake, wherein the identity, population and kinetics of homology-modeled human DAT (hDAT) substates were derived from molecular simulations. The complex morphology of axon terminals near active zones was observed to give rise to large variations in DA reuptake efficiency, and thereby in extracellular DA density. Comparison of the effect of different firing patterns showed that phasic firing would increase the probability of reaching local DA levels sufficiently high to activate low-affinity DA receptors, mainly owing to high DA levels transiently attained during the burst phase. The experimentally observed nonuniform surface distribution of DATs emerged as a major modulator of DA signaling: reuptake was slower, and the peaks/width of transient DA levels were sharper/wider under nonuniform distribution of DATs, compared with uniform. Overall, the study highlights the importance of accurate descriptions of extrasynaptic morphology, DAT distribution, and conformational kinetics for quantitative evaluation of dopaminergic transmission and for providing deeper understanding of the mechanisms that regulate DA transmission.
Collapse
|
16
|
|
17
|
Targeting of dopamine transporter to filopodia requires an outward-facing conformation of the transporter. Sci Rep 2017; 7:5399. [PMID: 28710426 PMCID: PMC5511133 DOI: 10.1038/s41598-017-05637-x] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/27/2017] [Accepted: 06/01/2017] [Indexed: 12/01/2022] Open
Abstract
Dopamine transporter (DAT) has been shown to accumulate in filopodia in neurons and non-neuronal cells. To examine the mechanisms of DAT filopodial targeting, we used quantitative live-cell fluorescence microscopy, and compared the effects of the DAT inhibitor cocaine and its fluorescent analog JHC1-64 on the plasma membrane distribution of wild-type DAT and two non-functional DAT mutants, R60A and W63A, that do not accumulate in filopodia. W63A did not bind JHC1-64, whereas R60A did, although less efficiently compared to the wild-type DAT. Molecular dynamics simulations predicted that R60A preferentially assumes an outward-facing (OF) conformation through compensatory intracellular salt bridge formation, which in turn favors binding of cocaine. Imaging analysis showed that JHC1-64-bound R60A mutant predominantly localized in filopodia, whereas free R60A molecules were evenly distributed within the plasma membrane. Cocaine binding significantly increased the density of R60A, but not that of W63A, in filopodia. Further, zinc binding, known to stabilize the OF state, also increased R60A concentration in filopodia. Finally, amphetamine, that is thought to disrupt DAT OF conformation, reduced the concentration of wild-type DAT in filopodia. Altogether, these data indicate that OF conformation is required for the efficient targeting of DAT to, and accumulation in, filopodia.
Collapse
|
18
|
Effective Cellular Morphology Analysis for Differentiation Processes by a Fluorescent 1,3a,6a-Triazapentalene Derivative Probe in Live Cells. PLoS One 2016; 11:e0160625. [PMID: 27490470 PMCID: PMC4973928 DOI: 10.1371/journal.pone.0160625] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/20/2016] [Accepted: 07/24/2016] [Indexed: 12/13/2022] Open
Abstract
Nuclear and cytoplasmic morphological changes provide important information about cell differentiation processes, cell functions, and signal responses. There is a strong desire to develop a rapid and simple method for visualizing cytoplasmic and nuclear morphology. Here, we developed a novel and rapid method for probing cellular morphological changes of live cell differentiation process by a fluorescent probe, TAP-4PH, a 1,3a,6a-triazapentalene derivative. TAP-4PH showed high fluorescence in cytoplasmic area, and visualized cytoplasmic and nuclear morphological changes of live cells during differentiation. We demonstrated that TAP-4PH visualized dendritic axon and spine formation in neuronal differentiation, and nuclear structural changes during neutrophilic differentiation. We also showed that the utility of TAP-4PH for visualization of cytoplasmic and nuclear morphologies of various type of live cells. Our visualizing method has no toxicity and no influence on the cellular differentiation and function. The cell morphology can be rapidly observed after addition of TAP-4PH and can continue to be observed in the presence of TAP-4PH in cell culture medium. Moreover, TAP-4PH can be easily removed after observation by washing for subsequent biological assay. Taken together, these results demonstrate that our visualization method is a powerful tool to probe differentiation processes before subsequent biological assay in live cells.
Collapse
|
19
|
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
|
20
|
Killinger BA, Moszczynska A. Epothilone D prevents binge methamphetamine-mediated loss of striatal dopaminergic markers. J Neurochem 2015; 136:510-25. [PMID: 26465779 DOI: 10.1111/jnc.13391] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/19/2015] [Revised: 09/12/2015] [Accepted: 09/21/2015] [Indexed: 02/06/2023]
Abstract
Exposure to binge methamphetamine (METH) can result in a permanent or transient loss of dopaminergic (DAergic) markers such as dopamine (DA), dopamine transporter, and tyrosine hydroxylase (TH) in the striatum. We hypothesized that the METH-induced loss of striatal DAergic markers was, in part, due to a destabilization of microtubules (MTs) in the nigrostriatal DA pathway that ultimately impedes anterograde axonal transport of these markers. To test this hypothesis, adult male Sprague-Dawley rats were treated with binge METH or saline in the presence or absence of epothilone D (EpoD), a MT-stabilizing compound, and assessed 3 days after the treatments for the levels of several DAergic markers as well as for the levels of tubulins and their post-translational modifications (PMTs). Binge METH induced a loss of stable long-lived MTs within the striatum but not within the substantia nigra pars compacta (SNpc). Treatment with a low dose of EpoD increased the levels of markers of stable MTs and prevented METH-mediated deficits in several DAergic markers in the striatum. In contrast, administration of a high dose of EpoD appeared to destabilize MTs and potentiated the METH-induced deficits in several DAergic markers. The low-dose EpoD also prevented the METH-induced increase in striatal DA turnover and increased behavioral stereotypy during METH treatment. Together, these results demonstrate that MT dynamics plays a role in the development of METH-induced losses of several DAergic markers in the striatum and may mediate METH-induced degeneration of terminals in the nigrostriatal DA pathway. Our study also demonstrates that MT-stabilizing drugs such as EpoD have a potential to serve as useful therapeutic agents to restore function of DAergic nerve terminals following METH exposure when administered at low doses. Administration of binge methamphetamine (METH) negatively impacts neurotransmission in the nigrostriatal dopamine (DA) system. The effects of METH include decreasing the levels of DAergic markers in the striatum. We have determined that high-dose METH destabilizes microtubules in this pathway, which is manifested by decreased levels of acetylated (Acetyl) and detyrosinated (Detyr) α-tubulin (I). A microtubule stabilizing agent epothilone D protects striatal microtubules form the METH-induced loss of DAergic markers (II). These findings provide a new strategy for protection form METH - restoration of proper axonal transport.
Collapse
Affiliation(s)
- Bryan A Killinger
- Department of Pharmaceutical Sciences, Eugene Applebaum College of Pharmacy and Health Sciences, Wayne State University, Detroit, Michigan, USA
| | - Anna Moszczynska
- Department of Pharmaceutical Sciences, Eugene Applebaum College of Pharmacy and Health Sciences, Wayne State University, Detroit, Michigan, USA
| |
Collapse
|
21
|
Beerepoot P, Lam VM, Salahpour A. A β-lactamase based assay to measure surface expression of membrane proteins. Methods Mol Biol 2015; 1270:107-14. [PMID: 25702112 DOI: 10.1007/978-1-4939-2309-0_8] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/05/2023]
Abstract
Measurement of cell surface expression is an essential part of studying membrane proteins. Traditional techniques for measuring surface expression depend on the availability of appropriate radioligands or antibodies towards extracellular epitopes of a protein of interest. The current protocol outlines the use of an assay to monitor surface expression of membrane proteins tagged with a bacterial β-lactamase in mammalian cell lines. The use of this technique allows for quick, quantitative, sensitive, and inexpensive measurement of surface expression, with the potential for high-throughput screening.
Collapse
Affiliation(s)
- Pieter Beerepoot
- Department of Pharmacology and Toxicology, University of Toronto, Toronto, ON, Canada
| | | | | |
Collapse
|
22
|
Frau L, Costa G, Porceddu PF, Khairnar A, Castelli MP, Ennas MG, Madeddu C, Wardas J, Morelli M. Influence of caffeine on 3,4-methylenedioxymethamphetamine-induced dopaminergic neuron degeneration and neuroinflammation is age-dependent. J Neurochem 2015; 136:148-62. [DOI: 10.1111/jnc.13377] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/01/2015] [Revised: 09/02/2015] [Accepted: 09/03/2015] [Indexed: 02/03/2023]
Affiliation(s)
- Lucia Frau
- Department of Biomedical Sciences; Section of Neuropsychopharmacology; University of Cagliari; Cagliari Italy
| | - Giulia Costa
- Department of Biomedical Sciences; Section of Neuropsychopharmacology; University of Cagliari; Cagliari Italy
| | - Pier Francesca Porceddu
- Department of Biomedical Sciences; Section of Neuropsychopharmacology; University of Cagliari; Cagliari Italy
| | - Amit Khairnar
- Applied Neuroscience Research Group; CEITEC - Central European Institute of Technology; Masaryk University; Brno Czech Republic
| | - Maria Paola Castelli
- Department of Biomedical Sciences; Section of Neuroscience and Clinical Pharmacology; University of Cagliari; Monserrato (CA) Italy
| | - Maria Grazia Ennas
- Department of Biomedical Sciences; Section of Neuroscience and Clinical Pharmacology; University of Cagliari; Monserrato (CA) Italy
| | - Camilla Madeddu
- Department of Biomedical Sciences; Section of Neuroscience and Clinical Pharmacology; University of Cagliari; Monserrato (CA) Italy
| | - Jadwiga Wardas
- Department of Neuropsychopharmacology; Institute of Pharmacology; Polish Academy of Sciences; Krakow Poland
| | - Micaela Morelli
- Department of Biomedical Sciences; Section of Neuropsychopharmacology; University of Cagliari; Cagliari Italy
- CNR; Institute of Neuroscience; Cagliari Italy
| |
Collapse
|
23
|
Moritz AE, Rastedt DE, Stanislowski DJ, Shetty M, Smith MA, Vaughan RA, Foster JD. Reciprocal Phosphorylation and Palmitoylation Control Dopamine Transporter Kinetics. J Biol Chem 2015; 290:29095-105. [PMID: 26424792 DOI: 10.1074/jbc.m115.667055] [Citation(s) in RCA: 43] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/21/2015] [Indexed: 11/06/2022] Open
Abstract
The dopamine transporter is a neuronal protein that drives the presynaptic reuptake of dopamine (DA) and is the major determinant of transmitter availability in the brain. Dopamine transporter function is regulated by protein kinase C (PKC) and other signaling pathways through mechanisms that are complex and poorly understood. Here we investigate the role of Ser-7 phosphorylation and Cys-580 palmitoylation in mediating steady-state transport kinetics and PKC-stimulated transport down-regulation. Using both mutational and pharmacological approaches, we demonstrate that these post-translational modifications are reciprocally regulated, leading to transporter populations that display high phosphorylation-low palmitoylation or low phosphorylation-high palmitoylation. The balance between the modifications dictates transport capacity, as conditions that promote high phosphorylation or low palmitoylation reduce transport Vmax and enhance PKC-stimulated down-regulation, whereas conditions that promote low phosphorylation or high palmitoylation increase transport Vmax and suppress PKC-stimulated down-regulation. Transitions between these functional states occur when endocytosis is blocked or undetectable, indicating that the modifications kinetically regulate the velocity of surface transporters. These findings reveal a novel mechanism for control of DA reuptake that may represent a point of dysregulation in DA imbalance disorders.
Collapse
Affiliation(s)
- Amy E Moritz
- From the Department of Basic Sciences, University of North Dakota School of Medicine and Health Sciences, Grand Forks, North Dakota 58202-9061
| | - Danielle E Rastedt
- From the Department of Basic Sciences, University of North Dakota School of Medicine and Health Sciences, Grand Forks, North Dakota 58202-9061
| | - Daniel J Stanislowski
- From the Department of Basic Sciences, University of North Dakota School of Medicine and Health Sciences, Grand Forks, North Dakota 58202-9061
| | - Madhur Shetty
- From the Department of Basic Sciences, University of North Dakota School of Medicine and Health Sciences, Grand Forks, North Dakota 58202-9061
| | - Margaret A Smith
- From the Department of Basic Sciences, University of North Dakota School of Medicine and Health Sciences, Grand Forks, North Dakota 58202-9061
| | - Roxanne A Vaughan
- From the Department of Basic Sciences, University of North Dakota School of Medicine and Health Sciences, Grand Forks, North Dakota 58202-9061
| | - James D Foster
- From the Department of Basic Sciences, University of North Dakota School of Medicine and Health Sciences, Grand Forks, North Dakota 58202-9061
| |
Collapse
|
24
|
Action potentials and amphetamine release antipsychotic drug from dopamine neuron synaptic VMAT vesicles. Proc Natl Acad Sci U S A 2015. [PMID: 26216995 DOI: 10.1073/pnas.1503766112] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/25/2023] Open
Abstract
Based on lysotracker red imaging in cultured hippocampal neurons, antipsychotic drugs (APDs) were proposed to accumulate in synaptic vesicles by acidic trapping and to be released in response to action potentials. Because many APDs are dopamine (DA) D2 receptor (D2R) antagonists, such a mechanism would be particularly interesting if it operated in midbrain DA neurons. Here, the APD cyamemazine (CYAM) is visualized directly by two-photon microscopy in substantia nigra and striatum brain slices. CYAM accumulated slowly into puncta based on vacuolar H(+)-ATPase activity and dispersed rapidly upon dissipating organelle pH gradients. Thus, CYAM is subject to acidic trapping and released upon deprotonation. In the striatum, Ca(2+)-dependent reduction of the CYAM punctate signal was induced by depolarization or action potentials. Striatal CYAM overlapped with the dopamine transporter (DAT). Furthermore, parachloroamphetamine (pCA), acting via vesicular monoamine transporter (VMAT), and a charged VMAT, substrate 1-methyl-4-phenylpyridinium (MPP(+)), reduced striatal CYAM. In vivo CYAM administration and in vitro experiments confirmed that clinically relevant CYAM concentrations result in vesicular accumulation and pCA-dependent release. These results show that some CYAM is in DA neuron VMAT vesicles and suggests a new drug interaction in which amphetamine induces CYAM deprotonation and release as a consequence of the H(+) countertransport by VMAT that accompanies vesicular uptake, but not by inducing exchange or acting as a weak base. Therefore, in the striatum, APDs are released with DA in response to action potentials and an amphetamine. This synaptic corelease is expected to enhance APD antagonism of D2Rs where and when dopaminergic transmission occurs.
Collapse
|
25
|
Dopamine transporter is enriched in filopodia and induces filopodia formation. Mol Cell Neurosci 2015; 68:120-30. [PMID: 25936602 DOI: 10.1016/j.mcn.2015.04.005] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/23/2014] [Revised: 04/03/2015] [Accepted: 04/21/2015] [Indexed: 11/24/2022] Open
Abstract
Dopamine transporter (DAT, SLC6A3) controls dopamine (DA) neurotransmission by mediating re-uptake of extracellular DA into DA neurons. DA uptake depends on the amount of DAT at the cell surface, and is therefore regulated by DAT subcellular distribution. Hence we used spinning disk confocal microscopy to demonstrate DAT localization in membrane protrusions that contained filamentous actin and myosin X (MyoX), a molecular motor located in filopodia tips, thus confirming that these protrusions are filopodia. DAT was enriched in filopodia. In contrast, R60A and W63A DAT mutants with disrupted outward-facing conformation were not accumulated in filopodia, suggesting that this conformation is necessary for DAT filopodia targeting. Three independent approaches of filopodia counting showed that DAT expression leads to an increase in the number of filopodia per cell, indicating that DAT can induce filopodia formation. Depletion of MyoX by RNA interference resulted in a significant loss of filopodia but did not completely eliminate filopodia, implying that DAT-enriched filopodia can be formed without MyoX. In cultured postnatal DA neurons MyoX was mainly localized to growth cones that displayed highly dynamic DAT-containing filopodia. We hypothesize that the concave shape of the DAT molecule functions as the targeting determinant for DAT accumulation in outward-curved membrane domains, and may also allow high local concentrations of DAT to induce an outward membrane bending. Such targeting and membrane remodeling capacities may be part of the mechanism responsible for DAT enrichment in the filopodia and its targeting to the axonal processes of DA neurons.
Collapse
|
26
|
Kovtun O, Sakrikar D, Tomlinson ID, Chang JC, Arzeta-Ferrer X, Blakely RD, Rosenthal SJ. Single-quantum-dot tracking reveals altered membrane dynamics of an attention-deficit/hyperactivity-disorder-derived dopamine transporter coding variant. ACS Chem Neurosci 2015; 6:526-34. [PMID: 25747272 PMCID: PMC5530757 DOI: 10.1021/cn500202c] [Citation(s) in RCA: 29] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/14/2023] Open
Abstract
The presynaptic, cocaine- and amphetamine-sensitive dopamine (DA) transporter (DAT, SLC6A3) controls the intensity and duration of synaptic dopamine signals by rapid clearance of DA back into presynaptic nerve terminals. Abnormalities in DAT-mediated DA clearance have been linked to a variety of neuropsychiatric disorders, including addiction, autism, and attention deficit/hyperactivity disorder (ADHD). Membrane trafficking of DAT appears to be an important, albeit incompletely understood, post-translational regulatory mechanism; its dysregulation has been recently proposed as a potential risk determinant of these disorders. In this study, we demonstrate a link between an ADHD-associated DAT mutation (Arg615Cys, R615C) and variation on DAT transporter cell surface dynamics, a combination only previously studied with ensemble biochemical and optical approaches that featured limited spatiotemporal resolution. Here, we utilize high-affinity, DAT-specific antagonist-conjugated quantum dot (QD) probes to establish the dynamic mobility of wild-type and mutant DATs at the plasma membrane of living cells. Single DAT-QD complex trajectory analysis revealed that the DAT 615C variant exhibited increased membrane mobility relative to DAT 615R, with diffusion rates comparable to those observed after lipid raft disruption. This phenomenon was accompanied by a loss of transporter mobilization triggered by amphetamine, a common component of ADHD medications. Together, our data provides the first dynamic imaging of single DAT proteins, providing new insights into the relationship between surface dynamics and trafficking of both wild-type and disease-associated transporters. Our approach should be generalizable to future studies that explore the possibilities of perturbed surface DAT dynamics that may arise as a consequence of genetic alterations, regulatory changes, and drug use that contribute to the etiology or treatment of neuropsychiatric disorders.
Collapse
Affiliation(s)
- Oleg Kovtun
- Department of Chemistry, Vanderbilt University, Nashville, Tennessee 37235, United States
- McCoy & McCoy Laboratories, Inc, Madisonville, Kentucky 42431, United States
| | - Dhananjay Sakrikar
- Department of Pharmacology, Vanderbilt University, Nashville, Tennessee 37235, United States
| | - Ian D. Tomlinson
- Department of Chemistry, Vanderbilt University, Nashville, Tennessee 37235, United States
| | - Jerry C. Chang
- Laboratory of Molecular and Cellular Neuroscience, The Rockefeller University, New York, New York 10065, United States
| | - Xochitl Arzeta-Ferrer
- Department of Chemistry, Vanderbilt University, Nashville, Tennessee 37235, United States
| | - Randy D. Blakely
- Department of Pharmacology, Vanderbilt University, Nashville, Tennessee 37235, United States
- Department of Psychiatry, Vanderbilt University, Nashville, Tennessee 37235, United States
- Silvio O. Conte Center for Neuroscience Research, Vanderbilt University, Nashville, Tennessee 37235, United States
| | - Sandra J. Rosenthal
- Department of Chemistry, Vanderbilt University, Nashville, Tennessee 37235, United States
- Department of Pharmacology, Vanderbilt University, Nashville, Tennessee 37235, United States
- Department of Chemical and Biomolecular Engineering, Vanderbilt University, Nashville, Tennessee 37235, United States
- Department of Physics and Astronomy, Vanderbilt University, Nashville, Tennessee 37235, United States
- Vanderbilt Institute of Nanoscale Science and Engineering, Vanderbilt University, Nashville, Tennessee 37235, United States
- Materials Science and Technology Division, Oak Ridge National Laboratory, Oak Ridge, Tennessee 37831, United States
| |
Collapse
|
27
|
Vecchio LM, Bermejo MK, Beerepoot P, Ramsey AJ, Salahpour A. N-terminal tagging of the dopamine transporter impairs protein expression and trafficking in vivo. Mol Cell Neurosci 2014; 61:123-32. [PMID: 24886986 DOI: 10.1016/j.mcn.2014.05.007] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/28/2013] [Revised: 05/22/2014] [Accepted: 05/29/2014] [Indexed: 01/26/2023] Open
Abstract
The dopamine transporter (DAT) is the primary protein responsible for the uptake of dopamine from the extracellular space back into presynaptic neurons. As such, it plays an important role in the cessation of dopaminergic neurotransmission and in the maintenance of extracellular dopamine homeostasis. Here, we report the development of a new BAC transgenic mouse line that expresses DAT with an N-terminal HA-epitope (HAD-Tg). In this line, two copies of the HA-DAT BAC are incorporated into the genome, increasing DAT mRNA levels by 47%. Despite the increase in mRNA levels, HAD-Tg mice show no significant increase in the level of DAT protein in the striatum, indicating a defect in protein trafficking or stability. By crossing HAD-Tg mice with DAT knockout mice (DAT-KO), we engineered mice that exclusively express HA-tagged DAT in the absence of endogenous DAT (DAT-KO/HAD-Tg). We show that DAT-KO/HAD-Tg mice express only 8.5% of WT DAT levels in the striatum. Importantly, the HA-tagged DAT that is present in DAT-KO/HAD-Tg mice is functional, as it is able to partially rescue the DAT-KO hyperactive phenotype. Finally, we provide evidence that the HA-tagged DAT is retained in the cell body based on a reduction in the striatum:midbrain protein ratio. These results demonstrate that the presence of the N-terminal tag leads to impaired DAT protein expression in vivo due in part to improper trafficking of the tagged transporter, and highlight the importance of the N-terminus in the transport of DAT to striatal terminals.
Collapse
Affiliation(s)
- Laura M Vecchio
- Department of Pharmacology, University of Toronto: Medical Sciences Building, Room 4302, 1 King's College Circle, Toronto, ON M5S 1A8, Canada.
| | - M Kristel Bermejo
- Department of Pharmacology, University of Toronto: Medical Sciences Building, Room 4302, 1 King's College Circle, Toronto, ON M5S 1A8, Canada.
| | - Pieter Beerepoot
- Department of Pharmacology, University of Toronto: Medical Sciences Building, Room 4302, 1 King's College Circle, Toronto, ON M5S 1A8, Canada.
| | - Amy J Ramsey
- Department of Pharmacology, University of Toronto: Medical Sciences Building, Room 4302, 1 King's College Circle, Toronto, ON M5S 1A8, Canada.
| | - Ali Salahpour
- Department of Pharmacology, University of Toronto: Medical Sciences Building, Room 4302, 1 King's College Circle, Toronto, ON M5S 1A8, Canada.
| |
Collapse
|
28
|
Dey A, Wu J, Kirkpatrick DS. Interrogation of in vivo protein-protein interactions using transgenic mouse models and stable isotope labeling. Methods Mol Biol 2014; 1176:179-190. [PMID: 25030928 DOI: 10.1007/978-1-4939-0992-6_15] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 06/03/2023]
Abstract
Methods in mass spectrometry have evolved in recent years, facilitating proteomic analyses that were previously beyond the limits of the technology. Transgenic mouse models, coupled with mass spectrometry proteomics, have served as valuable platform for elucidating the in vivo function of individual genes and proteins. Here we discuss the methods we have recently employed to characterize protein-protein interactions and posttranslational modifications in tagged knock-in mouse models. These methods can be broadly applied to other systems for various applications in both basic and translational science.
Collapse
Affiliation(s)
- Anwesha Dey
- Department of Molecular Biology, Genentech, Inc., 1 DNA Way, South San Francisco, CA, 94080, USA,
| | | | | |
Collapse
|
29
|
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
|
30
|
Costa G, Frau L, Wardas J, Pinna A, Plumitallo A, Morelli M. MPTP-induced dopamine neuron degeneration and glia activation is potentiated in MDMA-pretreated mice. Mov Disord 2013; 28:1957-65. [PMID: 24108425 DOI: 10.1002/mds.25646] [Citation(s) in RCA: 42] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/16/2013] [Revised: 07/08/2013] [Accepted: 07/23/2013] [Indexed: 12/13/2022] Open
Abstract
Clinical observations report a greater propensity to develop Parkinson's disease (PD) in amphetamine users. 3,4-Methylenedioxymethamphetamine (MDMA; "ecstasy") is an amphetamine-related drug that is largely consumed by adolescents and young adults, which may have neuroinflammatory and neurotoxic effects. Here, the objective was to evaluate in mice whether consumption of MDMA during adolescence might influence the neuroinflammatory and neurotoxic effects of 1-methyl-4-phenyl-1,2,3,6-tetrahydropyridine (MPTP), a toxin known to induce PD in humans. The activation of astroglia and microglia by glial fibrillary acidic protein (GFAP) and complement receptor type 3 (CD11b) immunohistochemistry and the degeneration of dopaminergic neurons by tyrosine hydroxylase (TH) immunohistochemistry were evaluated. MPTP (20 mg/kg × 4) was administered to mice treated from ages 8 weeks to 17 weeks with MDMA (10 mg/kg twice daily, two times a week). In mice that were chronically treated with MDMA, administration of MPTP induced a higher microglial and astroglial response in both the striatum and the substantia nigra pars compacta (SNc) compared with vehicle-treated or vehicle + MPTP-treated mice. Inflammatory changes were associated with a decrease in TH immunoreactivity in the SNc of MDMA-treated mice and with a further decrease in the striatum and the SNc of MDMA + MPTP-treated mice compared with vehicle-treated, MDMA-treated, and MPTP-treated mice. The results demonstrate that chronic administration of MDMA during late adolescence in mice exacerbates the neurodegeneration and neuroinflammation caused by MPTP, suggesting that MDMA may constitute a risk factor for dopaminergic neuron degeneration.
Collapse
Affiliation(s)
- Giulia Costa
- Department of Biomedical Sciences, Section of Neuropsychopharmacology, University of Cagliari, Cagliari, Italy
| | | | | | | | | | | |
Collapse
|
31
|
Rao A, Sorkin A, Zahniser NR. Mice expressing markedly reduced striatal dopamine transporters exhibit increased locomotor activity, dopamine uptake turnover rate, and cocaine responsiveness. Synapse 2013; 67:668-77. [PMID: 23564231 DOI: 10.1002/syn.21671] [Citation(s) in RCA: 24] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/04/2012] [Accepted: 03/23/2013] [Indexed: 02/06/2023]
Abstract
Variations in the expression levels of the dopamine transporter (DAT) can influence responsiveness to psychostimulant drugs like cocaine. To better understand this relationship, we studied a new DAT-low expresser (DAT-LE) mouse model and performed behavioral and biochemical studies with it. Immunoblotting and [(3) H]WIN 35,428 binding analyses revealed that these mice express ∼35% of wildtype (WT) mouse striatal DAT levels. Compared to WT mice, DAT-LE mice were hyperactive in a novel open-field environment. Despite their higher basal locomotor activity, cocaine (10 or 20 mg/kg, i.p.) induced greater locomotor activation in DAT-LE mice than in WT mice. The maximal velocity (Vmax ) of DAT-mediated [(3) H]DA uptake into striatal synaptosomes was reduced by 46% in DAT-LE mice, as compared to WT. Overall, considering the reduced number of DAT binding sites (Bmax ) along with the reduced Vmax in DAT-LE mice, a 2-fold increase in DA uptake turnover rate (Vmax /Bmax ) was found, relative to WT mice. This suggests that neuroadaptive changes have occurred in the DAT-LE mice that would help to compensate for their low DAT numbers. Interestingly, these changes do not include a reduction in tyrosine hydroxylase levels, as was previously reported in DAT knockout homozygous and heterozygous animals. Further, these changes are not sufficient to prevent elevated novelty- and cocaine-induced locomotor activity. Hence, these mice represent a unique model for studying changes of in vivo DAT function and regulation that result from markedly reduced levels of DAT expression.
Collapse
Affiliation(s)
- Anjali Rao
- Department of Pharmacology, School of Medicine, University of Colorado, Anschutz Medical Campus, Aurora, Colorado, 80045, USA.
| | | | | |
Collapse
|
32
|
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
|
33
|
Gallo G. Mechanisms underlying the initiation and dynamics of neuronal filopodia: from neurite formation to synaptogenesis. INTERNATIONAL REVIEW OF CELL AND MOLECULAR BIOLOGY 2013; 301:95-156. [PMID: 23317818 DOI: 10.1016/b978-0-12-407704-1.00003-8] [Citation(s) in RCA: 51] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/15/2022]
Abstract
Filopodia are finger-like cellular protrusions found throughout the metazoan kingdom and perform fundamental cellular functions during development and cell migration. Neurons exhibit a wide variety of extremely complex morphologies. In the nervous system, filopodia underlie many major morphogenetic events. Filopodia have roles spanning the initiation and guidance of neuronal processes, axons and dendrites to the formation of synaptic connections. This chapter addresses the mechanisms of the formation and dynamics of neuronal filopodia. Some of the major lessons learned from the study of neuronal filopodia are (1) there are multiple mechanisms that can regulate filopodia in a context-dependent manner, (2) that filopodia are specialized subcellular domains, (3) that filopodia exhibit dynamic membrane recycling which also controls aspects of filopodial dynamics, (4) that neuronal filopodia contain machinery for the orchestration of the actin and microtubule cytoskeleton, and (5) localized protein synthesis contributes to neuronal filopodial dynamics.
Collapse
Affiliation(s)
- Gianluca Gallo
- Shriners Hospitals Pediatric Research Center, Center for Neural Repair and Rehabilitation, Department of Anatomy and Cell Biology, Temple University, Philadelphia, PA, USA.
| |
Collapse
|
34
|
Abstract
Phenotypic analysis of gene-specific knockout (KO) mice has revolutionized our understanding of in vivo gene functions. As the use of mouse embryonic stem (ES) cells is inevitable for conventional gene targeting, the generation of knockout mice remains a very time-consuming and expensive process. To accelerate the large-scale production and phenotype analyses of KO mice, international efforts have organized global consortia such as the International Knockout Mouse Consortium (IKMC) and International Mouse Phenotype Consortium (IMPC), and they are persistently expanding the KO mouse catalogue that is publicly available for the researches studying specific genes of interests in vivo. However, new technologies, adopting zinc-finger nucleases (ZFNs) or Transcription Activator-Like Effector (TALE) Nucleases (TALENs) to edit the mouse genome, are now emerging as valuable and effective shortcuts alternative for the conventional gene targeting using ES cells. Here, we introduce the recent achievement of IKMC, and evaluate the significance of ZFN/TALEN technology in mouse genetics.
Collapse
Affiliation(s)
- Young Hoon Sung
- Department of Biochemistry, College of Life Science and Biotechnology, Laboratory Animal Research Center, Yonsei University, Seoul 120-749, Korea
| | - In-Jeoung Baek
- Asan Institute for Life Sciences, University of Ulsan College of Medicine, Seoul 138-736, Korea
| | - Je Kyung Seong
- College of Veterinary Medicine, Research Institute for Veterinary Science, Seoul National University, Korea
| | - Jin-Soo Kim
- Department of Chemistry, Seoul National University, Seoul 151-747, Korea
| | - Han-Woong Lee
- Department of Biochemistry, College of Life Science and Biotechnology, Laboratory Animal Research Center, Yonsei University, Seoul 120-749, Korea
| |
Collapse
|
35
|
Ruggiero A, Wright J, Ferguson SM, Lewis M, Emerson K, Iwamoto H, Ivy MT, Holmstrand EC, Ennis EA, Weaver CD, Blakely RD. Nonoisotopic assay for the presynaptic choline transporter reveals capacity for allosteric modulation of choline uptake. ACS Chem Neurosci 2012; 3:767-81. [PMID: 23077721 PMCID: PMC3474274 DOI: 10.1021/cn3000718] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/24/2012] [Accepted: 07/09/2012] [Indexed: 11/29/2022] Open
Abstract
Current therapies to enhance CNS cholinergic function rely primarily on extracellular acetylcholinesterase (AChE) inhibition, a pharmacotherapeutic strategy that produces dose-limiting side effects. The Na(+)-dependent, high-affinity choline transporter (CHT) is an unexplored target for cholinergic medication development. Although functional at the plasma membrane, CHT at steady-state is localized to synaptic vesicles such that vesicular fusion can support a biosynthetic response to neuronal excitation. To identify allosteric potentiators of CHT activity, we mapped endocytic sequences in the C-terminus of human CHT, identifying transporter mutants that exhibit significantly increased transport function. A stable HEK-293 cell line was generated from one of these mutants (CHT LV-AA) and used to establish a high-throughput screen (HTS) compatible assay based on the electrogenic nature of the transporter. We established that the addition of choline to these cells, at concentrations appropriate for high-affinity choline transport at presynaptic terminals, generates a hemicholinium-3 (HC-3)-sensitive, membrane depolarization that can be used for the screening of CHT inhibitors and activators. Using this assay, we discovered that staurosporine increased CHT LV-AA choline uptake activity, an effect mediated by a decrease in choline K(M) with no change in V(max). As staurosporine did not change surface levels of CHT, nor inhibit HC-3 binding, we propose that its action is directly or indirectly allosteric in nature. Surprisingly, staurosporine reduced choline-induced membrane depolarization, suggesting that increased substrate coupling to ion gradients, arising at the expense of nonstoichiometric ion flow, accompanies a shift of CHT to a higher-affinity state. Our findings provide a new approach for the identification of CHT modulators that is compatible with high-throughput screening approaches and presents a novel model by which small molecules can enhance substrate flux through enhanced gradient coupling.
Collapse
Affiliation(s)
- Alicia
M. Ruggiero
- Center for Molecular
Neuroscience,
Department of Pharmacology, Vanderbilt University School
of Medicine, Nashville, Tennessee 37232-8548, United
States
| | - Jane Wright
- Center for Molecular
Neuroscience,
Department of Pharmacology, Vanderbilt University School
of Medicine, Nashville, Tennessee 37232-8548, United
States
| | - Shawn M. Ferguson
- Center for Molecular
Neuroscience,
Department of Pharmacology, Vanderbilt University School
of Medicine, Nashville, Tennessee 37232-8548, United
States
| | - Michelle Lewis
- Vanderbilt Institute
of Chemical
Biology, Vanderbilt University School of Medicine, Nashville, Tennessee 37232-6304, United States
| | - Katie
S. Emerson
- Center for Molecular
Neuroscience,
Department of Pharmacology, Vanderbilt University School
of Medicine, Nashville, Tennessee 37232-8548, United
States
| | - Hideki Iwamoto
- Center for Molecular
Neuroscience,
Department of Pharmacology, Vanderbilt University School
of Medicine, Nashville, Tennessee 37232-8548, United
States
| | - Michael T. Ivy
- Department of Biological Sciences, Tennessee State University, Nashville, Tennessee 37209-1561,
United States
| | - Ericka C. Holmstrand
- Center for Molecular
Neuroscience,
Department of Pharmacology, Vanderbilt University School
of Medicine, Nashville, Tennessee 37232-8548, United
States
| | - Elizabeth. A. Ennis
- Center for Molecular
Neuroscience,
Department of Pharmacology, Vanderbilt University School
of Medicine, Nashville, Tennessee 37232-8548, United
States
| | - C. David Weaver
- Vanderbilt Institute
of Chemical
Biology, Vanderbilt University School of Medicine, Nashville, Tennessee 37232-6304, United States
- Department
of Pharmacology, Vanderbilt University School of Medicine, Nashville,
Tennessee 37232-6600, United States
| | - Randy D. Blakely
- Center for Molecular
Neuroscience,
Department of Pharmacology, Vanderbilt University School
of Medicine, Nashville, Tennessee 37232-8548, United
States
- Department of Psychiatry, Vanderbilt
University School of Medicine, Nashville,
Tennessee 37232-8548, United States
| |
Collapse
|