1
|
Shi Y, Feng X, Chung CY. Chronic adulthood exposure to bisphenol A causes behavioral changes via suppressing dopamine transporter trafficking. Sci Rep 2025; 15:13520. [PMID: 40253493 PMCID: PMC12009279 DOI: 10.1038/s41598-025-98084-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/26/2024] [Accepted: 04/09/2025] [Indexed: 04/21/2025] Open
Abstract
Evidence suggests that early life exposure to Bisphenol A (BPA) may impact neurobehavioral development in animals. BPA has been linked to changes in the dopamine level in the brain. However, molecular and cellular details of how BPA exposure causes these behavioral and cognitive outcomes are poorly understood. We examined how BPA affects the behaviors of adult mice and found that BPA induced hyperactivity and abnormal reward feedback in mice exposed at the early adult stage. We hypothesized that BPA might cause hyperactivity in mice by suppressing DAT trafficking. Fluorescence microscopy revealed that YFP-DAT remains in the perinuclear area when treated with BPA, compared to broader distribution throughout the cytoplasm in control cells. Results from MPTP toxicity and APP + uptake assays indicate that the surface expression of DAT was reduced by BPA treatment. Immunofluorescence staining of neurons in the Substantia nigra (SN) area of the mouse brain also revealed that DAT remains in the perinuclear region, indicating lower surface expression of DAT in the SN, playing important roles in reward and movement. We used another in vivo model, C. elegans, expressing GFP-tagged DAT-1 fusion protein and found that exposure to 50 µM BPA induced a significant increase in the frequency of body bends. However, the frequency of body bends was significantly reduced at 100 µM BPA, indicating biphasic effects of BPA. In conclusion, our results suggest that BPA contributes to the alterations of mice and worm behavior by reducing DAT expression on the surface of neurons via blocking of DAT trafficking.
Collapse
Affiliation(s)
- Yu Shi
- School of Pharmaceutical Science and Technology, Tianjin University, Tianjin, 300072, People's Republic of China
| | - Xiaoye Feng
- School of Pharmaceutical Science and Technology, Tianjin University, Tianjin, 300072, People's Republic of China
| | - Chang Y Chung
- School of Pharmaceutical Science and Technology, Tianjin University, Tianjin, 300072, People's Republic of China.
- Department of Biomedical Sciences, Mercer University School of Medicine, Columbus, GA, 31901, USA.
| |
Collapse
|
2
|
Kalra P, Grewal AK, Khan H, Singh TG. Unscrambling the cellular and molecular threads of Neuroplasticity: Insights into Alzheimer's disease pathogenesis. Neuroscience 2025; 571:74-88. [PMID: 39970983 DOI: 10.1016/j.neuroscience.2025.02.037] [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: 12/02/2024] [Revised: 01/14/2025] [Accepted: 02/15/2025] [Indexed: 02/21/2025]
Abstract
Alzheimer's disease (AD) is predominantly the most recurring and devastating neurological condition among the elderly population, characterized by the accumulation of amyloid-β (Aβ) and phosphorylated tau proteins, and is accompanied by progressive decline of learning and memory. Due to its complex and multifactorial etiology, a wide variety of therapeutic interventions have been developed. Despite constant advancements in the field, effective treatments that ameliorate the severity of Alzheimer's symptoms or cease their progression are still insufficient. Mounting evidence suggests that synaptic dysfunction could be an essential component of AD pathogenesis as synapse signaling is impaired in the aging brain, which contributes to synaptic decline. Therefore, improving neuroplasticity such as synaptic plasticity or neurogenesis could be a promising therapeutic approach for alleviating the effects of AD. This article reviews the cellular and molecular threads of neuroplasticity as well as targets that restore neuronal survival and plasticity to provide functional recoveries, including receptors, downstream signaling pathways, ion channels, transporters, enzymes, and neurotrophic factors.
Collapse
Affiliation(s)
- Palak Kalra
- Chitkara College of Pharmacy, Chitkara University, Rajpura, Punjab 140401, India; University School of Pharmaceutical Sciences, Rayat Bahra University, Mohali, Punjab 140103, India
| | - Amarjot Kaur Grewal
- Chitkara College of Pharmacy, Chitkara University, Rajpura, Punjab 140401, India.
| | - Heena Khan
- Chitkara College of Pharmacy, Chitkara University, Rajpura, Punjab 140401, India
| | - Thakur Gurjeet Singh
- Chitkara College of Pharmacy, Chitkara University, Rajpura, Punjab 140401, India.
| |
Collapse
|
3
|
Wang H, He H. Optical Precise Ablation of Targeted Individual Neurons In Vivo. ACS Chem Neurosci 2025; 16:374-383. [PMID: 39800970 DOI: 10.1021/acschemneuro.4c00538] [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] [Indexed: 02/06/2025] Open
Abstract
Targeted cell ablation is a powerful strategy for investigating the function of individual neurons within neuronal networks. Multiphoton ablation technology by a tightly focused femtosecond laser, with its significant advantages of noninvasiveness, high efficiency, and single-cell resolution, has been widely used in the study of neuroscience. However, the firing activity of the ablated neuron and its impact on the surrounding neurons and entire neuronal ensembles are still unclear. In this study, we describe the depolarization process of targeted neuron ablation by a femtosecond laser based on a standard two-photon microscope in vitro and in vivo. The photoporation damages the cell membrane, depolarizes the membrane potential, and thus disables the neuron's ability to fire action potentials. The dysfunctional neuron after laser ablation affects both the responses of surrounding neighbors and the functions of ensemble neurons in vivo. Although abnormal Ca2+ responses in spatially surrounding neurons are observed, the damage effect is confined to the focal volume. The function of the neuronal ensembles that associate with a specific visual stimulation is not influenced by the ablation of an individual member of the ensemble, indicating the redundancy of the ensemble organization. This study thus provides an insight into the targeted neuron ablation as well as the role of an individual neuron in an ensemble.
Collapse
Affiliation(s)
- Haipeng Wang
- School of Biomedical Engineering, Shanghai Jiao Tong University, Shanghai 200030, P. R. China
| | - Hao He
- School of Biomedical Engineering, Shanghai Jiao Tong University, Shanghai 200030, P. R. China
| |
Collapse
|
4
|
Li L, Ma R, Yuan Y, Yao Q, Han Y, Cao H, Qi J. Neurotoxicity induced by aged microplastics from plastic bowls: Abnormal neurotransmission in Caenorhabditis elegans. THE SCIENCE OF THE TOTAL ENVIRONMENT 2024; 952:175939. [PMID: 39218100 DOI: 10.1016/j.scitotenv.2024.175939] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/27/2024] [Revised: 07/30/2024] [Accepted: 08/29/2024] [Indexed: 09/04/2024]
Abstract
The use of plastic bowls (PB) has garnered increasing scrutiny due to the inevitable generation of microplastics (MPs) throughout their lifecycle. Despite this concern, there exists a limited understanding of the behaviors, toxicological effects, and mechanisms associated with aged PB (A-PB). This research investigated the photoaging properties of A-PB following ultraviolet irradiation and evaluated the neurotoxic impact of exposure to A-PB at environmentally relevant concentrations (0.001-1 mg/L) on Caenorhabditis elegans. Significant alterations in the crystallinity, elemental composition, and functional groups of A-PB were observed compared to virgin PB (V-PB), along with the emergence of environmentally persistent free radicals and reactive oxygen species. Toxicity assessments revealed that exposure to 0.1-1 mg/L A-PB induced greater neurotoxicity on locomotion behaviors compared to V-PB, as evidenced by marked reductions in head thrashes, body bends, wavelength, and mean amplitude. Exposure to A-PB also altered the fluorescence intensities and neurodegeneration percentage of dopaminergic, serotonergic, and GABAergic neurons, suggesting neuronal damage in the nematodes. Correspondingly, decreases in the levels of dopamine, serotonin, and GABA were noted together with significant drops in the expression of neurotransmitter-related genes (e.g., dat-1, tph-1, and unc-47). Correlation analyses established a significant positive relationship between these genes and locomotion behaviors. Further exploration showed the absence of locomotion behaviors in dat-1 (ok157), tph-1 (mg280), and unc-47 (e307) mutants, underscoring the pivotal roles of the dat-1, tph-1, and unc-47 genes in mediating neurotoxicity in C. elegans. This study sheds light on the photoaging characteristics and heightened toxicity of A-PB, elucidating the mechanisms driving A-PB-induced neurotoxicity.
Collapse
Affiliation(s)
- Liangzhong Li
- Guangdong Provincial Key Laboratory of High-Quality Recycling of End-of-Life New Energy Devices, Guangzhou Institute of Energy Conversion, Chinese Academy of Sciences, Guangzhou 510630, China
| | - Ruixue Ma
- State Environmental Protection Key Laboratory of Environmental Pollution Health Risk Assessment, South China Institute of Environmental Sciences, Ministry of Ecology and Environment, Guangzhou 510655, China
| | - Yuan Yuan
- Chengdu Research Academy of Environmental Protection Science, Chengdu 610072, China
| | - Qian Yao
- Guangdong Provincial Key Laboratory of High-Quality Recycling of End-of-Life New Energy Devices, Guangzhou Institute of Energy Conversion, Chinese Academy of Sciences, Guangzhou 510630, China
| | - Yajing Han
- State Environmental Protection Key Laboratory of Environmental Pollution Health Risk Assessment, South China Institute of Environmental Sciences, Ministry of Ecology and Environment, Guangzhou 510655, China.
| | - Hanlin Cao
- Technical Centre for Soil, Agriculture and Rural Ecology and Environment, Ministry of Ecology and Environment, Beijing 100012, China.
| | - Jianying Qi
- State Environmental Protection Key Laboratory of Environmental Pollution Health Risk Assessment, South China Institute of Environmental Sciences, Ministry of Ecology and Environment, Guangzhou 510655, China
| |
Collapse
|
5
|
Ambigapathy G, McCowan TJ, Carvelli L. Amphetamine exposure during embryogenesis changes expression and function of the dopamine transporter in Caenorhabditis elegans offspring. J Neurochem 2024; 168:2989-2998. [PMID: 38960397 PMCID: PMC11449651 DOI: 10.1111/jnc.16166] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/29/2024] [Revised: 06/12/2024] [Accepted: 06/13/2024] [Indexed: 07/05/2024]
Abstract
The dopamine transporter (DAT) is a transmembrane protein that regulates dopamine (DA) neurotransmission by binding to and moving DA from the synaptic cleft back into the neurons. Besides moving DA and other endogenous monoamines, DAT is also a neuronal carrier for exogenous compounds such as the psychostimulant amphetamine (Amph), and several studies have shown that Amph-induced behaviors require a functional DAT. Here, we demonstrate that exposure to Amph during early development causes behavioral, functional, and epigenetic modifications at the Caenorhabditis elegans DAT gene homolog, dat-1, in C. elegans offspring. Specifically, we show that, while embryos exposed to Amph generate adults that produce offspring with no obvious behavioral alterations, both adults and offspring exhibit an increased behavioral response when challenged with Amph. Our functional studies suggest that a decrease in DAT-1 expression underlies the increased behavioral response to Amph seen in offspring. Moreover, our epigenetic data suggest that histone methylation is a mechanism utilized by Amph to maintain changes in DAT-1 expression in offspring. Taken together, our data reveal that Amph, by altering the epigenetic landscape of DAT, propagates long-lasting functional and behavioral changes in offspring.
Collapse
Affiliation(s)
- Ganesh Ambigapathy
- Department of Biomedical Sciences, School of Medicine and Health Sciences, University of North Dakota, Grand Forks, North Dakota, USA
| | - Talus J McCowan
- Department of Biomedical Sciences, School of Medicine and Health Sciences, University of North Dakota, Grand Forks, North Dakota, USA
| | - Lucia Carvelli
- Harriet L. Wilkes Honors College Florida Atlantic University, Jupiter, Florida, USA
- Stiles-Nicholson Brain Institute, Florida Atlantic University, Jupiter, Florida, USA
| |
Collapse
|
6
|
Roustaee S, Sani M, Mehranpour M, Raee P, Moghaddam MH, Bahar R, Nourirad SN, Golzarian MJ, Beirami A, Jafary H, Aalipour MA, Taghizadeh M, Abdollahifar MA, Vakili K, Fathi M, Heidari MH, Abbaszadeh HA, Aliaghaei A, Nazarian H. Chronic Administration of Lisdexamfetamine Induces Apoptosis and Inflammation and Reduces Sperm Quality in Adult Male Rats. Reprod Sci 2024; 31:1278-1289. [PMID: 38228974 DOI: 10.1007/s43032-023-01449-9] [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: 09/11/2023] [Accepted: 12/28/2023] [Indexed: 01/18/2024]
Abstract
Concerns have been raised about potentially irreversible brain damage and damage to the neuroendocrine system during development when treating attention-deficit/hyperactivity disorder with lisdexamfetamine (LDX), a norepinephrine dopamine reuptake inhibitor. This study aims to elucidate the potential adverse effects of LDX on the male reproductive system due to its widespread use and potential for abuse. In this study, adult male rats were randomized into control and LDX groups. Thirty milligrams per kilogram LDX was administered orally for 3 weeks. After isolation of epididymal spermatozoa, the rats were euthanized and testicular tissues were collected for stereological and molecular analyses. The LDX group showed a decrease in sperm motility and an increase in DNA fragmentation compared to the control group. There was also a dramatic decrease in testosterone in the LDX group. Testicular expression of caspase-3 and TNF-α was significantly increased in the LDX group. According to our findings, prolonged use of LDX leads to reduced sperm quality. It also induces apoptosis, inflammatory response, and pathological changes in the testicular tissue. What we have observed in this study is noteworthy but requires further investigation, particularly in people who use LDX over a longer period of time.
Collapse
Affiliation(s)
- Susan Roustaee
- Men's Health and Reproductive Health Research Center, Shahid Beheshti University of Medical Sciences, Tehran, Iran
- Department of Biology and Anatomical Sciences, School of Medicine, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Mojtaba Sani
- Department of Educational Neuroscience, Aras International Campus, University of Tabriz, Tabriz, Iran
| | - Maryam Mehranpour
- Department of Genetics, Faculty of Biological Sciences, North Tehran Branch, Islamic Azad University, Tehran, Iran
| | - Pourya Raee
- Department of Biology and Anatomical Sciences, School of Medicine, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Meysam Hassani Moghaddam
- Department of Anatomical Sciences, Faculty of Medicine, AJA University of Medical Sciences, Tehran, Iran
| | - Reza Bahar
- Department of Biology and Anatomical Sciences, School of Medicine, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Seyedeh Naghmeh Nourirad
- Department of Biology and Anatomical Sciences, School of Medicine, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Mohammad-Jasim Golzarian
- Department of Biology and Anatomical Sciences, School of Medicine, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Amirreza Beirami
- Department of Biology and Anatomical Sciences, School of Medicine, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Hosein Jafary
- Department of Biology and Anatomical Sciences, School of Medicine, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Mohammad Amin Aalipour
- Department of Biology and Anatomical Sciences, School of Medicine, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Mohammad Taghizadeh
- Department of Biology and Anatomical Sciences, School of Medicine, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Mohammad-Amin Abdollahifar
- Department of Biology and Anatomical Sciences, School of Medicine, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Kimia Vakili
- Student Research Committee, Faculty of Medicine, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Mobina Fathi
- Student Research Committee, Faculty of Medicine, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Mohammad Hossain Heidari
- Department of Biology and Anatomical Sciences, School of Medicine, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Hojjat-Allah Abbaszadeh
- Department of Biology and Anatomical Sciences, School of Medicine, Shahid Beheshti University of Medical Sciences, Tehran, Iran
- Laser Application in Medical Sciences Research Center, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Abbas Aliaghaei
- Men's Health and Reproductive Health Research Center, Shahid Beheshti University of Medical Sciences, Tehran, Iran.
- Department of Biology and Anatomical Sciences, School of Medicine, Shahid Beheshti University of Medical Sciences, Tehran, Iran.
| | - Hamid Nazarian
- Men's Health and Reproductive Health Research Center, Shahid Beheshti University of Medical Sciences, Tehran, Iran.
- Department of Biology and Anatomical Sciences, School of Medicine, Shahid Beheshti University of Medical Sciences, Tehran, Iran.
| |
Collapse
|
7
|
Vaughan RA, Henry LK, Foster JD, Brown CR. Post-translational mechanisms in psychostimulant-induced neurotransmitter efflux. ADVANCES IN PHARMACOLOGY (SAN DIEGO, CALIF.) 2024; 99:1-33. [PMID: 38467478 DOI: 10.1016/bs.apha.2023.10.003] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 03/13/2024]
Abstract
The availability of monoamine neurotransmitters in the brain is under the control of dopamine, norepinephrine, and serotonin transporters expressed on the plasma membrane of monoaminergic neurons. By regulating transmitter levels these proteins mediate crucial functions including cognition, attention, and reward, and dysregulation of their activity is linked to mood and psychiatric disorders of these systems. Amphetamine-based transporter substrates stimulate non-exocytotic transmitter efflux that induces psychomotor stimulation, addiction, altered mood, hallucinations, and psychosis, thus constituting a major component of drug neurochemical and behavioral outcomes. Efflux is under the control of transporter post-translational modifications that synergize with other regulatory events, and this review will summarize our knowledge of these processes and their role in drug mechanisms.
Collapse
Affiliation(s)
- Roxanne A Vaughan
- Department of Biomedical Sciences, University of North Dakota School of Medicine and Health Sciences, Grand Forks, ND, United States.
| | - L Keith Henry
- Department of Biomedical Sciences, University of North Dakota School of Medicine and Health Sciences, Grand Forks, ND, United States
| | - James D Foster
- Department of Biomedical Sciences, University of North Dakota School of Medicine and Health Sciences, Grand Forks, ND, United States
| | - Christopher R Brown
- Department of Biomedical Sciences, University of North Dakota School of Medicine and Health Sciences, Grand Forks, ND, United States
| |
Collapse
|
8
|
Shaikh A, Ahmad F, Teoh SL, Kumar J, Yahaya MF. Targeting dopamine transporter to ameliorate cognitive deficits in Alzheimer's disease. Front Cell Neurosci 2023; 17:1292858. [PMID: 38026688 PMCID: PMC10679733 DOI: 10.3389/fncel.2023.1292858] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/12/2023] [Accepted: 10/27/2023] [Indexed: 12/01/2023] Open
Abstract
Alzheimer's disease (AD) is characterized by the pathologic deposition of amyloid and neurofibrillary tangles in the brain, leading to neuronal damage and defective synapses. These changes manifest as abnormalities in cognition and behavior. The functional deficits are also attributed to abnormalities in multiple neurotransmitter systems contributing to neuronal dysfunction. One such important system is the dopaminergic system. It plays a crucial role in modulating movement, cognition, and behavior while connecting various brain areas and influencing other neurotransmitter systems, making it relevant in neurodegenerative disorders like AD and Parkinson's disease (PD). Considering its significance, the dopaminergic system has emerged as a promising target for alleviating movement and cognitive deficits in PD and AD, respectively. Extensive research has been conducted on dopaminergic neurons, receptors, and dopamine levels as critical factors in cognition and memory in AD. However, the exact nature of movement abnormalities and other features of extrapyramidal symptoms are not fully understood yet in AD. Recently, a previously overlooked element of the dopaminergic system, the dopamine transporter, has shown significant promise as a more effective target for enhancing cognition while addressing dopaminergic system dysfunction in AD.
Collapse
Affiliation(s)
- Ammara Shaikh
- Department of Anatomy, Faculty of Medicine, Universiti Kebangsaan Malaysia, Cheras, Kuala Lumpur, Malaysia
| | - Fairus Ahmad
- Department of Anatomy, Faculty of Medicine, Universiti Kebangsaan Malaysia, Cheras, Kuala Lumpur, Malaysia
| | - Seong Lin Teoh
- Department of Anatomy, Faculty of Medicine, Universiti Kebangsaan Malaysia, Cheras, Kuala Lumpur, Malaysia
| | - Jaya Kumar
- Department of Physiology, Faculty of Medicine, Universiti Kebangsaan Malaysia, Cheras, Kuala Lumpur, Malaysia
| | - Mohamad Fairuz Yahaya
- Department of Anatomy, Faculty of Medicine, Universiti Kebangsaan Malaysia, Cheras, Kuala Lumpur, Malaysia
| |
Collapse
|
9
|
Nguyen VT, Harris AC, Eltit JM. Structural and functional perspectives on interactions between synthetic cathinones and monoamine transporters. ADVANCES IN PHARMACOLOGY (SAN DIEGO, CALIF.) 2023; 99:83-124. [PMID: 38467490 DOI: 10.1016/bs.apha.2023.09.001] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 03/13/2024]
Abstract
Synthetic cathinone derivatives comprise a family of psychoactive compounds structurally related to amphetamine. Over the last decade, clandestine chemists have synthesized a consistent stream of innovative cathinone derivatives to outpace governmental regulatory restrictions. Many of these unregulated substances are produced and distributed as designer drugs. Two of the principal chemical scaffolds exploited to expand the synthetic cathinone family are methcathinone and α-pyrrolidinopentiophenone (or α-pyrrolidinovalerophenone, α-PVP). These compounds' main physiological targets are monoamine transporters, where they promote addiction by potentiating dopaminergic neurotransmission. This chapter describes techniques used to study the pharmacodynamic properties of cathinones at monoamine transporters in vitro. Biochemical techniques described include uptake inhibition and release assays in rat brain synaptosomes and in mammalian expression systems. Electrophysiological techniques include current measurements using the voltage clamp technique. We describe a Ca2+ mobilization assay wherein voltage-gated Ca2+ channels function as reporters to study the action of synthetic cathinones at monoamine transporters. We discuss results from systematic structure-activity relationship studies on simple and complex cathinones at monoamine transporters with an emphasis on identifying structural moieties that modulate potency and selectivity at these transporters. Moreover, different profiles of selectivity at monoamine transporters directly predict compounds associated with behavioral and subjective effects within animals and humans. In conclusion, clarification of the structural aspects of compounds which modulate potency and selectivity at monoamine transporters is critical to identify and predict potential addictive drugs. This knowledge may allow prompt allocation of resources toward drugs that represent the greatest threats after drugs are identified by forensic laboratories.
Collapse
Affiliation(s)
- Vy T Nguyen
- Department of Physiology and Biophysics, School of Medicine, Virginia Commonwealth University, Richmond, VA, United States
| | - Alan C Harris
- Department of Physiology and Biophysics, School of Medicine, Virginia Commonwealth University, Richmond, VA, United States
| | - Jose M Eltit
- Department of Physiology and Biophysics, School of Medicine, Virginia Commonwealth University, Richmond, VA, United States.
| |
Collapse
|
10
|
Davis SE, Cirincione AB, Jimenez-Torres AC, Zhu J. The Impact of Neurotransmitters on the Neurobiology of Neurodegenerative Diseases. Int J Mol Sci 2023; 24:15340. [PMID: 37895020 PMCID: PMC10607327 DOI: 10.3390/ijms242015340] [Citation(s) in RCA: 9] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/26/2023] [Revised: 10/16/2023] [Accepted: 10/17/2023] [Indexed: 10/29/2023] Open
Abstract
Neurodegenerative diseases affect millions of people worldwide. Neurodegenerative diseases result from progressive damage to nerve cells in the brain or peripheral nervous system connections that are essential for cognition, coordination, strength, sensation, and mobility. Dysfunction of these brain and nerve functions is associated with Alzheimer's disease, Parkinson's disease, Huntington's disease, Amyotrophic lateral sclerosis, and motor neuron disease. In addition to these, 50% of people living with HIV develop a spectrum of cognitive, motor, and/or mood problems collectively referred to as HIV-Associated Neurocognitive Disorders (HAND) despite the widespread use of a combination of antiretroviral therapies. Neuroinflammation and neurotransmitter systems have a pathological correlation and play a critical role in developing neurodegenerative diseases. Each of these diseases has a unique pattern of dysregulation of the neurotransmitter system, which has been attributed to different forms of cell-specific neuronal loss. In this review, we will focus on a discussion of the regulation of dopaminergic and cholinergic systems, which are more commonly disturbed in neurodegenerative disorders. Additionally, we will provide evidence for the hypothesis that disturbances in neurotransmission contribute to the neuronal loss observed in neurodegenerative disorders. Further, we will highlight the critical role of dopamine as a mediator of neuronal injury and loss in the context of NeuroHIV. This review will highlight the need to further investigate neurotransmission systems for their role in the etiology of neurodegenerative disorders.
Collapse
Affiliation(s)
| | | | | | - Jun Zhu
- Department of Drug Discovery and Biomedical Sciences, College of Pharmacy, University of South Carolina, 715 Sumter Street, Columbia, SC 29208, USA; (S.E.D.); (A.B.C.); (A.C.J.-T.)
| |
Collapse
|
11
|
Delignat-Lavaud B, Kano J, Ducrot C, Massé I, Mukherjee S, Giguère N, Moquin L, Lévesque C, Burke S, Denis R, Bourque MJ, Tchung A, Rosa-Neto P, Lévesque D, De Beaumont L, Trudeau LÉ. Synaptotagmin-1-dependent phasic axonal dopamine release is dispensable for basic motor behaviors in mice. Nat Commun 2023; 14:4120. [PMID: 37433762 PMCID: PMC10336101 DOI: 10.1038/s41467-023-39805-7] [Citation(s) in RCA: 9] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/18/2021] [Accepted: 06/27/2023] [Indexed: 07/13/2023] Open
Abstract
In Parkinson's disease (PD), motor dysfunctions only become apparent after extensive loss of DA innervation. This resilience has been hypothesized to be due to the ability of many motor behaviors to be sustained through a diffuse basal tone of DA; but experimental evidence for this is limited. Here we show that conditional deletion of the calcium sensor synaptotagmin-1 (Syt1) in DA neurons (Syt1 cKODA mice) abrogates most activity-dependent axonal DA release in the striatum and mesencephalon, leaving somatodendritic (STD) DA release intact. Strikingly, Syt1 cKODA mice showed intact performance in multiple unconditioned DA-dependent motor tasks and even in a task evaluating conditioned motivation for food. Considering that basal extracellular DA levels in the striatum were unchanged, our findings suggest that activity-dependent DA release is dispensable for such tasks and that they can be sustained by a basal tone of extracellular DA. Taken together, our findings reveal the striking resilience of DA-dependent motor functions in the context of a near-abolition of phasic DA release, shedding new light on why extensive loss of DA innervation is required to reveal motor dysfunctions in PD.
Collapse
Affiliation(s)
- Benoît Delignat-Lavaud
- Department of Pharmacology and Physiology, Faculty of Medicine, Université de Montréal, Montreal, QC, Canada
- Department of Neurosciences, Faculty of Medicine, Université de Montréal, Montreal, QC, Canada
- SNC and CIRCA Research Groups, Université de Montréal, Montréal, QC, Canada
| | - Jana Kano
- Department of Pharmacology and Physiology, Faculty of Medicine, Université de Montréal, Montreal, QC, Canada
- Department of Neurosciences, Faculty of Medicine, Université de Montréal, Montreal, QC, Canada
- SNC and CIRCA Research Groups, Université de Montréal, Montréal, QC, Canada
| | - Charles Ducrot
- Department of Pharmacology and Physiology, Faculty of Medicine, Université de Montréal, Montreal, QC, Canada
- Department of Neurosciences, Faculty of Medicine, Université de Montréal, Montreal, QC, Canada
- SNC and CIRCA Research Groups, Université de Montréal, Montréal, QC, Canada
| | - Ian Massé
- Hôpital du Sacré-Cœur-de-Montréal, CIUSSS NIM, Université de Montréal, Montreal, QC, Canada
| | - Sriparna Mukherjee
- Department of Pharmacology and Physiology, Faculty of Medicine, Université de Montréal, Montreal, QC, Canada
- Department of Neurosciences, Faculty of Medicine, Université de Montréal, Montreal, QC, Canada
- SNC and CIRCA Research Groups, Université de Montréal, Montréal, QC, Canada
- Aligning Science Across Parkinson's (ASAP) Collaborative Research Network, Chevy Chase, MD, 20815, USA
| | - Nicolas Giguère
- Department of Pharmacology and Physiology, Faculty of Medicine, Université de Montréal, Montreal, QC, Canada
- Department of Neurosciences, Faculty of Medicine, Université de Montréal, Montreal, QC, Canada
- SNC and CIRCA Research Groups, Université de Montréal, Montréal, QC, Canada
| | - Luc Moquin
- Centre intégré universitaire de santé et de services sociaux (CIUSSS) de l'Ouest-de-l'Île-de-Montréal; Department of Neurology and Neurosurgery, Psychiatry and Pharmacology and Therapeutics, McGill University, Montreal, QC, Canada
| | | | - Samuel Burke
- Department of Pharmacology and Physiology, Faculty of Medicine, Université de Montréal, Montreal, QC, Canada
- Department of Neurosciences, Faculty of Medicine, Université de Montréal, Montreal, QC, Canada
- SNC and CIRCA Research Groups, Université de Montréal, Montréal, QC, Canada
| | - Raphaëlle Denis
- Department of Pharmacology and Physiology, Faculty of Medicine, Université de Montréal, Montreal, QC, Canada
- Department of Neurosciences, Faculty of Medicine, Université de Montréal, Montreal, QC, Canada
- SNC and CIRCA Research Groups, Université de Montréal, Montréal, QC, Canada
| | - Marie-Josée Bourque
- Department of Pharmacology and Physiology, Faculty of Medicine, Université de Montréal, Montreal, QC, Canada
- Department of Neurosciences, Faculty of Medicine, Université de Montréal, Montreal, QC, Canada
- SNC and CIRCA Research Groups, Université de Montréal, Montréal, QC, Canada
- Aligning Science Across Parkinson's (ASAP) Collaborative Research Network, Chevy Chase, MD, 20815, USA
| | - Alex Tchung
- Department of Pharmacology and Physiology, Faculty of Medicine, Université de Montréal, Montreal, QC, Canada
- Department of Neurosciences, Faculty of Medicine, Université de Montréal, Montreal, QC, Canada
- SNC and CIRCA Research Groups, Université de Montréal, Montréal, QC, Canada
| | - Pedro Rosa-Neto
- Centre intégré universitaire de santé et de services sociaux (CIUSSS) de l'Ouest-de-l'Île-de-Montréal; Department of Neurology and Neurosurgery, Psychiatry and Pharmacology and Therapeutics, McGill University, Montreal, QC, Canada
| | - Daniel Lévesque
- Faculty of Pharmacy, Université de Montréal, Montreal, QC, Canada
| | - Louis De Beaumont
- Hôpital du Sacré-Cœur-de-Montréal, CIUSSS NIM, Université de Montréal, Montreal, QC, Canada
| | - Louis-Éric Trudeau
- Department of Pharmacology and Physiology, Faculty of Medicine, Université de Montréal, Montreal, QC, Canada.
- Department of Neurosciences, Faculty of Medicine, Université de Montréal, Montreal, QC, Canada.
- SNC and CIRCA Research Groups, Université de Montréal, Montréal, QC, Canada.
- Aligning Science Across Parkinson's (ASAP) Collaborative Research Network, Chevy Chase, MD, 20815, USA.
| |
Collapse
|
12
|
Gibson SB, Ness-Cohn E, Andersen EC. Benzimidazoles cause lethality by inhibiting the function of Caenorhabditis elegans neuronal beta-tubulin. Int J Parasitol Drugs Drug Resist 2022; 20:89-96. [PMID: 36332489 PMCID: PMC9771835 DOI: 10.1016/j.ijpddr.2022.10.004] [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: 07/19/2022] [Revised: 10/17/2022] [Accepted: 10/18/2022] [Indexed: 11/08/2022]
Abstract
Parasitic nematode infections cause an enormous global burden to both humans and livestock. Resistance to the limited arsenal of anthelmintic drugs used to combat these infections is widespread, including benzimidazole (BZ) compounds. Previous studies using the free-living nematode Caenorhabditis elegans to model parasitic nematode resistance have shown that loss-of-function mutations in the beta-tubulin gene ben-1 confer resistance to BZ drugs. However, the mechanism of resistance and the tissue-specific susceptibility are not well known in any nematode species. To identify in which tissue(s) ben-1 function underlies BZ susceptibility, transgenic strains that express ben-1 in different tissues, including hypodermis, muscles, neurons, intestine, and ubiquitous expression were generated. High-throughput fitness assays were performed to measure and compare the quantitative responses to BZ compounds among different transgenic lines. Significant BZ susceptibility was observed in animals expressing ben-1 in neurons, comparable to expression using the ben-1 promoter. This result suggests that ben-1 function in neurons underlies susceptibility to BZ. Subsetting neuronal expression of ben-1 based on the neurotransmitter system further restricted ben-1 function in cholinergic neurons to cause BZ susceptibility. These results better inform our current understanding of the cellular mode of action of BZs and also suggest additional treatments that might potentiate the effects of BZs in neurons.
Collapse
Affiliation(s)
- Sophia B Gibson
- Department of Molecular Biosciences, Northwestern University, Evanston, IL, 60208, USA
| | - Elan Ness-Cohn
- Department of Molecular Biosciences, Northwestern University, Evanston, IL, 60208, USA; Driskill Graduate Program in Life Sciences, Northwestern University, Chicago, IL, 60611, USA
| | - Erik C Andersen
- Department of Molecular Biosciences, Northwestern University, Evanston, IL, 60208, USA.
| |
Collapse
|
13
|
Catale C, Lo Iacono L, Martini A, Heil C, Guatteo E, Mercuri NB, Viscomi MT, Palacios D, Carola V. Early Life Social Stress Causes Sex- and Region-Dependent Dopaminergic Changes that Are Prevented by Minocycline. Mol Neurobiol 2022; 59:3913-3932. [PMID: 35435618 PMCID: PMC9148283 DOI: 10.1007/s12035-022-02830-6] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/29/2021] [Accepted: 04/02/2022] [Indexed: 02/03/2023]
Abstract
Early life stress (ELS) is known to modify trajectories of brain dopaminergic development, but the mechanisms underlying have not been determined. ELS perturbs immune system and microglia reactivity, and inflammation and microglia influence dopaminergic transmission and development. Whether microglia mediate the effects of ELS on dopamine (DA) system development is still unknown. We explored the effects of repeated early social stress on development of the dopaminergic system in male and female mice through histological, electrophysiological, and transcriptomic analyses. Furthermore, we tested whether these effects could be mediated by ELS-induced altered microglia/immune activity through a pharmacological approach. We found that social stress in early life altered DA neurons morphology, reduced dopamine transporter (DAT) and tyrosine hydroxylase expression, and lowered DAT-mediated currents in the ventral tegmental area but not substantia nigra of male mice only. Notably, stress-induced DA alterations were prevented by minocycline, an inhibitor of microglia activation. Transcriptome analysis in the developing male ventral tegmental area revealed that ELS caused downregulation of dopaminergic transmission and alteration in hormonal and peptide signaling pathways. Results from this study offer new insight into the mechanisms of stress response and altered brain dopaminergic maturation after ELS, providing evidence of neuroimmune interaction, sex differences, and regional specificity.
Collapse
Affiliation(s)
- Clarissa Catale
- Division of Experimental Neuroscience, Neurobiology of Behavior Laboratory, IRCCS Santa Lucia Foundation, Rome, Italy
| | - Luisa Lo Iacono
- Department of Dynamic and Clinical Psychology, and Health Studies, Sapienza University of Rome, Via degli Apuli 1, Rome, Italy
| | - Alessandro Martini
- Division of Experimental Neuroscience, Experimental Neurology Laboratory, IRCCS Santa Lucia Foundation, Rome, Italy
| | - Constantin Heil
- Division of Experimental Neuroscience, Epigenetics and Signal Transduction Laboratory, IRCCS Santa Lucia Foundation, Rome, Italy
| | - Ezia Guatteo
- Division of Experimental Neuroscience, Experimental Neurology Laboratory, IRCCS Santa Lucia Foundation, Rome, Italy
- Department of Motor Science and Wellness, University of Naples Parthenope, Naples, Italy
| | - Nicola Biagio Mercuri
- Division of Experimental Neuroscience, Experimental Neurology Laboratory, IRCCS Santa Lucia Foundation, Rome, Italy
- Department of Systems Medicine, Università Degli Studi Di Roma Tor Vergata, Rome, Italy
| | - Maria Teresa Viscomi
- Department of Life Science and Public Health, Section of Histology and Embryology, Università Cattolica Del S. Cuore, Rome, Italy
- IRCCS Fondazione Policlinico Universitario A. Gemelli, Rome, Italy
| | - Daniela Palacios
- Division of Experimental Neuroscience, Epigenetics and Signal Transduction Laboratory, IRCCS Santa Lucia Foundation, Rome, Italy
- IRCCS Fondazione Policlinico Universitario A. Gemelli, Rome, Italy
- Department of Life Science and Public Health, Section of Biology, Università Cattolica Del S. Cuore, Rome, Italy
| | - Valeria Carola
- Division of Experimental Neuroscience, Neurobiology of Behavior Laboratory, IRCCS Santa Lucia Foundation, Rome, Italy.
- Department of Dynamic and Clinical Psychology, and Health Studies, Sapienza University of Rome, Via degli Apuli 1, Rome, Italy.
| |
Collapse
|
14
|
Chou SH, Chen YJ, Liao CP, Pan CL. A role for dopamine in C. elegans avoidance behavior induced by mitochondrial stress. Neurosci Res 2022; 178:87-92. [PMID: 35074444 DOI: 10.1016/j.neures.2022.01.005] [Citation(s) in RCA: 11] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/19/2021] [Revised: 12/21/2021] [Accepted: 01/17/2022] [Indexed: 01/11/2023]
Abstract
Physiological stress triggers aversive learning that profoundly alters animal behavior. Systemic mitochondrial disruption induces avoidance of C. elegans to non-pathogenic food bacteria. Mutations in cat-2 and dat-1, which control dopamine synthesis and reuptake, respectively, impair this learned bacterial avoidance, suggesting that dopaminergic modulation is essential. Cell-specific rescue experiments indicate that dopamine likely acts from the CEP and ADE neurons to regulate learned bacterial avoidance. We find that mutations in multiple dopamine receptor genes, including dop-1, dop-2 and dop-3, reduced learned bacterial avoidance. Our work reveals a role for dopamine signaling in C. elegans learned avoidance behavior induced by mitochondrial stress.
Collapse
Affiliation(s)
- Shih-Hua Chou
- Institute of Molecular Medicine, College of Medicine, National Taiwan University, Taipei 10002, Taiwan; Center of Precision Medicine, College of Medicine, National Taiwan University, Taipei 10002, Taiwan
| | - Yen-Ju Chen
- Institute of Molecular Medicine, College of Medicine, National Taiwan University, Taipei 10002, Taiwan; Center of Precision Medicine, College of Medicine, National Taiwan University, Taipei 10002, Taiwan
| | - Chien-Po Liao
- Institute of Molecular Medicine, College of Medicine, National Taiwan University, Taipei 10002, Taiwan
| | - Chun-Liang Pan
- Institute of Molecular Medicine, College of Medicine, National Taiwan University, Taipei 10002, Taiwan; Center of Precision Medicine, College of Medicine, National Taiwan University, Taipei 10002, Taiwan.
| |
Collapse
|
15
|
Juhaszova M, Kobrinsky E, Zorov DB, Nuss HB, Yaniv Y, Fishbein KW, de Cabo R, Montoliu L, Gabelli SB, Aon MA, Cortassa S, Sollott SJ. ATP Synthase K +- and H +-Fluxes Drive ATP Synthesis and Enable Mitochondrial K +-"Uniporter" Function: I. Characterization of Ion Fluxes. FUNCTION (OXFORD, ENGLAND) 2021; 3:zqab065. [PMID: 35229078 PMCID: PMC8867323 DOI: 10.1093/function/zqab065] [Citation(s) in RCA: 25] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/07/2021] [Revised: 12/06/2021] [Accepted: 12/07/2021] [Indexed: 01/07/2023]
Abstract
ATP synthase (F1Fo) synthesizes daily our body's weight in ATP, whose production-rate can be transiently increased several-fold to meet changes in energy utilization. Using purified mammalian F1Fo-reconstituted proteoliposomes and isolated mitochondria, we show F1Fo can utilize both ΔΨm-driven H+- and K+-transport to synthesize ATP under physiological pH = 7.2 and K+ = 140 mEq/L conditions. Purely K+-driven ATP synthesis from single F1Fo molecules measured by bioluminescence photon detection could be directly demonstrated along with simultaneous measurements of unitary K+ currents by voltage clamp, both blocked by specific Fo inhibitors. In the presence of K+, compared to osmotically-matched conditions in which this cation is absent, isolated mitochondria display 3.5-fold higher rates of ATP synthesis, at the expense of 2.6-fold higher rates of oxygen consumption, these fluxes being driven by a 2.7:1 K+: H+ stoichiometry. The excellent agreement between the functional data obtained from purified F1Fo single molecule experiments and ATP synthase studied in the intact mitochondrion under unaltered OxPhos coupling by K+ presence, is entirely consistent with K+ transport through the ATP synthase driving the observed increase in ATP synthesis. Thus, both K+ (harnessing ΔΨm) and H+ (harnessing its chemical potential energy, ΔμH) drive ATP generation during normal physiology.
Collapse
Affiliation(s)
| | | | | | | | | | - Kenneth W Fishbein
- Laboratory of Clinical Investigation, National Institute on Aging, NIH, Baltimore, MD 21224, USA
| | - Rafael de Cabo
- Translational Gerontology Branch, National Institute on Aging, NIH, Baltimore, MD 21224, USA
| | - Lluis Montoliu
- National Centre for Biotechnology (CNB-CSIC), Biomedical Research Networking Center on Rare Diseases (CIBERER-ISCIII), 28049, Madrid, Spain
| | - Sandra B Gabelli
- Department of Medicine, Johns Hopkins University School of Medicine, Baltimore, MD 21205, USA,Department of Oncology, Johns Hopkins University School of Medicine, Baltimore, MD 21205, USA,Department of Biophysics and Biophysical Chemistry, Johns Hopkins University School of Medicine, Baltimore, MD 21205, USA
| | - Miguel A Aon
- Laboratory of Cardiovascular Science, National Institute on Aging, NIH, Baltimore, MD 21224, USA,Translational Gerontology Branch, National Institute on Aging, NIH, Baltimore, MD 21224, USA
| | - Sonia Cortassa
- Laboratory of Cardiovascular Science, National Institute on Aging, NIH, Baltimore, MD 21224, USA
| | | |
Collapse
|
16
|
The gating pore blocker 1-(2,4-xylyl)guanidinium selectively inhibits pacemaking of midbrain dopaminergic neurons. Neuropharmacology 2021; 197:108722. [PMID: 34273387 DOI: 10.1016/j.neuropharm.2021.108722] [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: 03/27/2021] [Revised: 07/08/2021] [Accepted: 07/13/2021] [Indexed: 11/22/2022]
Abstract
Although several ionic mechanisms are known to control rate and regularity of the slow pacemaker in dopamine (DA) neurons, the core mechanism of pacing is controversial. Here we tested the hypothesis that pacemaking of SNc DA neurons is enabled by an unconventional conductance. We found that 1-(2,4-xylyl)guanidinium (XG), an established blocker of gating pore currents, selectively inhibits pacemaking of DA neurons. The compound inhibited all slow pacemaking DA neurons that were tested, both in the substantia nigra pars compacta, and in the ventral tegmental area. Interestingly, bursting behavior was not affected by XG. Furthermore, the drug did not affect fast pacemaking of GABAergic neurons from substantia nigra pars reticulata neurons or slow pacemaking of noradrenergic neurons. In DA neurons, current-clamp analysis revealed that XG did not appear to affect ion channels involved in the action potential. Its inhibitory effect persisted during blockade of all ion channels previously suggested to contribute to pacemaking. RNA sequencing and voltage-clamp recordings yielded no evidence for a gating pore current to underlie the conductance. However, we could isolate a small subthreshold XG-sensitive current, which was carried by both Na+ and Cl- ions. Although the molecular target of XG remains to be defined, these observations represent a step towards understanding pacemaking in DA neurons.
Collapse
|
17
|
Bhat S, Niello M, Schicker K, Pifl C, Sitte HH, Freissmuth M, Sandtner W. Handling of intracellular K + determines voltage dependence of plasmalemmal monoamine transporter function. eLife 2021; 10:67996. [PMID: 34061030 PMCID: PMC8192120 DOI: 10.7554/elife.67996] [Citation(s) in RCA: 21] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/01/2021] [Accepted: 05/30/2021] [Indexed: 12/16/2022] Open
Abstract
The concentrative power of the transporters for dopamine (DAT), norepinephrine (NET), and serotonin (SERT) is thought to be fueled by the transmembrane Na+ gradient, but it is conceivable that they can also tap other energy sources, for example, membrane voltage and/or the transmembrane K+ gradient. We have addressed this by recording uptake of endogenous substrates or the fluorescent substrate APP+(4-(4-dimethylamino)phenyl-1-methylpyridinium) under voltage control in cells expressing DAT, NET, or SERT. We have shown that DAT and NET differ from SERT in intracellular handling of K+. In DAT and NET, substrate uptake was voltage-dependent due to the transient nature of intracellular K+ binding, which precluded K+ antiport. SERT, however, antiports K+ and achieves voltage-independent transport. Thus, there is a trade-off between maintaining constant uptake and harvesting membrane potential for concentrative power, which we conclude to occur due to subtle differences in the kinetics of co-substrate ion binding in closely related transporters.
Collapse
Affiliation(s)
- Shreyas Bhat
- Institute of Pharmacology and the Gaston H. Glock Research Laboratories for Exploratory Drug Development, Center of Physiology and Pharmacology, Medical University of Vienna, Vienna, Austria
| | - Marco Niello
- Institute of Pharmacology and the Gaston H. Glock Research Laboratories for Exploratory Drug Development, Center of Physiology and Pharmacology, Medical University of Vienna, Vienna, Austria
| | - Klaus Schicker
- Division of Neurophysiology and Neuropharmacology, Centre for Physiology and Pharmacology, Medical University of Vienna, Vienna, Austria
| | - Christian Pifl
- Center for Brain Research, Medical University of Vienna, Vienna, Austria
| | - Harald H Sitte
- Institute of Pharmacology and the Gaston H. Glock Research Laboratories for Exploratory Drug Development, Center of Physiology and Pharmacology, Medical University of Vienna, Vienna, Austria
| | - Michael Freissmuth
- Institute of Pharmacology and the Gaston H. Glock Research Laboratories for Exploratory Drug Development, Center of Physiology and Pharmacology, Medical University of Vienna, Vienna, Austria
| | - Walter Sandtner
- Institute of Pharmacology and the Gaston H. Glock Research Laboratories for Exploratory Drug Development, Center of Physiology and Pharmacology, Medical University of Vienna, Vienna, Austria
| |
Collapse
|
18
|
Ryan RM, Ingram SL, Scimemi A. Regulation of Glutamate, GABA and Dopamine Transporter Uptake, Surface Mobility and Expression. Front Cell Neurosci 2021; 15:670346. [PMID: 33927596 PMCID: PMC8076567 DOI: 10.3389/fncel.2021.670346] [Citation(s) in RCA: 21] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/21/2021] [Accepted: 03/15/2021] [Indexed: 01/31/2023] Open
Abstract
Neurotransmitter transporters limit spillover between synapses and maintain the extracellular neurotransmitter concentration at low yet physiologically meaningful levels. They also exert a key role in providing precursors for neurotransmitter biosynthesis. In many cases, neurons and astrocytes contain a large intracellular pool of transporters that can be redistributed and stabilized in the plasma membrane following activation of different signaling pathways. This means that the uptake capacity of the brain neuropil for different neurotransmitters can be dynamically regulated over the course of minutes, as an indirect consequence of changes in neuronal activity, blood flow, cell-to-cell interactions, etc. Here we discuss recent advances in the mechanisms that control the cell membrane trafficking and biophysical properties of transporters for the excitatory, inhibitory and modulatory neurotransmitters glutamate, GABA, and dopamine.
Collapse
Affiliation(s)
- Renae M. Ryan
- School of Medical Sciences, Faculty of Medicine and Health, University of Sydney, Sydney, NSW, Australia
| | - Susan L. Ingram
- Department of Neurological Surgery, Oregon Health & Science University, Portland, OR, United States
| | | |
Collapse
|
19
|
Torres Valladares D, Kudumala S, Hossain M, Carvelli L. Caenorhabditis elegans as an in vivo Model to Assess Amphetamine Tolerance. BRAIN, BEHAVIOR AND EVOLUTION 2021; 95:247-255. [PMID: 33831863 DOI: 10.1159/000514858] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/24/2020] [Accepted: 01/27/2021] [Indexed: 11/19/2022]
Abstract
Amphetamine is a potent psychostimulant also used to treat attention deficit/hyperactivity disorder and narcolepsy. In vivo and in vitro data have demonstrated that amphetamine increases the amount of extra synaptic dopamine by both inhibiting reuptake and promoting efflux of dopamine through the dopamine transporter. Previous studies have shown that chronic use of amphetamine causes tolerance to the drug. Thus, since the molecular mechanisms underlying tolerance to amphetamine are still unknown, an animal model to identify the neurochemical mechanisms associated with drug tolerance is greatly needed. Here we took advantage of a unique behavior caused by amphetamine in Caenorhabditis elegans to investigate whether this simple, but powerful, genetic model develops tolerance following repeated exposure to amphetamine. We found that at least 3 treatments with 0.5 mM amphetamine were necessary to see a reduction in the amphetamine-induced behavior and, thus, to promote tolerance. Moreover, we found that, after intervals of 60/90 minutes between treatments, animals were more likely to exhibit tolerance than animals that underwent 10-minute intervals between treatments. Taken together, our results show that C. elegans is a suitable system to study tolerance to drugs of abuse such as amphetamines.
Collapse
Affiliation(s)
- Dayana Torres Valladares
- Department of Biology, Harriet L. Wilkes Honors College, Florida Atlantic University, Jupiter, Florida, USA
| | - Sirisha Kudumala
- Department of Biology, Harriet L. Wilkes Honors College, Florida Atlantic University, Jupiter, Florida, USA
| | - Murad Hossain
- Department of Pharmaceutical Sciences, School of Health and Life Sciences, North South University, Dhaka, Bangladesh
| | - Lucia Carvelli
- Department of Biology, Harriet L. Wilkes Honors College, Florida Atlantic University, Jupiter, Florida, USA.,Brain Institute, Florida Atlantic University, Jupiter, Florida, USA
| |
Collapse
|
20
|
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: 66] [Impact Index Per Article: 16.5] [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
|
21
|
Mannal N, Kleiner K, Fauler M, Dougalis A, Poetschke C, Liss B. Multi-Electrode Array Analysis Identifies Complex Dopamine Responses and Glucose Sensing Properties of Substantia Nigra Neurons in Mouse Brain Slices. Front Synaptic Neurosci 2021; 13:635050. [PMID: 33716704 PMCID: PMC7952765 DOI: 10.3389/fnsyn.2021.635050] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/29/2020] [Accepted: 01/08/2021] [Indexed: 12/16/2022] Open
Abstract
Dopaminergic (DA) midbrain neurons within the substantia nigra (SN) display an autonomous pacemaker activity that is crucial for dopamine release and voluntary movement control. Their progressive degeneration is a hallmark of Parkinson's disease. Their metabolically demanding activity-mode affects Ca2+ homeostasis, elevates metabolic stress, and renders SN DA neurons particularly vulnerable to degenerative stressors. Accordingly, their activity is regulated by complex mechanisms, notably by dopamine itself, via inhibitory D2-autoreceptors and the neuroprotective neuronal Ca2+ sensor NCS-1. Analyzing regulation of SN DA neuron activity-pattern is complicated by their high vulnerability. We studied this activity and its control by dopamine, NCS-1, and glucose with extracellular multi-electrode array (MEA) recordings from midbrain slices of juvenile and adult mice. Our tailored MEA- and spike sorting-protocols allowed high throughput and long recording times. According to individual dopamine-responses, we identified two distinct SN cell-types, in similar frequency: dopamine-inhibited and dopamine-excited neurons. Dopamine-excited neurons were either silent in the absence of dopamine, or they displayed pacemaker-activities, similar to that of dopamine-inhibited neurons. Inhibition of pacemaker-activity by dopamine is typical for SN DA neurons, and it can undergo prominent desensitization. We show for adult mice, that the number of SN DA neurons with desensitized dopamine-inhibition was increased (~60–100%) by a knockout of NCS-1, or by prevention of NCS-1 binding to D2-autoreceptors, while time-course and degrees of desensitization were not altered. The number of neurons with desensitized D2-responses was also higher (~65%) at high glucose-levels (25 mM), compared to lower glucose (2.5 mM), while again desensitization-kinetics were unaltered. However, spontaneous firing-rates were significantly higher at high glucose-levels (~20%). Moreover, transient glucose-deprivation (1 mM) induced a fast and fully-reversible pacemaker frequency reduction. To directly address and quantify glucose-sensing properties of SN DA neurons, we continuously monitored their electrical activity, while altering extracellular glucose concentrations stepwise from 0.5 mM up to 25 mM. SN DA neurons were excited by glucose, with EC50 values ranging from 0.35 to 2.3 mM. In conclusion, we identified a novel, common subtype of dopamine-excited SN neurons, and a complex, joint regulation of dopamine-inhibited neurons by dopamine and glucose, within the range of physiological brain glucose-levels.
Collapse
Affiliation(s)
- Nadja Mannal
- Institute of Applied Physiology, University of Ulm, Ulm, Germany
| | | | - Michael Fauler
- Institute of Applied Physiology, University of Ulm, Ulm, Germany
| | | | | | - Birgit Liss
- Institute of Applied Physiology, University of Ulm, Ulm, Germany.,Linacre and New College, University of Oxford, Oxford, United Kingdom
| |
Collapse
|
22
|
Chen YC, Huang HR, Hsu CH, Ou CY. CRMP/UNC-33 organizes microtubule bundles for KIF5-mediated mitochondrial distribution to axon. PLoS Genet 2021; 17:e1009360. [PMID: 33571181 PMCID: PMC7904166 DOI: 10.1371/journal.pgen.1009360] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/03/2020] [Revised: 02/24/2021] [Accepted: 11/11/2020] [Indexed: 12/13/2022] Open
Abstract
Neurons are highly specialized cells with polarized cellular processes and subcellular domains. As vital organelles for neuronal functions, mitochondria are distributed by microtubule-based transport systems. Although the essential components of mitochondrial transport including motors and cargo adaptors are identified, it is less clear how mitochondrial distribution among somato-dendritic and axonal compartment is regulated. Here, we systematically study mitochondrial motors, including four kinesins, KIF5, KIF17, KIF1, KLP-6, and dynein, and transport regulators in C. elegans PVD neurons. Among all these motors, we found that mitochondrial export from soma to neurites is mainly mediated by KIF5/UNC-116. Interestingly, UNC-116 is especially important for axonal mitochondria, while dynein removes mitochondria from all plus-end dendrites and the axon. We surprisingly found one mitochondrial transport regulator for minus-end dendritic compartment, TRAK-1, and two mitochondrial transport regulators for axonal compartment, CRMP/UNC-33 and JIP3/UNC-16. While JIP3/UNC-16 suppresses axonal mitochondria, CRMP/UNC-33 is critical for axonal mitochondria; nearly no axonal mitochondria present in unc-33 mutants. We showed that UNC-33 is essential for organizing the population of UNC-116-associated microtubule bundles, which are tracks for mitochondrial trafficking. Disarrangement of these tracks impedes mitochondrial transport to the axon. In summary, we identified a compartment-specific transport regulation of mitochondria by UNC-33 through organizing microtubule tracks for different kinesin motors other than microtubule polarity.
Collapse
Affiliation(s)
- Ying-Chun Chen
- Institute of Biochemistry and Molecular Biology, College of Medicine, National Taiwan University, Taipei, Taiwan
| | - Hao-Ru Huang
- Institute of Biochemistry and Molecular Biology, College of Medicine, National Taiwan University, Taipei, Taiwan
| | - Chia-Hao Hsu
- Institute of Biochemistry and Molecular Biology, College of Medicine, National Taiwan University, Taipei, Taiwan
| | - Chan-Yen Ou
- Institute of Biochemistry and Molecular Biology, College of Medicine, National Taiwan University, Taipei, Taiwan
- * E-mail:
| |
Collapse
|
23
|
Pandey P, Singh A, Kaur H, Ghosh-Roy A, Babu K. Increased dopaminergic neurotransmission results in ethanol dependent sedative behaviors in Caenorhabditis elegans. PLoS Genet 2021; 17:e1009346. [PMID: 33524034 PMCID: PMC7877767 DOI: 10.1371/journal.pgen.1009346] [Citation(s) in RCA: 13] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/24/2020] [Revised: 02/11/2021] [Accepted: 01/06/2021] [Indexed: 12/19/2022] Open
Abstract
Ethanol is a widely used drug, excessive consumption of which could lead to medical conditions with diverse symptoms. Ethanol abuse causes dysfunction of memory, attention, speech and locomotion across species. Dopamine signaling plays an essential role in ethanol dependent behaviors in animals ranging from C. elegans to humans. We devised an ethanol dependent assay in which mutants in the dopamine autoreceptor, dop-2, displayed a unique sedative locomotory behavior causing the animals to move in circles while dragging the posterior half of their body. Here, we identify the posterior dopaminergic sensory neuron as being essential to modulate this behavior. We further demonstrate that in dop-2 mutants, ethanol exposure increases dopamine secretion and functions in a DVA interneuron dependent manner. DVA releases the neuropeptide NLP-12 that is known to function through cholinergic motor neurons and affect movement. Thus, DOP-2 modulates dopamine levels at the synapse and regulates alcohol induced movement through NLP-12.
Collapse
Affiliation(s)
- Pratima Pandey
- Department of Biological Sciences, Indian Institute of Science Education and Research (IISER) Mohali, Mohali, India
| | - Anuradha Singh
- Department of Biological Sciences, Indian Institute of Science Education and Research (IISER) Mohali, Mohali, India
| | - Harjot Kaur
- National Brain Research Centre, Gurgaon, India
| | | | - Kavita Babu
- Department of Biological Sciences, Indian Institute of Science Education and Research (IISER) Mohali, Mohali, India
- Centre for Neuroscience, Indian Institute of Science (IISc), Bangalore, India
| |
Collapse
|
24
|
Wu ZQ, Li K, Tian X, Zhou MX, Li ZJ. Schisandra chinensis water extract protects ethanol-induced neurotoxicity in Caenorhabditis elegans. J Food Biochem 2020; 44:e13249. [PMID: 32524635 DOI: 10.1111/jfbc.13249] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/18/2019] [Revised: 03/16/2020] [Accepted: 04/01/2020] [Indexed: 12/12/2022]
Abstract
The protective effect of Schisandra chinensis water extract (SWE) on ethanol-induced neurotoxicity in Caenorhabditis elegans and the underlying mechanism were investigated. Young worms were exposed to ethanol or a mixture of ethanol and SWE for 24 hr. Locomotion ability, tissue ethanol concentration, free radical content, antioxidant enzyme activity, lifespan, and expression of key dopaminergic nervous system-related genes were evaluated. Ethanol affected the motion ability of worms and shortened their lifespan. Ethanol intake increased the tissue ethanol concentration, resulting in redox imbalance, and dopamine release and accumulation. SWE alleviated motility loss of C. elegans and extended their lifespan. It reduced the tissue ethanol concentration and free radical content, likely because it alleviated oxidative stress. Finally, SWE inhibited continuous dopamine excitement. These results suggest that SWE plays a protective role in dopaminergic neurons. It can be used to treat ethanol-induced neurotoxicity, and to investigate its potential mechanism. PRACTICAL APPLICATIONS: Schisandra chinensis is a traditional functional food that has protective effects on the liver and brain. Although S. chinensis is found in some anti-alcohol products, the effects of S. chinensis on neurological and behavioral disorders caused by alcohol are rarely reported. The manuscript explored the protective effect of SWE on ethanol-induced nerve injury in Caenorhabditis elegans, and we preliminarily discussed the underlying mechanism. The results suggested that SWE can alleviate ethanol-induced neurotoxicity. Meanwhile, the results provide a theoretical basis for better use of S. chinensis to develop products to antagonize the side effects of alcohol. In addition, the method of using C. elegans model to evaluate the protective effect of S. chinensis on ethanol-induced nerve injury can provide practical reference for the screening and utilization of other plant functional components.
Collapse
Affiliation(s)
- Zhong-Qin Wu
- Hunan Province Key Laboratory of Food Science and Biotechnology, College of Food Science and Technology, Hunan Agricultural University, Changsha, PR China
| | - Ke Li
- Hunan Province Key Laboratory of Food Science and Biotechnology, College of Food Science and Technology, Hunan Agricultural University, Changsha, PR China
| | - Xing Tian
- Hunan Province Key Laboratory of Food Science and Biotechnology, College of Food Science and Technology, Hunan Agricultural University, Changsha, PR China.,Department of Food and Drug Engineering, School of Pharmacy, Hunan University of Chinese Medicine, Changsha, PR China
| | - Ming-Xi Zhou
- Hunan Province Key Laboratory of Food Science and Biotechnology, College of Food Science and Technology, Hunan Agricultural University, Changsha, PR China
| | - Zong-Jun Li
- Hunan Province Key Laboratory of Food Science and Biotechnology, College of Food Science and Technology, Hunan Agricultural University, Changsha, PR China
| |
Collapse
|
25
|
Ferdous N, Kudumala S, Sossi S, Carvelli L. Prolonged Amphetamine Treatments Cause Long-Term Decrease of Dopamine Uptake in Cultured Cells. Neurochem Res 2019; 45:1399-1409. [PMID: 31883055 PMCID: PMC7260268 DOI: 10.1007/s11064-019-02938-7] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/16/2019] [Revised: 12/12/2019] [Accepted: 12/18/2019] [Indexed: 11/24/2022]
Abstract
Amphetamine (AMPH) is a systemic stimulant used to treat a variety of diseases including Attention Deficit Hyperactive Disorder, narcolepsy and obesity. Previous data showed that by binding to catecholamine transporters, AMPH prevents the reuptake of the neurotransmitters dopamine (DA) and norepinephrine (NE). Because AMPH, either used therapeutically at final concentrations of 1–10 µM or abused as recreational drug (50–200 µM), is taken over long periods of time, we investigated the prolonged effects of this drug on the uptake of DA. We found that, in LLC-PK1 cells stably expressing the human DA transporter (hDAT), pretreatments with 1 or 50 µM AMPH caused significant reduction in DA uptake right after the 15-h pretreatment. Remarkably, after 50 but not 1 µM AMPH pretreatment, we observed a significant reduction in DA uptake also after one, two or three cell divisions. To test whether these long-term effects induced by AMPH where conserved in a model comparable to primordial neuronal cells and native neurons, we used the human neuroblastoma cell line SH-SY5Y cells, which were reported to endogenously express both hDAT and the NE transporter. Pretreatments with 50 µM AMPH caused a significant reduction of DA uptake both right after 15 h and 3 cell divisions followed by neuro-differentiation with retinoic acid (RA) for 5 days. Under these same conditions, AMPH did not change the intracellular concentrations of ATP, ROS and cell viability suggesting, therefore, that the reduction in DA uptake was not cause by AMPH-induced toxicity. Interestingly, while 1 µM AMPH did not cause long-term effects in the LLC-PK1 cells, in the SH-SY5Y cells, it decreased the DA uptake after one, two, but not three, cell divisions and 5-day RA differentiation. These data show that besides the well-known acute effects, AMPH can also produce long-term effects in vitro that are maintained during cell division and transmitted to the daughter cells.
Collapse
Affiliation(s)
- Nafisa Ferdous
- Department of Biomedical Science, University of North Dakota, Grand Forks, ND, USA
| | - Sirisha Kudumala
- Harriet L. Wilkes Honors College, Florida Atlantic University, FL, Jupiter, USA
| | - Serena Sossi
- Integrated Biology Program, Florida Atlantic University, FL, Boca Raton, USA
| | - Lucia Carvelli
- Brain Institute, Florida Atlantic University, Jupiter, FL, USA. .,Harriet L. Wilkes Honors College, Florida Atlantic University, FL, Jupiter, USA. .,Integrated Biology Program, Florida Atlantic University, FL, Boca Raton, USA.
| |
Collapse
|
26
|
Plasticity in striatal dopamine release is governed by release-independent depression and the dopamine transporter. Nat Commun 2019; 10:4263. [PMID: 31537790 PMCID: PMC6753151 DOI: 10.1038/s41467-019-12264-9] [Citation(s) in RCA: 47] [Impact Index Per Article: 7.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/25/2019] [Accepted: 08/13/2019] [Indexed: 01/19/2023] Open
Abstract
Mesostriatal dopaminergic neurons possess extensively branched axonal arbours. Whether action potentials are converted to dopamine output in the striatum will be influenced dynamically and critically by axonal properties and mechanisms that are poorly understood. Here, we address the roles for mechanisms governing release probability and axonal activity in determining short‐term plasticity of dopamine release, using fast‐scan cyclic voltammetry in the ex vivo mouse striatum. We show that brief short‐term facilitation and longer short term depression are only weakly dependent on the level of initial release, i.e. are release insensitive. Rather, short-term plasticity is strongly determined by mechanisms which govern axonal activation, including K+‐gated excitability and the dopamine transporter, particularly in the dorsal striatum. We identify the dopamine transporter as a master regulator of dopamine short‐term plasticity, governing the balance between release‐dependent and independent mechanisms that also show region‐specific gating. Dopamine release in the striatum has important roles in action selection and in disorders such as Parkinson’s disease. The authors here show that short-term plasticity of dopamine release is strongly determined by axonal activation and dopamine transporters.
Collapse
|
27
|
Robinson SB, Refai O, Hardaway JA, Sturgeon S, Popay T, Bermingham DP, Freeman P, Wright J, Blakely RD. Dopamine-dependent, swimming-induced paralysis arises as a consequence of loss of function mutations in the RUNX transcription factor RNT-1. PLoS One 2019; 14:e0216417. [PMID: 31083672 PMCID: PMC6513266 DOI: 10.1371/journal.pone.0216417] [Citation(s) in RCA: 5] [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: 01/08/2019] [Accepted: 04/21/2019] [Indexed: 11/18/2022] Open
Abstract
Dopamine (DA) is a neurotransmitter with actions across phylogeny that modulate core behaviors such as motor activity, reward, attention, and cognition. Perturbed DA signaling in humans is associated with multiple disorders, including addiction, ADHD, schizophrenia, and Parkinson's disease. The presynaptic DA transporter exerts powerful control on DA signaling by efficient clearance of the neurotransmitter following release. As in vertebrates, Caenorhabditis elegans DAT (DAT-1) constrains DA signaling and loss of function mutations in the dat-1 gene result in slowed crawling on solid media and swimming-induced paralysis (Swip) in water. Previously, we identified a mutant line, vt34, that exhibits robust DA-dependent Swip. vt34 exhibits biochemical and behavioral phenotypes consistent with reduced DAT-1 function though vt34; dat-1 double mutants exhibit an enhanced Swip phenotype, suggesting contributions of the vt34-associated mutation to additional mechanisms that lead to excess DA signaling. SNP mapping and whole genome sequencing of vt34 identified the site of the molecular lesion in the gene B0412.2 that encodes the Runx transcription factor ortholog RNT-1. Unlike dat-1 animals, but similar to other loss of function rnt-1 mutants, vt34 exhibits altered male tail morphology and reduced body size. Deletion mutations in both rnt-1 and the bro-1 gene, which encodes a RNT-1 binding partner also exhibit Swip. Both vt34 and rnt-1 mutations exhibit reduced levels of dat-1 mRNA as well as the tyrosine hydroxylase ortholog cat-2. Although reporter studies indicate that rnt-1 is expressed in DA neurons, its re-expression in DA neurons of vt34 animals fails to fully rescue Swip. Moreover, as shown for vt34, rnt-1 mutation exhibits additivity with dat-1 in generating Swip, as do rnt-1 and bro-1 mutations, and vt34 exhibits altered capacity for acetylcholine signaling at the neuromuscular junction. Together, these findings identify a novel role for rnt-1 in limiting DA neurotransmission and suggest that loss of RNT-1 may disrupt function of both DA neurons and body wall muscle to drive Swip.
Collapse
Affiliation(s)
- Sarah B Robinson
- Department of Pharmacology, Vanderbilt University Medical Center, Nashville, TN, United States of America
| | - Osama Refai
- Department of Biomedical Science, Charles E. Schmidt College of Science, Florida Atlantic University, Jupiter, FL United States of America
| | - J Andrew Hardaway
- Department of Pharmacology, Vanderbilt University Medical Center, Nashville, TN, United States of America
| | - Sarah Sturgeon
- Department of Pharmacology, Vanderbilt University Medical Center, Nashville, TN, United States of America
| | - Tessa Popay
- Department of Pharmacology, Vanderbilt University Medical Center, Nashville, TN, United States of America
| | - Daniel P Bermingham
- Department of Pharmacology, Vanderbilt University Medical Center, Nashville, TN, United States of America
| | - Phyllis Freeman
- Department of Pharmacology, Vanderbilt University Medical Center, Nashville, TN, United States of America
- Department of Life and Physical Sciences, Fisk University, Nashville, TN, United States of America
| | - Jane Wright
- Department of Pharmacology, Vanderbilt University Medical Center, Nashville, TN, United States of America
| | - Randy D Blakely
- Department of Biomedical Science, Charles E. Schmidt College of Science, Florida Atlantic University, Jupiter, FL United States of America
- Brain Institute, Florida Atlantic University, Jupiter, FL, United States of America
| |
Collapse
|
28
|
Steele TWE, Eltit JM. Using Ca 2+-channel biosensors to profile amphetamines and cathinones at monoamine transporters: electro-engineering cells to detect potential new psychoactive substances. Psychopharmacology (Berl) 2019; 236:973-988. [PMID: 30448989 PMCID: PMC6525079 DOI: 10.1007/s00213-018-5103-5] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/07/2018] [Accepted: 11/02/2018] [Indexed: 01/20/2023]
Abstract
BACKGROUND The appearance of stimulant-class new psychoactive substances (NPS) is a frequent and significant problem in our society. Cathinone variants are often sold illegally as 3,4-methylenedioxy methamphetamine ("ecstasy") or disguised for legal sale using misleading names such as "bath salts" and carry the risk of promoting disruptive mental states, addiction, and fatal overdose. The principal targets of these recreational drugs are monoamine transporters expressed in catecholaminergic and serotonergic neurons. Some transporter ligands can be transported into cells, where they can promote a massive release of neurotransmitters through reverse transport, and others can block uptake. A ligand's dopamine vs. serotonin transporter selectivity, potency, and activity as a substrate or blocker can help elucidate the abuse liability and subjective effects of a drug. OBJECTIVES Here, we describe the discovery, development, and validation of an emerging methodology for compound activity assessment at monoamine transporters. KEY FINDINGS Substrates generate inward electrical currents through transporters and can depolarize the plasma membrane, whereas blockers work as a "cork in a bottle" and function as antagonists. Voltage-gated Ca2+ channels were co-expressed with monoamine transporters in cultured cells and used to measure fluctuations of the membrane electrical potential. In this system, substrates of monoamine transporters produce reliable dose-dependent Ca2+ signals, while blockers hinder them. DISCUSSION This system constitutes a novel use of voltage-gated Ca2+ channels as biosensors for the purpose of characterizing ligand activity at monoamine transporters using fluorimetry. This approach in combination with in vivo evaluations of drugs' abuse-related effects is a powerful strategy for anticipating potential stimulant-class NPS.
Collapse
Affiliation(s)
- Tyler W E Steele
- Department of Physiology and Biophysics, School of Medicine, Virginia Commonwealth University, 1101 E Marshall St. Rm# 3-038H, Richmond, VA, 23298, USA
| | - Jose M Eltit
- Department of Physiology and Biophysics, School of Medicine, Virginia Commonwealth University, 1101 E Marshall St. Rm# 3-038H, Richmond, VA, 23298, USA.
| |
Collapse
|
29
|
Abstract
There is a plethora of amphetamine derivatives exerting stimulant, euphoric, anti-fatigue, and hallucinogenic effects; all structural properties allowing these effects are contained within the amphetamine structure. In the first part of this review, the interaction of amphetamine with the dopamine transporter (DAT), crucially involved in its behavioral effects, is covered, as well as the role of dopamine synthesis, the vesicular monoamine transporter VMAT2, and organic cation 3 transporter (OCT3). The second part deals with requirements in amphetamine's effect on the kinases PKC, CaMKII, and ERK, whereas the third part focuses on where we are in developing anti-amphetamine therapeutics. Thus, treatments are discussed that target DAT, VMAT2, PKC, CaMKII, and OCT3. As is generally true for the development of therapeutics for substance use disorder, there are multiple preclinically promising specific compounds against (meth)amphetamine, for which further development and clinical trials are badly needed.
Collapse
Affiliation(s)
- Maarten E A Reith
- Department of Psychiatry, New York University School of Medicine, New York, NY, USA.
| | - Margaret E Gnegy
- Department of Pharmacology, University of Michigan School of Medicine, Ann Arbor, MI, USA
| |
Collapse
|
30
|
Silwal AP, Lu HP. Mode-Selective Raman Imaging of Dopamine-Human Dopamine Transporter Interaction in Live Cells. ACS Chem Neurosci 2018; 9:3117-3127. [PMID: 30024721 DOI: 10.1021/acschemneuro.8b00301] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/30/2022] Open
Abstract
Dopamine (DA) is the catecholamine neurotransmitter which interacts with dopamine receptors (DARs) to generate dopaminergic signals in the nervous system. Dopamine transporter (DAT) interacts with DA to maintain DA's homeostasis in synaptic and perisynaptic space. DAT and DARs have great importance in the central nervous system (CNS) because they are associated with the targeted binding of drugs. Interactions of DA, its analogue with DARs, or DAT have been studied extensively to understand the mechanism of the dopaminergic signaling process and several neurodegenerative diseases, including schizophrenia, Parkinson's diseases, addiction, attention deficit hyperactivity disorder, and bipolar disorder. However, there is still a lack of a risk-free, label-free, and minimally invasive imaging approach to probe the interaction between DA and DAT or DARs. Here, we probed the DA, human dopamine transporter (hDAT), and DA-hDAT interactions in live cells using combined approach of two-photon excited (2PE) fluorescence imaging and mode-selective Raman measurement. We utilized the signature Raman peak at 1287 cm-1 to probe the location of DA and 807 and 1076 cm-1 to probe the DA-hDAT interaction in live cells. We found that the combined approach of mode-selective Raman imaging, 2PE fluorescence imaging, and computational methods is successful to probe and confirm the DA-hDAT interactions in living cells. The probing of the interactions of DARs or DAT with DA or other targeting drugs is crucial for the diagnosis and cure of several neurodegenerative diseases. Also, this analytical approach could be extended to probe other types of protein-ligand interactions.
Collapse
Affiliation(s)
- Achut P. Silwal
- Department of Chemistry and Center for Photochemical Sciences, Bowling Green State University, Ohio 43403, United States
| | - H. Peter Lu
- Department of Chemistry and Center for Photochemical Sciences, Bowling Green State University, Ohio 43403, United States
| |
Collapse
|
31
|
Mulvihill KG. Presynaptic regulation of dopamine release: Role of the DAT and VMAT2 transporters. Neurochem Int 2018; 122:94-105. [PMID: 30465801 DOI: 10.1016/j.neuint.2018.11.004] [Citation(s) in RCA: 42] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/22/2018] [Revised: 10/28/2018] [Accepted: 11/08/2018] [Indexed: 01/23/2023]
Abstract
The signaling dynamics of the neurotransmitter dopamine has been established to have an important role in a variety of behavioural processes including motor control, cognition, and emotional processing. Key regulators of transmitter release and the signaling dynamics of dopamine are the plasma membrane reuptake transporter (DAT) and the vesicular monoamine transporter (VMAT2). These proteins serve to remove dopamine molecules from the extracellular and cytosolic space, respectively and both determine the amount of transmitter released from synaptic vesicles. This review provides an overview of how these transporter proteins are involved in molecular regulation and function together to govern the dynamics of vesicular release with opposing effects on the quantal size and extracellular concentration of dopamine. These transporter proteins are both focal points of convergence for a variety of regulatory molecular cascades as well as targets for many pharmacological agents. The ratio between these transporters is argued to be useful as a molecular marker for delineating dopamine functional subsystems that may differ in transmitter release patterns.
Collapse
Affiliation(s)
- Kevin G Mulvihill
- Department of Psychology, Brock University, St. Catharines, ON, L2S 3A1, Canada.
| |
Collapse
|
32
|
Essmann CL, Ryan KR, Elmi M, Bryon-Dodd K, Porter A, Vaughan A, McMullan R, Nurrish S. Activation of RHO-1 in cholinergic motor neurons competes with dopamine signalling to control locomotion. PLoS One 2018; 13:e0204057. [PMID: 30240421 PMCID: PMC6150489 DOI: 10.1371/journal.pone.0204057] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/18/2018] [Accepted: 08/31/2018] [Indexed: 12/14/2022] Open
Abstract
The small GTPase RhoA plays a crucial role in the regulation of neuronal signalling to generate behaviour. In the developing nervous system RhoA is known to regulate the actin cytoskeleton, however the effectors of RhoA-signalling in adult neurons remain largely unidentified. We have previously shown that activation of the RhoA ortholog (RHO-1) in C. elegans cholinergic motor neurons triggers hyperactivity of these neurons and loopy locomotion with exaggerated body bends. This is achieved in part through increased diacylglycerol (DAG) levels and the recruitment of the synaptic vesicle protein UNC-13 to synaptic release sites, however other pathways remain to be identified. Dopamine, which is negatively regulated by the dopamine re-uptake transporter (DAT), has a central role in modulating locomotion in both humans and C. elegans. In this study we identify a new pathway in which RHO-1 regulates locomotory behaviour by repressing dopamine signalling, via DAT-1, linking these two pathways together. We observed an upregulation of dat-1 expression when RHO-1 is activated and show that loss of DAT-1 inhibits the loopy locomotion phenotype caused by RHO-1 activation. Reducing dopamine signalling in dat-1 mutants through mutations in genes involved in dopamine synthesis or in the dopamine receptor DOP-1 restores the ability of RHO-1 to trigger loopy locomotion in dat-1 mutants. Taken together, we show that negative regulation of dopamine signalling via DAT-1 is necessary for the neuronal RHO-1 pathway to regulate locomotion.
Collapse
Affiliation(s)
- Clara L. Essmann
- MRC Laboratory for Molecular Cell Biology, University College London, London, United Kingdom
| | - Katie R. Ryan
- MRC Laboratory for Molecular Cell Biology, University College London, London, United Kingdom
| | - Muna Elmi
- MRC Laboratory for Molecular Cell Biology, University College London, London, United Kingdom
| | - Kimberley Bryon-Dodd
- MRC Laboratory for Molecular Cell Biology, University College London, London, United Kingdom
| | - Andrew Porter
- MRC Laboratory for Molecular Cell Biology, University College London, London, United Kingdom
| | - Andrew Vaughan
- MRC Laboratory for Molecular Cell Biology, University College London, London, United Kingdom
| | - Rachel McMullan
- MRC Laboratory for Molecular Cell Biology, University College London, London, United Kingdom
| | - Stephen Nurrish
- MRC Laboratory for Molecular Cell Biology, University College London, London, United Kingdom
| |
Collapse
|
33
|
Hovde MJ, Larson GH, Vaughan RA, Foster JD. Model systems for analysis of dopamine transporter function and regulation. Neurochem Int 2018; 123:13-21. [PMID: 30179648 DOI: 10.1016/j.neuint.2018.08.015] [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] [Received: 06/30/2018] [Revised: 08/23/2018] [Accepted: 08/31/2018] [Indexed: 02/07/2023]
Abstract
The dopamine transporter (DAT) plays a critical role in dopamine (DA) homeostasis by clearing transmitter from the extraneuronal space after vesicular release. DAT serves as a site of action for a variety of addictive and therapeutic reuptake inhibitors, and transport dysfunction is associated with transmitter imbalances in disorders such as schizophrenia, attention deficit hyperactive disorder, bipolar disorder, and Parkinson disease. In this review, we describe some of the model systems that have been used for in vitro analyses of DAT structure, function and regulation, and discuss a potential relationship between transporter kinetic values and membrane cholesterol.
Collapse
Affiliation(s)
- Moriah J Hovde
- Department of Biomedical Sciences, University of North Dakota, School of Medicine and Health Sciences, Grand Forks, ND, 58202, USA
| | - Garret H Larson
- Department of Biomedical Sciences, University of North Dakota, School of Medicine and Health Sciences, Grand Forks, ND, 58202, USA
| | - Roxanne A Vaughan
- Department of Biomedical Sciences, University of North Dakota, School of Medicine and Health Sciences, Grand Forks, ND, 58202, USA
| | - James D Foster
- Department of Biomedical Sciences, University of North Dakota, School of Medicine and Health Sciences, Grand Forks, ND, 58202, USA.
| |
Collapse
|
34
|
Aversa D, Martini A, Guatteo E, Pisani A, Mercuri NB, Berretta N. Reversal of dopamine-mediated firing inhibition through activation of the dopamine transporter in substantia nigra pars compacta neurons. Br J Pharmacol 2018; 175:3534-3547. [PMID: 29933497 DOI: 10.1111/bph.14422] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/22/2018] [Revised: 06/11/2018] [Accepted: 06/14/2018] [Indexed: 12/24/2022] Open
Abstract
BACKGROUND AND PURPOSE One of the hallmarks of ventral midbrain dopamine-releasing neurons is membrane hyperpolarization in response to stimulation of somato-dendritic D2 receptors. At early postnatal age, under sustained dopamine, this inhibitory response is followed by a slow recovery, resulting in dopamine inhibition reversal (DIR). In the present investigation, we aimed to get a better insight into the cellular mechanisms underlying DIR. EXPERIMENTAL APPROACH We performed single-unit extracellular recordings with a multi-electrode array device and conventional patch-clamp recordings on midbrain mouse slices. KEY RESULTS While continuous dopamine (100 μM) perfusion gave rise to firing inhibition that recovered in 10 to 15 min, the same effect was not obtained with the D2 receptor agonist quinpirole (100 nM). Moreover, firing inhibition caused by the GABAB receptor agonist baclofen (300 nM) was reversed by dopamine (100 μM), albeit D2 receptors had been blocked by sulpiride (10 μM). Conversely, the block of the dopamine transporter (DAT) with cocaine (30 μM) prevented firing recovery by dopamine under GABAB receptor stimulation. Accordingly, in whole-cell recordings from single cells, the baclofen-induced outward current was counteracted by dopamine (100 μM) in the presence of sulpiride (10 μM), and this effect was prevented by the DAT antagonists cocaine (30 μM) and GBR12909 (2 μM). CONCLUSIONS AND IMPLICATIONS Our results indicate that the DAT plays a major role in DIR, mediating it under conditions of sustained dopamine exposure, and point to DAT as an important target for pharmacological therapies leading to prolonged enhancement of the dopaminergic signal.
Collapse
Affiliation(s)
- Daniela Aversa
- Fondazione Santa Lucia IRCCS, Rome, Italy.,Dipartimento di Medicina dei Sistemi, Università di Roma Tor Vergata, Rome, Italy
| | - Alessandro Martini
- Fondazione Santa Lucia IRCCS, Rome, Italy.,Dipartimento di Medicina dei Sistemi, Università di Roma Tor Vergata, Rome, Italy
| | - Ezia Guatteo
- Fondazione Santa Lucia IRCCS, Rome, Italy.,Dipartimento di Scienze Motorie e del Benessere, Università 'Parthenope', Naples, Italy
| | - Antonio Pisani
- Fondazione Santa Lucia IRCCS, Rome, Italy.,Dipartimento di Medicina dei Sistemi, Università di Roma Tor Vergata, Rome, Italy
| | - Nicola Biagio Mercuri
- Fondazione Santa Lucia IRCCS, Rome, Italy.,Dipartimento di Medicina dei Sistemi, Università di Roma Tor Vergata, Rome, Italy
| | | |
Collapse
|
35
|
Battisti UM, Sitta R, Harris A, Sakloth F, Walther D, Ruchala I, Negus SS, Baumann MH, Glennon RA, Eltit JM. Effects of N-Alkyl-4-Methylamphetamine Optical Isomers on Plasma Membrane Monoamine Transporters and Abuse-Related Behavior. ACS Chem Neurosci 2018; 9:1829-1839. [PMID: 29697951 PMCID: PMC6051915 DOI: 10.1021/acschemneuro.8b00138] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022] Open
Abstract
4-Methylamphetamine (4-MA) is an emerging drug of abuse that acts as a substrate at plasma membrane transporters for dopamine (DAT), norepinephrine (NET), and serotonin (SERT), thereby causing nonexocytotic release of monoamine transmitters via reverse transport. Prior studies by us showed that increasing the N-alkyl chain length of N-substituted 4-MA analogues converts 4-MA from a transportable substrate (i.e., releaser) at DAT and NET to a nontransported blocker at these sites. Here, we studied the effects of the individual optical isomers of N-methyl-, N-ethyl-, and N- n-propyl 4-MA on monoamine transporters and abuse-related behavior in rats because action/function might be related to stereochemistry. Uptake inhibition and release assays were conducted in rat brain synaptosomes whereas electrophysiological assessments of drug-transporter interactions were examined using cell-based biosensors. Intracranial-self-stimulation in rats was employed to assess abuse potential in vivo. The experimental evidence demonstrates that S(+) N-methyl 4-MA is a potent and efficacious releaser at DAT, NET, and SERT with the highest abuse potential among the test drugs, whereas R(-) N-methyl 4-MA is a less potent releaser with reduced abuse potential. The S(+)ethyl analogue has decreased efficacy as a releaser at DAT but retains full release activity at NET and SERT with a reduction in abuse-related effects; the R(-)ethyl analogue has a similar profile but is less potent. S(+) N-Propyl 4-MA is a nontransported blocker at DAT and NET but an efficacious releaser at SERT, whereas the R enantiomer is almost inactive. In conclusion, the S enantiomers of the N-alkyl 4-MA analogues are most potent. Lengthening the N-alkyl chain converts compounds from potent nonselective releasers showing abuse-related effects to more selective SERT releasers with no apparent abuse potential.
Collapse
Affiliation(s)
- Umberto M Battisti
- Department of Medicinal Chemistry, School of Pharmacy, Box 980540 , Virginia Commonwealth University , Richmond , Virginia 23298 , United States
| | - Ramsey Sitta
- Department of Physiology and Biophysics, School of Medicine , Virginia Commonwealth University , Richmond , Virginia 23298 United States
| | - Alan Harris
- Department of Physiology and Biophysics, School of Medicine , Virginia Commonwealth University , Richmond , Virginia 23298 United States
| | - Farhana Sakloth
- Department of Pharmacology and Toxicology, School of Medicine , Virginia Commonwealth University , Richmond , Virginia 23298 , United States
| | - Donna Walther
- Designer Drug Research Unit, Intramural Research Program , National Institute on Drug Abuse, National Institutes of Health , Baltimore , Maryland 21224 , United States
| | - Iwona Ruchala
- Department of Physiology and Biophysics, School of Medicine , Virginia Commonwealth University , Richmond , Virginia 23298 United States
| | - S Stevens Negus
- Department of Pharmacology and Toxicology, School of Medicine , Virginia Commonwealth University , Richmond , Virginia 23298 , United States
| | - Michael H Baumann
- Designer Drug Research Unit, Intramural Research Program , National Institute on Drug Abuse, National Institutes of Health , Baltimore , Maryland 21224 , United States
| | - Richard A Glennon
- Department of Medicinal Chemistry, School of Pharmacy, Box 980540 , Virginia Commonwealth University , Richmond , Virginia 23298 , United States
| | - Jose M Eltit
- Department of Physiology and Biophysics, School of Medicine , Virginia Commonwealth University , Richmond , Virginia 23298 United States
| |
Collapse
|
36
|
Lanzo A, Safratowich BD, Kudumala SR, Gallotta I, Zampi G, Di Schiavi E, Carvelli L. Silencing of Syntaxin 1A in the Dopaminergic Neurons Decreases the Activity of the Dopamine Transporter and Prevents Amphetamine-Induced Behaviors in C. elegans. Front Physiol 2018; 9:576. [PMID: 29872404 PMCID: PMC5972276 DOI: 10.3389/fphys.2018.00576] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/19/2018] [Accepted: 05/01/2018] [Indexed: 11/15/2022] Open
Abstract
The dopamine transporter (DAT) is a cell membrane protein whose main function is to reuptake the dopamine (DA) released in the synaptic cleft back into the dopaminergic neurons. Previous studies suggested that the activity of DAT is regulated by allosteric proteins such as Syntaxin-1A and is altered by drugs of abuse such as amphetamine (Amph). Because Caenorhabditis elegans expresses both DAT (DAT-1) and Syntaxin-1A (UNC-64), we used this model system to investigate the functional and behavioral effects caused by lack of expression of unc-64 in cultured dopaminergic neurons and in living animals. Using an inheritable RNA silencing technique, we were able to knockdown unc-64 specifically in the dopaminergic neurons. This cell-specific knockdown approach avoids the pleiotropic phenotypes caused by knockout mutations of unc-64 and ensures the transmission of dopaminergic specific unc-64 silencing to the progeny. We found that, similarly to dat-1 knockouts and dat-1 silenced lines, animals with reduced unc-64 expression in the dopaminergic neurons did not respond to Amph treatment when tested for locomotor behaviors. Our in vitro data demonstrated that in neuronal cultures derived from animals silenced for unc-64, the DA uptake was reduced by 30% when compared to controls, and this reduction was similar to that measured in neurons isolated from animals silenced for dat-1 (40%). Moreover, reduced expression of unc-64 in the dopaminergic neurons significantly reduced the DA release elicited by Amph. Because in C. elegans DAT-1 is the only protein capable to reuptake DA, these data show that reduced expression of unc-64 in the dopaminergic neurons decreases the capability of DAT in re-accumulating synaptic DA. Moreover, these results demonstrate that decreased expression of unc-64 in the dopaminergic neurons abrogates the locomotor behavior induced by Amph. Taken together these data suggest that Syntaxin-1A plays an important role in both functional and behavioral effects caused by Amph.
Collapse
Affiliation(s)
- Ambra Lanzo
- Institute of Biosciences and Bioresources, National Research Council (CNR), Naples, Italy
| | - Bryan D Safratowich
- Department of Biomedical Sciences, University of North Dakota, Grand Forks, ND, United States
| | - Sirisha R Kudumala
- Brain Institute, Florida Atlantic University, Jupiter, FL, United States
| | - Ivan Gallotta
- Institute of Genetics and Biophysics, National Research Council (CNR), Naples, Italy
| | - Giuseppina Zampi
- Institute of Biosciences and Bioresources, National Research Council (CNR), Naples, Italy
| | - Elia Di Schiavi
- Institute of Biosciences and Bioresources, National Research Council (CNR), Naples, Italy.,Institute of Genetics and Biophysics, National Research Council (CNR), Naples, Italy
| | - Lucia Carvelli
- Brain Institute, Florida Atlantic University, Jupiter, FL, United States.,Harriet L. Wilkes Honors College, Florida Atlantic University, Jupiter, FL, United States
| |
Collapse
|
37
|
Gracida X, Calarco JA. Cell type-specific transcriptome profiling in C. elegans using the Translating Ribosome Affinity Purification technique. Methods 2017. [PMID: 28648677 DOI: 10.1016/j.ymeth.2017.06.023] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023] Open
Abstract
Organs and specific cell types execute specialized functions in multicellular organisms, in large part through customized gene expression signatures. Thus, profiling the transcriptomes of specific cell and tissue types remains an important tool for understanding how cells become specialized. Methodological approaches to detect gene expression differences have utilized samples from whole animals, dissected tissues, and more recently single cells. Despite these advances, there is still a challenge and a need in most laboratories to implement less invasive yet powerful cell-type specific transcriptome profiling methods. Here, we describe the use of the Translating Ribosome Affinity Purification (TRAP) method for C. elegans to detect cell type-specific gene expression patterns at the level of translating mRNAs. In TRAP, a ribosomal protein is fused to a tag (GFP) and is expressed under cell type-specific promoters to mark genetically defined cell types in vivo. Affinity purification of lysates of animals expressing the tag enriches for ribosome-associated mRNAs of the targeted tissue. The purified mRNAs are used for making cDNA libraries subjected to high-throughput sequencing to obtain genome-wide profiles of transcripts from the targeted cell type. The ease of exposing C. elegans to diverse stimuli, coupled with available cell type specific promoters, makes TRAP a useful approach to enable the discovery of molecular components in response to external or genetic perturbations.
Collapse
Affiliation(s)
- Xicotencatl Gracida
- FAS Center for Systems Biology, Harvard University, Cambridge, MA 02138, United States; Department of Organismal and Evolutionary Biology, Harvard University, Cambridge, MA 02138, United States.
| | - John A Calarco
- FAS Center for Systems Biology, Harvard University, Cambridge, MA 02138, United States; Department of Cell and Systems Biology, University of Toronto, Toronto M5S 3G5, Canada.
| |
Collapse
|
38
|
Abstract
Products containing psychoactive synthetic cathinones, such as mephedrone and 3,4-methylenedioxypyrovalerone (MDPV) are prevalent in our society. Synthetic cathinones are structurally similar to methamphetamine, and numerous synthetics have biological activity at dopamine, serotonin, and norepinephrine transporters. Importantly, monoamine transporters co-transport sodium ions along with their substrate, and movement of substrates and ions through the transporter can generate measurable ionic currents. Here we review how electrophysiological information has enabled us to determine how synthetic cathinones affect transporter-mediated currents in cells that express these transporters. Specifically, drugs that act as transporter substrates induce inward depolarizing currents when cells are held near their resting membrane potential, whereas drugs that act as transporter blockers induce apparent outward currents by blocking an inherent inward leak current. We have employed the two-electrode voltage-clamp technique in Xenopus laevis oocytes overexpressing monoamine transporters to determine whether synthetic cathinones found in the so-called bath salts products behave as blockers or substrates. We also examined the structure-activity relationships for synthetic cathinone analogs related to the widely abused compound MDPV, a common constituent in "bath salts" possessing potent actions at the dopamine transporter.
Collapse
Affiliation(s)
- Ernesto Solis
- In Vivo Electrophysiology Unit, Behavioral Neuroscience Research Branch, National Institute on Drug Abuse - Intramural Research Program, National Institutes of Health, Triad Technology Center, 333 Cassell Drive, Suite 2200, Baltimore, MD, 21224, USA.
| |
Collapse
|
39
|
Sequence determinants of the Caenhorhabditis elegans dopamine transporter dictating in vivo axonal export and synaptic localization. Mol Cell Neurosci 2016; 78:41-51. [PMID: 27913309 DOI: 10.1016/j.mcn.2016.11.011] [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: 08/31/2016] [Revised: 11/25/2016] [Accepted: 11/28/2016] [Indexed: 02/06/2023] Open
Abstract
The monoamine neurotransmitter dopamine (DA) acts across phylogeny to modulate both simple and complex behaviors. The presynaptic DA transporter (DAT) is a major determinant of DA signaling capacity in ensuring efficient extracellular DA clearance. In humans, DAT is also a major target for prescribed and abused psychostimulants. Multiple structural determinants of DAT function and regulation have been defined, though largely these findings have arisen from heterologous expression or ex vivo cell culture studies. Loss of function mutations in the gene encoding the Caenhorhabditis elegans DAT (dat-1) produces rapid immobility when animals are placed in water, a phenotype termed swimming-induced paralysis (Swip). The ability of a DA neuron-expressed, GFP-tagged DAT-1 fusion protein (GFP::DAT-1) to localize to synapses and rescue Swip in these animals provides a facile approach to define sequences supporting DAT somatic export and function in vivo. In prior studies, we found that truncation of the last 25 amino acids of the DAT-1 C-terminus (Δ25) precludes Swip rescue, supported by a deficit in GFP::DAT-1 synaptic localization. Here, we further defined the elements within Δ25 required for DAT-1 export and function in vivo. We identified two conserved motifs (584KW585 and 591PYRKR595) where mutation results in a failure of GFP::DAT-1 to be efficiently exported to synapses and restore DAT-1 function. The 584KW585 motif conforms to a sequence proposed to support SEC24 binding, ER export from the endoplasmic reticulum (ER), and surface expression of mammalian DAT proteins, whereas the 591PYRKR595 sequence conforms to a 3R motif identified as a SEC24 binding site in vertebrate G-protein coupled receptors. Consistent with a potential role of SEC24 orthologs in DAT-1 export, we demonstrated DA neuron-specific expression of a sec-24.2 transcriptional reporter. Mutations of the orthologous C-terminal sequences in human DAT (hDAT) significantly reduced transporter surface expression and DA uptake, despite normal hDAT protein expression. Although, hDAT mutants retained SEC24 interactions, as defined in co-immunoprecipitation studies. However, these mutations disrupted the ability of SEC24D to enhance hDAT surface expression. Our studies document an essential role of conserved DAT C-terminal sequences in transporter somatic export and synaptic localization in vivo, that add further support for important roles for SEC24 family members in efficient transporter trafficking.
Collapse
|
40
|
Illiano P, Lanzo A, Leo D, Paglione M, Zampi G, Gainetdinov RR, Di Schiavi E. ACaenorhabditis elegansmodel to study dopamine transporter deficiency syndrome. Eur J Neurosci 2016; 45:207-214. [DOI: 10.1111/ejn.13366] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/09/2016] [Revised: 07/21/2016] [Accepted: 08/09/2016] [Indexed: 12/20/2022]
Affiliation(s)
- Placido Illiano
- Department of Neuroscience and Brain Technologies; Fondazione Istituto Italiano di Tecnologia; Via Morego 30 16163 Genova Italy
| | - Ambra Lanzo
- Department of Biology, Agriculture and Food Science - National Research Council, CNR; Institute of Biosciences and BioResources, IBBR; Via Pietro Castellino 111 80131 Naples Italy
| | - Damiana Leo
- Department of Neuroscience and Brain Technologies; Fondazione Istituto Italiano di Tecnologia; Via Morego 30 16163 Genova Italy
| | - Maria Paglione
- Department of Biology, Agriculture and Food Science - National Research Council, CNR; Institute of Biosciences and BioResources, IBBR; Via Pietro Castellino 111 80131 Naples Italy
| | - Giuseppina Zampi
- Department of Biology, Agriculture and Food Science - National Research Council, CNR; Institute of Biosciences and BioResources, IBBR; Via Pietro Castellino 111 80131 Naples Italy
| | - Raul R. Gainetdinov
- Institute of Translational Biomedicine; St. Petersburg State University; 199034 St. Petersburg Russia
- Skolkovo Institute of Science and Technology; Skolkovo 143025 Moscow Russia
| | - Elia Di Schiavi
- Department of Biology, Agriculture and Food Science - National Research Council, CNR; Institute of Biosciences and BioResources, IBBR; Via Pietro Castellino 111 80131 Naples Italy
| |
Collapse
|
41
|
Abstract
Serotonin transporters (SERTs) are largely recognized for one aspect of their function—to transport serotonin back into the presynaptic terminal after its release. Another aspect of their function, however, may be to generate currents large enough to have physiological consequences. The standard model for electrogenic transport is the alternating access model, in which serotonin is transported with a fixed ratio of co-transported ions resulting in net charge per cycle. The alternating access model, however, cannot account for all the observed currents through SERT or other monoamine transporters. Furthermore, SERT agonists like ecstasy or antagonists like fluoxetine generate or suppress currents that the standard model cannot support. Here we survey evidence for a channel mode of transport in which transmitters and ions move through a pore. Available structures for dopamine and serotonin transporters, however, provide no evidence for a pore conformation, raising questions of whether the proposed channel mode actually exists or whether the structural data are perhaps missing a transient open state.
Collapse
Affiliation(s)
- Louis J De Felice
- Physiology and Biophysics, School of Medicine, Virginia Commonwealth University, Richmond, VA, USA
| |
Collapse
|
42
|
Chloride requirement for monoamine transporters. Pflugers Arch 2016; 468:503-11. [PMID: 26794730 DOI: 10.1007/s00424-015-1783-4] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/10/2015] [Revised: 12/20/2015] [Accepted: 12/22/2015] [Indexed: 12/18/2022]
Abstract
This review focuses on the Cl(-) requirement for dopamine, serotonin, and norepinephrine (DA, 5-HT, and NE) transport and induced current via the transporters for these transmitters, DAT, SERT, and NET. Indirect evidence exists for the passage of Cl(-) ions through monoamine transporters; however, direct evidence is sparse. An unanswered question is why in some preparations, notably native neurons, it appears that Cl(-) ions carry the current through DAT, whereas in heterologous expression systems Na(+) ions carry the current often referred to as the uncoupled current. It is suggested that different functional states in monoamine transporters represent conformational states that carry dominantly Cl(-) or Na(+). Structures of monoamine transporters contribute enormously to structure-function relationships; however, thus far no structural features support the functionally relevant ionic currents that are known to exist in monoamine transporters.
Collapse
|
43
|
Amphetamine activates calcium channels through dopamine transporter-mediated depolarization. Cell Calcium 2015; 58:457-66. [PMID: 26162812 DOI: 10.1016/j.ceca.2015.06.013] [Citation(s) in RCA: 27] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/04/2015] [Revised: 06/27/2015] [Accepted: 06/29/2015] [Indexed: 02/07/2023]
Abstract
Amphetamine (AMPH) and its more potent enantiomer S(+)AMPH are psychostimulants used therapeutically to treat attention deficit hyperactivity disorder and have significant abuse liability. AMPH is a dopamine transporter (DAT) substrate that inhibits dopamine (DA) uptake and is implicated in DA release. Furthermore, AMPH activates ionic currents through DAT that modify cell excitability presumably by modulating voltage-gated channel activity. Indeed, several studies suggest that monoamine transporter-induced depolarization opens voltage-gated Ca(2+) channels (CaV), which would constitute an additional AMPH mechanism of action. In this study we co-express human DAT (hDAT) with Ca(2+) channels that have decreasing sensitivity to membrane depolarization (CaV1.3, CaV1.2 or CaV2.2). Although S(+)AMPH is more potent than DA in transport-competition assays and inward-current generation, at saturating concentrations both substrates indirectly activate voltage-gated L-type Ca(2+) channels (CaV1.3 and CaV1.2) but not the N-type Ca(2+) channel (CaV2.2). Furthermore, the potency to achieve hDAT-CaV electrical coupling is dominated by the substrate affinity on hDAT, with negligible influence of L-type channel voltage sensitivity. In contrast, the maximal coupling-strength (defined as Ca(2+) signal change per unit hDAT current) is influenced by CaV voltage sensitivity, which is greater in CaV1.3- than in CaV1.2-expressing cells. Moreover, relative to DA, S(+)AMPH showed greater coupling-strength at concentrations that induced relatively small hDAT-mediated currents. Therefore S(+)AMPH is not only more potent than DA at inducing hDAT-mediated L-type Ca(2+) channel currents but is a better depolarizing agent since it produces tighter electrical coupling between hDAT-mediated depolarization and L-type Ca(2+) channel activation.
Collapse
|
44
|
Abstract
Amphetamine (AMPH) is a psychostimulant and the most prescribed drug to treat attention deficit hyperactive disorder (ADHD). Although therapeutically used doses are generally well tolerated, numerous side effects are still known to occur, such as jitteriness, loss of appetite and psychosis. Moreover, AMPH is liable to be abused by users looking for increased alertness, weight loss or athletic performance. A growing body of evidence indicates that drugs of abuse, including AMPH, control gene expression through chromatin modifications. However, while numerous studies have investigated the molecular mechanisms of AMPH action, only a small number of studies have explored changes in gene expression caused by AMPH. This review examines the epigenetic changes induced by chronic and acute treatments with AMPH and includes, where relevant, data obtained with other psychostimulants such as methamphetamine and cocaine.
Collapse
Affiliation(s)
- Talus J McCowan
- Department of Basic Sciences, University of North Dakota School of Medicine and Health Sciences, Grand Forks, North Dakota, USA
| | - Archana Dhasarathy
- Department of Basic Sciences, University of North Dakota School of Medicine and Health Sciences, Grand Forks, North Dakota, USA
| | - Lucia Carvelli
- Department of Basic Sciences, University of North Dakota School of Medicine and Health Sciences, Grand Forks, North Dakota, USA
| |
Collapse
|
45
|
Masoudi N, Ibanez-Cruceyra P, Offenburger SL, Holmes A, Gartner A. Tetraspanin (TSP-17) protects dopaminergic neurons against 6-OHDA-induced neurodegeneration in C. elegans. PLoS Genet 2014; 10:e1004767. [PMID: 25474638 PMCID: PMC4256090 DOI: 10.1371/journal.pgen.1004767] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/09/2014] [Accepted: 09/21/2014] [Indexed: 12/31/2022] Open
Abstract
Parkinson's disease (PD), the second most prevalent neurodegenerative disease after Alzheimer's disease, is linked to the gradual loss of dopaminergic neurons in the substantia nigra. Disease loci causing hereditary forms of PD are known, but most cases are attributable to a combination of genetic and environmental risk factors. Increased incidence of PD is associated with rural living and pesticide exposure, and dopaminergic neurodegeneration can be triggered by neurotoxins such as 6-hydroxydopamine (6-OHDA). In C. elegans, this drug is taken up by the presynaptic dopamine reuptake transporter (DAT-1) and causes selective death of the eight dopaminergic neurons of the adult hermaphrodite. Using a forward genetic approach to find genes that protect against 6-OHDA-mediated neurodegeneration, we identified tsp-17, which encodes a member of the tetraspanin family of membrane proteins. We show that TSP-17 is expressed in dopaminergic neurons and provide genetic, pharmacological and biochemical evidence that it inhibits DAT-1, thus leading to increased 6-OHDA uptake in tsp-17 loss-of-function mutants. TSP-17 also protects against toxicity conferred by excessive intracellular dopamine. We provide genetic and biochemical evidence that TSP-17 acts partly via the DOP-2 dopamine receptor to negatively regulate DAT-1. tsp-17 mutants also have subtle behavioral phenotypes, some of which are conferred by aberrant dopamine signaling. Incubating mutant worms in liquid medium leads to swimming-induced paralysis. In the L1 larval stage, this phenotype is linked to lethality and cannot be rescued by a dop-3 null mutant. In contrast, mild paralysis occurring in the L4 larval stage is suppressed by dop-3, suggesting defects in dopaminergic signaling. In summary, we show that TSP-17 protects against neurodegeneration and has a role in modulating behaviors linked to dopamine signaling. Parkinson's disease (PD) is characterized by the progressive loss of dopaminergic neurons. While hereditary forms are known, most cases are attributable to a combination of genetic and environmental risk factors. In PD models, dopaminergic neurodegeneration can be triggered by neurotoxins such as 6-hydroxydopamine (6-OHDA). This drug, which is taken up by the presynaptic dopamine reuptake transporter (DAT-1), also causes the selective death of C. elegans dopaminergic neurons. We found that TSP-17, a member of the tetraspanin family of membrane proteins, protects dopaminergic neurons from 6-OHDA-induced degeneration. We provide evidence that TSP-17 inhibits the C. elegans dopamine transporter DAT-1, leading to increased neuronal 6-OHDA uptake in tsp-17 mutants. TSP-17 also protects against toxicity conferred by excessive intracellular dopamine. TSP-17 interacts with the DOP-2 dopamine receptor, possibly as part of a pathway that negatively regulates DAT-1. tsp-17 mutants have subtle behavioral phenotypes that are partly conferred by aberrant dopamine signaling. In summary, we have used C. elegans genetics to model key aspects of PD.
Collapse
Affiliation(s)
- Neda Masoudi
- Centre for Gene Regulation and Expression, University of Dundee, Dow Street, Dundee, United Kingdom
| | - Pablo Ibanez-Cruceyra
- Centre for Gene Regulation and Expression, University of Dundee, Dow Street, Dundee, United Kingdom
| | - Sarah-Lena Offenburger
- Centre for Gene Regulation and Expression, University of Dundee, Dow Street, Dundee, United Kingdom
| | - Alexander Holmes
- Centre for Gene Regulation and Expression, University of Dundee, Dow Street, Dundee, United Kingdom
| | - Anton Gartner
- Centre for Gene Regulation and Expression, University of Dundee, Dow Street, Dundee, United Kingdom
- * E-mail:
| |
Collapse
|
46
|
Dwyer DS, Aamodt E, Cohen B, Buttner EA. Drug elucidation: invertebrate genetics sheds new light on the molecular targets of CNS drugs. Front Pharmacol 2014; 5:177. [PMID: 25120487 PMCID: PMC4112795 DOI: 10.3389/fphar.2014.00177] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/23/2014] [Accepted: 07/09/2014] [Indexed: 02/02/2023] Open
Abstract
Many important drugs approved to treat common human diseases were discovered by serendipity, without a firm understanding of their modes of action. As a result, the side effects and interactions of these medications are often unpredictable, and there is limited guidance for improving the design of next-generation drugs. Here, we review the innovative use of simple model organisms, especially Caenorhabditis elegans, to gain fresh insights into the complex biological effects of approved CNS medications. Whereas drug discovery involves the identification of new drug targets and lead compounds/biologics, and drug development spans preclinical testing to FDA approval, drug elucidation refers to the process of understanding the mechanisms of action of marketed drugs by studying their novel effects in model organisms. Drug elucidation studies have revealed new pathways affected by antipsychotic drugs, e.g., the insulin signaling pathway, a trace amine receptor and a nicotinic acetylcholine receptor. Similarly, novel targets of antidepressant drugs and lithium have been identified in C. elegans, including lipid-binding/transport proteins and the SGK-1 signaling pathway, respectively. Elucidation of the mode of action of anesthetic agents has shown that anesthesia can involve mitochondrial targets, leak currents, and gap junctions. The general approach reviewed in this article has advanced our knowledge about important drugs for CNS disorders and can guide future drug discovery efforts.
Collapse
Affiliation(s)
- Donard S. Dwyer
- Department of Psychiatry–Department of Pharmacology, Toxicology and Neuroscience, Louisiana State University Health Sciences Center-ShreveportShreveport, LA, USA
| | - Eric Aamodt
- Department of Biochemistry and Molecular Biology, Louisiana State University Health Sciences Center-ShreveportShreveport, LA, USA
| | - Bruce Cohen
- Department of Psychiatry, Harvard Medical SchoolBoston, MA, USA
- Mailman Research Center, McLean HospitalBelmont, MA, USA
| | - Edgar A. Buttner
- Mailman Research Center, McLean HospitalBelmont, MA, USA
- Department of Neurology–Department of Psychiatry, McLean Hospital, Harvard Medical SchoolBelmont, MA, USA
| |
Collapse
|
47
|
Borre L, Andreassen TF, Shi L, Weinstein H, Gether U. The second sodium site in the dopamine transporter controls cation permeation and is regulated by chloride. J Biol Chem 2014; 289:25764-73. [PMID: 25063810 DOI: 10.1074/jbc.m114.574269] [Citation(s) in RCA: 39] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/06/2022] Open
Abstract
The dopamine transporter (DAT) belongs to the family of neurotransmitter:sodium symporters and controls dopamine (DA) homeostasis by mediating Na(+)- and Cl(-)-dependent reuptake of DA. Here we used two-electrode voltage clamp measurements in Xenopus oocytes together with targeted mutagenesis to investigate the mechanistic relationship between DAT ion binding sites and transporter conductances. In Li(+), DAT displayed a cocaine-sensitive cation leak current ∼10-fold larger than the substrate-induced current in Na(+). Mutation of Na(+) coordinating residues in the first (Na1) and second (Na2) binding sites suggested that the Li(+) leak depends on Li(+) interaction with Na2 rather than Na1. DA caused a marked inhibition of the Li(+) leak, consistent with the ability of the substrate to interact with the Li(+)-occupied state of the transporter. The leak current in Li(+) was also potently inhibited by low millimolar concentrations of Na(+), which according to our mutational data conceivably depended on high affinity binding to Na1. The Li(+) leak was further regulated by Cl(-) that most likely increases Li(+) permeation by allosterically lowering Na2 affinity. Interestingly, mutational lowering of Na2 affinity by substituting Asp-420 with asparagine dramatically increased cation permeability in Na(+) to a level higher than seen in Li(+). In addition to reveal a functional link between the bound Cl(-) and the cation bound in the Na2 site, the data support a key role of Na2 in determining cation permeability of the transporter and thereby possibly in regulating the opening probability of the inner gate.
Collapse
Affiliation(s)
- Lars Borre
- From the Molecular Neuropharmacology Laboratory, Lundbeck Foundation Center for Biomembranes in Nanomedicine, Department of Neuroscience and Pharmacology, Faculty of Health and Medical Sciences, The Panum Institute 18.6, University of Copenhagen, 2200 Copenhagen N, Denmark and
| | - Thorvald F Andreassen
- From the Molecular Neuropharmacology Laboratory, Lundbeck Foundation Center for Biomembranes in Nanomedicine, Department of Neuroscience and Pharmacology, Faculty of Health and Medical Sciences, The Panum Institute 18.6, University of Copenhagen, 2200 Copenhagen N, Denmark and
| | - Lei Shi
- the Department of Physiology and Biophysics, Institute for Computational Biomedicine, Weill Medical College of Cornell University, New York, New York 10021
| | - Harel Weinstein
- the Department of Physiology and Biophysics, Institute for Computational Biomedicine, Weill Medical College of Cornell University, New York, New York 10021
| | - Ulrik Gether
- From the Molecular Neuropharmacology Laboratory, Lundbeck Foundation Center for Biomembranes in Nanomedicine, Department of Neuroscience and Pharmacology, Faculty of Health and Medical Sciences, The Panum Institute 18.6, University of Copenhagen, 2200 Copenhagen N, Denmark and
| |
Collapse
|
48
|
Fraser R, Chen Y, Guptaroy B, Luderman KD, Stokes SL, Beg A, DeFelice LJ, Gnegy ME. An N-terminal threonine mutation produces an efflux-favorable, sodium-primed conformation of the human dopamine transporter. Mol Pharmacol 2014; 86:76-85. [PMID: 24753048 PMCID: PMC4053999 DOI: 10.1124/mol.114.091926] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/03/2014] [Accepted: 04/21/2014] [Indexed: 01/27/2023] Open
Abstract
The dopamine transporter (DAT) reversibly transports dopamine (DA) through a series of conformational transitions. Alanine (T62A) or aspartate (T62D) mutagenesis of Thr62 revealed T62D-human (h)DAT partitions in a predominately efflux-preferring conformation. Compared with wild-type (WT), T62D-hDAT exhibits reduced [(3)H]DA uptake and enhanced baseline DA efflux, whereas T62A-hDAT and WT-hDAT function in an influx-preferring conformation. We now interrogate the basis of the mutants' altered function with respect to membrane conductance and Na(+) sensitivity. The hDAT constructs were expressed in Xenopus oocytes to investigate if heightened membrane potential would explain the efflux characteristics of T62D-hDAT. In the absence of substrate, all constructs displayed identical resting membrane potentials. Substrate-induced inward currents were present in oocytes expressing WT- and T62A-hDAT but not T62D-hDAT, suggesting equal bidirectional ion flow through T62D-hDAT. Utilization of the fluorescent DAT substrate ASP(+) [4-(4-(dimethylamino)styryl)-N-methylpyridinium] revealed that T62D-hDAT accumulates substrate in human embryonic kidney (HEK)-293 cells when the substrate is not subject to efflux. Extracellular sodium (Na(+) e) replacement was used to evaluate sodium gradient requirements for DAT transport functions. The EC50 for Na(+) e stimulation of [(3)H]DA uptake was identical in all constructs expressed in HEK-293 cells. As expected, decreasing [Na(+)]e stimulated [(3)H]DA efflux in WT- and T62A-hDAT cells. Conversely, the elevated [(3)H]DA efflux in T62D-hDAT cells was independent of Na(+) e and commensurate with [(3)H]DA efflux attained in WT-hDAT cells, either by removal of Na(+) e or by application of amphetamine. We conclude that T62D-hDAT represents an efflux-willing, Na(+)-primed orientation-possibly representing an experimental model of the conformational impact of amphetamine exposure to hDAT.
Collapse
Affiliation(s)
- Rheaclare Fraser
- Department of Pharmacology, University of Michigan, Ann Arbor, Michigan (R.F., B.G., K.D.L., S.L.S., A.B., M.E.G.); and Departments of Psychiatry (Y.C.) and Physiology and Biophysics (L.J.D.), Virginia Commonwealth University, Richmond, Virginia
| | - Yongyue Chen
- Department of Pharmacology, University of Michigan, Ann Arbor, Michigan (R.F., B.G., K.D.L., S.L.S., A.B., M.E.G.); and Departments of Psychiatry (Y.C.) and Physiology and Biophysics (L.J.D.), Virginia Commonwealth University, Richmond, Virginia
| | - Bipasha Guptaroy
- Department of Pharmacology, University of Michigan, Ann Arbor, Michigan (R.F., B.G., K.D.L., S.L.S., A.B., M.E.G.); and Departments of Psychiatry (Y.C.) and Physiology and Biophysics (L.J.D.), Virginia Commonwealth University, Richmond, Virginia
| | - Kathryn D Luderman
- Department of Pharmacology, University of Michigan, Ann Arbor, Michigan (R.F., B.G., K.D.L., S.L.S., A.B., M.E.G.); and Departments of Psychiatry (Y.C.) and Physiology and Biophysics (L.J.D.), Virginia Commonwealth University, Richmond, Virginia
| | - Stephanie L Stokes
- Department of Pharmacology, University of Michigan, Ann Arbor, Michigan (R.F., B.G., K.D.L., S.L.S., A.B., M.E.G.); and Departments of Psychiatry (Y.C.) and Physiology and Biophysics (L.J.D.), Virginia Commonwealth University, Richmond, Virginia
| | - Asim Beg
- Department of Pharmacology, University of Michigan, Ann Arbor, Michigan (R.F., B.G., K.D.L., S.L.S., A.B., M.E.G.); and Departments of Psychiatry (Y.C.) and Physiology and Biophysics (L.J.D.), Virginia Commonwealth University, Richmond, Virginia
| | - Louis J DeFelice
- Department of Pharmacology, University of Michigan, Ann Arbor, Michigan (R.F., B.G., K.D.L., S.L.S., A.B., M.E.G.); and Departments of Psychiatry (Y.C.) and Physiology and Biophysics (L.J.D.), Virginia Commonwealth University, Richmond, Virginia
| | - Margaret E Gnegy
- Department of Pharmacology, University of Michigan, Ann Arbor, Michigan (R.F., B.G., K.D.L., S.L.S., A.B., M.E.G.); and Departments of Psychiatry (Y.C.) and Physiology and Biophysics (L.J.D.), Virginia Commonwealth University, Richmond, Virginia
| |
Collapse
|
49
|
Saha K, Sambo D, Richardson BD, Lin LM, Butler B, Villarroel L, Khoshbouei H. Intracellular methamphetamine prevents the dopamine-induced enhancement of neuronal firing. J Biol Chem 2014; 289:22246-57. [PMID: 24962577 DOI: 10.1074/jbc.m114.563056] [Citation(s) in RCA: 33] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/06/2022] Open
Abstract
The dysregulation of the dopaminergic system is implicated in multiple neurological and neuropsychiatric disorders such as Parkinson disease and drug addiction. The primary target of psychostimulants such as amphetamine and methamphetamine is the dopamine transporter (DAT), the major regulator of extracellular dopamine levels in the brain. However, the behavioral and neurophysiological correlates of methamphetamine and amphetamine administration are unique from one another, thereby suggesting these two compounds impact dopaminergic neurotransmission differentially. We further examined the unique mechanisms by which amphetamine and methamphetamine regulate DAT function and dopamine neurotransmission; in the present study we examined the impact of extracellular and intracellular amphetamine and methamphetamine on the spontaneous firing of cultured midbrain dopaminergic neurons and isolated DAT-mediated current. In dopaminergic neurons the spontaneous firing rate was enhanced by extracellular application of amphetamine > dopamine > methamphetamine and was DAT-dependent. Amphetamine > methamphetamine similarly enhanced DAT-mediated inward current, which was sensitive to isosmotic substitution of Na(+) or Cl(-) ion. Although isosmotic substitution of extracellular Na(+) ions blocked amphetamine and methamphetamine-induced DAT-mediated inward current similarly, the removal of extracellular Cl(-) ions preferentially blocked amphetamine-induced inward current. The intracellular application of methamphetamine, but not amphetamine, prevented the dopamine-induced increase in the spontaneous firing of dopaminergic neurons and the corresponding DAT-mediated inward current. The results reveal a new mechanism for methamphetamine-induced dysregulation of dopaminergic neurons.
Collapse
Affiliation(s)
- Kaustuv Saha
- From the Department of Neuroscience and Department of Psychiatry, McKnight Brain Institute, University of Florida College of Medicine, Gainesville, Florida 32611
| | - Danielle Sambo
- From the Department of Neuroscience and Department of Psychiatry, McKnight Brain Institute, University of Florida College of Medicine, Gainesville, Florida 32611
| | - Ben D Richardson
- From the Department of Neuroscience and Department of Psychiatry, McKnight Brain Institute, University of Florida College of Medicine, Gainesville, Florida 32611
| | - Landon M Lin
- From the Department of Neuroscience and Department of Psychiatry, McKnight Brain Institute, University of Florida College of Medicine, Gainesville, Florida 32611
| | - Brittany Butler
- From the Department of Neuroscience and Department of Psychiatry, McKnight Brain Institute, University of Florida College of Medicine, Gainesville, Florida 32611
| | - Laura Villarroel
- From the Department of Neuroscience and Department of Psychiatry, McKnight Brain Institute, University of Florida College of Medicine, Gainesville, Florida 32611
| | - Habibeh Khoshbouei
- From the Department of Neuroscience and Department of Psychiatry, McKnight Brain Institute, University of Florida College of Medicine, Gainesville, Florida 32611
| |
Collapse
|
50
|
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: 0.8] [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
|