1
|
Zhao X, Gao F. Dynamic Mechanism of Norepinephrine Reuptake and Antidepressants Blockade Regulated by Membrane Potential. J Chem Theory Comput 2025; 21:2780-2797. [PMID: 39992630 DOI: 10.1021/acs.jctc.4c01775] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/26/2025]
Abstract
During nerve signaling, changes in membrane potential are key to regulating neuronal activity. The norepinephrine transporter (NET) plays a crucial role in the reuptake of norepinephrine (NE), which is essential for maintaining neurotransmitter homeostasis. However, the impact of membrane potential on NET function has long been understudied. Despite the great biological significance of NET, the dynamic molecular mechanisms of NE transport and the blockade effects of antidepressants on this process remain unclear. Here, we reveal the structural, electrostatic, and dynamic characteristics of the NET-NE/antidepressants systems, indicating the dynamic voltage dependence of the NET function. By analyzing the structure and electrostatic properties of the central binding pocket, we find that a hydrophobic network stabilizes the localization of NE, while the dynamic hydrogen bond and salt bridge network plays a crucial role in facilitating the inward transport of NE. Changes in membrane potential significantly affect the reuptake of NE through an electrostatically driven substrate transport pathway, primarily influencing the substrate entrance, the hydrophilic channel leading to the central site, and the exit region. The hyperpolarized state favors NE reuptake, exhibiting a marked preference for inward movement, which aligns with the physiological need for neurons to regulate neurotransmitter concentration in the synaptic cleft via reuptake. Conversely, in the depolarized state, which corresponds to the generation of nerve impulses, NE reuptake may not peak. Furthermore, antidepressants, with their larger molecular size and longer charged amino groups, initially anchor to the essential residue E382 required for NE reuptake. They subsequently occupy the same binding pathway as NE, creating spatial hindrance that effectively blocks NE binding to the central pocket. Additionally, their binding/dissociation behaviors exhibit significant voltage dependence. Under the hyperpolarized state, antidepressants can better block NE entry through more flexible electrostatic and hydrophobic interactions with NET, while the depolarized state raises the binding barrier for antidepressants, facilitating their dissociation. And with this work, a computational strategy for membrane protein-ligand is proposed to emphasize that considering the effects of electric fields in the calculations can reveal more underlying mechanisms and key interactions.
Collapse
Affiliation(s)
- Xiaoyu Zhao
- Department of Physics, School of Science, Tianjin University, Tianjin 300072, China
| | - Feng Gao
- Department of Physics, School of Science, Tianjin University, Tianjin 300072, China
- Frontiers Science Center for Synthetic Biology and Key Laboratory of Systems Bioengineering (Ministry of Education), Tianjin University, Tianjin 300072, China
- State Key Laboratory of Synthetic Biology, Tianjin University, Tianjin 300072, China
| |
Collapse
|
2
|
Rui Y, Tang Q, Chen L, Pu J, Wang W, Ding SN. Rapid electrochemical detection of L-lactate in Baijiu affecting serotonin and dopamine secretion in mice. Analyst 2024. [PMID: 39143937 DOI: 10.1039/d4an00880d] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 08/16/2024]
Abstract
Baijiu, a traditional Chinese alcoholic beverage, carries China's rich historical and cultural heritage. Consumers experience varying levels of relaxation and pleasure after consuming different types of Baijiu, with the biological basis of delectation influenced by serotonin and dopamine. In this study, we prepared carbon fiber electrodes modified with surface decorated gold nanoparticles to directly measure the electrochemical response signals in the serum of mice before and after gavage with different types of Baijiu. It was observed that the serum signal change in mice after consuming Baijiu sample 1 (J1) was higher than that of the other two types of Baijiu. Consequently, trace flavor compounds in the Baijiu samples were detected using gas chromatography-mass spectrometry (GC-MS) and liquid chromatography-mass spectrometry (LC-MS), revealing the highest content of L-lactic acid in J1. Mice were intraperitoneally injected with 200 mg kg-1 of L-lactic acid. The changes in dopamine and serotonin in the serum of the injected mice were monitored using a biosensor, and the results were compared with the results of high performance liquid chromatography-triple quadrupole mass spectrometry (HPLC-MS). The findings confirmed that L-lactic acid could indeed stimulate the secretion of both neurotransmitters in mice, suggesting that the trace components in J1 may even exhibit synergistic effects. This study contributes to a deeper understanding of the effects of Baijiu on the body and provides a scientific basis for the production and consumption of Baijiu.
Collapse
Affiliation(s)
- Yating Rui
- Jiangsu Province Hi-Tech Key Laboratory for Biomedical Research, School of Chemistry and Chemical Engineering, Southeast University, Nanjing 211189, China.
| | - Qunyong Tang
- Jiangsu King's Lucky Brewery Joint-Stock Co., Ltd, Lian shui 223411, China
| | - Liyi Chen
- Jiangsu King's Lucky Brewery Joint-Stock Co., Ltd, Lian shui 223411, China
| | - Juan Pu
- Lian shui Peoples Hospital, Huaian, 223400, China
| | - Wanpeng Wang
- Lian shui Peoples Hospital, Huaian, 223400, China
| | - Shou-Nian Ding
- Jiangsu Province Hi-Tech Key Laboratory for Biomedical Research, School of Chemistry and Chemical Engineering, Southeast University, Nanjing 211189, China.
| |
Collapse
|
3
|
Bonanno SL, Sanfilippo P, Eamani A, Sampson MM, Kandagedon B, Li K, Burns GD, Makar ME, Zipursky SL, Krantz DE. Constitutive and Conditional Epitope Tagging of Endogenous G-Protein-Coupled Receptors in Drosophila. J Neurosci 2024; 44:e2377232024. [PMID: 38937100 PMCID: PMC11326870 DOI: 10.1523/jneurosci.2377-23.2024] [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: 12/18/2023] [Revised: 04/30/2024] [Accepted: 06/06/2024] [Indexed: 06/29/2024] Open
Abstract
To visualize the cellular and subcellular localization of neuromodulatory G-protein-coupled receptors in Drosophila, we implement a molecular strategy recently used to add epitope tags to ionotropic receptors at their endogenous loci. Leveraging evolutionary conservation to identify sites more likely to permit insertion of a tag, we generated constitutive and conditional tagged alleles for Drosophila 5-HT1A, 5-HT2A, 5-HT2B, Oct β 1R, Oct β 2R, two isoforms of OAMB, and mGluR The conditional alleles allow for the restricted expression of tagged receptor in specific cell types, an option not available for any previous reagents to label these proteins. We show expression patterns for these receptors in female brains and that 5-HT1A and 5-HT2B localize to the mushroom bodies (MBs) and central complex, respectively, as predicted by their roles in sleep. By contrast, the unexpected enrichment of Octβ1R in the central complex and of 5-HT1A and 5-HT2A to nerve terminals in lobular columnar cells in the visual system suggest new hypotheses about their functions at these sites. Using an additional tagged allele of the serotonin transporter, a marker of serotonergic tracts, we demonstrate diverse spatial relationships between postsynaptic 5-HT receptors and presynaptic 5-HT neurons, consistent with the importance of both synaptic and volume transmission. Finally, we use the conditional allele of 5-HT1A to show that it localizes to distinct sites within the MBs as both a postsynaptic receptor in Kenyon cells and a presynaptic autoreceptor.
Collapse
Affiliation(s)
- Shivan L Bonanno
- Department of Psychiatry and Biobehavioral Sciences, David Geffen School of Medicine, University of California, Los Angeles, California 90095
| | - Piero Sanfilippo
- Department of Biological Chemistry, David Geffen School of Medicine, University of California, Los Angeles, California 90095
- Howard Hughes Medical Institute, David Geffen School of Medicine, University of California Los Angeles, Los Angeles, California 90095
| | - Aditya Eamani
- Department of Psychiatry and Biobehavioral Sciences, David Geffen School of Medicine, University of California, Los Angeles, California 90095
| | - Maureen M Sampson
- Department of Psychiatry and Biobehavioral Sciences, David Geffen School of Medicine, University of California, Los Angeles, California 90095
| | - Binu Kandagedon
- Department of Psychiatry and Biobehavioral Sciences, David Geffen School of Medicine, University of California, Los Angeles, California 90095
| | - Kenneth Li
- Department of Psychiatry and Biobehavioral Sciences, David Geffen School of Medicine, University of California, Los Angeles, California 90095
| | - Giselle D Burns
- Department of Psychiatry and Biobehavioral Sciences, David Geffen School of Medicine, University of California, Los Angeles, California 90095
| | - Marylyn E Makar
- Department of Psychiatry and Biobehavioral Sciences, David Geffen School of Medicine, University of California, Los Angeles, California 90095
| | - S Lawrence Zipursky
- Department of Biological Chemistry, David Geffen School of Medicine, University of California, Los Angeles, California 90095
- Howard Hughes Medical Institute, David Geffen School of Medicine, University of California Los Angeles, Los Angeles, California 90095
| | - David E Krantz
- Department of Psychiatry and Biobehavioral Sciences, David Geffen School of Medicine, University of California, Los Angeles, California 90095
| |
Collapse
|
4
|
Larnerd C, Kachewar N, Wolf FW. Drosophila learning and memory centers and the actions of drugs of abuse. Learn Mem 2024; 31:a053815. [PMID: 38862166 PMCID: PMC11199947 DOI: 10.1101/lm.053815.123] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/12/2024] [Accepted: 03/27/2024] [Indexed: 06/13/2024]
Abstract
Drug addiction and the circuitry for learning and memory are intimately intertwined. Drugs of abuse create strong, inappropriate, and lasting memories that contribute to many of their destructive properties, such as continued use despite negative consequences and exceptionally high rates of relapse. Studies in Drosophila melanogaster are helping us understand how drugs of abuse, especially alcohol, create memories at the level of individual neurons and in the circuits where they function. Drosophila is a premier organism for identifying the mechanisms of learning and memory. Drosophila also respond to drugs of abuse in ways that remarkably parallel humans and rodent models. An emerging consensus is that, for alcohol, the mushroom bodies participate in the circuits that control acute drug sensitivity, not explicitly associative forms of plasticity such as tolerance, and classical associative memories of their rewarding and aversive properties. Moreover, it is becoming clear that drugs of abuse use the mushroom body circuitry differently from other behaviors, potentially providing a basis for their addictive properties.
Collapse
Affiliation(s)
- Caleb Larnerd
- Quantitative and Systems Biology Graduate Group, University of California, Merced, California 95343, USA
| | - Neha Kachewar
- Department of Molecular and Cell Biology, University of California, Merced, California 95343, USA
- Health Sciences Research Institute, University of California, Merced, California 95343, USA
| | - Fred W Wolf
- Quantitative and Systems Biology Graduate Group, University of California, Merced, California 95343, USA
- Department of Molecular and Cell Biology, University of California, Merced, California 95343, USA
| |
Collapse
|
5
|
Knigge T. Antidepressants - The new endocrine disruptors? The case of crustaceans. Mol Cell Endocrinol 2024; 583:112155. [PMID: 38185462 DOI: 10.1016/j.mce.2024.112155] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/15/2023] [Revised: 12/31/2023] [Accepted: 01/05/2024] [Indexed: 01/09/2024]
Abstract
Antidepressants are high-volume pharmaceuticals that accumulate to concentrations in the μg·L-1 range in surface waters. The release of peptide hormones via neurosecretory cells appears as a natural target for antidepressants. Here I review research that suggests that antidepressants indeed disrupt endocrine signalling in crustaceans, by acting on the synthesis and release of neurohormones, such as crustacean hyperglycaemic hormone, moult inhibiting hormone and pigment dispersing hormone in decapods, as well as methyl farnesoate in Daphnids. Hence, antidepressants can affect hormonal regulation of physiological functions: increase in energy metabolism and activity, lowered ecdysteroid levels, potentially disrupting moult and somatic growth, reducing colour change capacity and compromising camouflage, as well as induction of male sex determination. Several studies further suggest effects of antidepressants on crustacean reproduction, but the hormonal regulation of these effects remains elusive. All things considered, a body of evidence strongly suggests that antidepressants are endocrine disrupting compounds in crustaceans.
Collapse
Affiliation(s)
- Thomas Knigge
- Normandie Univ, Unilehavre, FR CNRS 3730 Sciences Appliquées à L'Environnement, UMR-I02, Environmental Stress and Biomonitoring of Aquatic Environments, University of Le Havre Normandy, France.
| |
Collapse
|
6
|
Rand MD, Tennessen JM, Mackay TFC, Anholt RRH. Perspectives on the Drosophila melanogaster Model for Advances in Toxicological Science. Curr Protoc 2023; 3:e870. [PMID: 37639638 PMCID: PMC10463236 DOI: 10.1002/cpz1.870] [Citation(s) in RCA: 17] [Impact Index Per Article: 8.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 08/31/2023]
Abstract
The use of Drosophila melanogaster for studies of toxicology has grown considerably in the last decade. The Drosophila model has long been appreciated as a versatile and powerful model for developmental biology and genetics because of its ease of handling, short life cycle, low cost of maintenance, molecular genetic accessibility, and availability of a wide range of publicly available strains and data resources. These features, together with recent unique developments in genomics and metabolomics, make the fly model especially relevant and timely for the development of new approach methodologies and movements toward precision toxicology. Here, we offer a perspective on how flies can be leveraged to identify risk factors relevant to environmental exposures and human health. First, we review and discuss fundamental toxicologic principles for experimental design with Drosophila. Next, we describe quantitative and systems genetics approaches to resolve the genetic architecture and candidate pathways controlling susceptibility to toxicants. Finally, we summarize the current state and future promise of the emerging field of Drosophila metabolomics for elaborating toxic mechanisms. © 2023 The Authors. Current Protocols published by Wiley Periodicals LLC.
Collapse
Affiliation(s)
- Matthew D. Rand
- Department of Environmental Medicine, University of Rochester School of Medicine and Dentistry, Rochester, NY, USA
| | | | - Trudy F. C. Mackay
- Center for Human Genetics and Department of Genetics and Biochemistry, Clemson University, 114 Gregor Mendel Circle, Greenwood, South Carolina 29646, USA
| | - Robert R. H. Anholt
- Center for Human Genetics and Department of Genetics and Biochemistry, Clemson University, 114 Gregor Mendel Circle, Greenwood, South Carolina 29646, USA
| |
Collapse
|
7
|
Gajardo I, Guerra S, Campusano JM. Navigating Like a Fly: Drosophila melanogaster as a Model to Explore the Contribution of Serotonergic Neurotransmission to Spatial Navigation. Int J Mol Sci 2023; 24:ijms24054407. [PMID: 36901836 PMCID: PMC10002024 DOI: 10.3390/ijms24054407] [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: 01/02/2023] [Revised: 02/06/2023] [Accepted: 02/13/2023] [Indexed: 02/25/2023] Open
Abstract
Serotonin is a monoamine that acts in vertebrates and invertebrates as a modulator promoting changes in the structure and activity of brain areas relevant to animal behavior, ranging from sensory perception to learning and memory. Whether serotonin contributes in Drosophila to human-like cognitive abilities, including spatial navigation, is an issue little studied. Like in vertebrates, the serotonergic system in Drosophila is heterogeneous, meaning that distinct serotonergic neurons/circuits innervate specific fly brain regions to modulate precise behaviors. Here we review the literature that supports that serotonergic pathways modify different aspects underlying the formation of navigational memories in Drosophila.
Collapse
Affiliation(s)
- Ivana Gajardo
- Departamento de Biología Celular y Molecular, Facultad de Ciencias Biológicas, Pontificia Universidad Católica de Chile, Santiago 8331150, Chile
- Departamento de Neurociencia, Instituto Milenio de Neurociencia Biomédica (BNI), Facultad de Medicina, Universidad de Chile, Santiago 8380453, Chile
| | - Simón Guerra
- Departamento de Biología Celular y Molecular, Facultad de Ciencias Biológicas, Pontificia Universidad Católica de Chile, Santiago 8331150, Chile
| | - Jorge M. Campusano
- Departamento de Biología Celular y Molecular, Facultad de Ciencias Biológicas, Pontificia Universidad Católica de Chile, Santiago 8331150, Chile
- Correspondence: ; Tel.: +56-2-2354-2133
| |
Collapse
|
8
|
Soslau G. Cardiovascular serotonergic system: Evolution, receptors, transporter, and function. JOURNAL OF EXPERIMENTAL ZOOLOGY PART 2021; 337:115-127. [PMID: 34662506 DOI: 10.1002/jez.2554] [Citation(s) in RCA: 13] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/18/2021] [Revised: 10/01/2021] [Accepted: 10/07/2021] [Indexed: 01/22/2023]
Abstract
The serotonergic system, serotonin (5HT), serotonin transporter (SERT), and serotonin receptors (5HT-x), is an evolutionarily ancient system that has clear physiological advantages to all life forms from bacteria to humans. This review focuses on the role of platelet/plasma serotonin and the cardiovascular system with minor references to its significant neurotransmitter function. Platelets transport and store virtually all plasma serotonin in dense granules. Stored serotonin is released from activated platelets and can bind to serotonin receptors on platelets and cellular components of the vascular wall to augment aggregation and induce vasoconstriction or vasodilation. The vascular endothelium is critical to the maintenance of cardiovascular homeostasis. While there are numerous ligands, neurological components, and baroreceptors that effect vascular tone it is proposed that serotonin and nitric oxide (an endothelium relaxing factor) are major players in the regulation of systemic blood pressure. Signals not fully defined, to date, that direct serotonin binding to one of the 15 identified 5HT receptors versus the transporter, and the role platelet/plasma serotonin plays in regulating hypertension within the cardiovascular system remain important issues to better understand many diseases and to develop new drugs. Also, expanded research of these pathways in lower life-forms may serve as important model systems to further our understanding of the evolution and mechanisms of action of serotonin.
Collapse
Affiliation(s)
- Gerald Soslau
- Department of Biochemistry and Molecular Biology, Drexel University College of Medicine, Philadelphia, Pennsylvania, USA
| |
Collapse
|
9
|
Baker BM, Mokashi SS, Shankar V, Hatfield JS, Hannah RC, Mackay TFC, Anholt RRH. The Drosophila brain on cocaine at single-cell resolution. Genome Res 2021; 31:1927-1937. [PMID: 34035044 PMCID: PMC8494231 DOI: 10.1101/gr.268037.120] [Citation(s) in RCA: 14] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/29/2020] [Accepted: 02/02/2021] [Indexed: 11/24/2022]
Abstract
Whereas the neurological effects of cocaine have been well documented, effects of acute cocaine consumption on genome-wide gene expression across the brain remain largely unexplored. This question cannot be readily addressed in humans but can be approached using the Drosophila melanogaster model, where gene expression in the entire brain can be surveyed at once. Flies exposed to cocaine show impaired locomotor activity, including climbing behavior and startle response (a measure of sensorimotor integration), and increased incidence of seizures and compulsive grooming. To identify specific cell populations that respond to acute cocaine exposure, we analyzed single-cell transcriptional responses in duplicate samples of flies that consumed fixed amounts of sucrose or sucrose supplemented with cocaine, in both sexes. Unsupervised clustering of the transcriptional profiles of a total of 86,224 cells yielded 36 distinct clusters. Annotation of clusters based on gene markers revealed that all major cell types (neuronal and glial) as well as neurotransmitter types from most brain regions were represented. The brain transcriptional responses to cocaine showed profound sexual dimorphism and were considerably more pronounced in males than females. Differential expression analysis within individual clusters indicated cluster-specific responses to cocaine. Clusters corresponding to Kenyon cells of the mushroom bodies and glia showed especially large transcriptional responses following cocaine exposure. Cluster specific coexpression networks and global interaction networks revealed a diverse array of cellular processes affected by acute cocaine exposure. These results provide an atlas of sexually dimorphic cocaine-modulated gene expression in a model brain.
Collapse
Affiliation(s)
- Brandon M Baker
- Center for Human Genetics, Department of Genetics and Biochemistry, Clemson University, Greenwood, South Carolina 29646, USA
| | - Sneha S Mokashi
- Center for Human Genetics, Department of Genetics and Biochemistry, Clemson University, Greenwood, South Carolina 29646, USA
| | - Vijay Shankar
- Center for Human Genetics, Department of Genetics and Biochemistry, Clemson University, Greenwood, South Carolina 29646, USA
| | - Jeffrey S Hatfield
- Center for Human Genetics, Department of Genetics and Biochemistry, Clemson University, Greenwood, South Carolina 29646, USA
| | - Rachel C Hannah
- Center for Human Genetics, Department of Genetics and Biochemistry, Clemson University, Greenwood, South Carolina 29646, USA
| | - Trudy F C Mackay
- Center for Human Genetics, Department of Genetics and Biochemistry, Clemson University, Greenwood, South Carolina 29646, USA
| | - Robert R H Anholt
- Center for Human Genetics, Department of Genetics and Biochemistry, Clemson University, Greenwood, South Carolina 29646, USA
| |
Collapse
|
10
|
Duruz J, Kaltenrieder C, Ladurner P, Bruggmann R, Martìnez P, Sprecher SG. Acoel Single-Cell Transcriptomics: Cell Type Analysis of a Deep Branching Bilaterian. Mol Biol Evol 2021; 38:1888-1904. [PMID: 33355655 PMCID: PMC8097308 DOI: 10.1093/molbev/msaa333] [Citation(s) in RCA: 13] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023] Open
Abstract
Bilaterian animals display a wide variety of cell types, organized into defined anatomical structures and organ systems, which are mostly absent in prebilaterian animals. Xenacoelomorpha are an early-branching bilaterian phylum displaying an apparently relatively simple anatomical organization that have greatly diverged from other bilaterian clades. In this study, we use whole-body single-cell transcriptomics on the acoel Isodiametra pulchra to identify and characterize different cell types. Our analysis identifies the existence of ten major cell type categories in acoels all contributing to main biological functions of the organism: metabolism, locomotion and movements, behavior, defense, and development. Interestingly, although most cell clusters express core fate markers shared with other animal clades, we also describe a surprisingly large number of clade-specific marker genes, suggesting the emergence of clade-specific common molecular machineries functioning in distinct cell types. Together, these results provide novel insight into the evolution of bilaterian cell types and open the door to a better understanding of the origins of the bilaterian body plan and their constitutive cell types.
Collapse
Affiliation(s)
- Jules Duruz
- Department of Biology, Institute of Zoology, University of Fribourg, Fribourg, Switzerland
| | - Cyrielle Kaltenrieder
- Department of Biology, Institute of Zoology, University of Fribourg, Fribourg, Switzerland
| | - Peter Ladurner
- Institute of Zoology and Center of Molecular Bioscience Innsbruck, University of Innsbruck, Innsbruck, Austria
| | - Rémy Bruggmann
- Institute of Cell Biology, University of Bern, Bern, Switzerland.,Interfaculty Bioinformatics Unit, University of Bern, Bern, Switzerland
| | - Pedro Martìnez
- Departament de Genètica, Universitat de Barcelona, Barcelona, Catalonia, Spain.,Institut Català de Recerca i Estudis Avancats (ICREA), Passeig de Lluís Companys, Barcelona, Spain
| | - Simon G Sprecher
- Department of Biology, Institute of Zoology, University of Fribourg, Fribourg, Switzerland
| |
Collapse
|
11
|
Chvilicek MM, Titos I, Rothenfluh A. The Neurotransmitters Involved in Drosophila Alcohol-Induced Behaviors. Front Behav Neurosci 2020; 14:607700. [PMID: 33384590 PMCID: PMC7770116 DOI: 10.3389/fnbeh.2020.607700] [Citation(s) in RCA: 17] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/17/2020] [Accepted: 11/23/2020] [Indexed: 12/18/2022] Open
Abstract
Alcohol is a widely used and abused substance with numerous negative consequences for human health and safety. Historically, alcohol's widespread, non-specific neurobiological effects have made it a challenge to study in humans. Therefore, model organisms are a critical tool for unraveling the mechanisms of alcohol action and subsequent effects on behavior. Drosophila melanogaster is genetically tractable and displays a vast behavioral repertoire, making it a particularly good candidate for examining the neurobiology of alcohol responses. In addition to being experimentally amenable, Drosophila have high face and mechanistic validity: their alcohol-related behaviors are remarkably consistent with humans and other mammalian species, and they share numerous conserved neurotransmitters and signaling pathways. Flies have a long history in alcohol research, which has been enhanced in recent years by the development of tools that allow for manipulating individual Drosophila neurotransmitters. Through advancements such as the GAL4/UAS system and CRISPR/Cas9 mutagenesis, investigation of specific neurotransmitters in small subsets of neurons has become ever more achievable. In this review, we describe recent progress in understanding the contribution of seven neurotransmitters to fly behavior, focusing on their roles in alcohol response: dopamine, octopamine, tyramine, serotonin, glutamate, GABA, and acetylcholine. We chose these small-molecule neurotransmitters due to their conservation in mammals and their importance for behavior. While neurotransmitters like dopamine and octopamine have received significant research emphasis regarding their contributions to behavior, others, like glutamate, GABA, and acetylcholine, remain relatively unexplored. Here, we summarize recent genetic and behavioral findings concerning these seven neurotransmitters and their roles in the behavioral response to alcohol, highlighting the fitness of the fly as a model for human alcohol use.
Collapse
Affiliation(s)
- Maggie M. Chvilicek
- Department of Psychiatry, University of Utah, Salt Lake City, UT, United States
- Molecular Medicine Program, University of Utah, Salt Lake City, UT, United States
- Neuroscience Graduate Program, University of Utah, Salt Lake City, UT, United States
| | - Iris Titos
- Molecular Medicine Program, University of Utah, Salt Lake City, UT, United States
| | - Adrian Rothenfluh
- Department of Psychiatry, University of Utah, Salt Lake City, UT, United States
- Molecular Medicine Program, University of Utah, Salt Lake City, UT, United States
- Neuroscience Graduate Program, University of Utah, Salt Lake City, UT, United States
- Department of Neurobiology and Anatomy, University of Utah, Salt Lake City, UT, United States
- Department of Human Genetics, University of Utah, Salt Lake City, UT, United States
| |
Collapse
|
12
|
Scholz H. Unraveling the Mechanisms of Behaviors Associated With AUDs Using Flies and Worms. Alcohol Clin Exp Res 2019; 43:2274-2284. [PMID: 31529787 DOI: 10.1111/acer.14199] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/30/2019] [Accepted: 09/11/2019] [Indexed: 12/11/2022]
Abstract
Alcohol use disorders (AUDs) are very common worldwide and negatively affect both individuals and societies. To understand how normal behavior turns into uncontrollable use of alcohol, several approaches have been utilized in the last decades. However, we still do not completely understand how AUDs evolve or how they are maintained in the brains of affected individuals. In addition, efficient and effective treatment is still in need of development. This review focuses on alternative approaches developed over the last 20 years using Drosophila melanogaster (Drosophila) and Caenorhabditis elegans (C. elegans) as genetic model systems to determine the mechanisms underlying the action of ethanol (EtOH) and behaviors associated with AUDs. All the results and insights of studies over the last 20 years cannot be comprehensively summarized. Thus, a few prominent examples are provided highlighting the principles of the genes and mechanisms that have been uncovered and are involved in the action of EtOH at the cellular level. In addition, examples are provided of the genes and mechanisms that regulate behaviors relevant to acquiring and maintaining excessive alcohol intake, such as decision making, reward and withdrawal, and/or relapse regulation. How the insight gained from the results of Drosophila and C. elegans models can be translated to higher organisms, such as rodents and/or humans, is discussed, as well as whether these insights have any relevance or impact on our understanding of the mechanisms underlying AUDs in humans. Finally, future directions are presented that might facilitate the identification of drugs to treat AUDs.
Collapse
Affiliation(s)
- Henrike Scholz
- From the, Department of Biology, Institute for Zoology, Albertus-Magnus University of Cologne, Cologne, Germany
| |
Collapse
|
13
|
Deng B, Li Q, Liu X, Cao Y, Li B, Qian Y, Xu R, Mao R, Zhou E, Zhang W, Huang J, Rao Y. Chemoconnectomics: Mapping Chemical Transmission in Drosophila. Neuron 2019; 101:876-893.e4. [PMID: 30799021 DOI: 10.1016/j.neuron.2019.01.045] [Citation(s) in RCA: 171] [Impact Index Per Article: 28.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/07/2018] [Revised: 11/02/2018] [Accepted: 01/17/2019] [Indexed: 12/27/2022]
Abstract
We define the chemoconnectome (CCT) as the entire set of neurotransmitters, neuromodulators, neuropeptides, and their receptors underlying chemotransmission in an animal. We have generated knockout lines of Drosophila CCT genes for functional investigations and knockin lines containing Gal4 and other tools for examining gene expression and manipulating neuronal activities, with a versatile platform allowing genetic intersections and logic gates. CCT reveals the coexistence of specific transmitters but mutual exclusion of the major inhibitory and excitatory transmitters in the same neurons. One neuropeptide and five receptors were detected in glia, with octopamine β2 receptor functioning in glia. A pilot screen implicated 41 genes in sleep regulation, with the dopamine receptor Dop2R functioning in neurons expressing the peptides Dilp2 and SIFa. Thus, CCT is a novel concept, chemoconnectomics a new approach, and CCT tool lines a powerful resource for systematic investigations of chemical-transmission-mediated neural signaling circuits underlying behavior and cognition.
Collapse
Affiliation(s)
- Bowen Deng
- Peking-Tsinghua Center for Life Sciences, PKU-IDG/McGovern Institute for Brain Research, Advanced Innovation Center for Genomics, Peking University School of Life Sciences, Chinese Institute for Brain Research, Beijing, Zhongguangchun Life Sciences Park, Beijing, China
| | - Qi Li
- Peking-Tsinghua Center for Life Sciences, PKU-IDG/McGovern Institute for Brain Research, Advanced Innovation Center for Genomics, Peking University School of Life Sciences, Chinese Institute for Brain Research, Beijing, Zhongguangchun Life Sciences Park, Beijing, China
| | - Xinxing Liu
- Peking-Tsinghua Center for Life Sciences, PKU-IDG/McGovern Institute for Brain Research, Advanced Innovation Center for Genomics, Peking University School of Life Sciences, Chinese Institute for Brain Research, Beijing, Zhongguangchun Life Sciences Park, Beijing, China
| | - Yue Cao
- Peking-Tsinghua Center for Life Sciences, PKU-IDG/McGovern Institute for Brain Research, Advanced Innovation Center for Genomics, Peking University School of Life Sciences, Chinese Institute for Brain Research, Beijing, Zhongguangchun Life Sciences Park, Beijing, China
| | - Bingfeng Li
- Peking-Tsinghua Center for Life Sciences, PKU-IDG/McGovern Institute for Brain Research, Advanced Innovation Center for Genomics, Peking University School of Life Sciences, Chinese Institute for Brain Research, Beijing, Zhongguangchun Life Sciences Park, Beijing, China
| | - Yongjun Qian
- Peking-Tsinghua Center for Life Sciences, PKU-IDG/McGovern Institute for Brain Research, Advanced Innovation Center for Genomics, Peking University School of Life Sciences, Chinese Institute for Brain Research, Beijing, Zhongguangchun Life Sciences Park, Beijing, China
| | - Rui Xu
- School of Basic Medical Sciences, Nanjing Medical University, Nanjing, China
| | - Renbo Mao
- Peking-Tsinghua Center for Life Sciences, PKU-IDG/McGovern Institute for Brain Research, Advanced Innovation Center for Genomics, Peking University School of Life Sciences, Chinese Institute for Brain Research, Beijing, Zhongguangchun Life Sciences Park, Beijing, China
| | - Enxing Zhou
- Peking-Tsinghua Center for Life Sciences, PKU-IDG/McGovern Institute for Brain Research, Advanced Innovation Center for Genomics, Peking University School of Life Sciences, Chinese Institute for Brain Research, Beijing, Zhongguangchun Life Sciences Park, Beijing, China
| | - Wenxia Zhang
- Peking-Tsinghua Center for Life Sciences, PKU-IDG/McGovern Institute for Brain Research, Advanced Innovation Center for Genomics, Peking University School of Life Sciences, Chinese Institute for Brain Research, Beijing, Zhongguangchun Life Sciences Park, Beijing, China
| | - Juan Huang
- School of Basic Medical Sciences, Nanjing Medical University, Nanjing, China
| | - Yi Rao
- Peking-Tsinghua Center for Life Sciences, PKU-IDG/McGovern Institute for Brain Research, Advanced Innovation Center for Genomics, Peking University School of Life Sciences, Chinese Institute for Brain Research, Beijing, Zhongguangchun Life Sciences Park, Beijing, China.
| |
Collapse
|
14
|
Rillich J, Stevenson PA. Serotonin Mediates Depression of Aggression After Acute and Chronic Social Defeat Stress in a Model Insect. Front Behav Neurosci 2018; 12:233. [PMID: 30349464 PMCID: PMC6186776 DOI: 10.3389/fnbeh.2018.00233] [Citation(s) in RCA: 22] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/18/2018] [Accepted: 09/18/2018] [Indexed: 01/01/2023] Open
Abstract
In all animals, losers of a conflict against a conspecific exhibit reduced aggressiveness, often coupled with depression-like symptoms, particularly after multiple defeats. While serotonin (5HT) is involved, discovering its natural role in aggression and depression has proven elusive. We show how 5HT influences aggression in male crickets, before, and after single and multiple defeats using serotonergic drugs, at dosages that had no obvious deleterious effect on general motility: the 5HT synthesis inhibitor alpha-methyltryptophan (AMTP), the 5HT2 receptor blocker ketanserin, methiothepin which blocks 5HT receptor subtypes other than 5HT2, 5HT's precursor 5-hydroxytryptophan (5HTP) and re-uptake inhibitor fluoxetine. Contrasting reports for other invertebrates, none of the drugs influenced aggression at the first encounter. However, the recovery of aggression after single defeat, which normally requires 3 h in crickets, was severely affected. Losers that received ketanserin or AMTP regained their aggressiveness sooner, whereas those that received fluoxetine, 5HTP, or methiothepin failed to recover within 3 h. Furthermore, compared to controls, which show long term aggressive depression 24 h after 6 defeats at 1 h intervals, crickets that received AMTP or ketanserin regained their full aggressiveness and were thus more resilient to chronic defeat stress. In contrast, 5HTP and fluoxetine treated crickets showed long term aggressive depression 24 h after only 2 defeats, and were thus more susceptible to defeat stress. We conclude that 5HT acts after social defeat via a 5HT2 like receptor to maintain depressed aggressiveness after defeat, and to promote the susceptibility to and establishment of long-term depression after chronic social defeat. It is known that the decision to flee and establishment of loser depression in crickets is controlled by nitric oxide (NO), whereas dopamine (DA), but not octopamine (OA) is necessary for recovery after defeat. Here we show that blocking NO synthesis, just like ketanserin, affords resilience to multiple defeat stress, whereas blocking DA receptors, but not OA receptors, increases susceptibility, just like fluoxetine. We discuss the possible interplay between 5HT, NO, DA, and OA in controlling aggression after defeat, as well as similarities and differences to findings in mammals and other invertebrate model systems.
Collapse
Affiliation(s)
- Jan Rillich
- Institute for Biology, Leipzig University, Leipzig, Germany
| | | |
Collapse
|
15
|
Shipley AT, Imeh-Nathaniel A, Orfanakos VB, Wormack LN, Huber R, Nathaniel TI. The Sensitivity of the Crayfish Reward System to Mammalian Drugs of Abuse. Front Physiol 2017; 8:1007. [PMID: 29270131 PMCID: PMC5723678 DOI: 10.3389/fphys.2017.01007] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/26/2017] [Accepted: 11/21/2017] [Indexed: 12/27/2022] Open
Abstract
The idea that addiction occurs when the brain is not able to differentiate whether specific reward circuits were triggered by adaptive natural rewards or falsely activated by addictive drugs exist in several models of drug addiction. The suitability of crayfish (Orconectes rusticus) for drug addiction research arises from developmental variation of growth, life span, reproduction, behavior and some quantitative traits, especially among isogenic mates reared in the same environment. This broad spectrum of traits makes it easier to analyze the effect of mammalian drugs of abuse in shaping behavioral phenotype. Moreover, the broad behavioral repertoire allows the investigation of self-reinforcing circuitries involving appetitive and exploratory motor behavior, while the step-wise alteration of the phenotype by metamorphosis allows accurate longitudinal analysis of different behavioral states. This paper reviews a series of recent experimental findings that evidence the suitability of crayfish as an invertebrate model system for the study of drug addiction. Results from these studies reveal that unconditioned exposure to mammalian drugs of abuse produces a variety of stereotyped behaviors. Moreover, if presented in the context of novelty, drugs directly stimulate exploration and appetitive motor patterns along with molecular processes for drug conditioned reward. Findings from these studies indicate the existence of drug sensitive circuitry in crayfish that facilitates exploratory behavior and appetitive motor patterns via increased incentive salience of environmental stimuli or by increasing exploratory motor patterns. This work demonstrates the potential of crayfish as a model system for research into the neural mechanisms of addiction, by contributing an evolutionary, comparative context to our understanding of natural reward as an important life-sustaining process.
Collapse
Affiliation(s)
- Adam T Shipley
- Department of Biomedical Sciences, University of South Carolina School of Medicine, Greenville, SC, United States
| | | | - Vasiliki B Orfanakos
- Department of Biomedical Sciences, University of South Carolina School of Medicine, Greenville, SC, United States
| | - Leah N Wormack
- Department of Biomedical Sciences, University of South Carolina School of Medicine, Greenville, SC, United States
| | - Robert Huber
- J.P Scott Center for Neuroscience, Mind and Behavior, Bowling Green State University, Bowling Green, OH, United States
| | - Thomas I Nathaniel
- Department of Biomedical Sciences, University of South Carolina School of Medicine, Greenville, SC, United States
| |
Collapse
|
16
|
Xu L, He J, Kaiser A, Gräber N, Schläger L, Ritze Y, Scholz H. A Single Pair of Serotonergic Neurons Counteracts Serotonergic Inhibition of Ethanol Attraction in Drosophila. PLoS One 2016; 11:e0167518. [PMID: 27936023 PMCID: PMC5147910 DOI: 10.1371/journal.pone.0167518] [Citation(s) in RCA: 19] [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: 06/22/2016] [Accepted: 11/15/2016] [Indexed: 11/18/2022] Open
Abstract
Attraction to ethanol is common in both flies and humans, but the neuromodulatory mechanisms underlying this innate attraction are not well understood. Here, we dissect the function of the key regulator of serotonin signaling—the serotonin transporter–in innate olfactory attraction to ethanol in Drosophila melanogaster. We generated a mutated version of the serotonin transporter that prolongs serotonin signaling in the synaptic cleft and is targeted via the Gal4 system to different sets of serotonergic neurons. We identified four serotonergic neurons that inhibit the olfactory attraction to ethanol and two additional neurons that counteract this inhibition by strengthening olfactory information. Our results reveal that compensation can occur on the circuit level and that serotonin has a bidirectional function in modulating the innate attraction to ethanol. Given the evolutionarily conserved nature of the serotonin transporter and serotonin, the bidirectional serotonergic mechanisms delineate a basic principle for how random behavior is switched into targeted approach behavior.
Collapse
Affiliation(s)
- Li Xu
- Zoology, Albertus Magnus University Cologne, Köln, Germany
| | - Jianzheng He
- Zoology, Albertus Magnus University Cologne, Köln, Germany
| | - Andrea Kaiser
- Zoology, Albertus Magnus University Cologne, Köln, Germany
| | - Nikolas Gräber
- Zoology, Albertus Magnus University Cologne, Köln, Germany
| | - Laura Schläger
- Zoology, Albertus Magnus University Cologne, Köln, Germany
| | - Yvonne Ritze
- Institute of Genetics and Neurobiology, Julius Maximillian University Würzburg, Würzburg, Germany
| | - Henrike Scholz
- Zoology, Albertus Magnus University Cologne, Köln, Germany
- Institute of Genetics and Neurobiology, Julius Maximillian University Würzburg, Würzburg, Germany
- * E-mail:
| |
Collapse
|
17
|
Sealover NR, Felts B, Kuntz CP, Jarrard RE, Hockerman GH, Lamb PW, Barker EL, Henry LK. The external gate of the human and Drosophila serotonin transporters requires a basic/acidic amino acid pair for 3,4-methylenedioxymethamphetamine (MDMA) translocation and the induction of substrate efflux. Biochem Pharmacol 2016; 120:46-55. [PMID: 27638414 PMCID: PMC11851564 DOI: 10.1016/j.bcp.2016.09.006] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/15/2016] [Accepted: 09/08/2016] [Indexed: 11/24/2022]
Abstract
The substituted amphetamine, 3,4-methylenedioxy-methamphetamine (MDMA, ecstasy), is a widely used drug of abuse that induces non-exocytotic release of serotonin, dopamine, and norepinephrine through their cognate transporters as well as blocking the reuptake of neurotransmitter by the same transporters. The resulting dramatic increase in volume transmission and signal duration of neurotransmitters leads to psychotropic, stimulant, and entactogenic effects. The mechanism by which amphetamines drive reverse transport of the monoamines remains largely enigmatic, however, promising outcomes for the therapeutic utility of MDMA for post-traumatic stress disorder and the long-time use of the dopaminergic and noradrenergic-directed amphetamines in treatment of attention-deficit hyperactivity disorder and narcolepsy increases the importance of understanding this phenomenon. Previously, we identified functional differences between the human and Drosophila melanogaster serotonin transporters (hSERT and dSERT, respectively) revealing that MDMA is an effective substrate for hSERT but not dSERT even though serotonin is a potent substrate for both transporters. Chimeric dSERT/hSERT transporters revealed that the molecular components necessary for recognition of MDMA as a substrate was linked to regions of the protein flanking transmembrane domains (TM) V through IX. Here, we performed species-scanning mutagenesis of hSERT, dSERT and C. elegans SERT (ceSERT) along with biochemical and electrophysiological analysis and identified a single amino acid in TM10 (Glu394, hSERT; Asn484, dSERT, Asp517, ceSERT) that is primarily responsible for the differences in MDMA recognition. Our findings reveal that an acidic residue is necessary at this position for MDMA recognition as a substrate and serotonin releaser.
Collapse
Affiliation(s)
- Natalie R Sealover
- Department of Medicinal Chemistry and Molecular Pharmacology, Purdue University College of Pharmacy, West Lafayette, IN 47907, United States
| | - Bruce Felts
- Department of Biomedical Sciences, University of North Dakota School of Medicine and Health Sciences, Grand Forks, ND 58202, United States
| | - Charles P Kuntz
- Department of Medicinal Chemistry and Molecular Pharmacology, Purdue University College of Pharmacy, West Lafayette, IN 47907, United States
| | - Rachel E Jarrard
- Department of Medicinal Chemistry and Molecular Pharmacology, Purdue University College of Pharmacy, West Lafayette, IN 47907, United States
| | - Gregory H Hockerman
- Department of Medicinal Chemistry and Molecular Pharmacology, Purdue University College of Pharmacy, West Lafayette, IN 47907, United States
| | | | - Eric L Barker
- Department of Medicinal Chemistry and Molecular Pharmacology, Purdue University College of Pharmacy, West Lafayette, IN 47907, United States.
| | - L Keith Henry
- Department of Biomedical Sciences, University of North Dakota School of Medicine and Health Sciences, Grand Forks, ND 58202, United States.
| |
Collapse
|
18
|
Pyakurel P, Privman Champaloux E, Venton BJ. Fast-Scan Cyclic Voltammetry (FSCV) Detection of Endogenous Octopamine in Drosophila melanogaster Ventral Nerve Cord. ACS Chem Neurosci 2016; 7:1112-9. [PMID: 27326831 DOI: 10.1021/acschemneuro.6b00070] [Citation(s) in RCA: 35] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/29/2022] Open
Abstract
Octopamine is an endogenous biogenic amine neurotransmitter, neurohormone, and neuromodulator in invertebrates and has functional analogy with norepinephrine in vertebrates. Fast-scan cyclic voltammetry (FSCV) can detect rapid changes in neurotransmitters, but FSCV has not been optimized for octopamine detection in situ. The goal of this study was to characterize octopamine release in the ventral nerve cord of Drosophila larvae for the first time. A FSCV waveform was optimized so that the potential for octopamine oxidation would not be near the switching potential where interferences can occur. Endogenous octopamine release was stimulated by genetically inserting either the ATP sensitive channel, P2X2, or the red-light sensitive channelrhodopsin, CsChrimson, into cells expressing tyrosine decarboxylase (TDC), an octopamine synthesis enzyme. To ensure that release is due to octopamine and not the precursor tyramine, the octopamine synthesis inhibitor disulfiram was applied, and the signal decreased by 80%. Stimulated release was vesicular, and a 2 s continuous light stimulation of CsChrimson evoked 0.22 ± 0.03 μM of octopamine release in the larval ventral nerve cord. Repeated stimulations were stable with 2 or 5 min interstimulation times. With pulsed stimulations, the release was dependent on the frequency of applied light pulse. An octopamine transporter has not been identified, and blockers of the dopamine transporter and serotonin transporter had no significant effect on the clearance time of octopamine, suggesting that they do not take up octopamine. This study shows that octopamine can be monitored in Drosophila, facilitating future studies of how octopamine release functions in the insect brain.
Collapse
Affiliation(s)
- Poojan Pyakurel
- Department of Chemistry, University of Virginia, PO Box 400319, Charlottesville, Virginia 22904, United States
| | - Eve Privman Champaloux
- Department of Chemistry, University of Virginia, PO Box 400319, Charlottesville, Virginia 22904, United States
| | - B. Jill Venton
- Department of Chemistry, University of Virginia, PO Box 400319, Charlottesville, Virginia 22904, United States
| |
Collapse
|
19
|
Abstract
Biogenic amine transporters mediate two important steps in the reuptake and recycling of monoamines released by neurons in the central nervous system. First, high-affinity transporters found in the plasma membrane of neurons and glial cells mediate the removal of neurotransmitter from the extracellular space, thus terminating the action of the monoamines serotonin, norepinephrine, and dopamine. Within the cell, vesicular transporters repackage monoamines into vesicles for additional cycles of release. Two gene families are involved in the transport of the biogenic amines—the Na+/Cl--dependent plasma membrane carriers and the H+-dependent vesicular amine carriers. These transporters are known to regulate neurotransmitter con centrations in monoaminergic pathways and are the primary targets for a wide variety of clinically important antidepressants, antihypertensives, stimulants, and stimulant drugs of abuse. The Neuroscientist 1:259-267, 1995
Collapse
Affiliation(s)
- Susan G. Amara
- The Vollum Institute and Howard Hughes Medical Institute
Oregon Health Sciences University Portland, Oregon
| |
Collapse
|
20
|
Davis BA, Nagarajan A, Forrest LR, Singh SK. Mechanism of Paroxetine (Paxil) Inhibition of the Serotonin Transporter. Sci Rep 2016; 6:23789. [PMID: 27032980 PMCID: PMC4817154 DOI: 10.1038/srep23789] [Citation(s) in RCA: 40] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/19/2016] [Accepted: 03/15/2016] [Indexed: 11/11/2022] Open
Abstract
The serotonin transporter (SERT) is an integral membrane protein that exploits preexisting sodium-, chloride-, and potassium ion gradients to catalyze the thermodynamically unfavorable movement of synaptic serotonin into the presynaptic neuron. SERT has garnered significant clinical attention partly because it is the target of multiple psychoactive agents, including the antidepressant paroxetine (Paxil), the most potent selective serotonin reuptake inhibitor known. However, the binding site and orientation of paroxetine in SERT remain controversial. To provide molecular insight, we constructed SERT homology models based on the Drosophila melanogaster dopamine transporter and docked paroxetine to these models. We tested the predicted binding configurations with a combination of radioligand binding and flux assays on wild-type and mutant SERTs. Our data suggest that the orientation of paroxetine, specifically its fluorophenyl ring, in SERT’s substrate binding site directly depends on this pocket’s charge distribution, and thereby provide an avenue toward understanding and enhancing high-affinity antidepressant activity.
Collapse
Affiliation(s)
- Bruce A Davis
- Department of Cellular and Molecular Physiology, Yale University School of Medicine, 333 Cedar Street, New Haven, CT 06520 USA
| | - Anu Nagarajan
- Computational Structural Biology Section, National Institute of Neurological Disorders and Stroke, 35 Convent Drive, Bethesda, MD 20892 USA
| | - Lucy R Forrest
- Computational Structural Biology Section, National Institute of Neurological Disorders and Stroke, 35 Convent Drive, Bethesda, MD 20892 USA
| | - Satinder K Singh
- Department of Cellular and Molecular Physiology, Yale University School of Medicine, 333 Cedar Street, New Haven, CT 06520 USA
| |
Collapse
|
21
|
Modulatory Action by the Serotonergic System: Behavior and Neurophysiology in Drosophila melanogaster. Neural Plast 2016; 2016:7291438. [PMID: 26989517 PMCID: PMC4773565 DOI: 10.1155/2016/7291438] [Citation(s) in RCA: 41] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/23/2015] [Revised: 12/09/2015] [Accepted: 12/13/2015] [Indexed: 01/13/2023] Open
Abstract
Serotonin modulates various physiological processes and behaviors. This study investigates the role of 5-HT in locomotion and feeding behaviors as well as in modulation of sensory-motor circuits. The 5-HT biosynthesis was dysregulated by feeding Drosophila larvae 5-HT, a 5-HT precursor, or an inhibitor of tryptophan hydroxylase during early stages of development. The effects of feeding fluoxetine, a selective serotonin reuptake inhibitor, during early second instars were also examined. 5-HT receptor subtypes were manipulated using RNA interference mediated knockdown and 5-HT receptor insertional mutations. Moreover, synaptic transmission at 5-HT neurons was blocked or enhanced in both larvae and adult flies. The results demonstrate that disruption of components within the 5-HT system significantly impairs locomotion and feeding behaviors in larvae. Acute activation of 5-HT neurons disrupts normal locomotion activity in adult flies. To determine which 5-HT receptor subtype modulates the evoked sensory-motor activity, pharmacological agents were used. In addition, the activity of 5-HT neurons was enhanced by expressing and activating TrpA1 channels or channelrhodopsin-2 while recording the evoked excitatory postsynaptic potentials (EPSPs) in muscle fibers. 5-HT2 receptor activation mediates a modulatory role in a sensory-motor circuit, and the activation of 5-HT neurons can suppress the neural circuit activity, while fluoxetine can significantly decrease the sensory-motor activity.
Collapse
|
22
|
Majeed ZR, Ritter K, Robinson J, Blümich SLE, Brailoiu E, Cooper RL. New insights into the acute actions from a high dosage of fluoxetine on neuronal and cardiac function: Drosophila, crayfish and rodent models. Comp Biochem Physiol C Toxicol Pharmacol 2015; 176-177:52-61. [PMID: 26232582 DOI: 10.1016/j.cbpc.2015.07.010] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/09/2015] [Revised: 07/22/2015] [Accepted: 07/23/2015] [Indexed: 12/31/2022]
Abstract
The commonly used mood altering drug fluoxetine (Prozac) in humans has a low occurrence in reports of harmful effects from overdose; however, individuals with altered metabolism of the drug and accidental overdose have led to critical conditions and even death. We addressed direct actions of high concentrations on synaptic transmission at neuromuscular junctions (NMJs), neural properties, and cardiac function unrelated to fluoxetine's action as a selective 5-HT reuptake inhibitor. There appears to be action in blocking action potentials in crayfish axons, enhanced occurrences of spontaneous synaptic vesicle fusion events in the presynaptic terminals at NMJs of both Drosophila and crayfish. In rodent neurons, cytoplasmic Ca(2+) rises by fluoxetine and is thapsigargin dependent. The Drosophila larval heart showed a dose dependent effect in cardiac arrest. Acute paralytic behavior in crayfish occurred at a systemic concentration of 2mM. A high percentage of death as well as slowed development occurred in Drosophila larvae consuming food containing 100μM fluoxetine. The release of Ca(2+) from the endoplasmic reticulum in neurons and the cardiac tissue as well as blockage of voltage-gated Na(+) channels in neurons could explain the effects on the whole animal as well as the isolated tissues. The use of various animal models in demonstrating the potential mechanisms for the toxic effects with high doses of fluoxetine maybe beneficial for acute treatments in humans. Future studies in determining how fluoxetine is internalized in cells and if there are subtle effects of these mentioned mechanisms presented with chronic therapeutic doses are of general interest.
Collapse
Affiliation(s)
- Zana R Majeed
- Department of Biology, University of Kentucky, USA; Lexington, KY, USA; Department of Biology, University of Salahaddin, Erbil, Iraq
| | - Kyle Ritter
- Department of Biology, University of Kentucky, USA; Lexington, KY, USA; Centre College, Danville, KY, USA
| | - Jonathan Robinson
- Department of Biology, University of Kentucky, USA; Lexington, KY, USA; Morehead State University, Morehead, KY, USA
| | - Sandra L E Blümich
- Department of Biology, University of Kentucky, USA; Lexington, KY, USA; V.M.F., University of Leipzig, Leipzig, Germany
| | | | - Robin L Cooper
- Department of Biology, University of Kentucky, USA; Lexington, KY, USA.
| |
Collapse
|
23
|
Denno ME, Privman E, Venton BJ. Analysis of neurotransmitter tissue content of Drosophila melanogaster in different life stages. ACS Chem Neurosci 2015; 6:117-23. [PMID: 25437353 PMCID: PMC4304510 DOI: 10.1021/cn500261e] [Citation(s) in RCA: 31] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/28/2022] Open
Abstract
![]()
Drosophila melanogaster is a widely used model organism for studying neurological diseases
with similar neurotransmission to mammals. While both larva and adult Drosophila have central nervous systems, not much is known
about how neurotransmitter tissue content changes through development.
In this study, we quantified tyramine, serotonin, octopamine, and
dopamine in larval, pupal, and adult fly brains using capillary electrophoresis
coupled to fast-scan cyclic voltammetry. Tyramine and octopamine content
varied between life stages, with almost no octopamine being present
in the pupa, while tyramine levels in the pupa were very high. Adult
females had significantly higher dopamine content than males, but
no other neurotransmitters were dependent on sex in the adult. Understanding
the tissue content of different life stages will be beneficial for
future work comparing the effects of diseases on tissue content throughout
development.
Collapse
Affiliation(s)
- Madelaine E. Denno
- Department of Chemistry, ‡Neuroscience Graduate
Program, and §Medical Scientist Training Program, University of Virginia, Charlottesville, Virginia 22904, United States
| | - Eve Privman
- Department of Chemistry, ‡Neuroscience Graduate
Program, and §Medical Scientist Training Program, University of Virginia, Charlottesville, Virginia 22904, United States
| | - B. Jill Venton
- Department of Chemistry, ‡Neuroscience Graduate
Program, and §Medical Scientist Training Program, University of Virginia, Charlottesville, Virginia 22904, United States
| |
Collapse
|
24
|
Xiao N, Privman E, Venton BJ. Optogenetic control of serotonin and dopamine release in Drosophila larvae. ACS Chem Neurosci 2014; 5:666-73. [PMID: 24849718 DOI: 10.1021/cn500044b] [Citation(s) in RCA: 31] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/30/2022] Open
Abstract
Optogenetic control of neurotransmitter release is an elegant method to investigate neurobiological mechanisms with millisecond precision and cell type-specific resolution. Channelrhodopsin-2 (ChR2) can be expressed in specific neurons, and blue light used to activate those neurons. Previously, in Drosophila, neurotransmitter release and uptake have been studied after continuous optical illumination. In this study, we investigated the effects of pulsed optical stimulation trains on serotonin or dopamine release in larval ventral nerve cords. In larvae with ChR2 expressed in serotonergic neurons, low-frequency stimulations produced a distinct, steady-state response while high-frequency patterns were peak shaped. Evoked serotonin release increased with increasing stimulation frequency and then plateaued. The steady-state response and the frequency dependence disappeared after administering the uptake inhibitor fluoxetine, indicating that uptake plays a significant role in regulating the extracellular serotonin concentration. Pulsed stimulations were also used to evoke dopamine release in flies expressing ChR2 in dopaminergic neurons and similar frequency dependence was observed. Release due to pulsed optical stimulations was modeled to determine the uptake kinetics. For serotonin, Vmax was 0.54 ± 0.07 μM/s and Km was 0.61 ± 0.04 μM; and for dopamine, Vmax was 0.12 ± 0.03 μM/s and Km was 0.45 ± 0.13 μM. The amount of serotonin released per stimulation pulse was 4.4 ± 1.0 nM, and the amount of dopamine was 1.6 ± 0.3 nM. Thus, pulsed optical stimulations can be used to mimic neuronal firing patterns and will allow Drosophila to be used as a model system for studying mechanisms underlying neurotransmission.
Collapse
Affiliation(s)
- Ning Xiao
- Department of Chemistry, University of Virginia, P.O. Box 400319, Charlottesville, Virginia 22904, United States
| | - Eve Privman
- Department of Chemistry, University of Virginia, P.O. Box 400319, Charlottesville, Virginia 22904, United States
| | - B. Jill Venton
- Department of Chemistry, University of Virginia, P.O. Box 400319, Charlottesville, Virginia 22904, United States
| |
Collapse
|
25
|
Drosophila melanogaster as a genetic model system to study neurotransmitter transporters. Neurochem Int 2014; 73:71-88. [PMID: 24704795 DOI: 10.1016/j.neuint.2014.03.015] [Citation(s) in RCA: 70] [Impact Index Per Article: 6.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/13/2013] [Revised: 03/20/2014] [Accepted: 03/24/2014] [Indexed: 12/30/2022]
Abstract
The model genetic organism Drosophila melanogaster, commonly known as the fruit fly, uses many of the same neurotransmitters as mammals and very similar mechanisms of neurotransmitter storage, release and recycling. This system offers a variety of powerful molecular-genetic methods for the study of transporters, many of which would be difficult in mammalian models. We review here progress made using Drosophila to understand the function and regulation of neurotransmitter transporters and discuss future directions for its use.
Collapse
|
26
|
Beckman ML, Pramod AB, Perley D, Henry LK. Stereoselective inhibition of serotonin transporters by antimalarial compounds. Neurochem Int 2013; 73:98-106. [PMID: 24161619 DOI: 10.1016/j.neuint.2013.10.009] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/03/2013] [Revised: 10/11/2013] [Accepted: 10/15/2013] [Indexed: 10/26/2022]
Abstract
The serotonin (5-HT) transporter (SERT) is an integral membrane protein that functions to reuptake 5-HT released into the synapse following neurotransmission. This role serves an important regulatory mechanism in neuronal homeostasis. Previous studies have demonstrated that several clinically important antimalarial compounds inhibit serotonin (5-hydroxytryptamine, 5-HT) reuptake. In this study, we examined the details of antimalarial inhibition of 5-HT transport in both Drosophila (dSERT) and human SERT (hSERT) using electrophysiologic, biochemical and computational approaches. We found that the cinchona alkaloids quinidine and cinchonine, which have identical stereochemistry about carbons 8 and 9, exhibited the greatest inhibition of dSERT and hSERT transporter function whereas quinine and cinchonidine, enantiomers of quinidine and cinchonine, respectively, were weaker inhibitors of dSERT and hSERT. Furthermore, SERT mutations known to decrease the binding affinity of many antidepressants affected the cinchona alkaloids in a stereo-specific manner where the similar inhibitory profiles for quinine and cinchonidine (8S,9R) were distinct from quinidine and cinchonine (8R,9S). Small molecule docking studies with hSERT homology models predict that quinine and cinchonidine bind to the central 5-HT binding site (S1) whereas quinidine and cinchonine bind to the S2 site. Taken together, the data presented here support binding of cinchona alkaloids to two different sites on SERT defined by their stereochemistry which implies separate modes of transporter inhibition. Notably, the most potent antimalarial inhibitors of SERT appear to preferentially bind to the S2 site. Our findings provide important insight related to how this class of drugs can modulate the serotonergic system as well as identify compounds that may discriminate between the S1 and S2 binding sites and serve as lead compounds for novel SERT inhibitors.
Collapse
Affiliation(s)
- Matthew L Beckman
- Department of Biology, Augsburg College, 2211 Riverside Ave S, Minneapolis, MN 55454, United States.
| | - Akula Bala Pramod
- Department of Basic Sciences, University of North Dakota School of Medicine and Health Sciences, 501 North Columbia Road, Room 1700, Grand Forks, ND 58203, United States
| | - Danielle Perley
- Department of Basic Sciences, University of North Dakota School of Medicine and Health Sciences, 501 North Columbia Road, Room 1700, Grand Forks, ND 58203, United States
| | - L Keith Henry
- Department of Basic Sciences, University of North Dakota School of Medicine and Health Sciences, 501 North Columbia Road, Room 1700, Grand Forks, ND 58203, United States.
| |
Collapse
|
27
|
Abstract
Increasingly invertebrates are being used to investigate the molecular and cellular effects of drugs of abuse to explore basic mechanisms of addiction. However, in mammals the principle factors contributing to addiction are long-term adaptive responses to repeated drug use. Here we examined whether adaptive responses to cocaine are also seen in invertebrates using the honey bee model system. Repeated topical treatment with a low dose of cocaine rendered bees resistant to the deleterious motor effects of a higher cocaine dose, indicating the development of physiological tolerance to cocaine in bees. Cocaine inhibits biogenic amine reuptake transporters, but neither acute nor repeated cocaine treatments caused measurable changes in levels of biogenic amines measured in whole bee brains. Our data show clear short and long-term behavioural responses of bees to cocaine administration, but caution that, despite the small size of the bee brain, measures of biogenic amines conducted at the whole-brain level may not reveal neurochemical effects of the drug.
Collapse
|
28
|
Curran KP, Chalasani SH. Serotonin circuits and anxiety: what can invertebrates teach us? INVERTEBRATE NEUROSCIENCE : IN 2012; 12:81-92. [PMID: 22918570 PMCID: PMC3505513 DOI: 10.1007/s10158-012-0140-y] [Citation(s) in RCA: 26] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 04/03/2012] [Accepted: 07/26/2012] [Indexed: 11/08/2022]
Abstract
Fear, a reaction to a threatening situation, is a broadly adaptive feature crucial to the survival and reproductive fitness of individual organisms. By contrast, anxiety is an inappropriate behavioral response often to a perceived, not real, threat. Functional imaging, biochemical analysis, and lesion studies with humans have identified the HPA axis and the amygdala as key neuroanatomical regions driving both fear and anxiety. Abnormalities in these biological systems lead to misregulated fear and anxiety behaviors such as panic attacks and post-traumatic stress disorders. These behaviors are often treated by increasing serotonin levels at synapses, suggesting a role for serotonin signaling in ameliorating both fear and anxiety. Interestingly, serotonin signaling is highly conserved between mammals and invertebrates. We propose that genetically tractable invertebrate models organisms, such as Drosophila melanogaster and Caenorhabditis elegans, are ideally suited to unravel the complexity of the serotonin signaling pathways. These model systems possess well-defined neuroanatomies and robust serotonin-mediated behavior and should reveal insights into how serotonin can modulate human cognitive functions.
Collapse
Affiliation(s)
- Kevin P. Curran
- Molecular Neurobiology Lab, The Salk Institute for Biological Studies, 10010 N. Torrey Pines Road, La Jolla, CA 92037 USA
| | - Sreekanth H. Chalasani
- Molecular Neurobiology Lab, The Salk Institute for Biological Studies, 10010 N. Torrey Pines Road, La Jolla, CA 92037 USA
| |
Collapse
|
29
|
McCoole MD, Atkinson NJ, Graham DI, Grasser EB, Joselow AL, McCall NM, Welker AM, Wilsterman EJ, Baer KN, Tilden AR, Christie AE. Genomic analyses of aminergic signaling systems (dopamine, octopamine and serotonin) in Daphnia pulex. COMPARATIVE BIOCHEMISTRY AND PHYSIOLOGY D-GENOMICS & PROTEOMICS 2012; 7:35-58. [DOI: 10.1016/j.cbd.2011.10.005] [Citation(s) in RCA: 29] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/18/2011] [Revised: 10/26/2011] [Accepted: 10/29/2011] [Indexed: 01/24/2023]
|
30
|
Henry LK, Iwamoto H, Field JR, Kaufmann K, Dawson ES, Jacobs MT, Adams C, Felts B, Zdravkovic I, Armstrong V, Combs S, Solis E, Rudnick G, Noskov SY, DeFelice LJ, Meiler J, Blakely RD. A conserved asparagine residue in transmembrane segment 1 (TM1) of serotonin transporter dictates chloride-coupled neurotransmitter transport. J Biol Chem 2011; 286:30823-30836. [PMID: 21730057 DOI: 10.1074/jbc.m111.250308] [Citation(s) in RCA: 32] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/06/2022] Open
Abstract
Na(+)- and Cl(-)-dependent uptake of neurotransmitters via transporters of the SLC6 family, including the human serotonin transporter (SLC6A4), is critical for efficient synaptic transmission. Although residues in the human serotonin transporter involved in direct Cl(-) coordination of human serotonin transport have been identified, the role of Cl(-) in the transport mechanism remains unclear. Through a combination of mutagenesis, chemical modification, substrate and charge flux measurements, and molecular modeling studies, we reveal an unexpected role for the highly conserved transmembrane segment 1 residue Asn-101 in coupling Cl(-) binding to concentrative neurotransmitter uptake.
Collapse
Affiliation(s)
- L Keith Henry
- Departments of Pharmacology, Vanderbilt University School of Medicine, Nashville, Tennessee 37232-8548; Center for Structural Biology, Vanderbilt University School of Medicine, Nashville, Tennessee 37232-8548; Department of Pharmacology, Physiology, and Therapeutics, University of North Dakota, Grand Forks, North Dakota 58203.
| | - Hideki Iwamoto
- Departments of Pharmacology, Vanderbilt University School of Medicine, Nashville, Tennessee 37232-8548
| | - Julie R Field
- Departments of Pharmacology, Vanderbilt University School of Medicine, Nashville, Tennessee 37232-8548
| | - Kristian Kaufmann
- Chemistry, Vanderbilt University School of Medicine, Nashville, Tennessee 37232-8548
| | - Eric S Dawson
- Center for Structural Biology, Vanderbilt University School of Medicine, Nashville, Tennessee 37232-8548; Biochemistry, Vanderbilt University School of Medicine, Nashville, Tennessee 37232-8548; Vanderbilt Center for Neuroscience Drug Discovery, Vanderbilt University School of Medicine, Nashville, Tennessee 37232-8548
| | - Miriam T Jacobs
- Department of Pharmacology, Yale University School of Medicine, New Haven, Connecticut 06520-8066
| | - Chelsea Adams
- Department of Pharmacology, Physiology, and Therapeutics, University of North Dakota, Grand Forks, North Dakota 58203
| | - Bruce Felts
- Department of Pharmacology, Physiology, and Therapeutics, University of North Dakota, Grand Forks, North Dakota 58203
| | - Igor Zdravkovic
- Institute for Biocomplexity and Informatics, Department of Biological Sciences, University of Calgary, Calgary, Alberta T2N 1N4, Canada
| | - Vanessa Armstrong
- Department of Pharmacology, Physiology, and Therapeutics, University of North Dakota, Grand Forks, North Dakota 58203
| | - Steven Combs
- Chemistry, Vanderbilt University School of Medicine, Nashville, Tennessee 37232-8548
| | - Ernesto Solis
- Departments of Pharmacology, Vanderbilt University School of Medicine, Nashville, Tennessee 37232-8548
| | - Gary Rudnick
- Department of Pharmacology, Yale University School of Medicine, New Haven, Connecticut 06520-8066
| | - Sergei Y Noskov
- Institute for Biocomplexity and Informatics, Department of Biological Sciences, University of Calgary, Calgary, Alberta T2N 1N4, Canada
| | - Louis J DeFelice
- Departments of Pharmacology, Vanderbilt University School of Medicine, Nashville, Tennessee 37232-8548; Center for Molecular Neuroscience, Vanderbilt University School of Medicine, Nashville, Tennessee 37232-8548
| | - Jens Meiler
- Departments of Pharmacology, Vanderbilt University School of Medicine, Nashville, Tennessee 37232-8548; Center for Structural Biology, Vanderbilt University School of Medicine, Nashville, Tennessee 37232-8548; Chemistry, Vanderbilt University School of Medicine, Nashville, Tennessee 37232-8548
| | - Randy D Blakely
- Departments of Pharmacology, Vanderbilt University School of Medicine, Nashville, Tennessee 37232-8548; Center for Molecular Neuroscience, Vanderbilt University School of Medicine, Nashville, Tennessee 37232-8548; Psychiatry, Vanderbilt University School of Medicine, Nashville, Tennessee 37232-8548.
| |
Collapse
|
31
|
Larsen MB, Sonders MS, Mortensen OV, Larson GA, Zahniser NR, Amara SG. Dopamine transport by the serotonin transporter: a mechanistically distinct mode of substrate translocation. J Neurosci 2011; 31:6605-15. [PMID: 21525301 PMCID: PMC3107525 DOI: 10.1523/jneurosci.0576-11.2011] [Citation(s) in RCA: 79] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/02/2011] [Revised: 03/04/2011] [Accepted: 03/10/2011] [Indexed: 01/23/2023] Open
Abstract
The serotonin transporter (SERT) is the principal mechanism for terminating serotonin (5-HT) signals in the nervous system and is a site of action for a variety of psychoactive drugs including antidepressants, amphetamines, and cocaine. Here we show that human SERTs (hSERTs) and rat SERTs are capable of robust dopamine (DA) uptake through a process that differs mechanistically from 5-HT transport in several unanticipated ways. DA transport by hSERT has a higher maximum velocity than 5-HT transport, requires significantly higher Na(+) and Cl(-) concentrations to sustain transport, is inhibited noncompetitively by 5-HT, and is more sensitive to SERT inhibitors, including selective serotonin reuptake inhibitors. We use a thiol-reactive methane thiosulfonate (MTS) reagent to modify a conformationally sensitive cysteine residue to demonstrate that hSERT spends more time in an outward facing conformation when transporting DA than when transporting 5-HT. Cotransfection of an inactive or an MTS-sensitive SERT with wild-type SERT subunits reveals an absence of cooperative interactions between subunits during DA but not 5-HT transport. To establish the physiological relevance of this mechanism for DA clearance, we show using in vivo high-speed chronoamperometry that SERT has the capacity to clear extracellularly applied DA in the hippocampal CA3 region of anesthetized rats. Together, these observations suggest the possibility that SERT serves as a DA transporter in vivo and highlight the idea that there can be distinct modes of transport of alternative physiological substrates by SERT.
Collapse
Affiliation(s)
- Mads Breum Larsen
- Department of Neurobiology, University of Pittsburgh School of Medicine, Pittsburgh, Pennsylvania 15260
| | - Mark S. Sonders
- Center for Molecular Recognition, Columbia University and New York State Psychiatric Institute, New York, New York 10032, and
| | - Ole Valente Mortensen
- Department of Neurobiology, University of Pittsburgh School of Medicine, Pittsburgh, Pennsylvania 15260
| | - Gaynor A. Larson
- Department of Pharmacology, University of Colorado Denver, Aurora, Colorado 80045
| | - Nancy R. Zahniser
- Department of Pharmacology, University of Colorado Denver, Aurora, Colorado 80045
| | - Susan G. Amara
- Department of Neurobiology, University of Pittsburgh School of Medicine, Pittsburgh, Pennsylvania 15260
| |
Collapse
|
32
|
Giang T, Ritze Y, Rauchfuss S, Ogueta M, Scholz H. The serotonin transporter expression in Drosophila melanogaster. J Neurogenet 2011; 25:17-26. [PMID: 21314480 DOI: 10.3109/01677063.2011.553002] [Citation(s) in RCA: 31] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/13/2022]
Abstract
The serotonin transporter is an important regulator of serotonergic signaling. In order to analyze where the Drosophila melanogaster ortholog of the mammalian serotonin transporter (dSERT) is expressed in the nervous system, a dSERT antibody serum was used. Ectopic expression studies and loss of function analysis revealed that the dSERT antibody serum specifically recognizes dSERT. It was shown that in the embryonic nervous system dSERT is expressed in a subset of Engrailed-positive neurons. In the larval brain, dSERT is exclusively expressed in serotonergic neurons, all of which express dSERT. dSERT-positive neurons surround almost all brain neuropiles. In the mushroom body of the adult brain, extrinsic serotonergic neurons expressing dSERT engulf the mushroom body lobes. These neurons show regional differences in dSERT and serotonin expression. At the presynaptic terminals, serotonin release is sterically linked to serotonin reuptake. In contrast to this, there are other areas in serotonergic neurons where dSERT expression and/or function are uncoupled from synaptic neurotransmitter recycling and serotonin release. The localization pattern of dSERT can be employed to further understanding and analysis of serotonergic networks.
Collapse
Affiliation(s)
- Thomas Giang
- Department of Animal Physiology, University of Cologne, Köln, Germany
| | | | | | | | | |
Collapse
|
33
|
Chatterjee N, Rollins J, Mahowald AP, Bazinet C. Neurotransmitter Transporter-Like: a male germline-specific SLC6 transporter required for Drosophila spermiogenesis. PLoS One 2011; 6:e16275. [PMID: 21298005 PMCID: PMC3029318 DOI: 10.1371/journal.pone.0016275] [Citation(s) in RCA: 13] [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: 09/14/2010] [Accepted: 12/13/2010] [Indexed: 02/03/2023] Open
Abstract
The SLC6 class of membrane transporters, known primarily as neurotransmitter transporters, is increasingly appreciated for its roles in nutritional uptake of amino acids and other developmentally specific functions. A Drosophila SLC6 gene, Neurotransmitter transporter-like (Ntl), is expressed only in the male germline. Mobilization of a transposon inserted near the 3' end of the Ntl coding region yields male-sterile mutants defining a single complementation group. Germline transformation with Ntl cDNAs under control of male germline-specific control elements restores Ntl/Ntl homozygotes to normal fertility, indicating that Ntl is required only in the germ cells. In mutant males, sperm morphogenesis appears normal, with elongated, individualized and coiled spermiogenic cysts accumulating at the base of the testes. However, no sperm are transferred to the seminal vesicle. The level of polyglycylation of Ntl mutant sperm tubulin appears to be significantly lower than that of wild type controls. Glycine transporters are the most closely related SLC6 transporters to Ntl, suggesting that Ntl functions as a glycine transporter in developing sperm, where augmentation of the cytosolic pool of glycine may be required for the polyglycylation of the massive amounts of tubulin in the fly's giant sperm. The male-sterile phenotype of Ntl mutants may provide a powerful genetic system for studying the function of an SLC6 transporter family in a model organism.
Collapse
Affiliation(s)
- Nabanita Chatterjee
- Department of Biological Sciences, St. John's University, Jamaica, New York, United States of America
| | - Janet Rollins
- Department of Biological Sciences, St. John's University, Jamaica, New York, United States of America
| | - Anthony P. Mahowald
- Department of Molecular Genetics and Cell Biology, University of Chicago, Chicago, Illinois, United States of America
| | - Christopher Bazinet
- Department of Biological Sciences, St. John's University, Jamaica, New York, United States of America
| |
Collapse
|
34
|
Huber R, Panksepp JB, Nathaniel T, Alcaro A, Panksepp J. Drug-sensitive reward in crayfish: an invertebrate model system for the study of SEEKING, reward, addiction, and withdrawal. Neurosci Biobehav Rev 2010; 35:1847-53. [PMID: 21182861 DOI: 10.1016/j.neubiorev.2010.12.008] [Citation(s) in RCA: 45] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/08/2010] [Accepted: 12/14/2010] [Indexed: 11/17/2022]
Abstract
In mammals, rewarding properties of drugs depend on their capacity to activate appetitive motivational states. With the underlying mechanisms strongly conserved in evolution, invertebrates have recently emerged as a powerful new model in addiction research. In crayfish natural reward has proven surprisingly sensitive to human drugs of abuse, opening an unlikely avenue of research into the basic biological mechanisms of drug addiction. In a series of studies we first examined the presence of natural reward systems in crayfish, then characterized its sensitivity to a wide range of human drugs of abuse. A conditioned place preference (CPP) paradigm was used to demonstrate that crayfish seek out those environments that had previously been paired with the psychostimulants cocaine and amphetamine, and the opioid morphine. The administration of amphetamine exerted its effects at a number of sites, including the stimulation of circuits for active exploratory behaviors (i.e., SEEKING). A further study examined morphine-induced reward, extinction and reinstatement in crayfish. Repeated intra-circulatory infusions of morphine served as a reward when paired with distinct visual or tactile cues. Morphine-induced CPP was extinguished after repeated saline injections. Following this extinction phase, morphine-experienced crayfish were once again challenged with the drug. The priming injections of morphine reinstated CPP at all tested doses, suggesting that morphine-induced CPP is unrelenting. In an exploration of drug-associated behavioral sensitization in crayfish we concurrently mapped measures of locomotion and rewarding properties of morphine. Single and repeated intra-circulatory infusions of morphine resulted in persistent locomotory sensitization, even 5 days following the infusion. Moreover, a single dose of morphine was sufficient to induce long-term behavioral sensitization. CPP for morphine and context-dependent cues could not be disrupted over a drug free period of 5 days. This work demonstrates that crayfish offer a comparative and complementary approach in addiction research. Serving as an invertebrate animal model for the exposure to mammalian drugs of abuse, modularly organized and experimentally accessible nervous systems render crayfish uniquely suited for studying (1) the basic biological mechanisms of drug effects, (2) to explore how the appetitive/seeking disposition is implemented in a simple neural system, and (3) how such a disposition is related to the rewarding action of drugs of abuse. This work aimed to contribute an evolutionary, comparative context to our understanding of a key component in learning, and of natural reward as an important life-sustaining process.
Collapse
Affiliation(s)
- Robert Huber
- J.P. Scott Center for Neuroscience, Mind & Behavior, Department of Biological Sciences, Bowling Green State University, Bowling Green, OH 43403, USA.
| | | | | | | | | |
Collapse
|
35
|
Mercado CP, Kilic F. Molecular mechanisms of SERT in platelets: regulation of plasma serotonin levels. Mol Interv 2010; 10:231-41. [PMID: 20729489 DOI: 10.1124/mi.10.4.6] [Citation(s) in RCA: 176] [Impact Index Per Article: 11.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/16/2022]
Abstract
The serotonin transporter (SERT) on platelets is a primary mechanism for serotonin (5HT) uptake from the blood plasma. Alteration in plasma 5HT level is associated with a number of cardiovascular diseases and disorders. Therefore, the regulation of the transporter's activity represents a key mechanism to stabilize the concentration of plasma 5HT. There is a biphasic relationship between plasma 5HT elevation, loss of surface SERT, and depletion of platelet 5HT. Specifically, in platelets, plasma membrane SERT levels and platelet 5HT uptake initially rise as plasma 5HT levels are increased but then fall below normal as the plasma 5HT level continues to rise. Therefore, we propose that elevated plasma 5HT limits its own uptake in platelets by down-regulating SERT as well as modifying the characteristics of SERT partners in the membrane trafficking pathway. This review will summarize current findings regarding the biochemical mechanisms by which elevated 5HT downregulates the expression of SERT on the platelet membrane. Intriguing aspects of this regulation include the intracellular interplay of SERT with the small G protein Rab4 and the concerted 5HT-mediated phosphorylation of vimentin.
Collapse
Affiliation(s)
- Charles P Mercado
- Department of Biochemistry and Molecular Biology College of Medicine, The University of Arkansas for Medical Sciences Little Rock, Arkansas 72205, USA
| | | |
Collapse
|
36
|
Andersen J, Kristensen AS, Bang-Andersen B, Strømgaard K. Recent advances in the understanding of the interaction of antidepressant drugs with serotonin and norepinephrine transporters. Chem Commun (Camb) 2009:3677-92. [PMID: 19557250 DOI: 10.1039/b903035m] [Citation(s) in RCA: 83] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2022]
Abstract
The biogenic monoamine transporters are integral membrane proteins that perform active transport of extracellular dopamine, serotonin and norepinephrine into cells. These transporters are targets for therapeutic agents such as antidepressants, as well as addictive substances such as cocaine and amphetamine. Seminal advances in the understanding of the structure and function of this transporter family have recently been accomplished by structural studies of a bacterial transporter, as well as medicinal chemistry and pharmacological studies of mammalian transporters. This feature article focuses on antidepressant drugs that act on the serotonin and/or the norepinephrine transporters. Specifically, we focus on structure-activity relationships of these drugs with emphasis on relationships between their molecular properties and the current knowledge of transporter structure.
Collapse
Affiliation(s)
- Jacob Andersen
- Department of Medicinal Chemistry, University of Copenhagen, Universitetsparken 2, DK-2100 Copenhagen, Denmark.
| | | | | | | |
Collapse
|
37
|
Barron AB, Maleszka R, Helliwell PG, Robinson GE. Effects of cocaine on honey bee dance behaviour. ACTA ACUST UNITED AC 2009; 212:163-8. [PMID: 19112134 DOI: 10.1242/jeb.025361] [Citation(s) in RCA: 42] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/21/2022]
Abstract
The role of cocaine as an addictive drug of abuse in human society is hard to reconcile with its ecological role as a natural insecticide and plant-protective compound, preventing herbivory of coca plants (Erythroxylum spp.). This paradox is often explained by proposing a fundamental difference in mammalian and invertebrate responses to cocaine, but here we show effects of cocaine on honey bees (Apis mellifera L.) that parallel human responses. Forager honey bees perform symbolic dances to advertise the location and value of floral resources to their nest mates. Treatment with a low dose of cocaine increased the likelihood and rate of bees dancing after foraging but did not otherwise increase locomotor activity. This is consistent with cocaine causing forager bees to overestimate the value of the floral resources they collected. Further, cessation of chronic cocaine treatment caused a withdrawal-like response. These similarities likely occur because in both insects and mammals the biogenic amine neuromodulator systems disrupted by cocaine perform similar roles as modulators of reward and motor systems. Given these analogous responses to cocaine in insects and mammals, we propose an alternative solution to the paradox of cocaine reinforcement. Ecologically, cocaine is an effective plant defence compound via disruption of herbivore motor control but, because the neurochemical systems targeted by cocaine also modulate reward processing, the reinforcing properties of cocaine occur as a ;side effect'.
Collapse
Affiliation(s)
- Andrew B Barron
- ARC Centre for Molecular Genetics of Development, Research School of Biological Sciences, Australian National University, Canberra, ACT 2601, Australia.
| | | | | | | |
Collapse
|
38
|
Borue X, Cooper S, Hirsh J, Condron B, Venton BJ. Quantitative evaluation of serotonin release and clearance in Drosophila. J Neurosci Methods 2009; 179:300-8. [PMID: 19428541 DOI: 10.1016/j.jneumeth.2009.02.013] [Citation(s) in RCA: 62] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/13/2008] [Revised: 02/19/2009] [Accepted: 02/19/2009] [Indexed: 10/21/2022]
Abstract
Serotonin signaling plays a key role in the regulation of development, mood and behavior. Drosophila is well suited for the study of the basic mechanisms of serotonergic signaling, but the small size of its nervous system has previously precluded the direct measurements of neurotransmitters. This study demonstrates the first real-time measurements of changes in extracellular monoamine concentrations in a single larval Drosophila ventral nerve cord. Channelrhodopsin-2-mediated, neuronal type-specific stimulation is used to elicit endogenous serotonin release, which is detected using fast-scan cyclic voltammetry at an implanted microelectrode. Release is decreased when serotonin synthesis or packaging are pharmacologically inhibited, confirming that the detected substance is serotonin. Similar to tetanus-evoked serotonin release in mammals, evoked serotonin concentrations are 280-640nM in the fly, depending on the stimulation length. Extracellular serotonin signaling is prolonged after administering cocaine or fluoxetine, showing that transport regulates the clearance of serotonin from the extracellular space. When ChR2 is targeted to dopaminergic neurons, dopamine release is measured demonstrating that this method is broadly applicable to other neurotransmitter systems. This study shows that the dynamics of serotonin release and reuptake in Drosophila are analogous to those in mammals, making this simple organism more useful for the study of the basic physiological mechanisms of serotonergic signaling.
Collapse
Affiliation(s)
- Xenia Borue
- Medical Scientist Training Program, University of Virginia, Charlottesville, VA 22904, USA
| | | | | | | | | |
Collapse
|
39
|
Drosophila vesicular monoamine transporter mutants can adapt to reduced or eliminated vesicular stores of dopamine and serotonin. Genetics 2008; 181:525-41. [PMID: 19033154 DOI: 10.1534/genetics.108.094110] [Citation(s) in RCA: 42] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/18/2022] Open
Abstract
Physiologic and pathogenic changes in amine release induce dramatic behavioral changes, but the underlying cellular mechanisms remain unclear. To investigate these adaptive processes, we have characterized mutations in the Drosophila vesicular monoamine transporter (dVMAT), which is required for the vesicular storage of dopamine, serotonin, and octopamine. dVMAT mutant larvae show reduced locomotion and decreased electrical activity in motoneurons innervating the neuromuscular junction (NMJ) implicating central amines in the regulation of these activities. A parallel increase in evoked glutamate release by the motoneuron is consistent with a homeostatic adaptation at the NMJ. Despite the importance of aminergic signaling for regulating locomotion and other behaviors, adult dVMAT homozygous null mutants survive under conditions of low population density, thus allowing a phenotypic characterization of adult behavior. Homozygous mutant females are sterile and show defects in both egg retention and development; males also show reduced fertility. Homozygotes show an increased attraction to light but are mildly impaired in geotaxis and escape behaviors. In contrast, heterozygous mutants show an exaggerated escape response. Both hetero- and homozygous mutants demonstrate an altered behavioral response to cocaine. dVMAT mutants define potentially adaptive responses to reduced or eliminated aminergic signaling and will be useful to identify the underlying molecular mechanisms.
Collapse
|
40
|
Haydon PG, Blendy J, Moss SJ, Rob Jackson F. Astrocytic control of synaptic transmission and plasticity: a target for drugs of abuse? Neuropharmacology 2008; 56 Suppl 1:83-90. [PMID: 18647612 DOI: 10.1016/j.neuropharm.2008.06.050] [Citation(s) in RCA: 81] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/02/2008] [Revised: 06/24/2008] [Accepted: 06/25/2008] [Indexed: 02/03/2023]
Abstract
It is well recognized that drugs of abuse lead to plastic changes in synapses and that these long-term modifications have the potential to underlie adaptive changes of the brain that lead to substance abuse. However the variety of molecular mechanisms involved in these responses are not completely defined. We are just beginning to understand some of the roles of glial cells that are associated with synapses. At many synapses an astrocyte process is associated with pre- and postsynaptic neuron processes leading to the naming of this synaptic structure as the Tripartite Synapse. Therefore, these glial cells are positioned so that they influence synaptic transmission and thus could potentially regulate the actions of some drugs of abuse. In mammalian systems there are correlations between long-term structural changes in astrocytes and responses to drugs of abuse. However, whether such changes in glia impact brain function and subsequent behaviors associated with addiction is poorly understood. Studies using Drosophila show important roles of fly glia in mediating responses to cocaine pointing to the potential for the involvement of mammalian glia in the brain's responses to this as well as other drugs. In agreement with this possibility three receptor systems known to be important in substance abuse, mGluR5, GABA(B) and CB-1 receptors, are all expressed by astrocytes and the activation of these glial receptors is now known to impact neuronal excitability and synaptic transmission. Given our new knowledge about the presence of reciprocal signaling between astrocytes and synapses we are now at a time when it becomes appropriate to determine how glial cells respond to drugs of abuse and whether they contribute to the changes in brain function underlying substance abuse.
Collapse
Affiliation(s)
- Philip G Haydon
- Department of Neuroscience, University of Pennsylvania School of Medicine, Philadelphia, PA 19104, USA.
| | | | | | | |
Collapse
|
41
|
Hardie SL, Zhang JX, Hirsh J. Trace amines differentially regulate adult locomotor activity, cocaine sensitivity, and female fertility in Drosophila melanogaster. Dev Neurobiol 2007; 67:1396-405. [PMID: 17638385 DOI: 10.1002/dneu.20459] [Citation(s) in RCA: 49] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/11/2022]
Abstract
The trace biogenic amines tyramine and octopamine are found in the nervous systems of animals ranging in complexity from nematodes to mammals. In insects such as Drosophila melanogaster, the trace amine octopamine is a well-established neuromodulator that mediates a diverse range of physiological processes, but an independent role for tyramine is less clear. Tyramine is synthesized from tyrosine by the enzyme tyrosine decarboxylase (TDC). We previously reported the identification of two Tdc genes in Drosophila: the peripherally-expressed Tdc1 and the neurally-expressed Tdc2. To further clarify the neural functions of the trace amines in Drosophila, we examined normal and cocaine-induced locomotor activity in flies that lack both neural tyramine and octopamine because of mutation in Tdc2 (Tdc2(RO54)). Tdc2(RO54) flies have dramatically reduced basal locomotor activity levels and are hypersensitive to an initial dose of cocaine. Tdc2-targeted expression of the constitutively active inward rectifying potassium channel Kir2.1 replicates these phenotypes, and Tdc2-driven expression of Tdc1 rescues the phenotypes. However, flies that contain no measurable neural octopamine and an excess of tyramine due to a null mutation in the tyramine beta-hydroxylase gene (TbetaH(nM18)) exhibit normal locomotor activity and cocaine responses in spite of showing female sterility due to loss of octopamine. The ability of elevated levels of neural tyramine in TbetaH(nM18) flies to supplant the role of octopamine in adult locomotor and cocaine-induced behaviors, but not in functions related to female fertility, indicates mechanistic differences in the roles of trace amines in these processes.
Collapse
Affiliation(s)
- Shannon L Hardie
- Department of Biology and Neuroscience Graduate Program, University of Virginia, Charlottesville, Virginia 22903, USA
| | | | | |
Collapse
|
42
|
Yang GB, Qiu CL, Aye P, Shao Y, Lackner AA. Expression of serotonin transporters by peripheral blood mononuclear cells of rhesus monkeys (Macaca mulatta). Cell Immunol 2007; 248:69-76. [PMID: 17980355 DOI: 10.1016/j.cellimm.2007.09.001] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/22/2007] [Revised: 09/16/2007] [Accepted: 09/18/2007] [Indexed: 12/16/2022]
Abstract
It has been well established that serotonin (5-hydroxytryptamine, 5-HT) plays a key role in neuro-endocrine-immune networks, mostly through its receptors and/or transporters. Although the presence of 5-HT receptor mRNAs in peripheral blood mononuclear cells (PBMCs) of rhesus monkeys has been reported, there is little information about serotonin transporter (SERT) expression by these cells. To examine SERT expression at the transcription and translation level, one-step RT-PCR, confocal microscopy and flow cytometry were used to detect SERT mRNA and protein expression by rhesus monkey PBMCs. It was found that SERT mRNA could be detected by RT-PCR from all of the rhesus macaque PBMC RNA samples and the nucleotide sequence of the amplicons was identical to the published SERT mRNA sequence. Low level SERT immunoreactivity was also demonstrated on the surface of rhesus PBMCs by confocal microscopy. Almost all lymphocytes and most monocytes were positive for SERT by flow cytometry. In the 2 rhesus macaques examined by multicolor flow cytometry, SERT(bright) cells were more than 84%, 94%, and 96% among CD20+, CD3+, and CD3+CD4+ lymphocytes respectively. These data demonstrate expression of SERT by rhesus macaque PBMCs, and indicate that rhesus macaques would be suitable models to test the in vivo immune regulatory effects of 5-HT or drugs targeting SERT.
Collapse
Affiliation(s)
- G B Yang
- State Key Laboratory for Infectious Disease Prevention and Control, National Center for AIDS/STD Control and Prevention, China-CDC, 27 Nanwei Road, Xuanwu District, Beijing 100050, PR China.
| | | | | | | | | |
Collapse
|
43
|
Romero-Calderón R, Shome RM, Simon AF, Daniels RW, DiAntonio A, Krantz DE. A screen for neurotransmitter transporters expressed in the visual system of Drosophila melanogaster identifies three novel genes. Dev Neurobiol 2007; 67:550-69. [PMID: 17443808 DOI: 10.1002/dneu.20342] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/07/2022]
Abstract
The fly eye provides an attractive substrate for genetic studies, and critical transport activities for synaptic transmission and pigment biogenesis in the insect visual system remain unknown. We therefore screened for transporters in Drosophila melanogaster that are down-regulated by genetically ablating the eye. Using a large panel of transporter specific probes on Northern blots, we identified three transcripts that are down-regulated in flies lacking eye tissue. Two of these, CG13794 and CG13795, are part of a previously unknown subfamily of putative solute carriers within the neurotransmitter transporter family. The third, CG4476, is a member of a related subfamily that includes characterized nutrient transporters expressed in the insect gut. Using imprecise excision of a nearby transposable P element, we have generated a series of deletions in the CG4476 gene. In fast phototaxis assays, CG4476 mutants show a decreased behavioral response to light, and the most severe mutant behaves as if it were blind. These data suggest an unforeseen role for the "nutrient amino acid transporter" subfamily in the nervous system, and suggest new models to study transport function using the fly eye.
Collapse
Affiliation(s)
- Rafael Romero-Calderón
- Interdepartmental Ph.D. Program in Neuroscience, University of California, Los Angeles, California 90095-1761, USA
| | | | | | | | | | | |
Collapse
|
44
|
Vieira R, Mancebo MJ, Aldegunde M. Effects of reserpine and p-chloroamphetamine on 5-HT metabolism and release in the cerebral ganglia of Inachis io (Lepidoptera). J Physiol Biochem 2007; 63:129-41. [DOI: 10.1007/bf03168224] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/20/2022]
|
45
|
Li C, Zhong H, Wang Y, Wang H, Yang Z, Zheng Y, Liu K, Liu Y. Voltage and ionic regulation of human serotonin transporter in Xenopus oocytes. Clin Exp Pharmacol Physiol 2007; 33:1088-92. [PMID: 17042919 DOI: 10.1111/j.1440-1681.2006.04491.x] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/30/2022]
Abstract
1. The serotoninergic system is known to be involved in the control of multiple behavioural and physiological functions. The serotonin (5-hydroxtryptamine; 5-HT) transporter (SERT), which controls the synaptic 5-HT concentration through re-uptake of this neurotransmitter into presynaptic terminals, has been a primary therapeutic target for various psychiatric and peripheral disorders. The aim of the present study was to identify the regulatory mechanism(s) of the human SERT (hSERT) in heterologously expressed oocytes. 2. The hSERT cRNA was transcribed in vitro and injected into Xenopus oocytes. The 5-HT-induced transporter currents were measured by voltage clamp. The effects of extracellular sodium or chloride were studied by replacement perfusion with tetramethylammonium-chloride (96 mmol/L) or sodium acetate (96 mmol/L). In addition, to alter the internal calcium concentration, CaCl2 (50 micromol/L) and inositol triphosphate (IP3; 50 micromol/L), with or without EGTA (2.5 mmol/L), were injected into oocytes. The specificity of 5-HT-sensitive currents was determined by the use of the SERT antagonist desipramine and niflumic acid to block background chloride currents. 3. The hSERT-expressing oocytes displayed voltage-dependent, 5-HT-induced currents that increased at negative potentials. Replacing extracellular sodium or chloride significantly decreased the hSERT currents by 89 and 45%, respectively (P < 0.05, n = 7 each). Injection of IP3 or CaCl2 increased the hSERT currents by approximately 65% (P < 0.05; n = 10 each) and the effect of IP3 was abolished by preinjection of EGTA. 4. These results demonstrate that hSERT activity is not only voltage dependent, but is also affected by intracellular calcium and extracellular sodium and chloride.
Collapse
Affiliation(s)
- Cizhen Li
- Department of Physiology, Shanghai Jiao Tong University Medical School, Shanghai, China
| | | | | | | | | | | | | | | |
Collapse
|
46
|
Thimgan MS, Berg JS, Stuart AE. Comparative sequence analysis and tissue localization of members of the SLC6 family of transporters in adult Drosophila melanogaster. ACTA ACUST UNITED AC 2006; 209:3383-404. [PMID: 16916974 DOI: 10.1242/jeb.02328] [Citation(s) in RCA: 49] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/09/2023]
Abstract
The SLC6 family comprises proteins that move extracellular neurotransmitters, amino acids and osmolytes across the plasma membrane into the cytosol. In mammals, deletion of SLC6 family members has dramatic physiologic consequences, but in the model organism Drosophila melanogaster, little is known about this family of proteins. Therefore, in this study we carried out an initial analysis of 21 known or putative SLC6 family members from the Drosophila genome. Protein sequences from these genes segregated into either well-defined subfamilies, including the novel insect amino acid transporter subfamily, or into a group of weakly related sequences not affiliated with a recognized subfamily. Reverse transcription-polymerase chain reaction analysis and in situ hybridization showed that seven of these genes are expressed in the CNS. In situ hybridization revealed that two previously cloned SLC6 members, the serotonin and dopamine transporters, were localized to presumptive presynaptic neurons that previously immunolabelled for these transmitters. RNA for CG1732 (the putative GABA transporter) and CG15088 (a member of the novel insect amino acid transporter family) was localized in cells likely to be subtypes of glia, while RNA for CG5226, CG10804 (both members of the orphan neurotransmitter transporter subfamily) and CG5549 (a putative glycine transporter) were expressed broadly throughout the cellular cortex of the CNS. Eight of the 21 sequences were localized outside the CNS in the alimentary canal, Malpighian tubules and reproductive organs. Localization for six sequences was not found or not attempted in the adult fly. We used the Drosophila ortholog of the mammalian vesicular monoamine transporter 2, CG33528, to independently identify monoaminergic neurons in the adult fly. RNA for CG33528 was detected in a limited number of cells in the central brain and in a beaded stripe at the base of the photoreceptors in the position of glia, but not in the photoreceptors themselves. The SLC6 localization observations in conjunction with likely substrates based on phylogenetic inferences are a first step in defining the role of Na/Cl-dependent transporters in Drosophila physiology.
Collapse
Affiliation(s)
- Matthew S Thimgan
- Department of Cell and Molecular Physiology, University of North Carolina School of Medicine, Chapel Hill, 27599, USA.
| | | | | |
Collapse
|
47
|
Park SK, George R, Cai Y, Chang HY, Krantz DE, Friggi-Grelin F, Birman S, Hirsh J. Cell-type-specific limitation on in vivo serotonin storage following ectopic expression of the Drosophila serotonin transporter, dSERT. ACTA ACUST UNITED AC 2006; 66:452-62. [PMID: 16470720 DOI: 10.1002/neu.20222] [Citation(s) in RCA: 11] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/10/2022]
Abstract
The synaptic machinery for neurotransmitter storage is cell-type specific. Although most elements of biosynthesis and transport have been identified, it remains unclear whether additional factors may be required to maintain this specificity. The Drosophila serotonin transporter (dSERT) is normally expressed exclusively in serotonin (5-HT) neurons in the CNS. Here we examine the effects of ectopic transcriptional expression of dSERT in the Drosophila larval CNS. We find a surprising limitation on 5-HT storage following ectopic expression of dSERT and green fluorescence protein-tagged dSERT (GFP-dSERT). When dSERT transcription is driven ectopically in the CNS, 5-HT is detectable only in 5-HT, dopamine (DA), and a very limited number of additional neurons. Addition of exogenous 5-HT does not dramatically broaden neuronal storage sites, so this limitation is only partly due to restricted intercellular diffusion of 5-HT. Furthermore, this limitation is not due to gross mislocalization of dSERT, because cells lacking or containing 5-HT show similar levels and subcellular distribution of GFP-dSERT protein, nor is it due to lack of the vesicular transporter, dVMAT. These data suggest that a small number of neurons selectively express factor(s) required for 5-HT storage, and potentially for function of dSERT.
Collapse
Affiliation(s)
- Sang Ki Park
- Department of Biology, University of Virginia, Charlottesville, Virginia 22904, USA
| | | | | | | | | | | | | | | |
Collapse
|
48
|
Quick M, Yano H, Goldberg NR, Duan L, Beuming T, Shi L, Weinstein H, Javitch JA. State-dependent conformations of the translocation pathway in the tyrosine transporter Tyt1, a novel neurotransmitter:sodium symporter from Fusobacterium nucleatum. J Biol Chem 2006; 281:26444-54. [PMID: 16798738 DOI: 10.1074/jbc.m602438200] [Citation(s) in RCA: 78] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/06/2022] Open
Abstract
The gene of a novel prokaryotic member (Tyt1) of the neurotransmitter:sodium symporter (NSS) family has been cloned from Fusobacterium nucleatum. In contrast to eukaryotic and some prokaryotic NSSs, which contain 12 transmembrane domains (TMs), Tyt1 contains only 11 TMs, a characteristic shared by approximately 70% of prokaryotic NSS homologues. Nonetheless upon heterologous expression in an engineered Escherichia coli host, Tyt1 catalyzes robust Na+-dependent, highly selective l-tyrosine transport. Genetic engineering of Tyt1 variants devoid of cysteines or with individually retained endogenous cysteines at positions 18 or 238, at the cytoplasmic ends of TM1 and TM6, respectively, preserved normal transport activity. Whereas cysteine-less Tyt1 was resistant to the inhibitory effect of sulfhydryl-alkylating reagents, N-ethylmaleimide inhibited transport by Tyt1 variants containing either one or both of the endogenous cysteines, and this inhibition was altered by the substrates sodium and tyrosine, consistent with substrate-induced dynamics in the transport pathway. Our findings support a binding model of Tyt1 function in which an ordered sequence of substrate-induced structural changes reflects distinct conformational states of the transporter. This work identifies Tyt1 as the first functional bacterial NSS member putatively consisting of only 11 TMs and shows that Tyt1 is a suitable model for the study of NSS dynamics with relevance to structure/function relationships of human NSSs, including the dopamine, norepinephrine, serotonin, and gamma-aminobutyric acid transporters.
Collapse
Affiliation(s)
- Matthias Quick
- Center for Molecular Recognition, Columbia University College of Physicians and Surgeons, New York, New York 10032, USA
| | | | | | | | | | | | | | | |
Collapse
|
49
|
Schwartz JW, Piston D, DeFelice LJ. Molecular microfluorometry: converting arbitrary fluorescence units into absolute molecular concentrations to study binding kinetics and stoichiometry in transporters. Handb Exp Pharmacol 2006:23-57. [PMID: 16722229 DOI: 10.1007/3-540-29784-7_2] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 05/09/2023]
Abstract
Cotransporters use energy stored in Na+ or H+ gradients to transport neurotransmitters or other substrates against their own gradient. Cotransport is rapid and efficient, and at synapses it helps terminate signaling. Cotransport in norepinephrine (NET), epinephrine (EpiT), dopamine (DAT), and serotonin (SERT) transporters couples downhill Na+ flux to uphill transmitter flux. NETs, for example, attenuate signaling at adrenergic synapses by efficiently clearing NE from the synaptic cleft, thus preparing the synapse for the next signal. Transport inhibition with tricyclic antidepressants prolongs neurotransmitter presence in the synaptic cleft, potentially alleviating symptoms of depression. Transport inhibition with cocaine or amphetamine, which respectively block or replace normal transport, may result in hyperactivity. Little is known about the kinetic interactions of substrates or drugs with transporters, largely because the techniques that have been successful in discovering trans- porter agonists and antagonists do not yield detailed kinetic information. Mechanistic data are for the most part restricted to global parameters, such as Km and Vmax, measured from large populations of transporter molecules averaged over thousands of cells. Three relatively new techniques used in transporter research are electrophysiology, amperometry, and microfluorometry. This review focuses on fluorescence-based methodologies, which--unlike any other technique-permit the simultaneous measurement of binding and transport. Microfluorometry provides unique insights into binding kinetics and transport mechanisms from a quantitative analysis of fluorescence data. Here we demonstrate how to quantify the number of bound substrate molecules, the number of transported substrate molecules, and the kinetics of substrate binding to individual transporters. Although we describe experiments on a specific neurotransmitter transporter, these methods are applicable to other membrane proteins.
Collapse
Affiliation(s)
- J W Schwartz
- Imaging Center, Stowers Institute for Medical Research, 1000 E 50th St., Kansas City, MO 64110, USA
| | | | | |
Collapse
|
50
|
Chang HY, Grygoruk A, Brooks ES, Ackerson LC, Maidment NT, Bainton RJ, Krantz DE. Overexpression of the Drosophila vesicular monoamine transporter increases motor activity and courtship but decreases the behavioral response to cocaine. Mol Psychiatry 2006; 11:99-113. [PMID: 16189511 DOI: 10.1038/sj.mp.4001742] [Citation(s) in RCA: 92] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/08/2022]
Abstract
Aminergic signaling pathways have been implicated in a variety of neuropsychiatric illnesses, but the mechanisms by which these pathways influence complex behavior remain obscure. Vesicular monoamine transporters (VMATs) have been shown to regulate the amount of monoamine neurotransmitter that is stored and released from synaptic vesicles in mammalian systems, and an increase in their expression has been observed in bipolar patients. The model organism Drosophila melanogaster provides a powerful, but underutilized genetic system for studying how dopamine (DA) and serotonin (5HT) may influence behavior. We show that a Drosophila isoform of VMAT (DVMAT-A) is expressed in both dopaminergic and serotonergic neurons in the adult Drosophila brain. Overexpression of DVMAT-A in these cells potentiates stereotypic grooming behaviors and locomotion and can be reversed by reserpine, which blocks DVMAT activity, and haloperidol, a DA receptor antagonist. We also observe a prolongation of courtship behavior, a decrease in successful mating and a decrease in fertility, suggesting a role for aminergic circuits in the modulation of sexual behaviors. Finally, we find that DMVAT-A overexpression decreases the fly's sensitivity to cocaine, suggesting that the synaptic machinery responsible for this behavior may be downregulated. DVMAT transgenes may be targeted to additional neuronal pathways using standard Drosophila techniques, and our results provide a novel paradigm to study the mechanisms by which monoamines regulate complex behaviors relevant to neuropsychiatric illness.
Collapse
Affiliation(s)
- H-Y Chang
- Department of Psychiatry and Biobehavioral Sciences, Gonda (Goldschmied) Center for Genetic and Neuroscience Research, Geffen School of Medicine-UCLA, University of California at Los Angeles, 695 Charles Young Drive, Los Angeles, CA 90095-1761, USA
| | | | | | | | | | | | | |
Collapse
|