1
|
Szebik H, Miskolczi C, Bruzsik B, Balla G, Szabó S, Biró L, Mikics É. Dynamic changes of serotonin transporter expression in the prefrontal cortex evoked by aggressive social interactions. Neurobiol Stress 2025; 36:100722. [PMID: 40230625 PMCID: PMC11994973 DOI: 10.1016/j.ynstr.2025.100722] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/20/2024] [Revised: 03/17/2025] [Accepted: 03/25/2025] [Indexed: 04/16/2025] Open
Abstract
Aggression is a complex behavior influenced by developmental experiences, internal state, and social context, yet its neurobiological underpinnings remain insufficiently understood. The serotonergic system, particularly the serotonin transporter (SERT), plays a crucial role in aggression regulation. Here, we investigated region-specific, dynamic changes in SERT expression following aggressive interactions and in mice subjected to early-life social adversity. We found that aggressive encounters (resident-intruder test) triggered a significant, rapid increase in SERT immunoreactivity within 90 min, accompanied by neuronal activation in aggression-related brain regions, including the medial prefrontal cortex (mPFC), lateral septum (LS), medial amygdala (MeA), ventromedial hypothalamus (VMHvl), lateral habenula (LH), and dorsal raphe (DR), but not in the paraventricular thalamus (PVT). Notably, this SERT upregulation occurred across the aggression circuitry but was accompanied by a significant increase in 5-HT levels only in the mPFC, a key region in top-down regulation of social and aggressive behavior. This SERT upregulation was not observed following exposure to a non-social challenge, suggesting that it may be more specifically associated with social contexts. Using super-resolution microscopy, we identified an increased density of SERT localization points within serotonergic mPFC axons after an aggressive encounter. Social isolation during adolescence, a model of early social neglect, impaired this rapid SERT response, particularly in the ventral and medial orbitofrontal regions, and altered the relationship between SERT levels and aggression-related behaviors. These findings demonstrate that SERT expression undergoes rapid, experience-dependent plasticity in response to social aggression, and that early-life adversity disrupts this adaptive mechanism, providing new insights into the serotonergic regulation of aggression and its potential relevance for stress-related social dysfunctions.
Collapse
Affiliation(s)
- Huba Szebik
- Translational Behavioral Neuroscience Research Group, HUN-REN Institute of Experimental Medicine, Budapest, Hungary
- Semmelweis University, Doctoral School, Budapest, Hungary
| | - Christina Miskolczi
- Translational Behavioral Neuroscience Research Group, HUN-REN Institute of Experimental Medicine, Budapest, Hungary
- Semmelweis University, Doctoral School, Budapest, Hungary
| | - Bíborka Bruzsik
- Translational Behavioral Neuroscience Research Group, HUN-REN Institute of Experimental Medicine, Budapest, Hungary
| | - Gyula Balla
- Translational Behavioral Neuroscience Research Group, HUN-REN Institute of Experimental Medicine, Budapest, Hungary
| | - Soma Szabó
- Translational Behavioral Neuroscience Research Group, HUN-REN Institute of Experimental Medicine, Budapest, Hungary
| | - László Biró
- Translational Behavioral Neuroscience Research Group, HUN-REN Institute of Experimental Medicine, Budapest, Hungary
- Thalamus Research Group, HUN-REN Institute of Experimental Medicine, Budapest, Hungary
| | - Éva Mikics
- Translational Behavioral Neuroscience Research Group, HUN-REN Institute of Experimental Medicine, Budapest, Hungary
| |
Collapse
|
2
|
Fagundes KRC, Kasica N, Potoczna M, Okitsu-Sakurayama S, Podlasz P, de Britto Mari R. Disruptive ecotoxicological effects of fluoxetine on serotoninergic signaling and enteric neurogenesis in early zebrafish larvae (Danio rerio). ENVIRONMENTAL TOXICOLOGY AND PHARMACOLOGY 2025; 116:104698. [PMID: 40216344 DOI: 10.1016/j.etap.2025.104698] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/27/2024] [Revised: 04/04/2025] [Accepted: 04/09/2025] [Indexed: 04/18/2025]
Abstract
This study investigated the multilevel effects of environmentally relevant concentrations of fluoxetine on serotonergic signaling and enteric neurogenesis in early zebrafish larvae (Danio rerio). To this end, zebrafish were exposed to various concentrations of fluoxetine for four days, from the 1,000-cell stage to 4 days post-fertilization (dpf).Following exposure, whole larvae were subjected to molecular, morphological, and behavioral analyses. All tested concentrations led to upregulation of the serotonin transporter (slc6a4a). At intermediate concentrations, overexpression of the serotonin receptor htr1aa was observed. The highest concentration caused a reduced total enteric neurons density, while the intermediate concentration reduced the density of serotonergic enteric neurons. Additionally, the highest concentration decreased larval locomotion and impaired their ability to differentiate between light and dark phases.Across all tested concentrations, fluoxetine disrupted serotonergic signaling, impaired enteric neurogenesis, and induced sedative-like behavioral effects.
Collapse
Affiliation(s)
- Kainã Rocha Cabrera Fagundes
- Laboratory of Animal Morphophysiology, Biosciences Institute, Sao Paulo State University, Praça Infante Dom Henrique, s/n, São Vicente, SP 11330-900, Brazil.
| | - Natalia Kasica
- Department of Animal Anatomy, Faculty of Veterinary Medicine, University of Warmia and Mazury, Olsztyn, Poland
| | - Małgorzata Potoczna
- Department of Pathophysiology, Forensic Veterinary Medicine and Administration, Faculty of Veterinary Medicine, University of Warmia and Mazury, Olsztyn, Poland; Transphamation Poland Ltd., Olsztyn, Poland
| | - Shiho Okitsu-Sakurayama
- Department of Pathophysiology, Forensic Veterinary Medicine and Administration, Faculty of Veterinary Medicine, University of Warmia and Mazury, Olsztyn, Poland
| | - Piotr Podlasz
- Department of Pathophysiology, Forensic Veterinary Medicine and Administration, Faculty of Veterinary Medicine, University of Warmia and Mazury, Olsztyn, Poland
| | - Renata de Britto Mari
- Laboratory of Animal Morphophysiology, Biosciences Institute, Sao Paulo State University, Praça Infante Dom Henrique, s/n, São Vicente, SP 11330-900, Brazil
| |
Collapse
|
3
|
Alexander C, Jeon J, Nickerson K, Hassler S, Vasefi M. CBD and the 5-HT1A receptor: A medicinal and pharmacological review. Biochem Pharmacol 2025; 233:116742. [PMID: 39778776 DOI: 10.1016/j.bcp.2025.116742] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/30/2024] [Revised: 11/25/2024] [Accepted: 01/03/2025] [Indexed: 01/11/2025]
Abstract
Cannabidiol (CBD), a phytocannabinoid, has emerged as a promising candidate for addressing a wide array of symptoms. It has the ability to bind to multiple proteins and receptors, including 5-HT1AR, transient receptor potential vanilloid 1 (TRPV1), and cannabinoid receptors. However, CBD's pharmacodynamic interaction with 5-HT1AR and its medicinal outcomes are still debated. This review explores recent literature to elucidate these questions, highlighting the neurotherapeutic outcomes of this pharmacodynamic interaction and proposing a signaling pathway underlying the mechanism by which CBD desensitizes 5-HT1AR signaling. A comprehensive survey of the literature underscores CBD's multifaceted neurotherapeutic effects, which include antidepressant, anxiolytic, neuroprotective, antipsychotic, antiemetic, anti-allodynic, anti-epileptic, anti-degenerative, and addiction-treating properties, attributable in part to its interactions with 5-HT1AR. Furthermore, evidence suggests that the pharmacodynamic interaction between CBD and 5-HT1AR is contingent upon dosage. Moreover, we propose that CBD can induce desensitization of 5-HT1AR via both homologous and heterologous mechanisms. Homologous desensitization involves the recruitment of G protein-coupled receptor kinase 2 (GRK2) and β-arrestin, leading to receptor endocytosis. In contrast, heterologous desensitization is mediated by an elevated intracellular calcium level or activation of protein kinases, such as c-Jun N-terminal kinase (JNK), through the activity of other receptors.
Collapse
Affiliation(s)
- Claire Alexander
- Tulane Brain Institute, Tulane University, New Orleans, LA, 70118, USA
| | - Jiyoon Jeon
- Department of Biology, Lamar University, Beaumont, TX, 77710, USA
| | - Kyle Nickerson
- Department of Biology, Baylor University, Waco, TX, 76706, USA
| | - Shayne Hassler
- Department of Biomedical Sciences, Tilman J. Fertitta Family College of Medicine, University of Houton, Houston, TX, 77204, USA
| | - Maryam Vasefi
- Department of Biomedical Sciences, Tilman J. Fertitta Family College of Medicine, University of Houton, Houston, TX, 77204, USA.
| |
Collapse
|
4
|
Li X, Feng D, Ma S, Li M, Zhao S, Tang M. Ventral hippocampus is more sensitive to fluoxetine-induced changes in extracellular 5-HT concentration, membrane 5-HT transporter level and immobility times. Neuropharmacology 2024; 242:109766. [PMID: 37858884 DOI: 10.1016/j.neuropharm.2023.109766] [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: 07/06/2023] [Revised: 09/26/2023] [Accepted: 10/15/2023] [Indexed: 10/21/2023]
Abstract
Hippocampal responses to selective 5-HT reuptake inhibitor (SSRI) have long been studied. However, its sub-regional involvements in mediating SSRI's pharmacological effects have not been fully addressed. The current study sought to investigate neurochemical, neurobiological and neurobehavioral changes in response to direct fluoxetine perfusion into the ventral and dorsal sub-regions of the hippocampus in C57BL/6 mice. Following fluoxetine perfusion, time courses of dialysate 5-HT, 5-HT transporter (5-HTT) protein (total, membrane and cytoplasmic fractions), locomotion, and immobility times in the forced swim test (FST) and tail suspension test (TST) were determined. At baseline, 5-HT uptake efficiency assessed by the no-net-flux microdialysis, and 5-HTT protein were measured as well. Results show that fluoxetine dose-dependently increased dialysate 5-HT, lowered membrane 5-HTT protein and increased cytoplasmic fraction without changing the total level, decreased immobility times in both the FST and TST, with greater responses all detected in the ventral sub-region compared to the dorsal sub-region. Fluoxetine didn't affect locomotor activity, ruling out the possibility that fluoxetine's effects on immobility maybe due to alteration in locomotion. Besides, lower 5-HT uptake efficiency and lower membrane 5-HTT protein level were found in the ventral sub-region at baseline. Together, the sub-regional differences at baseline and in responses to fluoxetine added powerful evidence to support the existence of two distinct 5-HT sub-systems in the hippocampus, with greater changes to fluoxetine detected in the ventral sub-system.
Collapse
Affiliation(s)
- Xiang Li
- Department of Pharmacy, The Fourth Affiliated Hospital of China Medical University, Shenyang, 110032, China
| | - Dan Feng
- Department of Clinical Pharmacology, College of Pharmacy, China Medical University, Shenyang, 110122, China
| | - Shenglu Ma
- Department of Clinical Pharmacology, College of Pharmacy, China Medical University, Shenyang, 110122, China
| | - Mingxing Li
- Department of Clinical Pharmacology, College of Pharmacy, China Medical University, Shenyang, 110122, China
| | - Shulei Zhao
- Center for Devices and Radiological Health, U.S. Food and Drug Administration, Silver Spring, MD, 20993, USA
| | - Man Tang
- Department of Clinical Pharmacology, College of Pharmacy, China Medical University, Shenyang, 110122, China.
| |
Collapse
|
5
|
Holmes J, Lau T, Saylor R, Fernández-Novel N, Hersey M, Keen D, Hampel L, Horschitz S, Ladewig J, Parke B, Reed MC, Nijhout HF, Best J, Koch P, Hashemi P. Voltammetric Approach for Characterizing the Biophysical and Chemical Functionality of Human Induced Pluripotent Stem Cell-Derived Serotonin Neurons. Anal Chem 2022; 94:8847-8856. [PMID: 35713335 DOI: 10.1021/acs.analchem.1c05082] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/30/2022]
Abstract
Depression is quickly becoming one of the world's most pressing public health crises, and there is an urgent need for better diagnostics and therapeutics. Behavioral models in animals and humans have not adequately addressed the diagnosis and treatment of depression, and biomarkers of mental illnesses remain ill-defined. It has been very difficult to identify biomarkers of depression because of in vivo measurement challenges. While our group has made important strides in developing in vivo tools to measure such biomarkers (e.g., serotonin) in mice using voltammetry, these tools cannot be easily applied for depression diagnosis and drug screening in humans due to the inaccessibility of the human brain. In this work, we take a chemical approach, ex vivo, to introduce a human-derived system to investigate brain serotonin. We utilize human induced pluripotent stem cells differentiated into serotonin neurons and establish a new ex vivo model of real-time serotonin neurotransmission measurements. We show that evoked serotonin release responds to stimulation intensity and tryptophan preloading, and that serotonin release and reuptake kinetics resemble those found in vivo in rodents. Finally, after selective serotonin reuptake inhibitor (SSRI) exposure, we find dose-dependent internalization of the serotonin reuptake transporters (a signature of the in vivo response to SSRI). Our new human-derived chemical model has great potential to provide an ex vivo chemical platform as a translational tool for in vivo neuropsychopharmacology.
Collapse
Affiliation(s)
- Jordan Holmes
- Department of Chemistry and Biochemistry, University of South Carolina, Columbia, South Carolina 29208, United States
| | - Thorsten Lau
- Department of Translational Brain Research, Central Institute of Mental Health, University of Heidelberg, Medical Faculty Mannheim, 68159 Mannheim, Germany.,German Cancer Research Center, 69120 Heidelberg, Germany.,HITBR Hector Institute for Translational Brain Research gGmbH, 68159 Mannheim, Germany
| | - Rachel Saylor
- Department of Chemistry and Biochemistry, University of South Carolina, Columbia, South Carolina 29208, United States
| | - Nadine Fernández-Novel
- Department of Translational Brain Research, Central Institute of Mental Health, University of Heidelberg, Medical Faculty Mannheim, 68159 Mannheim, Germany.,German Cancer Research Center, 69120 Heidelberg, Germany.,HITBR Hector Institute for Translational Brain Research gGmbH, 68159 Mannheim, Germany
| | - Melinda Hersey
- Department of Chemistry and Biochemistry, University of South Carolina, Columbia, South Carolina 29208, United States.,Department of Pharmacology, Physiology, & Neuroscience, University of South Carolina, Columbia, South Carolina 29209, United States
| | - Deanna Keen
- Department of Chemistry and Biochemistry, University of South Carolina, Columbia, South Carolina 29208, United States
| | - Lena Hampel
- Department of Translational Brain Research, Central Institute of Mental Health, University of Heidelberg, Medical Faculty Mannheim, 68159 Mannheim, Germany.,German Cancer Research Center, 69120 Heidelberg, Germany.,HITBR Hector Institute for Translational Brain Research gGmbH, 68159 Mannheim, Germany
| | - Sandra Horschitz
- Department of Translational Brain Research, Central Institute of Mental Health, University of Heidelberg, Medical Faculty Mannheim, 68159 Mannheim, Germany.,German Cancer Research Center, 69120 Heidelberg, Germany.,HITBR Hector Institute for Translational Brain Research gGmbH, 68159 Mannheim, Germany
| | - Julia Ladewig
- Department of Translational Brain Research, Central Institute of Mental Health, University of Heidelberg, Medical Faculty Mannheim, 68159 Mannheim, Germany.,German Cancer Research Center, 69120 Heidelberg, Germany.,HITBR Hector Institute for Translational Brain Research gGmbH, 68159 Mannheim, Germany
| | - Brenna Parke
- Department of Bioengineering, Imperial College London, London SW7 2AZ, U.K
| | - Michael C Reed
- Department of Mathematics, Duke University, Durham, North Carolina 27708, United States
| | - H Frederik Nijhout
- Department of Biology, Duke University, Durham, North Carolina 27708, United States
| | - Janet Best
- Department of Mathematics, The Ohio State University, Columbus, Ohio 43210, United States
| | - Philipp Koch
- Department of Translational Brain Research, Central Institute of Mental Health, University of Heidelberg, Medical Faculty Mannheim, 68159 Mannheim, Germany.,German Cancer Research Center, 69120 Heidelberg, Germany.,HITBR Hector Institute for Translational Brain Research gGmbH, 68159 Mannheim, Germany
| | - Parastoo Hashemi
- Department of Chemistry and Biochemistry, University of South Carolina, Columbia, South Carolina 29208, United States.,Department of Bioengineering, Imperial College London, London SW7 2AZ, U.K
| |
Collapse
|
6
|
Armand S, Ozenne B, Svart N, Frøkjaer VG, Knudsen GM, Fisher PM, Stenbaek DS. Brain serotonin transporter is associated with cognitive-affective biases in healthy individuals. Hum Brain Mapp 2022; 43:4174-4184. [PMID: 35607850 PMCID: PMC9374883 DOI: 10.1002/hbm.25946] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/03/2022] [Revised: 04/21/2022] [Accepted: 05/02/2022] [Indexed: 12/26/2022] Open
Abstract
Cognitive affective biases describe the tendency to process negative information or positive information over the other. These biases can be modulated by changing extracellular serotonin (5-HT) levels in the brain, for example, by pharmacologically blocking and downregulating the 5-HT transporter (5-HTT), which remediates negative affective bias. This suggests that higher levels of 5-HTT are linked to a priority of negative information over positive, but this link remains to be tested in vivo in healthy individuals. We, therefore, evaluated the association between 5-HTT levels, as measured with [11 C]DASB positron emission tomography (PET), and affective biases, hypothesising that higher 5-HTT levels are associated with a more negative bias. We included 98 healthy individuals with measures of [11 C]DASB binding potential (BPND ) and affective biases using The Emotional Faces Identification Task by subtracting the per cent hit rate for happy from that of sad faces (EFITAB ). We evaluated the association between [11 C]DASB BPND and EFITAB in a linear latent variable model, with the latent variable (5-HTTLV ) modelled from [11 C]DASB BPND in the fronto-striatal and fronto-limbic networks implicated in affective cognition. We observed an inverse association between 5-HTTLV and EFITAB (β = -8% EFITAB per unit 5-HTTLV , CI = -14% to -3%, p = .002). These findings show that higher 5-HTT levels are linked to a more negative bias in healthy individuals. High 5-HTT supposedly leads to high clearance of 5-HT, and thus, a negative bias could result from low extracellular 5-HT. Future studies must reveal if a similar inverse association exists in individuals with affective disorders.
Collapse
Affiliation(s)
- Sophia Armand
- Neurobiology Research Unit, Copenhagen University Hospital Rigshospitalet, Copenhagen, Denmark.,Department of Psychology, University of Copenhagen, Copenhagen, Denmark
| | - Brice Ozenne
- Neurobiology Research Unit, Copenhagen University Hospital Rigshospitalet, Copenhagen, Denmark.,Department of Public Health, Section of Biostatistics, University of Copenhagen, Copenhagen, Denmark.,BrainDrugs, Neurobiology Research Unit, Copenhagen University Hospital Rigshospitalet, Copenhagen, Denmark
| | - Nanna Svart
- Neurobiology Research Unit, Copenhagen University Hospital Rigshospitalet, Copenhagen, Denmark.,Department of Psychology, University of Copenhagen, Copenhagen, Denmark
| | - Vibe G Frøkjaer
- Neurobiology Research Unit, Copenhagen University Hospital Rigshospitalet, Copenhagen, Denmark.,BrainDrugs, Neurobiology Research Unit, Copenhagen University Hospital Rigshospitalet, Copenhagen, Denmark.,Psychiatric Center Copenhagen, Copenhagen University Hospital Rigshospitalet, Copenhagen, Denmark
| | - Gitte M Knudsen
- Neurobiology Research Unit, Copenhagen University Hospital Rigshospitalet, Copenhagen, Denmark.,BrainDrugs, Neurobiology Research Unit, Copenhagen University Hospital Rigshospitalet, Copenhagen, Denmark.,Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark
| | - Patrick M Fisher
- Neurobiology Research Unit, Copenhagen University Hospital Rigshospitalet, Copenhagen, Denmark.,BrainDrugs, Neurobiology Research Unit, Copenhagen University Hospital Rigshospitalet, Copenhagen, Denmark
| | - Dea S Stenbaek
- Neurobiology Research Unit, Copenhagen University Hospital Rigshospitalet, Copenhagen, Denmark.,Department of Psychology, University of Copenhagen, Copenhagen, Denmark.,BrainDrugs, Neurobiology Research Unit, Copenhagen University Hospital Rigshospitalet, Copenhagen, Denmark
| |
Collapse
|
7
|
Meinke C, Quinlan MA, Paffenroth KC, Harrison FE, Fenollar-Ferrer C, Katamish RM, Stillman I, Ramamoorthy S, Blakely RD. Serotonin Transporter Ala276 Mouse: Novel Model to Assess the Neurochemical and Behavioral Impact of Thr276 Phosphorylation In Vivo. Neurochem Res 2022; 47:37-60. [PMID: 33830406 PMCID: PMC11574550 DOI: 10.1007/s11064-021-03299-w] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/20/2020] [Revised: 02/21/2021] [Accepted: 03/17/2021] [Indexed: 11/30/2022]
Abstract
The serotonin (5-HT) transporter (SERT) is a key regulator of 5-HT signaling and is a major target for antidepressants and psychostimulants. Human SERT coding variants have been identified in subjects with obsessive-compulsive disorder (OCD) and autism spectrum disorder (ASD) that impact transporter phosphorylation, cell surface trafficking and/or conformational dynamics. Prior to an initial description of a novel mouse line expressing the non-phosphorylatable SERT substitution Thr276Ala, we review efforts made to elucidate the structure and conformational dynamics of SERT with a focus on research implicating phosphorylation at Thr276 as a determinant of SERT conformational dynamics. Using the high-resolution structure of human SERT in inward- and outward-open conformations, we explore the conformation dependence of SERT Thr276 exposure, with results suggesting that phosphorylation is likely restricted to an inward-open conformation, consistent with prior biochemical studies. Assessment of genotypes from SERT/Ala276 heterozygous matings revealed a deviation from Mendelian expectations, with reduced numbers of Ala276 offspring, though no genotype differences were seen in growth or physical appearance. Similarly, no genotype differences were evident in midbrain or hippocampal 5-HT levels, midbrain and hippocampal SERT mRNA or midbrain protein levels, nor in midbrain synaptosomal 5-HT uptake kinetics. Behaviorally, SERT Ala276 homozygotes appeared normal in measures of anxiety and antidepressant-sensitive stress coping behavior. However, these mice displayed sex-dependent alterations in repetitive and social interactions, consistent with circuit-dependent requirements for Thr276 phosphorylation underlying these behaviors. Our findings indicate the utility of SERT Ala276 mice in evaluation of developmental, functional and behavioral consequences of regulatory SERT phosphorylation in vivo.
Collapse
Affiliation(s)
- Carina Meinke
- International Max Planck Research School for Brain and Behavior, Max Planck Florida Institute for Neuroscience, Jupiter, FL, USA
- Department of Biomedical Science, Charles E. Schmidt College of Medicine, Florida Atlantic University, Jupiter, FL, USA
| | - Meagan A Quinlan
- Department of Biomedical Science, Charles E. Schmidt College of Medicine, Florida Atlantic University, Jupiter, FL, USA
- Department of Psychiatry and Behavioral Sciences, University of Washington, Seattle, WA, USA
| | | | - Fiona E Harrison
- Department of Medicine, Vanderbilt University School of Medicine, Nashville, TN, USA
| | - Cristina Fenollar-Ferrer
- Laboratories of Molecular Genetics and Molecular Biology, National Institute On Deafness and Other Communication Disorders, National Institutes of Health, Bethesda, MD, USA
| | - Rania M Katamish
- Department of Biomedical Science, Charles E. Schmidt College of Medicine, Florida Atlantic University, Jupiter, FL, USA
| | - Isabel Stillman
- Department of Biomedical Science, Charles E. Schmidt College of Medicine, Florida Atlantic University, Jupiter, FL, USA
| | | | - Randy D Blakely
- Department of Biomedical Science, Charles E. Schmidt College of Medicine, Florida Atlantic University, Jupiter, FL, USA.
- Florida Atlantic University Brain Institute, Rm 109, MC-17, 5353 Parkside Dr, Jupiter, FL, 35348, USA.
| |
Collapse
|
8
|
Tomlinson ID, Kovtun O, Torres R, Bellocchio LG, Josephs T, Rosenthal SJ. A Novel Biotinylated Homotryptamine Derivative for Quantum Dot Imaging of Serotonin Transporter in Live Cells. Front Cell Neurosci 2021; 15:667044. [PMID: 34867196 PMCID: PMC8637195 DOI: 10.3389/fncel.2021.667044] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/11/2021] [Accepted: 10/19/2021] [Indexed: 11/21/2022] Open
Abstract
The serotonin transporter (SERT) is the primary target for selective serotonin reuptake inhibitor (SSRI) antidepressants that are thought to exert their therapeutic effects by increasing the synaptic concentration of serotonin. Consequently, probes that can be utilized to study cellular trafficking of SERT are valuable research tools. We have developed a novel ligand (IDT785) that is composed of a SERT antagonist (a tetrahydro pyridyl indole derivative) conjugated to a biotinylated poly ethylene glycol (PEG) via a phenethyl linker. This compound was determined to be biologically active and inhibited SERT-mediated reuptake of IDT307 with the half-maximal inhibitory concentration of 7.2 ± 0.3 μM. We demonstrated that IDT785 enabled quantum dot (QD) labeling of membrane SERT in transfected HEK-293 cultures that could be blocked using the high affinity serotonin reuptake inhibitor paroxetine. Molecular docking studies suggested that IDT785 might be binding to the extracellular vestibule binding site rather than the orthosteric substrate binding site, which could be attributable to the hydrophilicity of the PEG chain and the increased loss of degrees of freedom that would be required to penetrate into the orthosteric binding site. Using IDT785, we were able to study the membrane localization and membrane dynamics of YFP-SERT heterologously expressed in HEK-293 cells and demonstrated that SERT expression was enriched in the membrane edge and in thin cellular protrusions.
Collapse
Affiliation(s)
- Ian D. Tomlinson
- Department of Chemistry, Vanderbilt University, Nashville, TN, United States
| | - Oleg Kovtun
- Department of Chemistry, Vanderbilt University, Nashville, TN, United States
| | - Ruben Torres
- Department of Chemistry, Vanderbilt University, Nashville, TN, United States
- Vanderbilt Institute of Chemical Biology, Vanderbilt University, Nashville, TN, United States
| | | | - Travis Josephs
- Neuroscience Program, Vanderbilt University, Nashville, TN, United States
| | - Sandra J. Rosenthal
- Department of Chemistry, Vanderbilt University, Nashville, TN, United States
- Vanderbilt Institute of Chemical Biology, Vanderbilt University, Nashville, TN, United States
- Department of Pharmacology, Vanderbilt University, Nashville, TN, United States
- Department of Chemical and Biomolecular Engineering, Vanderbilt University, Nashville, TN, United States
- Department of Physics and Astronomy, Vanderbilt University, Nashville, TN, United States
- Vanderbilt Institute of Nanoscale Science and Engineering, Vanderbilt University, Nashville, TN, United States
| |
Collapse
|
9
|
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
|
10
|
Cisneros IE, Cunningham KA. Covid-19 interface with drug misuse and substance use disorders. Neuropharmacology 2021; 198:108766. [PMID: 34454912 PMCID: PMC8388132 DOI: 10.1016/j.neuropharm.2021.108766] [Citation(s) in RCA: 13] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/25/2021] [Revised: 08/09/2021] [Accepted: 08/23/2021] [Indexed: 12/26/2022]
Abstract
The coronavirus disease 2019 (Covid-19) pandemic intensified the already catastrophic drug overdose and substance use disorder (SUD) epidemic, signaling a syndemic as social isolation, economic and mental health distress, and disrupted treatment services disproportionally impacted this vulnerable population. Along with these social and societal factors, biological factors triggered by intense stress intertwined with incumbent overactivity of the immune system and the resulting inflammatory outcomes may impact the functional status of the central nervous system (CNS). We review the literature concerning SARS-CoV2 infiltration and infection in the CNS and the prospects of synergy between stress, inflammation, and kynurenine pathway function during illness and recovery from Covid-19. Taken together, inflammation and neuroimmune signaling, a consequence of Covid-19 infection, may dysregulate critical pathways and underlie maladaptive changes in the CNS, to exacerbate the development of neuropsychiatric symptoms and in the vulnerability to develop SUD. This article is part of the special Issue on 'Vulnerabilities to Substance Abuse'.
Collapse
Affiliation(s)
- I E Cisneros
- Center for Addiction Research, University of Texas Medical Branch, Galveston, TX, USA; Department of Pathology, University of Texas Medical Branch, Galveston, TX, USA; Department of Pharmacology and Toxicology, University of Texas Medical Branch, Galveston, TX, USA; Institute for Human Infections and Immunity, University of Texas Medical Branch, Galveston, TX, USA; Center for Biodefense and Emerging Infectious Diseases, University of Texas Medical Branch, Galveston, TX, USA.
| | - K A Cunningham
- Center for Addiction Research, University of Texas Medical Branch, Galveston, TX, USA; Department of Pharmacology and Toxicology, University of Texas Medical Branch, Galveston, TX, USA; Institute for Human Infections and Immunity, University of Texas Medical Branch, Galveston, TX, USA
| |
Collapse
|
11
|
Increased release of serotonin from rat primary isolated adult cardiac myofibroblasts. Sci Rep 2021; 11:20376. [PMID: 34645867 PMCID: PMC8514503 DOI: 10.1038/s41598-021-99632-y] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/03/2021] [Accepted: 09/22/2021] [Indexed: 11/08/2022] Open
Abstract
Elevated blood serotonin levels have been observed in patients with heart failure and serotonin has a role in pathological cardiac function. The serotonin receptor system was examined in adult rat isolated cardiac fibroblast and myofibroblast cells. This is one of the first studies that has investigated serotonin receptors and other proteins involved in the serotonin receptor system in rat cardiac fibroblast and myofibroblast cells. Rat primary cardiac fibroblasts were isolated and transformed into myofibroblasts using 5 ng/ml TGF-β1. Transformation of cells to myofibroblasts was confirmed with the presence of α-smooth muscle actin using Western blot. Serotonin metabolism and receptor protein expression was assessed using Western blot techniques and serotonin levels measured using ELISA. The 5-HT1A, 5-HT2A and 5-HT2B receptors were found to be present in both rat cardiac fibroblasts and myofibroblast cells, however no significance in protein expression between the two cell types was found (P > 0.05). In this study a significant increase in the serotonin transporter (SERT), tryptophan hydroxylase 1 and extracellular serotonin levels was observed in rat cardiac myofibroblasts when compared to fibroblasts (P < 0.05). These results suggest that serotonin levels may rise in parallel with cardiac myofibroblast populations and contribute to the pathogenesis of heart failure via serotonin receptors.
Collapse
|
12
|
Haase J, Jones AKC, Mc Veigh CJ, Brown E, Clarke G, Ahnert-Hilger G. Sex and brain region-specific regulation of serotonin transporter activity in synaptosomes in guanine nucleotide-binding protein G(q) alpha knockout mice. J Neurochem 2021; 159:156-171. [PMID: 34309872 DOI: 10.1111/jnc.15482] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/22/2020] [Revised: 07/15/2021] [Accepted: 07/21/2021] [Indexed: 10/20/2022]
Abstract
The regulation of the serotonin transporter (SERT) by guanine nucleotide-binding protein alpha (Gα) q was investigated using Gαq knockout mice. In the absence of Gαq, SERT-mediated uptake of 5-hydroxytryptamine (5HT) was enhanced in midbrain and frontal cortex synaptosomes, but only in female mice. The mechanisms underlying this sexual dimorphism were investigated using quantitative western blot analysis revealing brain region-specific differences. In the frontal cortex, SERT protein expression was decreased in male knockout mice, seemingly explaining the sex-dependent variation in SERT activity. The differential expression of Gαi1 in female mice contributes to the sex differences in the midbrain. In fact, Gαi1 levels inversely correlate with 5HT uptake rates across both sexes and genotypes. Likely due to differential SERT regulation as well as sex differences in the expression of tryptophan hydroxylase 2, Gαq knockout mice also displayed sex- and genotype-dependent alterations in total 5HT tissue levels as determined by high-performance liquid chromatography. Gαq inhibitors, YM-254890 and BIM-46187, differentially affected SERT activity in both, synaptosomes and cultured cells. YM-254890 treatment mimicked the effect of Gαq knockout in the frontal cortex. BIM-46187, which promotes the nucleotide-free form of Gα proteins, substantially inhibited 5HT uptake, prompting us to hypothesise that Gαq interacts with SERT similarly as with G-protein-coupled receptors and inhibits SERT activity by modulating transport-associated conformational changes. Taken together, our findings reveal a novel mechanism of SERT regulation and impact our understanding of sex differences in diseases associated with dysregulation of serotonin transmission, such as depression and anxiety.
Collapse
Affiliation(s)
- Jana Haase
- UCD School of Biomolecular and Biomedical Science, UCD Conway Institute, University College Dublin, Dublin, Ireland
| | - Aimée K C Jones
- UCD School of Biomolecular and Biomedical Science, UCD Conway Institute, University College Dublin, Dublin, Ireland
| | - Conor J Mc Veigh
- UCD School of Biomolecular and Biomedical Science, UCD Conway Institute, University College Dublin, Dublin, Ireland
| | - Eric Brown
- UCD School of Biomolecular and Biomedical Science, UCD Conway Institute, University College Dublin, Dublin, Ireland
| | - Gerard Clarke
- APC Microbiome Ireland and Department of Psychiatry and Neurobehavioural Science, University College Cork, Cork, Ireland
| | - Gudrun Ahnert-Hilger
- Institute of Integrative Neuroanatomy, Charité University Medicine Berlin and Max-Planck-Institute for Biophysical Chemistry Göttingen, Göttingen, Germany
| |
Collapse
|
13
|
Chiu YC, Yang BH, Yang KC, Liu MN, Hu LY, Liou YJ, Chan LY, Chou YH. A study of tryptophan, kynurenine and serotonin transporter in first-episode drug-naïve major depressive disorder. Psychiatry Res Neuroimaging 2021; 312:111296. [PMID: 33945927 DOI: 10.1016/j.pscychresns.2021.111296] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/18/2020] [Revised: 04/16/2021] [Accepted: 04/22/2021] [Indexed: 11/24/2022]
Abstract
Major depressive disorder (MDD) is associated with the disharmonic functioning of the serotonin system. The serotonin system is mainly modulated by the serotonin transporter (SERT) which regulates serotonin uptake and the metabolism of its precursor, tryptophan and following kynurenine pathway. Currently, there is a lack of research examining both markers concurrently in MDD. This study evaluated the alterations and inter-relationships of both markers in first-episode drug-naïve MDD patients. Thirty-three MDD patients and 33 age- and sex-matched healthy controls (HC) were recruited. The SERT availability were comparable between two groups in the midbrain, thalamus, caudate, and putamen. The kynurenine/tryptophan ratio which indicates tryptophan metabolism was lower in MDD than HC with no group difference in the tryptophan or kynurenine concentration. A negative correlation between the midbrain SERT availability and kynurenine concentration in HC was found. For the subgroup of HC with high kynurenine/tryptophan ratio, the SERT availability was positively associated with the kynurenine/tryptophan ratio and negatively correlated with tryptophan or kynurenine concentration. This study demonstrated the altered tryptophan metabolism and the relationship between tryptophan metabolism and the SERT availability in first-episode drug-naïve MDD patients, which gave a new insight towards the future investigation of the pathophysiology of MDD.
Collapse
Affiliation(s)
- Yen-Chen Chiu
- Department of Psychiatry, Taipei Veterans General Hospital, Taipei, Taiwan; Center for Quality Management, Taipei Veterans General Hospital, Taipei, Taiwan
| | - Bang-Hung Yang
- Department of Nuclear Medicine, Taipei Veterans General Hospital, Taipei, Taiwan; Department of Biomedical Imaging and Radiological Sciences, National Yang Ming Chiao Tung University, Taipei, Taiwan
| | - Kai-Chin Yang
- Department of Psychiatry, Taipei Veterans General Hospital, Taipei, Taiwan; Department of Psychiatry, Faculty of Medicine, National Yang Ming Chiao Tung University, Taipei, Taiwan
| | - Mu-N Liu
- Department of Psychiatry, Taipei Veterans General Hospital, Taipei, Taiwan; Department of Psychiatry, Faculty of Medicine, National Yang Ming Chiao Tung University, Taipei, Taiwan
| | - Li-Yu Hu
- Department of Psychiatry, Taipei Veterans General Hospital, Taipei, Taiwan; Department of Psychiatry, Faculty of Medicine, National Yang Ming Chiao Tung University, Taipei, Taiwan
| | - Ying-Jay Liou
- Department of Psychiatry, Taipei Veterans General Hospital, Taipei, Taiwan; Department of Psychiatry, Faculty of Medicine, National Yang Ming Chiao Tung University, Taipei, Taiwan
| | - Li-Yi Chan
- Center for Quality Management, Taipei Veterans General Hospital, Taipei, Taiwan
| | - Yuan-Hwa Chou
- Department of Psychiatry, Taipei Veterans General Hospital, Taipei, Taiwan; Center for Quality Management, Taipei Veterans General Hospital, Taipei, Taiwan; Department of Psychiatry, Faculty of Medicine, National Yang Ming Chiao Tung University, Taipei, Taiwan.
| |
Collapse
|
14
|
Ji MJ, Yang J, Gao ZQ, Zhang L, Liu C. The Role of the Kappa Opioid System in Comorbid Pain and Psychiatric Disorders: Function and Implications. Front Neurosci 2021; 15:642493. [PMID: 33716658 PMCID: PMC7943636 DOI: 10.3389/fnins.2021.642493] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/16/2020] [Accepted: 01/27/2021] [Indexed: 01/25/2023] Open
Abstract
Both pain and psychiatric disorders, such as anxiety and depression, significantly impact quality of life for the sufferer. The two also share a strong pathological link: chronic pain-induced negative affect drives vulnerability to psychiatric disorders, while patients with comorbid psychiatric disorders tend to experience exacerbated pain. However, the mechanisms responsible for the comorbidity of pain and psychiatric disorders remain unclear. It is well established that the kappa opioid system contributes to depressive and dysphoric states. Emerging studies of chronic pain have revealed the role and mechanisms of the kappa opioid system in pain processing and, in particular, in the associated pathological alteration of affection. Here, we discuss the key findings and summarize compounds acting on the kappa opioid system that are potential candidates for therapeutic strategies against comorbid pain and psychiatric disorders.
Collapse
Affiliation(s)
- Miao-Jin Ji
- Jiangsu Province Key Laboratory of Anesthesiology and Jiangsu Province Key Laboratory of Anesthesia and Analgesia Application Technology, School of Anesthesiology, Xuzhou Medical University, Xuzhou, China
| | - Jiao Yang
- Jiangsu Province Key Laboratory of Anesthesiology and Jiangsu Province Key Laboratory of Anesthesia and Analgesia Application Technology, School of Anesthesiology, Xuzhou Medical University, Xuzhou, China
| | - Zhi-Qiang Gao
- Jiangsu Province Key Laboratory of New Drug Research and Clinical Pharmacy, School of Pharmacy, Xuzhou Medical University, Xuzhou, China
| | - Liang Zhang
- Department of Neurology, Affiliated Hospital of Qingdao University, Qingdao, China
| | - Chao Liu
- Jiangsu Province Key Laboratory of Anesthesiology and Jiangsu Province Key Laboratory of Anesthesia and Analgesia Application Technology, School of Anesthesiology, Xuzhou Medical University, Xuzhou, China
| |
Collapse
|
15
|
Sex and the serotonergic underpinnings of depression and migraine. HANDBOOK OF CLINICAL NEUROLOGY 2020; 175:117-140. [PMID: 33008520 DOI: 10.1016/b978-0-444-64123-6.00009-6] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Subscribe] [Scholar Register] [Indexed: 11/20/2022]
Abstract
Most psychiatric disorders demonstrate sex differences in their prevalence and symptomatology, and in their response to treatment. These differences are particularly pronounced in mood disorders. Differences in sex hormone levels are among the most overt distinctions between males and females and are thus an intuitive underpinning for these clinical observations. In fact, treatment with estrogen and testosterone was shown to exert antidepressant effects, which underscores this link. Changes to monoaminergic signaling in general, and serotonergic transmission in particular, are understood as central components of depressive pathophysiology. Thus, modulation of the serotonin system may serve as a mechanism via which sex hormones exert their clinical effects in mental health disorders. Over the past 20 years, various experimental approaches have been applied to identify modes of influence of sex and sex hormones on the serotonin system. This chapter provides an overview of different molecular components of the serotonin system, followed by a review of studies performed in animals and in humans with the purpose of elucidating sex hormone effects. Particular emphasis will be placed on studies performed with positron emission tomography, a method that allows for human in vivo molecular imaging and, therefore, assessment of effects in a clinically representative context. The studies addressed in this chapter provide a wealth of information on the interaction between sex, sex hormones, and serotonin in the brain. In general, they offer evidence for the concept that the influence of sex hormones on various components of the serotonin system may serve as an underpinning for the clinical effects these hormones demonstrate.
Collapse
|
16
|
Ragu Varman D, Jayanthi LD, Ramamoorthy S. Glycogen synthase kinase-3ß supports serotonin transporter function and trafficking in a phosphorylation-dependent manner. J Neurochem 2020; 156:445-464. [PMID: 32797733 DOI: 10.1111/jnc.15152] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/23/2020] [Revised: 07/23/2020] [Accepted: 08/10/2020] [Indexed: 12/13/2022]
Abstract
Serotonin (5-HT) transporter (SERT) plays a crucial role in serotonergic transmission in the central nervous system, and any aberration causes serious mental illnesses. Nevertheless, the cellular mechanisms that regulate SERT function and trafficking are not entirely understood. Growing evidence suggests that several protein kinases act as modulators. Here, we delineate the molecular mechanisms by which glycogen synthase kinase-3ß (GSK3ß) regulates SERT. When mouse striatal synaptosomes were treated with the GSK3α/ß inhibitor CHIR99021, we observed a significant increase in SERT function, Vmax , surface expression with a reduction in 5-HT Km and SERT phosphorylation. To further study how the SERT molecule is affected by GSK3α/ß, we used HEK-293 cells as a heterologous expression system. As in striatal synaptosomes, CHIR99021 treatment of cells expressing wild-type hSERT (hSERT-WT) resulted in a time and dose-dependent elevation of hSERT function with a concomitant increase in the Vmax and surface transporters because of reduced internalization and enhanced membrane insertion; silencing GSK3α/ß in these cells with siRNA also similarly affected hSERT. Converting putative GSK3α/ß phosphorylation site serine at position 48 to alanine in hSERT (hSERT-S48A) completely abrogated the effects of both the inhibitor CHIR99021 and GSK3α/ß siRNA. Substantiating these findings, over-expression of constitutively active GSK3ß with hSERT-WT, but not with hSERT-S48A, reduced SERT function, Vmax , surface density, and enhanced transporter phosphorylation. Both hSERT-WT and hSERT-S48A were inhibited similarly by PKC activation or by inhibition of Akt, CaMKII, p38 MAPK, or Src kinase. These findings provide new evidence that GSK3ß supports basal SERT function and trafficking via serine-48 phosphorylation.
Collapse
Affiliation(s)
- Durairaj Ragu Varman
- Department of Pharmacology and Toxicology, Virginia Commonwealth University, Richmond, VA, USA
| | - Lankupalle D Jayanthi
- Department of Pharmacology and Toxicology, Virginia Commonwealth University, Richmond, VA, USA
| | - Sammanda Ramamoorthy
- Department of Pharmacology and Toxicology, Virginia Commonwealth University, Richmond, VA, USA
| |
Collapse
|
17
|
Quinlan MA, Robson MJ, Ye R, Rose KL, Schey KL, Blakely RD. Ex vivo Quantitative Proteomic Analysis of Serotonin Transporter Interactome: Network Impact of the SERT Ala56 Coding Variant. Front Mol Neurosci 2020; 13:89. [PMID: 32581705 PMCID: PMC7295033 DOI: 10.3389/fnmol.2020.00089] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/04/2020] [Accepted: 04/28/2020] [Indexed: 12/15/2022] Open
Abstract
Altered serotonin (5-HT) signaling is associated with multiple brain disorders, including major depressive disorder (MDD), obsessive-compulsive disorder (OCD), and autism spectrum disorder (ASD). The presynaptic, high-affinity 5-HT transporter (SERT) tightly regulates 5-HT clearance after release from serotonergic neurons in the brain and enteric nervous systems, among other sites. Accumulating evidence suggests that SERT is dynamically regulated in distinct activity states as a result of environmental and intracellular stimuli, with regulation perturbed by disease-associated coding variants. Our lab identified a rare, hypermorphic SERT coding substitution, Gly56Ala, in subjects with ASD, finding that the Ala56 variant stabilizes a high-affinity outward-facing conformation (SERT∗) that leads to elevated 5-HT uptake in vitro and in vivo. Hyperactive SERT Ala56 appears to preclude further activity enhancements by p38α mitogen-activated protein kinase (MAPK) and can be normalized by pharmacological p38α MAPK inhibition, consistent with SERT Ala56 mimicking, constitutively, a high-activity conformation entered into transiently by p38α MAPK activation. We hypothesize that changes in SERT-interacting proteins (SIPs) support the shift of SERT into the SERT∗ state which may be captured by comparing the composition of SERT Ala56 protein complexes with those of wildtype (WT) SERT, defining specific interactions through comparisons of protein complexes recovered using preparations from SERT–/– (knockout; KO) mice. Using quantitative proteomic-based approaches, we identify a total of 459 SIPs, that demonstrate both SERT specificity and sensitivity to the Gly56Ala substitution, with a striking bias being a loss of SIP interactions with SERT Ala56 compared to WT SERT. Among this group are previously validated SIPs, such as flotillin-1 (FLOT1) and protein phosphatase 2A (PP2A), whose functions are believed to contribute to SERT microdomain localization and regulation. Interestingly, our studies nominate a number of novel SIPs implicated in ASD, including fragile X mental retardation 1 protein (FMR1) and SH3 and multiple ankyrin repeat domains protein 3 (SHANK3), of potential relevance to long-standing evidence of serotonergic contributions to ASD. Further investigation of these SIPs, and the broader networks they engage, may afford a greater understanding of ASD as well as other brain and peripheral disorders associated with perturbed 5-HT signaling.
Collapse
Affiliation(s)
- Meagan A Quinlan
- Department of Psychiatry and Behavioral Sciences, University of Washington, Seattle, WA, United States.,Department of Pharmacology, Vanderbilt University, Nashville, TN, United States.,Department of Biomedical Science, Charles E. Schmidt College of Medicine, Florida Atlantic University, Jupiter, FL, United States
| | - Matthew J Robson
- Division of Pharmaceutical Sciences, James L. Winkle College of Pharmacy, University of Cincinnati, Cincinnati, OH, United States
| | - Ran Ye
- Department of Pharmacology, Vanderbilt University, Nashville, TN, United States
| | - Kristie L Rose
- Department of Biochemistry, Vanderbilt University, Nashville, TN, United States
| | - Kevin L Schey
- Department of Biochemistry, Vanderbilt University, Nashville, TN, United States
| | - Randy D Blakely
- Department of Pharmacology, Vanderbilt University, Nashville, TN, United States.,Brain Institute, Florida Atlantic University, Jupiter, FL, United States
| |
Collapse
|
18
|
Kranz GS, Zhang BBB, Handschuh P, Ritter V, Lanzenberger R. Gender-affirming hormone treatment - A unique approach to study the effects of sex hormones on brain structure and function. Cortex 2020; 129:68-79. [PMID: 32438011 DOI: 10.1016/j.cortex.2020.04.005] [Citation(s) in RCA: 21] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/19/2020] [Revised: 03/23/2020] [Accepted: 04/09/2020] [Indexed: 12/22/2022]
Abstract
Investigating the effects of the gender-affirming hormone treatment of transgender people using neuroimaging provides a unique opportunity to study the impact of high dosages of sex hormones on human brain structure and function. This line of research is of relevance from a basic neuroscientific as well as from a psychiatric viewpoint. Prevalence rates, etiopathology, and disease course of many psychiatric disorders exhibit sex differences which are linked to differences in sex hormone levels. Here, we review recent neuroimaging studies from others and our group that investigate the effects of gender-affirming hormone treatment in a longitudinal design utilizing structural and functional magnetic resonance imaging and positron emission tomography. Studies point to a general anabolic and anticatabolic effect of testosterone on grey and white matter structure, whereas estradiol and antiandrogen treatment seems to have partly opposite effects. Moreover, preliminary research indicates that gender-affirming hormone treatment influences serotonergic neurotransmission, a finding that is especially interesting for psychiatry. A clear picture of a hormonal influence on brain activity has yet to emerge. In conclusion, the available evidence reviewed here clearly indicates that sex hormone applications influence brain structure and function in the adult human brain.
Collapse
Affiliation(s)
- Georg S Kranz
- Department of Rehabilitation Sciences, The Hong Kong Polytechnic University, Hong Kong, SAR, China; Department of Psychiatry and Psychotherapy, Medical University of Vienna, Vienna, Austria; The State Key Laboratory of Brain and Cognitive Sciences, The University of Hong Kong, Hong Kong, SAR, China.
| | - Bella B B Zhang
- Department of Rehabilitation Sciences, The Hong Kong Polytechnic University, Hong Kong, SAR, China
| | - Patricia Handschuh
- Department of Psychiatry and Psychotherapy, Medical University of Vienna, Vienna, Austria
| | - Vera Ritter
- Department of Psychiatry and Psychotherapy, Medical University of Vienna, Vienna, Austria
| | - Rupert Lanzenberger
- Department of Psychiatry and Psychotherapy, Medical University of Vienna, Vienna, Austria
| |
Collapse
|
19
|
Manzella C, Singhal M, Ackerman M, Alrefai WA, Saksena S, Dudeja PK, Gill RK. Serotonin transporter untranslated regions influence mRNA abundance and protein expression. GENE REPORTS 2020; 18. [PMID: 34113740 DOI: 10.1016/j.genrep.2019.100513] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/06/2023]
Abstract
The serotonin transporter (SERT, SLC6A4) is a Na+-dependent transporter that regulates the availability of serotonin (5-HT, 5-hydroxytryptamine), a key neurotransmitter and hormone in the brain and the intestine. The human SERT gene consists of two alternate promoters that drive expression of an identical SERT protein. However, there are different mRNA transcript variants derived from these two promoters that differ in their 5' untranslated region (5'UTR), which is the region of the mRNA upstream from the protein-coding region. Two of these transcripts contain exon-1a and are abundant in neuronal tissue, whereas the third transcript contains exon-1c and is abundant in the intestine. The 3'UTR is nearly identical among the transcripts. Current studies tested the hypothesis that the UTRs of SERT influence its expression in intestinal epithelial cells (IECs) by controlling mRNA or protein levels. The SERT UTRs were cloned into luciferase reporter plasmids and luciferase mRNA and activity were measured following transient transfection of the UTR constructs into the model IEC Caco-2. Luciferase activity and mRNA abundance were higher than the empty vector for two of the three 5'UTR variants. Calculation of translation index (luciferase activity divided by the relative luciferase mRNA level) revealed that the exon-1a containing 5'UTRs had enhanced translation when compared to the exon-1c containing 5'UTR which exhibited a low translation efficiency. Compared to the empty vector, the SERT 3'UTR markedly decreased luciferase activity. In silico analysis of the SERT 3'UTR revealed many conserved potential miRNA binding sites that may be responsible for this decrease. In conclusion, we have shown that the UTRs of SERT regulate mRNA abundance and protein expression. Delineating the molecular basis by which the UTRs of SERT influence its expression will lead to an increased understanding of post-transcriptional regulation of SERT in GI disorders associated with altered 5-HT availability.
Collapse
Affiliation(s)
- Christopher Manzella
- Department of Physiology & Biophysics, University of Illinois at Chicago, Chicago, IL, United States of America
| | - Megha Singhal
- Division of Gastroenterology & Hepatology, University of Illinois at Chicago, Chicago, IL, United States of America
| | - Max Ackerman
- University of Illinois at Chicago, College of Liberal Arts and Sciences, United States of America
| | - Waddah A Alrefai
- Division of Gastroenterology & Hepatology, University of Illinois at Chicago, Chicago, IL, United States of America.,Jesse Brown VA Medical Center, Chicago, IL, United States of America
| | - Seema Saksena
- Division of Gastroenterology & Hepatology, University of Illinois at Chicago, Chicago, IL, United States of America.,Jesse Brown VA Medical Center, Chicago, IL, United States of America
| | - Pradeep K Dudeja
- Division of Gastroenterology & Hepatology, University of Illinois at Chicago, Chicago, IL, United States of America.,Jesse Brown VA Medical Center, Chicago, IL, United States of America
| | - Ravinder K Gill
- Division of Gastroenterology & Hepatology, University of Illinois at Chicago, Chicago, IL, United States of America.,Jesse Brown VA Medical Center, Chicago, IL, United States of America
| |
Collapse
|
20
|
Kokhan VS, Lebedeva-Georgievskaya KB, Kudrin VS, Bazyan AS, Maltsev AV, Shtemberg AS. An investigation of the single and combined effects of hypogravity and ionizing radiation on brain monoamine metabolism and rats' behavior. LIFE SCIENCES IN SPACE RESEARCH 2019; 20:12-19. [PMID: 30797429 DOI: 10.1016/j.lssr.2018.11.003] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/25/2018] [Revised: 11/23/2018] [Accepted: 11/28/2018] [Indexed: 06/09/2023]
Abstract
BACKGROUND Ionizing radiation and hypogravity can cause central nervous system (CNS) dysfunctions. This is a key limiting factor for deep space missions. Up until now, the mechanisms through which they affect the neural tissue are not completely understood. OBJECTIVES We studied how the combination of hypogravity (antiorthostatic suspension model, AS) and ionizing radiations (γ-quanta and 1H+ together, R) affects the CNS. METHODS We applied separately and in combination AS and R to determine the influence of these factors on behavior and metabolism of monoamines in Wistar rat's brain. RESULTS We found out that R has a slight effect on both the behavior and metabolism of monoamines. However, when applied in combination with AS the former was able to reduce the negative effects of the latter. The combined effect of ionizing radiation and hypogravity led to the recovery of locomotor activity, orientation and exploratory behavior, and long-term context memory impaired under the impact of hypogravity only. These changes came together with an increase in the serotonin and dopamine turnover in all of the brain structures that were studied. CONCLUSIONS We received the first evidence of interferential interaction between the effects of ionizing radiation and hypogravity factors with regard to a behavior and monoamine turnover in the brain. Further studies with heavy nuclei at relevant doses (<0.5 Gy) are needed.
Collapse
Affiliation(s)
- Viktor S Kokhan
- Laboratory of Radiation and Extreme Neurophysiology, Institute of Biomedical Problems RAS, Khoroshevskoe shosse 76A, Moscow 123007, Russia.
| | - Kseniya B Lebedeva-Georgievskaya
- Laboratory of Radiation and Extreme Neurophysiology, Institute of Biomedical Problems RAS, Khoroshevskoe shosse 76A, Moscow 123007, Russia
| | - Vladimir S Kudrin
- Laboratory of Radiation and Extreme Neurophysiology, Institute of Biomedical Problems RAS, Khoroshevskoe shosse 76A, Moscow 123007, Russia
| | - Ara S Bazyan
- Laboratory of Radiation and Extreme Neurophysiology, Institute of Biomedical Problems RAS, Khoroshevskoe shosse 76A, Moscow 123007, Russia; Institute of Higher Nervous Activity and Neurophysiology RAS, Moscow, Russia
| | - Andrey V Maltsev
- Institute of Physiologically Active Compounds RAS, Chernogolovka, Russia
| | - Andrey S Shtemberg
- Laboratory of Radiation and Extreme Neurophysiology, Institute of Biomedical Problems RAS, Khoroshevskoe shosse 76A, Moscow 123007, Russia
| |
Collapse
|
21
|
Bailey DM, Catron MA, Kovtun O, Macdonald RL, Zhang Q, Rosenthal SJ. Single Quantum Dot Tracking Reveals Serotonin Transporter Diffusion Dynamics are Correlated with Cholesterol-Sensitive Threonine 276 Phosphorylation Status in Primary Midbrain Neurons. ACS Chem Neurosci 2018; 9:2534-2541. [PMID: 29787674 DOI: 10.1021/acschemneuro.8b00214] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/16/2023] Open
Abstract
Serotonin transporter (SERT) terminates serotonin signaling in the brain by enabling rapid clearance of the neurotransmitter. SERT dysfunction has been associated with a variety of psychiatric disorders, including depression, anxiety, and autism. Visualizing SERT behavior at the single molecule level in endogenous systems remains a challenge. In this study, we utilize quantum dot (QD) single particle tracking (SPT) to capture SERT dynamics in primary rat midbrain neurons. Membrane microenvironment, specifically membrane cholesterol, plays a key role in SERT regulation and has been found to affect SERT conformational state. We sought to determine how reduced cholesterol content affects both lateral mobility and phosphorylation of conformationally sensitive threonine 276 (Thr276) in endogenous SERT using two different methods of cholesterol manipulation, statins and methyl-β-cyclodextrin. Both chronic and acute cholesterol depletion increased SERT lateral diffusion, radial displacement along the membrane, mobile fraction, and Thr276 phosphorylation levels. Overall, this work has provided new insights about endogenous neuronal SERT mobility and its associations with membrane cholesterol and SERT phosphorylation status.
Collapse
|
22
|
p38α MAPK signaling drives pharmacologically reversible brain and gastrointestinal phenotypes in the SERT Ala56 mouse. Proc Natl Acad Sci U S A 2018; 115:E10245-E10254. [PMID: 30297392 PMCID: PMC6205438 DOI: 10.1073/pnas.1809137115] [Citation(s) in RCA: 38] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/18/2022] Open
Abstract
Autism spectrum disorder (ASD) is a common neurobehavioral disorder with limited treatment options. Activation of p38 MAPK signaling networks has been identified in ASD, and p38 MAPK signaling elevates serotonin (5-HT) transporter (SERT) activity, effects mimicked by multiple, hyperfunctional SERT coding variants identified in ASD subjects. Mice expressing the most common of these variants (SERT Ala56) exhibit hyperserotonemia, a biomarker observed in ASD subjects, as well as p38 MAPK-dependent SERT hyperphosphorylation, elevated hippocampal 5-HT clearance, hypersensitivity of CNS 5-HT1A and 5-HT2A/2C receptors, and behavioral and gastrointestinal perturbations reminiscent of ASD. As the α-isoform of p38 MAPK drives SERT activation, we tested the hypothesis that CNS-penetrant, α-isoform-specific p38 MAPK inhibitors might normalize SERT Ala56 phenotypes. Strikingly, 1-week treatment of adult SERT Ala56 mice with MW150, a selective p38α MAPK inhibitor, normalized hippocampal 5-HT clearance, CNS 5-HT1A and 5-HT2A/2C receptor sensitivities, social interactions, and colonic motility. Conditional elimination of p38α MAPK in 5-HT neurons of SERT Ala56 mice restored 5-HT1A and 5-HT2A/2C receptor sensitivities as well as social interactions, mirroring effects of MW150. Our findings support ongoing p38α MAPK activity as an important determinant of the physiological and behavioral perturbations of SERT Ala56 mice and, more broadly, supports consideration of p38α MAPK inhibition as a potential treatment for core and comorbid phenotypes present in ASD subjects.
Collapse
|
23
|
Brown E, Mc Veigh CJ, Santos L, Gogarty M, Müller HK, Elfving B, Brayden DJ, Haase J. TNFα-dependent anhedonia and upregulation of hippocampal serotonin transporter activity in a mouse model of collagen-induced arthritis. Neuropharmacology 2018; 137:211-220. [DOI: 10.1016/j.neuropharm.2018.04.023] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/19/2017] [Revised: 04/19/2018] [Accepted: 04/22/2018] [Indexed: 01/02/2023]
|
24
|
Kruk JS, Bermeo S, Skarratt KK, Fuller SJ, Duque G. The Effect of Antidepressants on Mesenchymal Stem Cell Differentiation. J Bone Metab 2018; 25:43-51. [PMID: 29564305 PMCID: PMC5854822 DOI: 10.11005/jbm.2018.25.1.43] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/11/2018] [Revised: 02/27/2018] [Accepted: 02/28/2018] [Indexed: 11/25/2022] Open
Abstract
Background Use of antidepressant medications has been linked to detrimental impacts on bone mineral density and osteoporosis; however, the cellular basis behind these observations remains poorly understood. The effect does not appear to be homogeneous across the whole class of drugs and may be linked to affinity for the serotonin transporter system. In this study, we hypothesized that antidepressants have a class- and dose-dependent effect on mesenchymal stem cell (MSC) differentiation, which may affect bone metabolism. Methods Human MSCs (hMSCs) were committed to differentiate when either adipogenic or osteogenic media was added, supplemented with five increasing concentrations of amitriptyline (0.001–10 µM), venlafaxine (0.01–25 µM), or fluoxetine (0.001–10 µM). Alizarin red staining (mineralization), alkaline phosphatase (osteoblastogenesis), and oil red O (adipogenesis) assays were performed at timed intervals. In addition, cell viability was assessed using a MTT. Results We found that fluoxetine had a significant inhibitory effect on mineralization. Furthermore, adipogenic differentiation of hMSC was affected by the addition of amitriptyline, venlafaxine, and fluoxetine to the media. Finally, none of the tested medications significantly affected cell survival. Conclusions This study showed a divergent effect of three antidepressants on hMSC differentiation, which appears to be independent of class and dose. As fluoxetine and amitriptyline, but not venlafaxine, affected both osteoblastogenesis and adipogenesis, this inhibitory effect could be associated to the high affinity of fluoxetine to the serotonin transporter system.
Collapse
Affiliation(s)
- Jeffrey S Kruk
- Sydney Medical School Nepean, The University of Sydney, Penrith, Australia
| | - Sandra Bermeo
- Sydney Medical School Nepean, The University of Sydney, Penrith, Australia.,Facultad de Ciencias Básicas y Biomédicas, Universidad Simón Bolívar, Barranquilla, Colombia
| | - Kristen K Skarratt
- Sydney Medical School Nepean, The University of Sydney, Penrith, Australia
| | - Stephen J Fuller
- Sydney Medical School Nepean, The University of Sydney, Penrith, Australia
| | - Gustavo Duque
- Sydney Medical School Nepean, The University of Sydney, Penrith, Australia.,Australian Institute for Musculoskeletal Science (AIMSS), The University of Melbourne and Western Health, Melbourne, Australia.,Department of Medicine-Western Health, Melbourne Medical School, The University of Melbourne, Melbourne, Australia
| |
Collapse
|
25
|
Komorowski A, James GM, Philippe C, Gryglewski G, Bauer A, Hienert M, Spies M, Kautzky A, Vanicek T, Hahn A, Traub-Weidinger T, Winkler D, Wadsak W, Mitterhauser M, Hacker M, Kasper S, Lanzenberger R. Association of Protein Distribution and Gene Expression Revealed by PET and Post-Mortem Quantification in the Serotonergic System of the Human Brain. Cereb Cortex 2018; 27:117-130. [PMID: 27909009 PMCID: PMC5939202 DOI: 10.1093/cercor/bhw355] [Citation(s) in RCA: 27] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/26/2016] [Indexed: 12/12/2022] Open
Abstract
Regional differences in posttranscriptional mechanisms may influence in vivo protein densities. The association of positron emission tomography (PET) imaging data from 112 healthy controls and gene expression values from the Allen Human Brain Atlas, based on post-mortem brains, was investigated for key serotonergic proteins. PET binding values and gene expression intensities were correlated for the main inhibitory (5-HT1A) and excitatory (5-HT2A) serotonin receptor, the serotonin transporter (SERT) as well as monoamine oxidase-A (MAO-A), using Spearman's correlation coefficients (rs) in a voxel-wise and region-wise analysis. Correlations indicated a strong linear relationship between gene and protein expression for both the 5-HT1A (voxel-wise rs = 0.71; region-wise rs = 0.93) and the 5-HT2A receptor (rs = 0.66; 0.75), but only a weak association for MAO-A (rs = 0.26; 0.66) and no clear correlation for SERT (rs = 0.17; 0.29). Additionally, region-wise correlations were performed using mRNA expression from the HBT, yielding comparable results (5-HT1Ars = 0.82; 5-HT2Ars = 0.88; MAO-A rs = 0.50; SERT rs = -0.01). The SERT and MAO-A appear to be regulated in a region-specific manner across the whole brain. In contrast, the serotonin-1A and -2A receptors are presumably targeted by common posttranscriptional processes similar in all brain areas suggesting the applicability of mRNA expression as surrogate parameter for density of these proteins.
Collapse
Affiliation(s)
- A Komorowski
- Department of Psychiatry and Pychotherapy, Division of Biological Psychiatry, Medical University of Vienna, Währinger Gürtel 18-20, 1090 Vienna, Austria
| | - G M James
- Department of Psychiatry and Pychotherapy, Division of Biological Psychiatry, Medical University of Vienna, Währinger Gürtel 18-20, 1090 Vienna, Austria
| | - C Philippe
- Department of Biomedical Imaging and Image-guided Therapy, Division of Nuclear Medicine, Medical University of Vienna, Währinger Gürtel 18-20, 1090 Vienna, Austria
| | - G Gryglewski
- Department of Psychiatry and Pychotherapy, Division of Biological Psychiatry, Medical University of Vienna, Währinger Gürtel 18-20, 1090 Vienna, Austria
| | - A Bauer
- Institute of Neuroscience and Medicine (INM-2), Research Centre Jülich, 52425 Jülich, Germany
| | - M Hienert
- Department of Psychiatry and Pychotherapy, Division of Biological Psychiatry, Medical University of Vienna, Währinger Gürtel 18-20, 1090 Vienna, Austria
| | - M Spies
- Department of Psychiatry and Pychotherapy, Division of Biological Psychiatry, Medical University of Vienna, Währinger Gürtel 18-20, 1090 Vienna, Austria
| | - A Kautzky
- Department of Psychiatry and Pychotherapy, Division of Biological Psychiatry, Medical University of Vienna, Währinger Gürtel 18-20, 1090 Vienna, Austria
| | - T Vanicek
- Department of Psychiatry and Pychotherapy, Division of Biological Psychiatry, Medical University of Vienna, Währinger Gürtel 18-20, 1090 Vienna, Austria
| | - A Hahn
- Department of Psychiatry and Pychotherapy, Division of Biological Psychiatry, Medical University of Vienna, Währinger Gürtel 18-20, 1090 Vienna, Austria
| | - T Traub-Weidinger
- Department of Biomedical Imaging and Image-guided Therapy, Division of Nuclear Medicine, Medical University of Vienna, Währinger Gürtel 18-20, 1090 Vienna, Austria
| | - D Winkler
- Department of Psychiatry and Pychotherapy, Division of Biological Psychiatry, Medical University of Vienna, Währinger Gürtel 18-20, 1090 Vienna, Austria
| | - W Wadsak
- Department of Biomedical Imaging and Image-guided Therapy, Division of Nuclear Medicine, Medical University of Vienna, Währinger Gürtel 18-20, 1090 Vienna, Austria
| | - M Mitterhauser
- Department of Biomedical Imaging and Image-guided Therapy, Division of Nuclear Medicine, Medical University of Vienna, Währinger Gürtel 18-20, 1090 Vienna, Austria
| | - M Hacker
- Department of Biomedical Imaging and Image-guided Therapy, Division of Nuclear Medicine, Medical University of Vienna, Währinger Gürtel 18-20, 1090 Vienna, Austria
| | - S Kasper
- Department of Psychiatry and Pychotherapy, Division of Biological Psychiatry, Medical University of Vienna, Währinger Gürtel 18-20, 1090 Vienna, Austria
| | - R Lanzenberger
- Department of Psychiatry and Pychotherapy, Division of Biological Psychiatry, Medical University of Vienna, Währinger Gürtel 18-20, 1090 Vienna, Austria
| |
Collapse
|
26
|
Bodnar CN, Morganti JM, Bachstetter AD. Depression following a traumatic brain injury: uncovering cytokine dysregulation as a pathogenic mechanism. Neural Regen Res 2018; 13:1693-1704. [PMID: 30136679 PMCID: PMC6128046 DOI: 10.4103/1673-5374.238604] [Citation(s) in RCA: 38] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022] Open
Abstract
A substantial number of individuals have long-lasting adverse effects from a traumatic brain injury (TBI). Depression is one of these long-term complications that influences many aspects of life. Depression can limit the ability to return to work, and even worsen cognitive function and contribute to dementia. The mechanistic cause for the increased depression risk associated with a TBI remains to be defined. As TBI results in chronic neuroinflammation, and priming of glia to a secondary challenge, the inflammatory theory of depression provides a promising framework for investigating the cause of depression following a TBI. Increases in cytokines similar to those seen in depression in the general population are also increased following a TBI. Biomarker levels of cytokines peak within hours-to-days after the injury, yet pro-inflammatory cytokines may still be elevated above physiological levels months-to-years following TBI, which is the time frame in which post-TBI depression can persist. As tumor necrosis factor α and interleukin 1 can signal directly at the neuronal synapse, pathophysiological levels of these cytokines can detrimentally alter neuronal synaptic physiology. The purpose of this review is to outline the current evidence for the inflammatory hypothesis of depression specifically as it relates to depression following a TBI. Moreover, we will illustrate the potential synaptic mechanisms by which tumor necrosis factor α and interleukin 1 could contribute to depression. The association of inflammation with the development of depression is compelling; however, in the context of post-TBI depression, the role of inflammation is understudied. This review attempts to highlight the need to understand and treat the psychological complications of a TBI, potentially by neuroimmune modulation, as the neuropsychiatric disabilities can have a great impact on the rehabilitation from the injury, and overall quality of life.
Collapse
Affiliation(s)
- Colleen N Bodnar
- Spinal Cord & Brain Injury Research Center, University of Kentucky; Department of Neuroscience, University of Kentucky, Lexington, KY, USA
| | - Josh M Morganti
- Department of Neuroscience, University of Kentucky; Sanders-Brown Center on Aging, University of Kentucky, Lexington, KY, USA
| | - Adam D Bachstetter
- Spinal Cord & Brain Injury Research Center, University of Kentucky; Department of Neuroscience, University of Kentucky, Lexington, KY, USA
| |
Collapse
|
27
|
Haase J, Grudzinska-Goebel J, Müller HK, Münster-Wandowski A, Chow E, Wynne K, Farsi Z, Zander JF, Ahnert-Hilger G. Serotonin Transporter Associated Protein Complexes Are Enriched in Synaptic Vesicle Proteins and Proteins Involved in Energy Metabolism and Ion Homeostasis. ACS Chem Neurosci 2017; 8:1101-1116. [PMID: 28362488 DOI: 10.1021/acschemneuro.6b00437] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022] Open
Abstract
The serotonin transporter (SERT) mediates Na+-dependent high-affinity serotonin uptake and plays a key role in regulating extracellular serotonin concentration in the brain and periphery. To gain novel insight into SERT regulation, we conducted a comprehensive proteomics screen to identify components of SERT-associated protein complexes in the brain by employing three independent approaches. In vivo SERT complexes were purified from rat brain using an immobilized high-affinity SERT ligand, amino-methyl citalopram. This approach was combined with GST pulldown and yeast two-hybrid screens using N- and C-terminal cytoplasmic transporter domains as bait. Potential SERT associated proteins detected by at least two of the interaction methods were subjected to gene ontology analysis resulting in the identification of functional protein clusters that are enriched in SERT complexes. Prominent clusters include synaptic vesicle proteins, as well as proteins involved in energy metabolism and ion homeostasis. Using subcellular fractionation and electron microscopy we provide further evidence that SERT is indeed associated with synaptic vesicle fractions, and colocalizes with small vesicular structures in axons and axon terminals. We also show that SERT is found in close proximity to mitochondrial membranes in both, hippocampal and neocortical regions. We propose a model of the SERT interactome, in which SERT is distributed between different subcellular compartments through dynamic interactions with site-specific protein complexes. Finally, our protein interaction data suggest novel hypotheses for the regulation of SERT activity and trafficking, which ultimately impact on serotonergic neurotransmission and serotonin dependent brain functions.
Collapse
Affiliation(s)
- Jana Haase
- UCD School of Biomolecular and Biomedical Science, UCD Conway Institute, University College Dublin, Dublin 4, Ireland
| | - Joanna Grudzinska-Goebel
- UCD School of Biomolecular and Biomedical Science, UCD Conway Institute, University College Dublin, Dublin 4, Ireland
| | - Heidi Kaastrup Müller
- UCD School of Biomolecular and Biomedical Science, UCD Conway Institute, University College Dublin, Dublin 4, Ireland
- Department
of Clinical Medicine, Translational Neuropsychiatry Unit, Aarhus University, Risskov DK-8240, Denmark
| | | | - Elysian Chow
- UCD School of Biomolecular and Biomedical Science, UCD Conway Institute, University College Dublin, Dublin 4, Ireland
| | - Kieran Wynne
- Proteomic Core Facility, UCD Conway Institute, School
of Medicine and Medical Sciences, University College Dublin, Dublin 4, Ireland
| | - Zohreh Farsi
- Department of Neurobiology, Max-Planck-Institute for Biophysical Chemistry, 37077 Göttingen, Germany
| | | | - Gudrun Ahnert-Hilger
- Institute of Integrative Neuroanatomy, Charité University Medicine Berlin, 10117 Berlin, Germany
| |
Collapse
|
28
|
Robson MJ, Quinlan MA, Blakely RD. Immune System Activation and Depression: Roles of Serotonin in the Central Nervous System and Periphery. ACS Chem Neurosci 2017; 8:932-942. [PMID: 28345868 DOI: 10.1021/acschemneuro.6b00412] [Citation(s) in RCA: 63] [Impact Index Per Article: 7.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/18/2022] Open
Abstract
Serotonin (5-hydroxytryptamine, 5-HT) has long been recognized as a key contributor to the regulation of mood and anxiety and is strongly associated with the etiology of major depressive disorder (MDD). Although more known for its roles within the central nervous system (CNS), 5-HT is recognized to modulate several key aspects of immune system function that may contribute to the development of MDD. Copious amounts of research have outlined a connection between alterations in immune system function, inflammation status, and MDD. Supporting this connection, peripheral immune activation results in changes in the function and/or expression of many components of 5-HT signaling that are associated with depressive-like phenotypes. How 5-HT is utilized by the immune system to effect CNS function and ultimately behaviors related to depression is still not well understood. This Review summarizes the evidence that immune system alterations related to depression affect CNS 5-HT signaling that can alter MDD-relevant behaviors and that 5-HT regulates immune system signaling within the CNS and periphery. We suggest that targeting the interrelationships between immune and 5-HT signaling may provide more effective treatments for subsets of those suffering from inflammation-associated MDD.
Collapse
Affiliation(s)
- Matthew J Robson
- Department of Biomedical Science, Charles E. Schmidt College of Medicine, Florida Atlantic University , Jupiter, Florida 33458, United States
| | - Meagan A Quinlan
- Department of Biomedical Science, Charles E. Schmidt College of Medicine, Florida Atlantic University , Jupiter, Florida 33458, United States
- Department of Pharmacology, Vanderbilt University , Nashville, Tennessee 37240-7933, United States
| | - Randy D Blakely
- Department of Biomedical Science, Charles E. Schmidt College of Medicine, Florida Atlantic University , Jupiter, Florida 33458, United States
| |
Collapse
|
29
|
Bermingham DP, Blakely RD. Kinase-dependent Regulation of Monoamine Neurotransmitter Transporters. Pharmacol Rev 2016; 68:888-953. [PMID: 27591044 PMCID: PMC5050440 DOI: 10.1124/pr.115.012260] [Citation(s) in RCA: 73] [Impact Index Per Article: 8.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023] Open
Abstract
Modulation of neurotransmission by the monoamines dopamine (DA), norepinephrine (NE), and serotonin (5-HT) is critical for normal nervous system function. Precise temporal and spatial control of this signaling in mediated in large part by the actions of monoamine transporters (DAT, NET, and SERT, respectively). These transporters act to recapture their respective neurotransmitters after release, and disruption of clearance and reuptake has significant effects on physiology and behavior and has been linked to a number of neuropsychiatric disorders. To ensure adequate and dynamic control of these transporters, multiple modes of control have evolved to regulate their activity and trafficking. Central to many of these modes of control are the actions of protein kinases, whose actions can be direct or indirectly mediated by kinase-modulated protein interactions. Here, we summarize the current state of our understanding of how protein kinases regulate monoamine transporters through changes in activity, trafficking, phosphorylation state, and interacting partners. We highlight genetic, biochemical, and pharmacological evidence for kinase-linked control of DAT, NET, and SERT and, where applicable, provide evidence for endogenous activators of these pathways. We hope our discussion can lead to a more nuanced and integrated understanding of how neurotransmitter transporters are controlled and may contribute to disorders that feature perturbed monoamine signaling, with an ultimate goal of developing better therapeutic strategies.
Collapse
Affiliation(s)
- Daniel P Bermingham
- Department of Pharmacology (D.P.B., R.D.B.) and Psychiatry (R.D.B.), Vanderbilt University Medical Center, Nashville, Tennessee; and Department of Biomedical Sciences, Charles E. Schmidt College of Medicine and Brain Institute, Florida Atlantic University, Jupiter, Florida (R.D.B.)
| | - Randy D Blakely
- Department of Pharmacology (D.P.B., R.D.B.) and Psychiatry (R.D.B.), Vanderbilt University Medical Center, Nashville, Tennessee; and Department of Biomedical Sciences, Charles E. Schmidt College of Medicine and Brain Institute, Florida Atlantic University, Jupiter, Florida (R.D.B.)
| |
Collapse
|
30
|
Schwendt M, Olive MF. Protein kinase Cɛ activity regulates mGluR5 surface expression in the rat nucleus accumbens. J Neurosci Res 2016; 95:1079-1090. [PMID: 27546836 DOI: 10.1002/jnr.23868] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/29/2016] [Revised: 06/29/2016] [Accepted: 07/12/2016] [Indexed: 12/11/2022]
Abstract
Type 5 metabotropic glutamate receptors (mGluR5) activate protein kinase C (PKC) via coupling to Gαq/11 protein signaling. We have previously demonstrated that the epsilon isoform of PKC (PKCɛ) is a critical downstream target of mGluR5 in regulating behavioral and biochemical responses to alcohol. Recent evidence suggests that PKC-mediated phosphorylation of mGluR5 can lead to receptor desensitization and internalization. We therefore sought to examine the specific involvement of PKCɛ in the regulation of mGluR5 surface expression in the nucleus accumbens (NAc), a key regulator of alcohol-associated behaviors. Coronal brain sections from male Wistar rats were analyzed for either colocalization of mGluR5 and PKCɛ via immunohistochemistry or changes in mGluR5 surface expression and PKCɛ phosphorylation following local application of PKCɛ translocation activator or inhibitor peptides and/or an orthosteric mGluR5 agonist. We observed colocalization of mGluR5 and PKCɛ in the NAc. We also showed that intra-NAc infusion of the PKCɛ translocation inhibitor ɛV1-2 increased mGluR5 surface expression under baseline conditions. Stimulation of mGluR5 with an orthosteric agonist DHPG, dose dependently increased ERK1/2 and PKCɛ phosphorylation as well as mGluR5 internalization in acute NAc slices. Finally, we observed that activation of PKCɛ translocation with Tat-ΨɛRACK peptide mediates agonist-independent mGluR5 internalization, whereas PKCɛ translocation inhibitor ɛV1-2 prevents agonist-dependent internalization of mGluR5 in NAc slice preparations. These findings suggest that the subcellular localization of mGluR5 in the NAc is regulated by PKCɛ under basal and stimulation conditions, which may influence the role of mGluR5-PKCɛ signaling in alcohol-related behaviors. © 2016 Wiley Periodicals, Inc.
Collapse
Affiliation(s)
- Marek Schwendt
- Psychology Department, University of Florida, Gainesville, Florida
| | - M Foster Olive
- Psychology Department, Arizona State University, Tempe, Arizona
| |
Collapse
|
31
|
Physical and functional interactions between the serotonin transporter and the neutral amino acid transporter ASCT2. Biochem J 2016; 473:1953-65. [DOI: 10.1042/bcj20160315] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/08/2016] [Accepted: 05/03/2016] [Indexed: 01/23/2023]
Abstract
The activity of serotonergic systems depends on the reuptake of extracellular serotonin via its plasma membrane serotonin [5-HT (5-hydroxytryptamine)] transporter (SERT), a member of the Na+/Cl−-dependent solute carrier 6 family. SERT is finely regulated by multiple molecular mechanisms including its physical interaction with intracellular proteins. The majority of previously identified SERT partners that control its functional activity are soluble proteins, which bind to its intracellular domains. SERT also interacts with transmembrane proteins, but its association with other plasma membrane transporters remains to be established. Using a proteomics strategy, we show that SERT associates with ASCT2 (alanine–serine–cysteine–threonine 2), a member of the solute carrier 1 family co-expressed with SERT in serotonergic neurons and involved in the transport of small neutral amino acids across the plasma membrane. Co-expression of ASCT2 with SERT in HEK (human embryonic kidney)-293 cells affects glycosylation and cell-surface localization of SERT with a concomitant reduction in its 5-HT uptake activity. Conversely, depletion of cellular ASCT2 by RNAi enhances 5-HT uptake in both HEK-293 cells and primary cultured mesencephalon neurons. Mimicking the effect of ASCT2 down-regulation, treatment of HEK-293 cells and neurons with the ASCT2 inhibitor D-threonine also increases 5-HT uptake. Moreover, D-threonine does not enhance further the maximal velocity of 5-HT uptake in cells depleted of ASCT2. Collectively, these findings provide evidence for a complex assembly involving SERT and a member of another solute carrier family, which strongly influences the subcellular distribution of SERT and the reuptake of 5-HT.
Collapse
|
32
|
Schwamborn R, Brown E, Haase J. Elevation of cortical serotonin transporter activity upon peripheral immune challenge is regulated independently of p38 mitogen-activated protein kinase activation and transporter phosphorylation. J Neurochem 2016; 137:423-35. [DOI: 10.1111/jnc.13596] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/09/2015] [Revised: 02/12/2016] [Accepted: 02/23/2016] [Indexed: 12/27/2022]
Affiliation(s)
- Robert Schwamborn
- UCD School of Biomolecular and Biomedical Science; UCD Conway Institute; University College Dublin; Dublin Ireland
| | - Eric Brown
- UCD School of Biomolecular and Biomedical Science; UCD Conway Institute; University College Dublin; Dublin Ireland
| | - Jana Haase
- UCD School of Biomolecular and Biomedical Science; UCD Conway Institute; University College Dublin; Dublin Ireland
| |
Collapse
|
33
|
Ye R, Quinlan MA, Iwamoto H, Wu HH, Green NH, Jetter CS, McMahon DG, Veestra-VanderWeele J, Levitt P, Blakely RD. Physical Interactions and Functional Relationships of Neuroligin 2 and Midbrain Serotonin Transporters. Front Synaptic Neurosci 2016; 7:20. [PMID: 26793096 PMCID: PMC4707279 DOI: 10.3389/fnsyn.2015.00020] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/10/2015] [Accepted: 12/14/2015] [Indexed: 12/31/2022] Open
Abstract
The neurotransmitter serotonin [5-hydroxytryptamine (5-HT)] modulates many key brain functions including those subserving sensation, emotion, reward, and cognition. Efficient clearance of 5-HT after release is achieved by the antidepressant-sensitive 5-HT transporter (SERT, SLC6A4). To identify novel SERT regulators, we pursued a proteomic analysis of mouse midbrain SERT complexes, evaluating findings in the context of prior studies that established a SERT-linked transcriptome. Remarkably, both efforts converged on a relationship of SERT with the synaptic adhesion protein neuroligin 2 (NLGN2), a post-synaptic partner for presynaptic neurexins, and a protein well-known to organize inhibitory GABAergic synapses. Western blots of midbrain reciprocal immunoprecipitations confirmed SERT/NLGN2 associations, and also extended to other NLGN2 associated proteins [e.g., α-neurexin (NRXN), gephyrin]. Midbrain SERT/NLGN2 interactions were found to be Ca(2+)-independent, supporting cis vs. trans-synaptic interactions, and were absent in hippocampal preparations, consistent with interactions arising in somatodendritic compartments. Dual color in situ hybridization confirmed co-expression of Tph2 and Nlgn2 mRNA in the dorsal raphe, with immunocytochemical studies confirming SERT:NLGN2 co-localization in raphe cell bodies but not axons. Consistent with correlative mRNA expression studies, loss of NLGN2 expression in Nlgn2 null mice produced significant reductions in midbrain and hippocampal SERT expression and function. Additionally, dorsal raphe 5-HT neurons from Nlgn2 null mice exhibit reduced excitability, a loss of GABAA receptor-mediated IPSCs, and increased 5-HT1A autoreceptor sensitivity. Finally, Nlgn2 null mice display significant changes in behaviors known to be responsive to SERT and/or 5-HT receptor manipulations. We discuss our findings in relation to the possible coordination of intrinsic and extrinsic regulation afforded by somatodendritic SERT:NLGN2 complexes.
Collapse
Affiliation(s)
- Ran Ye
- Department of Pharmacology, Vanderbilt University School of Medicine, Nashville TN, USA
| | - Meagan A Quinlan
- Department of Pharmacology, Vanderbilt University School of Medicine, Nashville TN, USA
| | - Hideki Iwamoto
- Department of Pharmacology, Vanderbilt University School of Medicine, Nashville TN, USA
| | - Hsiao-Huei Wu
- Department of Psychiatry, Vanderbilt University School of Medicine, Nashville TN, USA
| | - Noah H Green
- Department of Biological Sciences, Vanderbilt University School of Medicine, Nashville TN, USA
| | - Christopher S Jetter
- Department of Psychiatry, Vanderbilt University School of Medicine, Nashville TN, USA
| | - Douglas G McMahon
- Department of Pharmacology, Vanderbilt University School of Medicine, NashvilleTN, USA; Department of Biological Sciences, Vanderbilt University School of Medicine, NashvilleTN, USA
| | - Jeremy Veestra-VanderWeele
- Department of Psychiatry, NYS Psychiatric Institute, Columbia University Medical Center, New York NY, USA
| | - Pat Levitt
- Department of Psychiatry, Vanderbilt University School of Medicine, Nashville TN, USA
| | - Randy D Blakely
- Department of Pharmacology, Vanderbilt University School of Medicine, NashvilleTN, USA; Department of Psychiatry, Vanderbilt University School of Medicine, NashvilleTN, USA
| |
Collapse
|
34
|
Baganz NL, Lindler KM, Zhu CB, Smith JT, Robson MJ, Iwamoto H, Deneris ES, Hewlett WA, Blakely RD. A requirement of serotonergic p38α mitogen-activated protein kinase for peripheral immune system activation of CNS serotonin uptake and serotonin-linked behaviors. Transl Psychiatry 2015; 5:e671. [PMID: 26529424 PMCID: PMC5068761 DOI: 10.1038/tp.2015.168] [Citation(s) in RCA: 21] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/17/2015] [Accepted: 09/19/2015] [Indexed: 12/22/2022] Open
Abstract
Alterations in central serotonin (5-hydroxytryptamine, 5-HT) neurotransmission and peripheral immune activation have been linked to multiple neuropsychiatric disorders, including depression, schizophrenia and autism. The antidepressant-sensitive 5-HT transporter (SERT, SLC6A4), a critical determinant of synaptic 5-HT inactivation, can be regulated by pro-inflammatory cytokine signaling. Systemic innate immune system activation via intraperitoneal lipopolysaccharide (LPS) injection rapidly elevates brain SERT activity and 5-HT clearance. Moreover, the pro-inflammatory cytokine interleukin (IL)-1β rapidly stimulates SERT activity in raphe nerve terminal preparations ex vivo, effects that are attenuated by pharmacological p38 MAPK inhibition. To establish a role of serotonergic p38α MAPK signaling in LPS/IL-1β-induced SERT regulation and attendant behavioral responses, we pursued studies in mice that afford conditional elimination of p38α MAPK in 5-HT neurons (p38α(5HT-)). We found p38α(5HT-) and control (p38α(5HT+)) littermates to be indistinguishable in viability and growth and to express equivalent levels of SERT protein and synaptosomal 5-HT transport activity. Consistent with pharmacological studies, however, IL-1β fails to increase SERT activity in midbrain synaptosomes prepared from p38α(5HT-) animals. Moreover, although LPS elevated plasma corticosterone and central/peripheral pro-inflammatory cytokines in p38α(5HT-) animals, elevations in midbrain SERT activity were absent nor were changes in depressive and anxiety-like behaviors observed. Our studies support an obligate role of p38α MAPK signaling in 5-HT neurons for the translation of immune activation to SERT regulation and 5-HT-modulated behaviors.
Collapse
Affiliation(s)
- N L Baganz
- Department of Pharmacology, Vanderbilt University School of Medicine, Nashville, TN, USA
| | - K M Lindler
- Department of Pharmacology, Vanderbilt University School of Medicine, Nashville, TN, USA
| | - C B Zhu
- Osher Center for Integrative Medicine, Vanderbilt University School of Medicine, Nashville, TN, USA
| | - J T Smith
- Department of Pharmacology, Vanderbilt University School of Medicine, Nashville, TN, USA
| | - M J Robson
- Department of Pharmacology, Vanderbilt University School of Medicine, Nashville, TN, USA
| | - H Iwamoto
- Department of Pharmacology, Vanderbilt University School of Medicine, Nashville, TN, USA
| | - E S Deneris
- Department of Neuroscience, Case Western Reserve University, Cleveland, OH, USA
| | - W A Hewlett
- Institute for Psychiatric Neuroscience, Nashville, TN, USA
| | - R D Blakely
- Department of Pharmacology, Vanderbilt University School of Medicine, Nashville, TN, USA
- Department of Psychiatry, Vanderbilt University School of Medicine, Nashville, TN, USA
| |
Collapse
|
35
|
Moritz AE, Rastedt DE, Stanislowski DJ, Shetty M, Smith MA, Vaughan RA, Foster JD. Reciprocal Phosphorylation and Palmitoylation Control Dopamine Transporter Kinetics. J Biol Chem 2015; 290:29095-105. [PMID: 26424792 DOI: 10.1074/jbc.m115.667055] [Citation(s) in RCA: 47] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/21/2015] [Indexed: 11/06/2022] Open
Abstract
The dopamine transporter is a neuronal protein that drives the presynaptic reuptake of dopamine (DA) and is the major determinant of transmitter availability in the brain. Dopamine transporter function is regulated by protein kinase C (PKC) and other signaling pathways through mechanisms that are complex and poorly understood. Here we investigate the role of Ser-7 phosphorylation and Cys-580 palmitoylation in mediating steady-state transport kinetics and PKC-stimulated transport down-regulation. Using both mutational and pharmacological approaches, we demonstrate that these post-translational modifications are reciprocally regulated, leading to transporter populations that display high phosphorylation-low palmitoylation or low phosphorylation-high palmitoylation. The balance between the modifications dictates transport capacity, as conditions that promote high phosphorylation or low palmitoylation reduce transport Vmax and enhance PKC-stimulated down-regulation, whereas conditions that promote low phosphorylation or high palmitoylation increase transport Vmax and suppress PKC-stimulated down-regulation. Transitions between these functional states occur when endocytosis is blocked or undetectable, indicating that the modifications kinetically regulate the velocity of surface transporters. These findings reveal a novel mechanism for control of DA reuptake that may represent a point of dysregulation in DA imbalance disorders.
Collapse
Affiliation(s)
- Amy E Moritz
- From the Department of Basic Sciences, University of North Dakota School of Medicine and Health Sciences, Grand Forks, North Dakota 58202-9061
| | - Danielle E Rastedt
- From the Department of Basic Sciences, University of North Dakota School of Medicine and Health Sciences, Grand Forks, North Dakota 58202-9061
| | - Daniel J Stanislowski
- From the Department of Basic Sciences, University of North Dakota School of Medicine and Health Sciences, Grand Forks, North Dakota 58202-9061
| | - Madhur Shetty
- From the Department of Basic Sciences, University of North Dakota School of Medicine and Health Sciences, Grand Forks, North Dakota 58202-9061
| | - Margaret A Smith
- From the Department of Basic Sciences, University of North Dakota School of Medicine and Health Sciences, Grand Forks, North Dakota 58202-9061
| | - Roxanne A Vaughan
- From the Department of Basic Sciences, University of North Dakota School of Medicine and Health Sciences, Grand Forks, North Dakota 58202-9061
| | - James D Foster
- From the Department of Basic Sciences, University of North Dakota School of Medicine and Health Sciences, Grand Forks, North Dakota 58202-9061
| |
Collapse
|
36
|
Raiteri L, Raiteri M. Multiple functions of neuronal plasma membrane neurotransmitter transporters. Prog Neurobiol 2015; 134:1-16. [PMID: 26300320 DOI: 10.1016/j.pneurobio.2015.08.002] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/01/2015] [Revised: 07/09/2015] [Accepted: 08/18/2015] [Indexed: 12/11/2022]
Abstract
Removal from receptors of neurotransmitters just released into synapses is one of the major steps in neurotransmission. Transporters situated on the plasma membrane of nerve endings and glial cells perform the process of neurotransmitter (re)uptake. Because the density of transporters in the membranes can fluctuate, transporters can determine the transmitter concentrations at receptors, thus modulating indirectly the excitability of neighboring neurons. Evidence is accumulating that neurotransmitter transporters can exhibit multiple functions. Being bidirectional, neurotransmitter transporters can mediate transmitter release by working in reverse, most often under pathological conditions that cause ionic gradient dysregulations. Some transporters reverse to release transmitters, like dopamine or serotonin, when activated by 'indirectly acting' substrates, like the amphetamines. Some transporters exhibit as one major function the ability to capture transmitters into nerve terminals that perform insufficient synthesis. Transporter activation can generate conductances that regulate directly neuronal excitability. Synaptic and non-synaptic transporters play different roles. Cytosolic Na(+) elevations accompanying transport can interact with plasmalemmal or/and mitochondrial Na(+)/Ca(2+) exchangers thus generating calcium signals. Finally, neurotransmitter transporters can behave as receptors mediating releasing stimuli able to cause transmitter efflux through multiple mechanisms. Neurotransmitter transporters are therefore likely to play hitherto unknown roles in multiple therapeutic treatments.
Collapse
Affiliation(s)
- Luca Raiteri
- Department of Pharmacy, Pharmacology and Toxicology Section, University of Genoa, Genoa, Italy; Center of Excellence for Biomedical Research, University of Genoa, Genoa, Italy; National Institute of Neuroscience, Genoa, Italy
| | - Maurizio Raiteri
- Department of Pharmacy, Pharmacology and Toxicology Section, University of Genoa, Genoa, Italy; Center of Excellence for Biomedical Research, University of Genoa, Genoa, Italy; National Institute of Neuroscience, Genoa, Italy.
| |
Collapse
|
37
|
Spies M, Knudsen GM, Lanzenberger R, Kasper S. The serotonin transporter in psychiatric disorders: insights from PET imaging. Lancet Psychiatry 2015; 2:743-755. [PMID: 26249305 DOI: 10.1016/s2215-0366(15)00232-1] [Citation(s) in RCA: 93] [Impact Index Per Article: 9.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/04/2015] [Revised: 05/05/2015] [Accepted: 05/07/2015] [Indexed: 01/09/2023]
Abstract
Over the past 20 years, psychotropics affecting the serotonergic system have been used extensively in the treatment of psychiatric disorders. Molecular imaging, in particular PET, has allowed for elucidation of the essential contribution of the serotonin transporter to the pathophysiology of various psychiatric disorders and their treatment. We review studies that use PET to measure cerebral serotonin transporter activity in psychiatric disorders, focusing on major depressive disorder and antidepressant treatment. We also discuss opportunities and limitations in the application of this neuroimaging method in clinical practice. Although results from individual studies diverge, meta-analysis indicates a trend towards reduced serotonin transporter availability in patients with major depressive disorder. Inconsistencies in results might suggest symptom heterogeneity in major depressive disorder and might therefore be relevant for stratification of patients into clinical subsets. PET has enabled the elucidation of mechanisms of response to selective serotonin reuptake inhibitors (SSRIs) and hence provides a basis for rational pharmacological treatment of major depressive disorder. Such imaging studies have also suggested that the pattern of serotonin transporter binding before treatment might predict response to antidepressant treatment, which could potentially be clinically useful in the future. Additionally, this Review discusses PET studies investigating the serotonin transporter in anxiety, obsessive-compulsive disorder, and eating disorders. Few studies have shown changes in serotonin transporter activity in schizophrenia and attention deficit hyperactivity disorder. By showing the scarcity of data in these psychiatric disorders, we highlight the potential for further investigation in this field.
Collapse
Affiliation(s)
- Marie Spies
- Department of Psychiatry and Psychotherapy, Medical University of Vienna, Vienna, Austria
| | - Gitte M Knudsen
- Neurobiology Research Unit, Copenhagen University Hospital Rigshospitalet, Copenhagen, Denmark
| | - Rupert Lanzenberger
- Department of Psychiatry and Psychotherapy, Medical University of Vienna, Vienna, Austria
| | - Siegfried Kasper
- Department of Psychiatry and Psychotherapy, Medical University of Vienna, Vienna, Austria.
| |
Collapse
|
38
|
Nazir S, Kumar A, Chatterjee I, Anbazhagan AN, Gujral T, Priyamvada S, Saksena S, Alrefai WA, Dudeja PK, Gill RK. Mechanisms of Intestinal Serotonin Transporter (SERT) Upregulation by TGF-β1 Induced Non-Smad Pathways. PLoS One 2015; 10:e0120447. [PMID: 25954931 PMCID: PMC4425666 DOI: 10.1371/journal.pone.0120447] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/22/2014] [Accepted: 01/22/2015] [Indexed: 01/01/2023] Open
Abstract
TGF-β1 is an important multifunctional cytokine with numerous protective effects on intestinal mucosa. The influence of TGF-β1 on serotonin transporter (SERT) activity, the critical mechanism regulating the extracellular availability of serotonin (5-HT), is not known. Current studies were designed to examine acute effects of TGF-β1 on SERT. Model human intestinal Caco-2 cells grown as monolayer’s or as cysts in 3D culture and ex vivo mouse model were utilized. Treatment of Caco-2 cells with TGF-β1 (10 ng/ml, 60 min) stimulated SERT activity (~2 fold, P<0.005). This stimulation of SERT function was dependent upon activation of TGF-β1 receptor (TGFRI) as SB-431542, a specific TGF-βRI inhibitor blocked the SERT stimulation. SERT activation in response to TGF-β1 was attenuated by inhibition of PI3K and occurred via enhanced recruitment of SERT-GFP to apical surface in a PI3K dependent manner. The exocytosis inhibitor brefeldin A (2.5 μM) attenuated the TGF-β1-mediated increase in SERT function. TGF-β1 increased the association of SERT with the soluble N-ethylmaleimide-sensitive factor attachment protein receptor (SNARE) syntaxin 3 (STX3) and promoted exocytosis of SERT. Caco-2 cells grown as cysts in 3D culture recapitulated the effects of TGF-β1 showing increased luminal staining of SERT. Ussing chamber studies revealed increase in 3H-5-HT uptake in mouse ileum treated ex vivo with TGF-β1 (10 ng/ml, 1h). These data demonstrate a novel mechanism rapidly regulating intestinal SERT via PI3K and STX3. Since decreased SERT is implicated in various gastro-intestinal disorders e.g IBD, IBS and diarrhea, understanding mechanisms stimulating SERT function by TGF-β1 offers a novel therapeutic strategy to treat GI disorders.
Collapse
Affiliation(s)
- Saad Nazir
- Division of Gastroenterology & Hepatology, Dept. of Medicine, University of Illinois at Chicago, Chicago, Illinois, United States of America
| | - Anoop Kumar
- Division of Gastroenterology & Hepatology, Dept. of Medicine, University of Illinois at Chicago, Chicago, Illinois, United States of America
| | - Ishita Chatterjee
- Division of Gastroenterology & Hepatology, Dept. of Medicine, University of Illinois at Chicago, Chicago, Illinois, United States of America
| | - Arivarasu N. Anbazhagan
- Division of Gastroenterology & Hepatology, Dept. of Medicine, University of Illinois at Chicago, Chicago, Illinois, United States of America
| | - Tarunmeet Gujral
- Division of Gastroenterology & Hepatology, Dept. of Medicine, University of Illinois at Chicago, Chicago, Illinois, United States of America
| | - Shubha Priyamvada
- Division of Gastroenterology & Hepatology, Dept. of Medicine, University of Illinois at Chicago, Chicago, Illinois, United States of America
| | - Seema Saksena
- Division of Gastroenterology & Hepatology, Dept. of Medicine, University of Illinois at Chicago, Chicago, Illinois, United States of America
| | - Waddah A. Alrefai
- Division of Gastroenterology & Hepatology, Dept. of Medicine, University of Illinois at Chicago, Chicago, Illinois, United States of America
- Jesse Brown VA Medical Center, Chicago, Illinois, United States of America
| | - Pradeep K. Dudeja
- Division of Gastroenterology & Hepatology, Dept. of Medicine, University of Illinois at Chicago, Chicago, Illinois, United States of America
- Jesse Brown VA Medical Center, Chicago, Illinois, United States of America
| | - Ravinder K. Gill
- Division of Gastroenterology & Hepatology, Dept. of Medicine, University of Illinois at Chicago, Chicago, Illinois, United States of America
- * E-mail:
| |
Collapse
|
39
|
Maximino C, Gemaque J, Benzecry R, Lima MG, Batista EDJO, Picanço-Diniz DW, Oliveira KRM, Herculano AM. Role of nitric oxide in the behavioral and neurochemical effects of IB-MECA in zebrafish. Psychopharmacology (Berl) 2015; 232:1671-80. [PMID: 25388291 DOI: 10.1007/s00213-014-3799-4] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/20/2014] [Accepted: 10/31/2014] [Indexed: 11/29/2022]
Abstract
RATIONALE The adenosine A3 receptor and the nitric oxide (NO) pathway regulate the function and localization of serotonin transporters (SERTs). These transporters regulate extracellular serotonin levels, which are correlated with defensive behavior. OBJECTIVE The purpose of this study was to understand the role of the A3AR on anxiety and arousal models in zebrafish, and whether this role is mediated by the nitrergic modulation of serotonin uptake. METHODS The effects of IB-MECA (0.01 and 0.1 mg/kg) were assessed in a series of behavioral tasks in adult zebrafish, as well as on extracellular serotonin levels in vivo and serotonin uptake in brain homogenates. Finally, the interaction between IB-MECA and drugs blocking voltage-dependent calcium channels (VDCCs), NO synthase, and SERT was analyzed. RESULTS At the lowest dose, IB-MECA decreased bottom dwelling and scototaxis, while at the highest dose, it also decreased shoaling, startle probability, and melanophore responses. These effects were accompanied by an increase in brain extracellular serotonin levels. IB-MECA also concentration-dependently increased serotonin uptake in vitro. The effects of IB-MECA on extracellular 5-HT, scototaxis, and geotaxis were blocked by L-NAME, while only the effects on 5-HT and scototaxis were blocked by verapamil. In vitro, the increase in 5-HT uptake was dependent on VDCCs and NO. Finally, fluoxetine blocked the effect of IB-MECA on scototaxis, but not geotaxis. CONCLUSION These results suggest that the effect of IB-MECA on scototaxis are mediated by a VDCC-NO-SERT pathway. While NO seems to mediate the effects of IB-MECA on geotaxis, neither VDCCs nor SERT seems to be involved in this process.
Collapse
Affiliation(s)
- Caio Maximino
- Laboratório de Neurociências e Comportamento, Universidade do Estado do Pará, Departamento de Morfologia e Ciências Fisiológicas, Núcleo Universitário de Marabá, Marabá, PA, Brazil,
| | | | | | | | | | | | | | | |
Collapse
|
40
|
Baldinger P, Kraus C, Rami-Mark C, Gryglewski G, Kranz GS, Haeusler D, Hahn A, Spies M, Wadsak W, Mitterhauser M, Rujescu D, Kasper S, Lanzenberger R. Interaction between 5-HTTLPR and 5-HT1B genotype status enhances cerebral 5-HT1A receptor binding. Neuroimage 2015; 111:505-12. [PMID: 25652393 DOI: 10.1016/j.neuroimage.2015.01.049] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/03/2014] [Revised: 01/18/2015] [Accepted: 01/26/2015] [Indexed: 10/24/2022] Open
Abstract
Serotonergic neurotransmission is thought to underlie a dynamic interrelation between different key structures of the serotonin system. The serotonin transporter (SERT), which is responsible for the reuptake of serotonin from the synaptic cleft into the neuron, as well as the serotonin-1A (5-HT1A) and -1B (5-HT1B) receptors, inhibitory auto-receptors in the raphe region and projection areas, respectively, are likely to determine serotonin release. Thereby, they are involved in the regulation of extracellular serotonin concentrations and the extent of serotonergic effects in respective projection areas. Complex receptor interactions can be assessed in vivo with positron emission tomography (PET) and single-nucleotide-polymorphisms, which are thought to alter protein expression levels. Due to the complexity of the serotonergic system, gene × gene interactions are likely to regulate transporter and receptor expression and therefore subsequently serotonergic transmission. In this context, we measured 51 healthy subjects (mean age 45.5 ± 12.9, 38 female) with PET using [carbonyl-(11)C]WAY-100635 to determine 5-HT1A receptor binding potential (5-HT1A BPND). Genotyping for rs6296 (HTR1B) and 5-HTTLPR (SERT gene promoter polymorphism) was performed using DNA isolated from whole blood. Voxel-wise whole-brain ANOVA revealed a positive interaction effect of genotype groups (5-HTTLPR: LL, LS+SS and HTR1B: rs6296: CC, GC+GG) on 5-HT1A BPND with peak t-values in the bilateral parahippocampal gyrus. More specifically, highest 5-HT1A BPND was identified for individuals homozygous for both the L-allele of 5-HTTLPR and the C-allele of rs6296. This finding suggests that the interaction between two major serotonergic structures involved in serotonin release, specifically the SERT and 5-HT1B receptor, results in a modification of the inhibitory serotonergic tone mediated via 5-HT1A receptors.
Collapse
Affiliation(s)
- Pia Baldinger
- Department of Psychiatry and Psychotherapy, Division of Biological Psychiatry, Medical University of Vienna, Austria
| | - Christoph Kraus
- Department of Psychiatry and Psychotherapy, Division of Biological Psychiatry, Medical University of Vienna, Austria
| | - Christina Rami-Mark
- Department of Biomedical Imaging und Image-guided Therapy, Division of Nuclear Medicine, Medical University of Vienna, Austria
| | - Gregor Gryglewski
- Department of Psychiatry and Psychotherapy, Division of Biological Psychiatry, Medical University of Vienna, Austria
| | - Georg S Kranz
- Department of Psychiatry and Psychotherapy, Division of Biological Psychiatry, Medical University of Vienna, Austria
| | - Daniela Haeusler
- Department of Biomedical Imaging und Image-guided Therapy, Division of Nuclear Medicine, Medical University of Vienna, Austria
| | - Andreas Hahn
- Department of Psychiatry and Psychotherapy, Division of Biological Psychiatry, Medical University of Vienna, Austria
| | - Marie Spies
- Department of Psychiatry and Psychotherapy, Division of Biological Psychiatry, Medical University of Vienna, Austria
| | - Wolfgang Wadsak
- Department of Biomedical Imaging und Image-guided Therapy, Division of Nuclear Medicine, Medical University of Vienna, Austria
| | - Markus Mitterhauser
- Department of Biomedical Imaging und Image-guided Therapy, Division of Nuclear Medicine, Medical University of Vienna, Austria
| | - Dan Rujescu
- Genetics Research Center, Department of Psychiatry and Psychotherapy, Ludwig-Maximilians-University Munich, Germany; Department of Psychiatry, Medical University of Halle, Germany
| | - Siegfried Kasper
- Department of Psychiatry and Psychotherapy, Division of Biological Psychiatry, Medical University of Vienna, Austria
| | - Rupert Lanzenberger
- Department of Psychiatry and Psychotherapy, Division of Biological Psychiatry, Medical University of Vienna, Austria.
| |
Collapse
|
41
|
Zhao R, Wang S, Huang Z, Zhang L, Yang X, Bai X, Zhou D, Qin Z, Du G. Lipopolysaccharide-induced serotonin transporter up-regulation involves PKG-I and p38MAPK activation partially through A3 adenosine receptor. Biosci Trends 2015; 9:367-76. [DOI: 10.5582/bst.2015.01168] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/05/2022]
Affiliation(s)
- Rui Zhao
- Beijing Key Laboratory of Drug Target and Screening Research, Institute of Materia Medica, Chinese Academy of MedicalSciences & Peking Union Medical College
| | - Shoubao Wang
- Beijing Key Laboratory of Drug Target and Screening Research, Institute of Materia Medica, Chinese Academy of MedicalSciences & Peking Union Medical College
| | | | - Li Zhang
- Beijing Key Laboratory of Drug Target and Screening Research, Institute of Materia Medica, Chinese Academy of MedicalSciences & Peking Union Medical College
| | - Xiuying Yang
- Beijing Key Laboratory of Drug Target and Screening Research, Institute of Materia Medica, Chinese Academy of MedicalSciences & Peking Union Medical College
| | - Xiaoyu Bai
- Beijing Key Laboratory of Drug Target and Screening Research, Institute of Materia Medica, Chinese Academy of MedicalSciences & Peking Union Medical College
| | - Dan Zhou
- Beijing Key Laboratory of Drug Target and Screening Research, Institute of Materia Medica, Chinese Academy of MedicalSciences & Peking Union Medical College
| | - Zhizhen Qin
- Beijing Key Laboratory of Drug Target and Screening Research, Institute of Materia Medica, Chinese Academy of MedicalSciences & Peking Union Medical College
| | - Guanhua Du
- Beijing Key Laboratory of Drug Target and Screening Research, Institute of Materia Medica, Chinese Academy of MedicalSciences & Peking Union Medical College
| |
Collapse
|
42
|
Integrating the monoamine, neurotrophin and cytokine hypotheses of depression--a central role for the serotonin transporter? Pharmacol Ther 2014; 147:1-11. [PMID: 25444754 DOI: 10.1016/j.pharmthera.2014.10.002] [Citation(s) in RCA: 115] [Impact Index Per Article: 10.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/20/2014] [Accepted: 10/20/2014] [Indexed: 01/01/2023]
Abstract
Monoamine, in particular serotonergic neurotransmission has long been recognized as an important factor in the aetiology of depression. The serotonin transporter (SERT) is the primary regulator of serotonin levels in the brain and a key target for widely used antidepressant drugs, such as selective serotonin reuptake inhibitors (SSRIs). In realising the limitations of current antidepressant therapy, depression research has branched out to encompass other areas such as synaptic plasticity, neurogenesis and brain structural remodelling as factors which influence mood and behaviour. More recently, the immune system has been implicated in the development of depression and various intriguing observations have inspired the cytokine hypothesis of depression. Over the past two decades evidence of in vitro and in vivo regulation of SERT function by pro-inflammatory cytokines as well as by mechanisms of synaptic plasticity has been accumulating, offering a mechanistic link between the monoamine, neurotrophin and cytokine theories of depression. This review will focus firstly on the interconnected roles of serotonin and neurotrophins in depression and antidepressant therapy, secondly on the impact of the immune system on serotonin transporter regulation and neurotrophin signalling and finally we propose a model of reciprocal regulation of serotonin and neurotrophin signalling in the context of inflammation-induced depression.
Collapse
|
43
|
Nikolaus S, Hautzel H, Müller HW. Neurochemical dysfunction in treated and nontreated schizophrenia - a retrospective analysis of in vivo imaging studies. Rev Neurosci 2014; 25:25-96. [PMID: 24486731 DOI: 10.1515/revneuro-2013-0063] [Citation(s) in RCA: 21] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/10/2013] [Accepted: 12/26/2013] [Indexed: 12/22/2022]
Abstract
To evaluate the contribution of individual synaptic constituents, all available in vivo imaging studies on schizophrenic patients were subjected to a retrospective analysis. For the pool of drug-naïve, drug-free, and acutely medicated patients, major findings were increases in neostriatal dopamine (DA) synthesis and release and decreases in neostriatal DA transporters and D1 receptors, neostriatal, thalamic, frontal, and parietal D2 receptors, mesencephalic/pontine and temporal 5-HT1A receptors, frontal and temporal HT2A and μ-amino butyric acid (GABA)A receptors. Based on the findings on drug-naïve and drug-free patients, it may be hypothesized that schizophrenia initially is characterized by an impaired mechanism of D2 autoreceptor and heteroreceptor sensitization leading to sensitization instead of desensitization in response to increased levels of neostriatal DA. Neuroleptic medication blocks neostriatal D2 autoreceptor and heteroreceptors, reducing neostriatal DA and disinhibiting DA action mediated by D2 heteroreceptor binding sites. Ultimately, this may result in a restitution of GABA function, leading to a recovery of inhibitory input to the target regions of the descending corticothalamostriatal efferents. Furthermore, a blockade of inhibitory and excitatory neocortical 5-HT function may be inferred, which is likely to reduce (excitatory) DAergic input to the mesolimbic target regions of corticothalamostriatal projections.
Collapse
|
44
|
Müller HK, Kragballe M, Fjorback AW, Wiborg O. Differential regulation of the serotonin transporter by vesicle-associated membrane protein 2 in cells of neuronal versus non-neuronal origin. PLoS One 2014; 9:e97540. [PMID: 24878716 PMCID: PMC4039532 DOI: 10.1371/journal.pone.0097540] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/29/2014] [Accepted: 04/21/2014] [Indexed: 11/30/2022] Open
Abstract
The serotonin transporter (SERT) is a key regulator of serotonergic signalling as it mediates the re-uptake of synaptic serotonin into nerve terminals, thereby terminating or modulating its signal. It is well-known that SERT regulation is a dynamic process orchestrated by a wide array of proteins and mechanisms. However, molecular details on possible coordinated regulation of SERT activity and 5-HT release are incomplete. Here, we report that vesicle-associated membrane protein 2 (VAMP2), a SNARE protein that mediates vesicle fusion with the plasma membrane, interacts with SERT. This was documented in vitro, through GST pull-down assays, by co-immunoprecipitation experiments on heterologous cells and rat hippocampal synaptosomes, and with FRET analysis in live transfected HEK-293 MSR cells. The related isoforms VAMP1 and VAMP3 also physically interact with SERT. However, comparison of the three VAMP isoforms shows that only VAMP2 possesses a functionally distinct role in relation to SERT. VAMP2 influences 5-HT uptake, cell surface expression and the delivery rate of SERT to the plasma membrane differentially in HEK-293 MSR and PC12 cells. Moreover, siRNA-mediated knock-down of endogenous VAMP2 reduces 5-HT uptake in CAD cells stably expressing low levels of heterologous SERT. Deletion and mutant analysis suggest a role for the isoform specific C-terminal domain of VAMP2 in regulating SERT function. Our data identify a novel interaction between SERT and a synaptic vesicle protein and support a link between 5-HT release and re-uptake.
Collapse
Affiliation(s)
- Heidi Kaastrup Müller
- Translational Neuropsychiatry Unit, Department of Clinical Medicine, Aarhus University Hospital, Risskov, Denmark
| | - Marie Kragballe
- Translational Neuropsychiatry Unit, Department of Clinical Medicine, Aarhus University Hospital, Risskov, Denmark
| | - Anja Winther Fjorback
- Stereology and Electron Microscopy Laboratory, Centre for Stochastic Geometry and Advanced Bioimaging, Department of Clinical Medicine, University of Aarhus, Aarhus, Denmark
| | - Ove Wiborg
- Translational Neuropsychiatry Unit, Department of Clinical Medicine, Aarhus University Hospital, Risskov, Denmark
| |
Collapse
|
45
|
Benmansour S, Privratsky AA, Adeniji OS, Frazer A. Signaling mechanisms involved in the acute effects of estradiol on 5-HT clearance. Int J Neuropsychopharmacol 2014; 17:765-77. [PMID: 24423185 PMCID: PMC3969768 DOI: 10.1017/s146114571300165x] [Citation(s) in RCA: 29] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/07/2022] Open
Abstract
Estradiol was found previously to have an antidepressant-like effect and to block the ability of selective serotonin reuptake inhibitors (SSRIs) to have an antidepressant-like effect. The antidepressant-like effect of estradiol was due to estrogen receptor β (ERβ) and/or GPR30 activation, whereas estradiol's blockade of the effect of an SSRI was mediated by ERα. This study focuses on investigating signaling pathways as well as interacting receptors associated with these two effects of estradiol. In vivo chronoamperometry was used to measure serotonin transporter (SERT) function. The effect of local application of estradiol or selective agonists for ERα (PPT) or ERβ (DPN) into the CA3 region of the hippocampus of ovariectomized (OVX) rats on 5-hydroxytryptamine (5-HT) clearance as well as on the ability of fluvoxamine to slow 5-HT clearance was examined after selective blockade of signaling pathways or that of interacting receptors. Estradiol- or DPN-induced slowing of 5-HT clearance mediated by ERβ was blocked after inhibition of MAPK/ERK1/2 but not of PI3K/Akt signaling pathways. This effect also involved interactions with TrkB, and IGF-1 receptors. Estradiol's or PPT's inhibition of the fluvoxamine-induced slowing of 5-HT clearance mediated by ERα, was blocked after inhibition of either MAPK/ERK1/2 or PI3K/Akt signaling pathways. This effect involved interactions with the IGF-1 receptor and with the metabotropic glutamate receptor 1, but not with TrkB. This study illustrates some of the signaling pathways required for the effects of estradiol on SERT function, and particularly shows that ER subtypes elicit different as well as common signaling pathways for their actions.
Collapse
Affiliation(s)
- Saloua Benmansour
- Department of Pharmacology, University of Texas Health Science Center at San Antonio, Texas 78229
| | - Anthony A. Privratsky
- Department of Pharmacology, University of Texas Health Science Center at San Antonio, Texas 78229
| | - Opeyemi S. Adeniji
- Department of Pharmacology, University of Texas Health Science Center at San Antonio, Texas 78229
| | - Alan Frazer
- Department of Pharmacology, University of Texas Health Science Center at San Antonio, Texas 78229
- South Texas Veterans Health Care System, San Antonio, Texas 78284, USA
| |
Collapse
|
46
|
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
|
47
|
Ye R, Carneiro AMD, Han Q, Airey D, Sanders-Bush E, Zhang B, Lu L, Williams R, Blakely RD. Quantitative trait loci mapping and gene network analysis implicate protocadherin-15 as a determinant of brain serotonin transporter expression. GENES BRAIN AND BEHAVIOR 2014; 13:261-75. [PMID: 24405699 DOI: 10.1111/gbb.12119] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/03/2013] [Revised: 12/23/2013] [Accepted: 01/02/2014] [Indexed: 12/15/2022]
Abstract
Presynaptic serotonin (5-hydroxytryptamine, 5-HT) transporters (SERT) regulate 5-HT signaling via antidepressant-sensitive clearance of released neurotransmitter. Polymorphisms in the human SERT gene (SLC6A4) have been linked to risk for multiple neuropsychiatric disorders, including depression, obsessive-compulsive disorder and autism. Using BXD recombinant inbred mice, a genetic reference population that can support the discovery of novel determinants of complex traits, merging collective trait assessments with bioinformatics approaches, we examine phenotypic and molecular networks associated with SERT gene and protein expression. Correlational analyses revealed a network of genes that significantly associated with SERT mRNA levels. We quantified SERT protein expression levels and identified region- and gender-specific quantitative trait loci (QTLs), one of which associated with male midbrain SERT protein expression, centered on the protocadherin-15 gene (Pcdh15), overlapped with a QTL for midbrain 5-HT levels. Pcdh15 was also the only QTL-associated gene whose midbrain mRNA expression significantly associated with both SERT protein and 5-HT traits, suggesting an unrecognized role of the cell adhesion protein in the development or function of 5-HT neurons. To test this hypothesis, we assessed SERT protein and 5-HT traits in the Pcdh15 functional null line (Pcdh15(av-) (3J) ), studies that revealed a strong, negative influence of Pcdh15 on these phenotypes. Together, our findings illustrate the power of multidimensional profiling of recombinant inbred lines in the analysis of molecular networks that support synaptic signaling, and that, as in the case of Pcdh15, can reveal novel relationships that may underlie risk for mental illness.
Collapse
Affiliation(s)
- R Ye
- Department of Pharmacology, Vanderbilt University School of Medicine, Nashville, TN, USA
| | | | | | | | | | | | | | | | | |
Collapse
|
48
|
Ulloa RE, Díaz-Valderrama A, Herrera-Pérez J, León-Olea M, Martínez-Mota L. Age differences in the impact of forced swimming test on serotonin transporter levels in lateral septum and dorsal raphe. BEHAVIORAL AND BRAIN FUNCTIONS : BBF 2014; 10:3. [PMID: 24490994 PMCID: PMC3922148 DOI: 10.1186/1744-9081-10-3] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 08/08/2013] [Accepted: 01/28/2014] [Indexed: 12/19/2022]
Abstract
BACKGROUND Forced swimming test (FST) is an animal model which evaluates behavioral despair and the effect of antidepressants such as the selective serotonin reuptake inhibitors; the FST modifies the expression of some receptors related to antidepressant response, but it is not known whether serotonin transporter (SERT), their main target, is affected by this test in animals of different ages. Antidepressant response has shown age-dependent variations which could be associated with SERT expression. The aim of the present study was to analyze changes in the SERT immunoreactivity (SERT-IR) in dorsal raphe and lateral septum of male rats from different age groups with or without behavioral despair induced by their exposure to the FST, since these two structures are related to the expression of this behavior. METHODS Prepubertal (24 PN), pubertal (40 PN), young adult (3-5 months) and middle-aged (12 months) male rats were assigned to a control group (non-FST) or depressed group (FST, two sessions separated by 24 h). Changes in SERT-IR in dorsal raphe and lateral septum were determined with immunofluorescence. RESULTS Pubertal and middle-aged rats showed higher levels of immobility behavior compared to prepubertal rats on the FST. SERT-IR showed an age-dependent increase followed by a moderate decrease in middle-aged rats in both structures; a decreased in SERT-IR in lateral septum and dorsal raphe of pubertal rats was observed after the FST. CONCLUSIONS Age differences were observed in the SERT-IR of structures related to behavioral despair; SERT expression was modified by the FST in lateral septum and dorsal raphe of pubertal rats.
Collapse
Affiliation(s)
- Rosa-Elena Ulloa
- Hospital Psiquiátrico Infantil “Dr. Juan N Navarro”, San Buenaventura 86, Col. Belisario Domínguez, Delegación Tlalpan, Mexico City 14080, Mexico
| | - Aliyeri Díaz-Valderrama
- Instituto Nacional de Psiquiatría Ramón de la Fuente Muñiz, Calzada México-Xochimilco 101, Col. San Lorenzo Huipulco, Delegación Tlalpan, Mexico City 14370, Mexico
| | - Jaime Herrera-Pérez
- Instituto Nacional de Psiquiatría Ramón de la Fuente Muñiz, Calzada México-Xochimilco 101, Col. San Lorenzo Huipulco, Delegación Tlalpan, Mexico City 14370, Mexico
| | - Martha León-Olea
- Instituto Nacional de Psiquiatría Ramón de la Fuente Muñiz, Calzada México-Xochimilco 101, Col. San Lorenzo Huipulco, Delegación Tlalpan, Mexico City 14370, Mexico
| | - Lucía Martínez-Mota
- Instituto Nacional de Psiquiatría Ramón de la Fuente Muñiz, Calzada México-Xochimilco 101, Col. San Lorenzo Huipulco, Delegación Tlalpan, Mexico City 14370, Mexico
| |
Collapse
|
49
|
Hahn A, Haeusler D, Kraus C, Höflich AS, Kranz GS, Baldinger P, Savli M, Mitterhauser M, Wadsak W, Karanikas G, Kasper S, Lanzenberger R. Attenuated serotonin transporter association between dorsal raphe and ventral striatum in major depression. Hum Brain Mapp 2014; 35:3857-66. [PMID: 24443158 DOI: 10.1002/hbm.22442] [Citation(s) in RCA: 43] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/16/2013] [Revised: 10/21/2013] [Accepted: 11/25/2013] [Indexed: 11/06/2022] Open
Abstract
Suffering from anhedonia, patients with major depressive disorder (MDD) exhibit alterations in several parts of the serotonergic neurotransmitter system, which are in turn involved in reward processing. However, previous investigations of the serotonin transporter (SERT) focused on regional differences with varying results depending on the clinical syndrome. Here, we aimed to describe the serotonergic system of MDD patients on a network level by evaluating SERT associations across brain regions. Twenty medication free patients with major depression and 20 healthy controls underwent positron emission tomography using the radioligand [(11) C]DASB. SERT binding potentials (BPND ) were quantified voxel-wise with the multilinear reference tissue model 2. In addition, SERT BPND was extracted from the dorsal raphe nucleus (DRN) as an indicator of midbrain serotonergic neurotransmission. Whole-brain linear regression analysis was applied to evaluate the association of DRN SERT bindings to those in projection areas, which was followed by ANCOVA to assess differences in interregional relationships between patients and controls. Although both groups showed widespread positive correlations, group differences were restricted to decreased SERT associations between the DRN and the ventral striatum (right and left respectively: t=5.85, P<0.05 corrected and t=5.07, P<0.1 corrected) when comparing MDD patients (R(2)=0.11 and 0.24) to healthy subjects (R(2)=0.72 and 0.66, P<0.01 and 0.05 corrected). Adjusting for age and sex did not change these findings. This study indicates a disturbed regulation between key regions involved in reward processing via the SERT. Our interregional approach highlights the importance of evaluating pathophysiological alterations on a network level to gain complementary information in addition to regional investigations.
Collapse
Affiliation(s)
- Andreas Hahn
- Department of Psychiatry and Psychotherapy, Medical University of Vienna, Austria
| | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
50
|
The SLC6 transporters: perspectives on structure, functions, regulation, and models for transporter dysfunction. Pflugers Arch 2013; 466:25-42. [PMID: 24337881 DOI: 10.1007/s00424-013-1410-1] [Citation(s) in RCA: 121] [Impact Index Per Article: 10.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/16/2013] [Revised: 11/20/2013] [Accepted: 11/23/2013] [Indexed: 10/25/2022]
Abstract
The human SLC6 family is composed of approximately 20 structurally related symporters (co-transporters) that use the transmembrane electrochemical gradient to actively import their substrates into cells. Approximately half of the substrates of these transporters are amino acids, with others transporting biogenic amines and/or closely related compounds, such as nutrients and compatible osmolytes. In this short review, five leaders in the field discuss a number of currently important research themes that involve SLC6 transporters, highlighting the integrative role they play across a wide spectrum of different functions. The first essay, by Gary Rudnick, describes the molecular mechanism of their coupled transport which is being progressively better understood based on new crystal structures, functional studies, and modeling. Next, the question of multiple levels of transporter regulation is discussed by Reinhard Krämer, in the context of osmoregulation and stress response by the related bacterial betaine transporter BetP. The role of selected members of the human SLC6 family that function as nutrient amino acid transporters is then reviewed by François Verrey. He discusses how some of these transporters mediate the active uptake of (essential) amino acids into epithelial cells of the gut and the kidney tubule to support systemic amino acid requirements, whereas others are expressed in specific cells to support their specialized metabolism and/or growth. The most extensively studied members of the human SLC6 family are neurotransmitter reuptake transporters, many of which are important drug targets for the treatment of neuropsychiatric disorders. Randy Blakely discusses the role of posttranscriptional modifications of these proteins in regulating transporter subcellular localization and activity state. Finally, Dennis Murphy reviews how natural gene variants and mouse genetic models display consistent behavioral alterations that relate to altered extracellular neurotransmitter levels.
Collapse
|