Gabab Receptor-Mediated Modulation of Synaptic

Total Page:16

File Type:pdf, Size:1020Kb

Gabab Receptor-Mediated Modulation of Synaptic GABAB RECEPTOR-MEDIATED MODULATION OF SYNAPTIC PLASTICITY IN THE LATERAL AMYG DALA Inaugurald issertation zu r Erlangung der Würde eines Doktors der Philosophie vorgele gt der Philosophisch-Naturwissen schaftlichen Fakultät der Universität Basel vo n Hamdy Shaban aus Alexandria, Aegypten Genehmigt von der Philosophisch-Naturwissenschaftlichen Fakultät auf Antrag von Prof. Dr. Andreas Lüthi , Prof. Dr.Bernhard Bettler Basel, den 24.5.2005 Dekan Prof. Dr. Hans-Jakob Wirz Table of contents 3 TABLE OF CONTENTS 1. Summary………………...……………………………………………….…. 5 2. Introduction ……………………..……………………………………….… 7 2.1. Fear emotion and memory formation……………………………...... 7 2.1.1. Early thoughts about emotion……………….………..………….… 7 2.1.2. Fear conditioning and the amygdala………………………….….… 8 2.1.3. The amygaloid complex: structure and connectivity………………. 11 2.1.4. Fear conditioning and synaptic plasticity……………..…………… 13 2.2. Synaptic plasticity…………………………………..………………… 16 2.2.1. Short-term plasticity……………………………………...………… 16 2.2.2. Long-term plasticity………………………………………….….…. 18 2.2.3. Presynaptic long-term potentiation: hippocampal mossy fiber LTP. 19 2.2.4. Postsynaptic signal cascade of LTP induction and expression…….. 20 2.2.5. The Hebb rule of synaptic plasticity………………………….……. 21 2.2.6. LTD, a different form of synaptic plasticity………………………. 22 2.2.7. Metaplasticity and the switch between LTP and LTD………..……. 23 2.3. Inhibition in the amygdala …………………………………….…….. 25 2.3.1. Interneurons in the amygdala ……………………………..……….. 25 2.3.2. Ionotropic GABAA receptors ……………………………..……….. 27 2.3.3. Metabotropic GABAB receptors ………………………..……..….. 28 2.3.4. Molecular structure of GABAB receptors…………………………. 29 2.3.5. GABAB receptor-mediated signaling……………………………… 31 2.3.6. GABAB receptor-mediated inhibition in the LA……………..……. 33 2.3.7. GABAergic modulation of synaptic plasticity in the LA….……… 35 2.3.8. The loss of inhibition and anxiety…………………………….……. 37 3. The aim of the study …………………..………………….…………....… 38 4. Materials and methods ………………………………….………….…… 39 4.1. Mouse brain slice preparation……………………………………….….… 39 4.2. Electrophysiology………………………………………………………… 39 4.3. Data analysis…………………………………………..…………….....… 41 4.4. Behavior experiments……………………………………..................…… 42 4.5. Drugs……………………………………………………………………… 43 5. Results…………………………………………..…………………………… 44 5.1. Presynaptic induction of heterosynaptic associative plasticity in the mammalian brain…………………………………………….…….….… 44 5.2. GABAB(1a) heteroreceptors modulate associative properties of presynaptic LTP and learning………………………………………...…57 5.2.1. Summary………………….……………….………………….…… 57 5.2.2. Introduction…………………………………...…………………… 58 5.2.3. Methods……………………………………………………………..59 5.2.4. Results…………………………..…………………….……….…… 61 5.2.5. Discussion…………….……………………………………….…… 74 -3- Table of contents 4 5.3. Postsynaptic GABAB(1b) receptors modulate the induction of homosynaptic LTP at thalamic afferents………….……..………..... 77 5.3.1. Summary………………………………………..…………………. 77 5.3.2. Introduction……………………………………...………………… 78 5.3.3. Methods………..………………………………………..………… 79 5.3.4. Results…………..………………………………………..………… 81 5.3.5. Discussion………………………………………….…………….… 93 5.4. Redistribution of GABAB(1) Protein and Atypical GABAB Responses in GABAB(2)-Deficient Mice …………..………………………..………. 98 5.4.1. Summary……………………………………….……………..…… 98 5.4.2. Introduction……………………...…………………….…………… 99 5.4.3. Methods………………..……………………………………….…. 100 5.4.4. Results…………………..……………………………………….… 105 5.4.5. Discussion…………………….……………………………….…… 124 6. Discussion………………………………..…………….…………………… 128 6.1. Pre- and postsynaptic GABABR-mediated inhibition in the LA…... 128 6.2. GABAB heteroreceptor-mediated inhibition at thalamic and cortical afferents is impaired in GABAB(1a) -/- mice ………………….……… 132 6.3. GABAB-mediated modulation of homosynaptic and heterosynaptic LTP at cortical afferents ………………..………………………….. 134 6.4. Homosynaptic LTP at thalamic afferents is postsynaptically induced………………………….………………………..……….…… 137 6.5. Intrinsic properties of GABAB receptors …………...……………… 139 6.6. Relevance of the GABABR-modulation of synaptic plasticity in LA in anxiety treatment ………………………………….……………… 140 6.7. Outlook and future experiments …………………………..…...…… 144 7. List of abbreviations……………………………………..….………… 145 8. References……………………………..……………………………………. 147 9. Acknowledgements ………………………..…………………………….. 172 10. Curriculum Vitae ….…………………………...………………………… 173 -4- Summary 5 1. SUMMARY Fear conditioning, one of the most powerful and widely used methods to investigate the mechanisms of associative learning in animals, involves the pairing of an aversive stimulus such as a foot-shock (the unconditioned stimulus; US) with a neutral stimulus such as a tone (the conditioned stimulus; CS). The tone acquires aversive properties and, on subsequent exposure, will elicit a fear response. Behavioral and in vivo electrophysiological experiments indicate that NMDA receptor-mediated long-term potentiation (LTP) in the lateral amygdala (LA), a key structure for emotional learning, underlies the acquisition of Pavlovian fear conditioning. Neuronal activity in the LA is tightly controlled by local inhibitory interneurons. Interneurons exert their inhibitory effect by releasing the neurotransmitter GABA acting on ionotropic GABAA and metabotropic GABAB receptors. There is accumulating evidence suggesting a role for GABAA and GABAB receptors in regulating amygdala- dependent fear and anxiety behavior. However, whereas the role of GABAA receptors for postsynaptic integration and gating of LTP induction is well documented, nothing is known about the role of GABAB receptors in the LA. GABABRs are G-protein-coupled receptors that are localized both pre- and + postsynaptically. Postsynaptic GABABRs are coupled to inwardly rectifying K channels. 2+ Presynaptic GABABRs inhibit neurotransmitter release by decreasing Ca influx at both GABAergic terminals and glutamatergic terminals. Functional GABAB receptors are generally thought to be heterodimers containing GABAB(1) and GABAB(2) subunits. The GABAB(1) subunit exists in two differentially expressed isoforms, GABAB(1a) and GABAB(1b), differing by the presence of two N-terminal “sushi” domains in the GABAB(1a) isoform. In the main study of the present thesis, using a combined electrophysiological and genetic approach in mice, I found that presynaptic GABAB heteroreceptors on glutamatergic cortical afferents are predominantly comprised of GABAB(1a) subunits, and critically determine associative properties of presynaptic cortical LTP. In the absence of functional presynaptic GABAB heteroreceptors, an NMDA receptor-independent, non-associative -5- Summary 6 form of presynaptic LTP is unmasked. Strikingly, the loss of associativity of cortico- amygdala LTP is accompanied by a generalization of conditioned fear at the behavioral level. This indicates that the specificity of information processing in the LA can be set by activity-dependent presynaptic inhibition mediated by specific GABAB receptors. In contrast to synaptic plasticity at cortico-amygdala afferents, I found that at thalamic afferents, GABAB receptors facilitate LTP induction by a postsynaptic mechanism. Moreover, this effect could be attributed to GABAB(1b) containing receptors. Thus, in the LA specific subtypes of pre- and postsynaptic GABAB receptors control induction pre- or postsynaptic LTP in an afferent-specific manner. Taken together, the present findings indicate that GABAB receptors are playing a key role in controlling associative plasticity in the LA, and suggest that GABAB receptors could be a pharmacological target for treatment of psychiatric conditions like anxiety and post traumatic stress disorder. -6- Introduction 7 2. INTRODUCTION 2. Overview In this introduction, I will go through the historical development and the early hypothesis about the formation of emotional memory. Later, I will outline the anatomical features of one of the key structures in fear memory formation, namely the amygdaloid nucleus, and its connectivity to other brain areas. Then I will discuss the different cellular mechanisms of synaptic plasticity implicated in fear memory formation. Finally, I will elucidate the important role of inhibition in the lateral amygdala. 2.1. Fear emotion and memory formation Our memories are our identity. All information is stored in the brain by an unknown encoding mechanism. However, not all information is stored in the same intensity. The reason why some memories remain in our mind forever and others not, is embedded in the emotional information accompanying this memory formation. Whether it is a pleasant emotion or an aversive one, both reinforce memory formation in the brain. Here, I will focus only on the formation of fear memory on the brain. Fear associative learning in mammalian is organized into separate anatomically defined functional systems. The amygdala serves as the neuroanatomical hub of fear memory formation. Pathways that convey information about signals for biologically important events arrive at these hubs by circuitry that depends on stimulus modality and complexity. Within the amygdala, neural plasticity occurs because of convergence of these stimuli and the biologically important information they predict. This neural plasticity is the physical basis of associative memory formation 2.1.1. Early thoughts about emotion Charles Darwin, in 1872, was the first who described that the expression of emotions in humans and animals is similar (Darwin 1872/1965). By comparing and analysing several -7- Introduction 8 sketches
Recommended publications
  • The Roles Played by Highly Truncated Splice Variants of G Protein-Coupled Receptors Helen Wise
    Wise Journal of Molecular Signaling 2012, 7:13 http://www.jmolecularsignaling.com/content/7/1/13 REVIEW Open Access The roles played by highly truncated splice variants of G protein-coupled receptors Helen Wise Abstract Alternative splicing of G protein-coupled receptor (GPCR) genes greatly increases the total number of receptor isoforms which may be expressed in a cell-dependent and time-dependent manner. This increased diversity of cell signaling options caused by the generation of splice variants is further enhanced by receptor dimerization. When alternative splicing generates highly truncated GPCRs with less than seven transmembrane (TM) domains, the predominant effect in vitro is that of a dominant-negative mutation associated with the retention of the wild-type receptor in the endoplasmic reticulum (ER). For constitutively active (agonist-independent) GPCRs, their attenuated expression on the cell surface, and consequent decreased basal activity due to the dominant-negative effect of truncated splice variants, has pathological consequences. Truncated splice variants may conversely offer protection from disease when expression of co-receptors for binding of infectious agents to cells is attenuated due to ER retention of the wild-type co-receptor. In this review, we will see that GPCRs retained in the ER can still be functionally active but also that highly truncated GPCRs may also be functionally active. Although rare, some truncated splice variants still bind ligand and activate cell signaling responses. More importantly, by forming heterodimers with full-length GPCRs, some truncated splice variants also provide opportunities to generate receptor complexes with unique pharmacological properties. So, instead of assuming that highly truncated GPCRs are associated with faulty transcription processes, it is time to reassess their potential benefit to the host organism.
    [Show full text]
  • GABAB Receptors and Pain
    King’s Research Portal DOI: 10.1016/j.neuropharm.2017.05.012 Document Version Peer reviewed version Link to publication record in King's Research Portal Citation for published version (APA): Malcangio, M. (2017). GABAB receptors and pain. Neuropharmacology. https://doi.org/10.1016/j.neuropharm.2017.05.012 Citing this paper Please note that where the full-text provided on King's Research Portal is the Author Accepted Manuscript or Post-Print version this may differ from the final Published version. If citing, it is advised that you check and use the publisher's definitive version for pagination, volume/issue, and date of publication details. And where the final published version is provided on the Research Portal, if citing you are again advised to check the publisher's website for any subsequent corrections. General rights Copyright and moral rights for the publications made accessible in the Research Portal are retained by the authors and/or other copyright owners and it is a condition of accessing publications that users recognize and abide by the legal requirements associated with these rights. •Users may download and print one copy of any publication from the Research Portal for the purpose of private study or research. •You may not further distribute the material or use it for any profit-making activity or commercial gain •You may freely distribute the URL identifying the publication in the Research Portal Take down policy If you believe that this document breaches copyright please contact [email protected] providing details, and we will remove access to the work immediately and investigate your claim.
    [Show full text]
  • Functioning of the Dimeric GABAB Receptor Extracellular Domain Revealed by Glycan Wedge Scanning
    Functioning of the dimeric GABAB receptor extracellular domain revealed by glycan wedge scanning Philippe Rondard1§, Siluo Huang2§, Carine Monnier1, Haijun Tu2, Bertrand Blanchard1, Nadia Oueslati1, Fanny Malhaire1, Ying Li2, Eric Trinquet3, Gilles Labesse4,5, Jean-Philippe Pin1 & Jianfeng Liu2 1 CNRS, UMR 5203, Institut de Génomique Fonctionnelle, Montpellier, France and INSERM, U661, Montpellier, France and Université Montpellier 1,2, Montpellier F-34000, France. 2, Sino-France Laboratory for Drug Screening, Key Laboratory of Molecular Biophysics of Ministry of Education, Huazhong University of Science and Technology, Wuhan, Hubei, China 3 CisBio International, Parc technologique Marcel Boiteux, Bagnols/Cèze cedex F-30204, France 4, Centre de Biochimie Structurale, CNRS UMR5048, Université Montpellier 1, Montpellier F-34060, France 5 INSERM U414, Montpellier, F-34094 France § P.R. and S.H. contributed equally to this work Correspondence should be addressed to J.P.P ([email protected]) and J.L. ([email protected]). Running title : Functional analysis of GABAB receptor VFTs using N-glycans Total character count (including spaces) : 55,058 1 Abstract The G-protein coupled receptor activated by the neurotransmitter GABA is made up of two subunits, GABAB1 and GABAB2. While GABAB1 binds agonists, GABAB2 is required for trafficking GABAB1 to the cell surface, increasing agonist affinity to GABAB1, and activating associated G-proteins. These subunits each comprise two domains, a Venus flytrap (VFT) domain and a heptahelical (7TM) domain. How agonist binding to the GABAB1 VFT leads to GABAB2 7TM activation remains unknown. Here, we used a glycan wedge scanning approach to investigate how the GABAB VFT dimer controls receptor activity.
    [Show full text]
  • Exploring the Heteromeric Interface of the 5-HT2A-Mglu2 Receptor Complex
    Virginia Commonwealth University VCU Scholars Compass Theses and Dissertations Graduate School 2020 Exploring the Heteromeric Interface of the 5-HT2A-mGlu2 Receptor Complex Mohamed Aarif Abdul Kareem Virginia Commonwealth University Follow this and additional works at: https://scholarscompass.vcu.edu/etd Part of the Nervous System Diseases Commons, Other Psychiatry and Psychology Commons, Physiological Processes Commons, and the Translational Medical Research Commons © The Author Downloaded from https://scholarscompass.vcu.edu/etd/6235 This Thesis is brought to you for free and open access by the Graduate School at VCU Scholars Compass. It has been accepted for inclusion in Theses and Dissertations by an authorized administrator of VCU Scholars Compass. For more information, please contact [email protected]. Exploring the Heteromeric Interface of the 5-HT2A-mGlu2 Receptor Complex A Thesis submitted in partial fulfillment of the requirements for the degree of Master of Science in Physiology and Biophysics at Virginia Commonwealth University By: Mohamed Aarif Abdul Kareem B.A. Neurobiology, Boston University, 2018 Mentor: Javier González-Maeso Associate Professor Department of Physiology and Biophysics Virginia Commonwealth University Richmond, Virginia April 30, 2020 Acknowledgments: Thank you to my peers for their continued support of my dreams and aspirations. Thank you to my mentors for pushing and supporting me every step of the way. Thank you to Virginia Commonwealth University for providing opportunities which foster my passion for science and allow it to continue to flourish. Indeed, I owe much gratitude to my parents, Abdul & Jasmine Kareem, who have guided me in becoming a strong, independent student, and encourage me to take calculated risks and face challenges head on.
    [Show full text]
  • G Protein-Coupled Receptors
    S.P.H. Alexander et al. The Concise Guide to PHARMACOLOGY 2015/16: G protein-coupled receptors. British Journal of Pharmacology (2015) 172, 5744–5869 THE CONCISE GUIDE TO PHARMACOLOGY 2015/16: G protein-coupled receptors Stephen PH Alexander1, Anthony P Davenport2, Eamonn Kelly3, Neil Marrion3, John A Peters4, Helen E Benson5, Elena Faccenda5, Adam J Pawson5, Joanna L Sharman5, Christopher Southan5, Jamie A Davies5 and CGTP Collaborators 1School of Biomedical Sciences, University of Nottingham Medical School, Nottingham, NG7 2UH, UK, 2Clinical Pharmacology Unit, University of Cambridge, Cambridge, CB2 0QQ, UK, 3School of Physiology and Pharmacology, University of Bristol, Bristol, BS8 1TD, UK, 4Neuroscience Division, Medical Education Institute, Ninewells Hospital and Medical School, University of Dundee, Dundee, DD1 9SY, UK, 5Centre for Integrative Physiology, University of Edinburgh, Edinburgh, EH8 9XD, UK Abstract The Concise Guide to PHARMACOLOGY 2015/16 provides concise overviews of the key properties of over 1750 human drug targets with their pharmacology, plus links to an open access knowledgebase of drug targets and their ligands (www.guidetopharmacology.org), which provides more detailed views of target and ligand properties. The full contents can be found at http://onlinelibrary.wiley.com/doi/ 10.1111/bph.13348/full. G protein-coupled receptors are one of the eight major pharmacological targets into which the Guide is divided, with the others being: ligand-gated ion channels, voltage-gated ion channels, other ion channels, nuclear hormone receptors, catalytic receptors, enzymes and transporters. These are presented with nomenclature guidance and summary information on the best available pharmacological tools, alongside key references and suggestions for further reading.
    [Show full text]
  • Multi-Functionality of Proteins Involved in GPCR and G Protein Signaling: Making Sense of Structure–Function Continuum with In
    Cellular and Molecular Life Sciences (2019) 76:4461–4492 https://doi.org/10.1007/s00018-019-03276-1 Cellular andMolecular Life Sciences REVIEW Multi‑functionality of proteins involved in GPCR and G protein signaling: making sense of structure–function continuum with intrinsic disorder‑based proteoforms Alexander V. Fonin1 · April L. Darling2 · Irina M. Kuznetsova1 · Konstantin K. Turoverov1,3 · Vladimir N. Uversky2,4 Received: 5 August 2019 / Revised: 5 August 2019 / Accepted: 12 August 2019 / Published online: 19 August 2019 © Springer Nature Switzerland AG 2019 Abstract GPCR–G protein signaling system recognizes a multitude of extracellular ligands and triggers a variety of intracellular signal- ing cascades in response. In humans, this system includes more than 800 various GPCRs and a large set of heterotrimeric G proteins. Complexity of this system goes far beyond a multitude of pair-wise ligand–GPCR and GPCR–G protein interactions. In fact, one GPCR can recognize more than one extracellular signal and interact with more than one G protein. Furthermore, one ligand can activate more than one GPCR, and multiple GPCRs can couple to the same G protein. This defnes an intricate multifunctionality of this important signaling system. Here, we show that the multifunctionality of GPCR–G protein system represents an illustrative example of the protein structure–function continuum, where structures of the involved proteins represent a complex mosaic of diferently folded regions (foldons, non-foldons, unfoldons, semi-foldons, and inducible foldons). The functionality of resulting highly dynamic conformational ensembles is fne-tuned by various post-translational modifcations and alternative splicing, and such ensembles can undergo dramatic changes at interaction with their specifc partners.
    [Show full text]
  • Complex GABAB Receptor Complexes
    Complex GABAB receptor complexes: how to generate multiple functionally distinct units from a single receptor Chanjuan Xu, Wenhua Zhang, Philippe Rondard, Jean-Philippe Pin, Jianfeng Liu To cite this version: Chanjuan Xu, Wenhua Zhang, Philippe Rondard, Jean-Philippe Pin, Jianfeng Liu. Complex GABAB receptor complexes: how to generate multiple functionally distinct units from a single receptor. Fron- tiers in Pharmacology, Frontiers, 2014, 5, pp.12. 10.3389/fphar.2014.00012. hal-01942936 HAL Id: hal-01942936 https://hal.archives-ouvertes.fr/hal-01942936 Submitted on 3 Dec 2018 HAL is a multi-disciplinary open access L’archive ouverte pluridisciplinaire HAL, est archive for the deposit and dissemination of sci- destinée au dépôt et à la diffusion de documents entific research documents, whether they are pub- scientifiques de niveau recherche, publiés ou non, lished or not. The documents may come from émanant des établissements d’enseignement et de teaching and research institutions in France or recherche français ou étrangers, des laboratoires abroad, or from public or private research centers. publics ou privés. REVIEW ARTICLE published: 11 February 2014 doi: 10.3389/fphar.2014.00012 Complex GABAB receptor complexes: how to generate multiple functionally distinct units from a single receptor Chanjuan Xu1,Wenhua Zhang1, Philippe Rondard 2 , Jean-Philippe Pin 2 , and Jianfeng Liu1* 1 Cellular Signaling Laboratory, Key Laboratory of Molecular Biophysics of Ministry of Education, College of Life Science and Technology, Huazhong University of Science and Technology, Wuhan, China 2 Institut de Génomique Fonctionnelle, CNRS UMR5203, INSERM U661, Universités de Montpellier I & II, Montpellier, France Edited by: The main inhibitory neurotransmitter, GABA, acts on both ligand-gated and G protein- Pietro Marini, University of Aberdeen, coupled receptors, the GABAA/C and GABAB receptors, respectively.
    [Show full text]
  • Elucidating Agonist-Induced Signaling Patterns of Human G Protein-Coupled Receptor GPR17 and Uncovering Pranlukast As a Biased Mixed Agonist-Antagonist at GPR17
    Elucidating agonist-induced signaling patterns of human G protein-coupled receptor GPR17 and uncovering pranlukast as a biased mixed agonist-antagonist at GPR17 Dissertation zur Erlangung des Doktorgrades (Dr. rer. nat.) der Mathematisch-Naturwissenschaftlichen Fakultät der Rheinischen Friedrich-Wilhelms-Universität Bonn vorgelegt von Stephanie Monika Hennen aus Saarburg Bonn 2011 Angefertigt mit Genehmigung der Mathematisch-Naturwissenschaftlichen Fakultät der Rheinischen Friedrich-Wilhelms Universität Bonn. 1. Gutachter: Prof. Dr. Evi Kostenis 2. Gutachter: Prof. Dr. Klaus Mohr Tag der Promotion: 15.09.2011 Erscheinungsjahr: 2011 Die vorliegende Arbeit wurde in der Zeit von April 2008 bis März 2011 am Institut für Pharmazeutische Biologie der Rheinischen Friedrich-Wilhelms Universität Bonn unter der Leitung von Frau Prof. Dr. Evi Kostenis durchgeführt. Meinen Eltern Abstract I Abstract The progress of human genome sequencing has revealed the existence of several hundred orphan G protein-coupled receptors (GPCRs), whose endogenous ligands are not yet identified, thus their deorphanization and characterization is fundamental in order to clarify their physiological and pathological role as well as their relevance as new drug targets. Recently, the orphan GPCR GPR17 that is phylogenetically and structurally related to the known P2Y and CysLT receptors has been identified as a dual uracil nucleotide/cysteinyl-leukotriene receptor. In spite of this, these deorphanization efforts could not be verified yet by independent laboratories, thus this classification remains a controversial matter and GPR17 most likely still represents an orphan GPCR. Additionally, a subsequent study revealed a ligand-independent regulatory role for GPR17 suppressing CysLT1 receptor function via GPCR-GPCR interactions. By means of a high throughput pharmacogenomic approach our group has identified a small molecule agonist for GPR17 that is used as pharmacological tool for characterization of ligand-dependent behaviors triggered by this receptor.
    [Show full text]
  • The Selective Antagonist EPPTB Reveals TAAR1-Mediated Regulatory Mechanisms in Dopaminergic Neurons of the Mesolimbic System
    The selective antagonist EPPTB reveals TAAR1-mediated regulatory mechanisms in dopaminergic neurons of the mesolimbic system Amyaouch Bradaiaa,b, Gerhard Trubec, Henri Stalderc, Roger D. Norcrossc, Laurence Ozmenc, Joseph G. Wettsteinc, Audre´ e Pinarda, Danie` le Buchyc, Martin Gassmanna, Marius C. Hoenerc, and Bernhard Bettlera,1 aDepartment of Biomedicine, Institute of Physiology, Pharmazentrum, University of Basel, CH-4056 Basel, Switzerland; bNeuroservice, 13593 Aix en Provence, Cedex 03, France; and cPharmaceuticals Division, Neuroscience Research, F. Hoffmann-La Roche Ltd., CH-4070 Basel, Switzerland Edited by Shigetada Nakanishi, Osaka Bioscience Institute, Osaka, Japan, and approved September 29, 2009 (received for review June 11, 2009) Trace amine-associated receptor 1 (TAAR1) is a G protein-coupled receptor-mediated pathway. The genetic absence of TAAR1 receptor (GPCR) that is nonselectively activated by endogenous clearly increased the spontaneous firing rate of DA neurons but metabolites of amino acids. TAAR1 is considered a promising drug the underlying signaling mechanism remained unclear (7). Taar1 target for the treatment of psychiatric and neurodegenerative knockout mice also display behavioral and neurochemical signs disorders. However, no selective ligand to identify TAAR1-specific of DA supersensitivity, a feature thought to relate to positive signaling mechanisms is available yet. Here we report a selective symptoms of schizophrenia (8). In addition, TAs were implicated TAAR1 antagonist, EPPTB, and characterize its physiological effects in the etiology of depression, addiction, attention-deficit/ at dopamine (DA) neurons of the ventral tegmental area (VTA). We hyperactivity disorder, and Parkinson’s disease (5, 9, 10). How- show that EPPTB prevents the reduction of the firing frequency of ever, validation of therapeutic concepts was hampered by the DA neurons induced by p-tyramine (p-tyr), a nonselective TAAR1 lack of a ligand that specifically regulates TAAR1 activity in agonist.
    [Show full text]
  • Activation of Presynaptic 5-Hydroxytryptamine 2A Receptors Facilitates Excitatory Synaptic Transmission Via Protein Kinase C in the Dorsolateral Septal Nucleus
    The Journal of Neuroscience, September 1, 2002, 22(17):7509–7517 Activation of Presynaptic 5-Hydroxytryptamine 2A Receptors Facilitates Excitatory Synaptic Transmission via Protein Kinase C in the Dorsolateral Septal Nucleus Hiroshi Hasuo,1 Toshimasa Matsuoka,1,2 and Takashi Akasu1 Departments of 1Physiology and 2Neuropsychiatry, Kurume University School of Medicine, Kurume 830-0011, Japan Effects of 5-hydroxytryptamine (5-HT) on EPSPs and EPSCs in by 5-HT. 5-HT (10 ␮M) and ␣-methyl-5-HT (10 ␮M) increased the the rat dorsolateral septal nucleus (DLSN) were examined in the frequency of miniature EPSPs (mEPSPs) without changing the presence of GABAA and GABAB receptor antagonists. Bath ap- mEPSP amplitude. The ratio of the paired pulse facilitation was plication of 5-HT (10 ␮M) for 5–10 min increased the amplitude of significantly decreased by 5-HT and ␣-methyl-5-HT. The 5-HT- the EPSP and EPSC. (Ϯ)-8-Hydroxy-2-(di-N-propylamino)tetralin induced facilitation of the EPSP was blocked by calphostin C (100 ␮ hydrobromide (10 M), an agonist for 5-HT1A and 5-HT7 receptors, nM), a specific protein kinase C (PKC) inhibitor, but not by N-[2- ␣ ␮ did not facilitate the EPSP. -Methyl-5-HT (10 M), a 5-HT2 recep- ( p-bromocinnamylamino)ethyl]-5-isoquinolinesulfonamide (10 tor agonist, increased the amplitude of the EPSC. ␣-Methyl-5-(2- ␮M), a protein kinase A inhibitor. Phorbol 12,13-dibutyrate (3 ␮M) thienylmethoxy)-1H-indole-3-ethanamine (10 ␮M) and 6-chloro-2- mimicked the facilitatory effects of 5-HT. These results suggest ␮ (1-piperazinyl)pyrazine (10 M), selective 5-HT2B and 5-HT2C that 5-HT enhances the EPSP by increasing the release of gluta- receptor agonists, respectively, had no effect on the EPSP.
    [Show full text]
  • Mechanisms of Cannabinoid Inhibition of Gabaasynaptic Transmission In
    The Journal of Neuroscience, April 1, 2000, 20(7):2470–2479 Mechanisms of Cannabinoid Inhibition of GABAA Synaptic Transmission in the Hippocampus Alexander F. Hoffman and Carl R. Lupica Cellular Neurobiology Branch, National Institute on Drug Abuse, Intramural Research Program, National Institutes of Health, Baltimore, Maryland 21224 The localization of cannabinoid (CB) receptors to GABAergic sured while sodium channels were blocked by tetrodotoxin interneurons in the hippocampus indicates that CBs may mod- (TTX). Blockade of voltage-dependent calcium channels (VD- ulate GABAergic function and thereby mediate some of the CCs) by cadmium also eliminated the effect of WIN 55,212–2 on disruptive effects of marijuana on spatial memory and sensory sIPSCs. Depolarization of inhibitory terminals with elevated processing. To investigate the possible mechanisms through extracellular potassium caused a large increase in the fre- which CB receptors may modulate GABAergic neurotransmis- quency of mIPSCs that was inhibited by both cadmium and sion in the hippocampus, whole-cell voltage-clamp recordings WIN 55,212–2. The presynaptic effect of WIN 55,212–2 was were performed on CA1 pyramidal neurons in rat brain slices. also investigated using the potassium channel blockers barium Stimulus-evoked GABAA receptor-mediated IPSCs were re- and 4-aminopyridine. Neither of these agents significantly al- duced in a concentration-dependent manner by the CB recep- tered the effect of WIN 55,212–2 on evoked IPSCs. Together, tor agonist WIN 55,212–2 (EC50 of 138 nM). This effect was these data suggest that presynaptic CB1 receptors reduce ␮ blocked by the CB1 receptor antagonist SR141716A (1 M) but GABAA- but not GABAB-mediated synaptic inhibition of CA1 not by the opioid antagonist naloxone.
    [Show full text]
  • The G Protein-Coupled Receptor Heterodimer Network (GPCR-Hetnet) and Its Hub Components
    Int. J. Mol. Sci. 2014, 15, 8570-8590; doi:10.3390/ijms15058570 OPEN ACCESS International Journal of Molecular Sciences ISSN 1422-0067 www.mdpi.com/journal/ijms Article The G Protein-Coupled Receptor Heterodimer Network (GPCR-HetNet) and Its Hub Components Dasiel O. Borroto-Escuela 1,†,*, Ismel Brito 1,2,†, Wilber Romero-Fernandez 1, Michael Di Palma 1,3, Julia Oflijan 4, Kamila Skieterska 5, Jolien Duchou 5, Kathleen Van Craenenbroeck 5, Diana Suárez-Boomgaard 6, Alicia Rivera 6, Diego Guidolin 7, Luigi F. Agnati 1 and Kjell Fuxe 1,* 1 Department of Neuroscience, Karolinska Institutet, Retzius väg 8, 17177 Stockholm, Sweden; E-Mails: [email protected] (I.B.); [email protected] (W.R.-F.); [email protected] (M.D.P.); [email protected] (L.F.A.) 2 IIIA-CSIC, Artificial Intelligence Research Institute, Spanish National Research Council, 08193 Barcelona, Spain 3 Department of Earth, Life and Environmental Sciences, Section of Physiology, Campus Scientifico Enrico Mattei, Urbino 61029, Italy 4 Department of Physiology, Faculty of Medicine, University of Tartu, Tartu 50411, Estonia; E-Mail: [email protected] 5 Laboratory of Eukaryotic Gene Expression and Signal Transduction (LEGEST), Ghent University, 9000 Ghent, Belgium; E-Mails: [email protected] (K.S.); [email protected] (J.D.); [email protected] (K.V.C.) 6 Department of Cell Biology, School of Science, University of Málaga, 29071 Málaga, Spain; E-Mails: [email protected] (D.S.-B.); [email protected] (A.R.) 7 Department of Molecular Medicine, University of Padova, Padova 35121, Italy; E-Mail: [email protected] † These authors contributed equally to this work.
    [Show full text]