1
|
Bian X, Zhu J, Jia X, Liang W, Yu S, Li Z, Zhang W, Rao Y. Suggestion of creatine as a new neurotransmitter by approaches ranging from chemical analysis and biochemistry to electrophysiology. eLife 2023; 12:RP89317. [PMID: 38126335 PMCID: PMC10735228 DOI: 10.7554/elife.89317] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/23/2023] Open
Abstract
The discovery of a new neurotransmitter, especially one in the central nervous system, is both important and difficult. We have been searching for new neurotransmitters for 12 y. We detected creatine (Cr) in synaptic vesicles (SVs) at a level lower than glutamate and gamma-aminobutyric acid but higher than acetylcholine and 5-hydroxytryptamine. SV Cr was reduced in mice lacking either arginine:glycine amidinotransferase (a Cr synthetase) or SLC6A8, a Cr transporter with mutations among the most common causes of intellectual disability in men. Calcium-dependent release of Cr was detected after stimulation in brain slices. Cr release was reduced in Slc6a8 and Agat mutants. Cr inhibited neocortical pyramidal neurons. SLC6A8 was necessary for Cr uptake into synaptosomes. Cr was found by us to be taken up into SVs in an ATP-dependent manner. Our biochemical, chemical, genetic, and electrophysiological results are consistent with the possibility of Cr as a neurotransmitter, though not yet reaching the level of proof for the now classic transmitters. Our novel approach to discover neurotransmitters is to begin with analysis of contents in SVs before defining their function and physiology.
Collapse
Affiliation(s)
- Xiling Bian
- Laboratory of Neurochemical Biology, PKU-IDG/McGovern Institute for Brain Research, Peking-Tsinghua Center for Life Sciences, School of Life Sciences, Department of Chemical Biology, College of Chemistry and Chemical Engineering, Department of Molecular and Cellular Pharmacology, School of Pharmaceutical Sciences, Peking UniversityBeijingChina
- Chinese Institute for Brain Research (CIBR)BeijingChina
| | - Jiemin Zhu
- Laboratory of Neurochemical Biology, PKU-IDG/McGovern Institute for Brain Research, Peking-Tsinghua Center for Life Sciences, School of Life Sciences, Department of Chemical Biology, College of Chemistry and Chemical Engineering, Department of Molecular and Cellular Pharmacology, School of Pharmaceutical Sciences, Peking UniversityBeijingChina
- Chinese Institute for Brain Research (CIBR)BeijingChina
| | - Xiaobo Jia
- Laboratory of Neurochemical Biology, PKU-IDG/McGovern Institute for Brain Research, Peking-Tsinghua Center for Life Sciences, School of Life Sciences, Department of Chemical Biology, College of Chemistry and Chemical Engineering, Department of Molecular and Cellular Pharmacology, School of Pharmaceutical Sciences, Peking UniversityBeijingChina
- Chinese Institute for Brain Research (CIBR)BeijingChina
| | - Wenjun Liang
- Chinese Institutes of Medical Research, Capital Medical UniversityBeijingChina
- Changping Laboratory, Yard 28, Science Park Road, Changping DistrictBeijingChina
| | - Sihan Yu
- Laboratory of Neurochemical Biology, PKU-IDG/McGovern Institute for Brain Research, Peking-Tsinghua Center for Life Sciences, School of Life Sciences, Department of Chemical Biology, College of Chemistry and Chemical Engineering, Department of Molecular and Cellular Pharmacology, School of Pharmaceutical Sciences, Peking UniversityBeijingChina
- Changping Laboratory, Yard 28, Science Park Road, Changping DistrictBeijingChina
| | - Zhiqiang Li
- Laboratory of Neurochemical Biology, PKU-IDG/McGovern Institute for Brain Research, Peking-Tsinghua Center for Life Sciences, School of Life Sciences, Department of Chemical Biology, College of Chemistry and Chemical Engineering, Department of Molecular and Cellular Pharmacology, School of Pharmaceutical Sciences, Peking UniversityBeijingChina
| | - Wenxia Zhang
- Laboratory of Neurochemical Biology, PKU-IDG/McGovern Institute for Brain Research, Peking-Tsinghua Center for Life Sciences, School of Life Sciences, Department of Chemical Biology, College of Chemistry and Chemical Engineering, Department of Molecular and Cellular Pharmacology, School of Pharmaceutical Sciences, Peking UniversityBeijingChina
- Chinese Institutes of Medical Research, Capital Medical UniversityBeijingChina
- Institute of Molecular Physiology, Shenzhen Bay LaboratoryShenzhenChina
| | - Yi Rao
- Laboratory of Neurochemical Biology, PKU-IDG/McGovern Institute for Brain Research, Peking-Tsinghua Center for Life Sciences, School of Life Sciences, Department of Chemical Biology, College of Chemistry and Chemical Engineering, Department of Molecular and Cellular Pharmacology, School of Pharmaceutical Sciences, Peking UniversityBeijingChina
- Chinese Institute for Brain Research (CIBR)BeijingChina
- Chinese Institutes of Medical Research, Capital Medical UniversityBeijingChina
- Changping Laboratory, Yard 28, Science Park Road, Changping DistrictBeijingChina
- Institute of Molecular Physiology, Shenzhen Bay LaboratoryShenzhenChina
- Research Unit of Medical Neurobiology, Chinese Academy of Medical SciencesBeijingChina
| |
Collapse
|
2
|
Jia X, Zhu J, Bian X, Liu S, Yu S, Liang W, Jiang L, Mao R, Zhang W, Rao Y. Importance of glutamine in synaptic vesicles revealed by functional studies of SLC6A17 and its mutations pathogenic for intellectual disability. eLife 2023; 12:RP86972. [PMID: 37440432 PMCID: PMC10393021 DOI: 10.7554/elife.86972] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 07/15/2023] Open
Abstract
Human mutations in the gene encoding the solute carrier (SLC) 6A17 caused intellectual disability (ID). The physiological role of SLC6A17 and pathogenesis of SLC6A17-based-ID were both unclear. Here, we report learning deficits in Slc6a17 knockout and point mutant mice. Biochemistry, proteomic, and electron microscopy (EM) support SLC6A17 protein localization in synaptic vesicles (SVs). Chemical analysis of SVs by liquid chromatography coupled to mass spectrometry (LC-MS) revealed glutamine (Gln) in SVs containing SLC6A17. Virally mediated overexpression of SLC6A17 increased Gln in SVs. Either genetic or virally mediated targeting of Slc6a17 reduced Gln in SVs. One ID mutation caused SLC6A17 mislocalization while the other caused defective Gln transport. Multidisciplinary approaches with seven types of genetically modified mice have shown Gln as an endogenous substrate of SLC6A17, uncovered Gln as a new molecule in SVs, established the necessary and sufficient roles of SLC6A17 in Gln transport into SVs, and suggested SV Gln decrease as the key pathogenetic mechanism in human ID.
Collapse
Affiliation(s)
- Xiaobo Jia
- Chinese Institute for Brain ResearchBeijingChina
- Changping LaboratoryBeijingChina
- Research Unit of Medical Neurobiology, Chinese Academy of Medical SciencesBeijingChina
| | - Jiemin Zhu
- Laboratory of Neurochemical Biology, PKU-IDG/McGovern Institute for Brain Research, Peking-Tsinghua Center for Life Sciences, School of Life Sciences, Department of Molecular and Cellular Pharmacology, School of Pharmaceutical Sciences, School of Chemistry and Chemical Engineering, Peking UniversityBeijingChina
| | - Xiling Bian
- Laboratory of Neurochemical Biology, PKU-IDG/McGovern Institute for Brain Research, Peking-Tsinghua Center for Life Sciences, School of Life Sciences, Department of Molecular and Cellular Pharmacology, School of Pharmaceutical Sciences, School of Chemistry and Chemical Engineering, Peking UniversityBeijingChina
| | | | - Sihan Yu
- Chinese Institute for Brain ResearchBeijingChina
| | | | - Lifen Jiang
- Institute of Molecular Physiology, Shenzhen Bay LaboratoryShenzhenChina
| | - Renbo Mao
- Chinese Institute for Brain ResearchBeijingChina
| | - Wenxia Zhang
- Laboratory of Neurochemical Biology, PKU-IDG/McGovern Institute for Brain Research, Peking-Tsinghua Center for Life Sciences, School of Life Sciences, Department of Molecular and Cellular Pharmacology, School of Pharmaceutical Sciences, School of Chemistry and Chemical Engineering, Peking UniversityBeijingChina
| | - Yi Rao
- Chinese Institute for Brain ResearchBeijingChina
- Changping LaboratoryBeijingChina
- Research Unit of Medical Neurobiology, Chinese Academy of Medical SciencesBeijingChina
- Laboratory of Neurochemical Biology, PKU-IDG/McGovern Institute for Brain Research, Peking-Tsinghua Center for Life Sciences, School of Life Sciences, Department of Molecular and Cellular Pharmacology, School of Pharmaceutical Sciences, School of Chemistry and Chemical Engineering, Peking UniversityBeijingChina
- Institute of Molecular Physiology, Shenzhen Bay LaboratoryShenzhenChina
- Capital Medical UniversityBeijingChina
| |
Collapse
|
3
|
Schary N, Novak B, Kämper L, Yousf A, Lübbert H. Identification and pharmacological modification of resistance mechanisms to protoporphyrin-mediated photodynamic therapy in human cutaneous squamous cell carcinoma cell lines. Photodiagnosis Photodyn Ther 2022; 39:103004. [PMID: 35811052 DOI: 10.1016/j.pdpdt.2022.103004] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/07/2022] [Revised: 06/21/2022] [Accepted: 07/06/2022] [Indexed: 10/17/2022]
Abstract
BACKGROUND Photodynamic therapy (PDT) is clinically approved to treat neoplastic skin diseases such as precursors of cutaneous squamous cell carcinoma (cSCC). In PDT, 5-aminolevulinic acid (5-ALA) drives the selective formation of the endogenous photosensitizer protoporphyrin IX (PpIX). Although 5-ALA PDT is clinically highly effective, resistance might occur due to decreased accumulation of PpIX in certain tumors. Such resistance may be caused by any fundamental step of PpIX accumulation: 5-ALA uptake, PpIX synthesis and PpIX efflux. METHODS We investigated PpIX accumulation and photodynamically induced cell death in PDT refractory SCC-13, PDT susceptible A431, and normal human epidermal keratinocytes (NHEK). Expression of genes associated with cellular PpIX kinetics was investigated on mRNA and protein level. PpIX accumulation and cell death upon illumination were pharmacologically manipulated using drugs targeting 5-ALA uptake, PpIX synthesis or efflux. RESULTS The experiments indicate that taurine transporter (SLC6A6) is the major pathway for 5-ALA uptake in cSCC cells, while being less important in NHEK. Downregulation of PpIX synthesis enzymes in SCC-13 was counteracted by methotrexate (MTX) treatment, which restored PpIX formation and cell death. PpIX efflux inhibitors targeting ABC transporters led to significantly increased PpIX accumulation in SCC-13, thereby fully overcoming resistance. CONCLUSIONS The results indicate a conserved threshold for PpIX accumulation with respect to PDT-resistance. Cells showed increased viability after PDT at PpIX concentrations below 1.5 nM. Selective uptake of 5-ALA via taurine transporter SLC6A6 in cutaneous tumor cells is novel but unrelated to resistance. MTX can partially abrogate resistance by PpIX synthesis enzyme induction, while efflux mechanisms via ABC transporters seem the main driving force and promising drug targets.
Collapse
Affiliation(s)
- Nicole Schary
- Department of Animal Physiology, Ruhr-University Bochum, Germany
| | - Ben Novak
- Department of Animal Physiology, Ruhr-University Bochum, Germany; Biofrontera Bioscience GmbH, Leverkusen, Germany.
| | - Laura Kämper
- Department of Animal Physiology, Ruhr-University Bochum, Germany
| | - Aisha Yousf
- Department of Animal Physiology, Ruhr-University Bochum, Germany
| | - Hermann Lübbert
- Department of Animal Physiology, Ruhr-University Bochum, Germany
| |
Collapse
|
4
|
Danbolt NC, López-Corcuera B, Zhou Y. Reconstitution of GABA, Glycine and Glutamate Transporters. Neurochem Res 2022; 47:85-110. [PMID: 33905037 PMCID: PMC8763731 DOI: 10.1007/s11064-021-03331-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/05/2021] [Revised: 04/13/2021] [Accepted: 04/15/2021] [Indexed: 10/25/2022]
Abstract
In contrast to water soluble enzymes which can be purified and studied while in solution, studies of solute carrier (transporter) proteins require both that the protein of interest is situated in a phospholipid membrane and that this membrane forms a closed compartment. An additional challenge to the study of transporter proteins has been that the transport depends on the transmembrane electrochemical gradients. Baruch I. Kanner understood this early on and first developed techniques for studying plasma membrane vesicles. This advanced the field in that the experimenter could control the electrochemical gradients. Kanner, however, did not stop there, but started to solubilize the membranes so that the transporter proteins were taken out of their natural environment. In order to study them, Kanner then had to find a way to reconstitute them (reinsert them into phospholipid membranes). The scope of the present review is both to describe the reconstitution method in full detail as that has never been done, and also to reveal the scientific impact that this method has had. Kanner's later work is not reviewed here although that also deserves a review because it too has had a huge impact.
Collapse
Affiliation(s)
- Niels Christian Danbolt
- Neurotransporter Group, Department of Molecular Medicine, Institute of Basic Medical Sciences, University of Oslo, 0317, Oslo, Norway.
| | - Beatriz López-Corcuera
- Departamento de Biología Molecular, Universidad Autónoma de Madrid, Madrid, Spain
- Centro de Biología Molecular "Severo Ochoa" Consejo Superior de Investigaciones Científicas, Universidad Autónoma de Madrid, Madrid, Spain
- IdiPAZ, Hospital Universitario La Paz, Madrid, Spain
| | - Yun Zhou
- Neurotransporter Group, Department of Molecular Medicine, Institute of Basic Medical Sciences, University of Oslo, 0317, Oslo, Norway
| |
Collapse
|
5
|
Kubrusly RCC, da Rosa Valli T, Ferreira MNMR, de Moura P, Borges-Martins VPP, Martins RS, Ferreira DDP, Sathler MF, de Melo Reis RA, Ferreira GC, Manhães AC, Dos Santos Pereira M. Caffeine Improves GABA Transport in the Striatum of Spontaneously Hypertensive Rats (SHR). Neurotox Res 2021; 39:1946-1958. [PMID: 34637050 DOI: 10.1007/s12640-021-00423-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/09/2021] [Revised: 09/25/2021] [Accepted: 09/27/2021] [Indexed: 11/28/2022]
Abstract
The spontaneously hypertensive rat (SHR) is an excellent animal model that mimics the behavioral and neurochemical phenotype of attention-deficit/hyperactivity disorder (ADHD). Here, we characterized the striatal GABA transport of SHR and investigated whether caffeine, a non-selective antagonist of adenosine receptors, could influence GABAergic circuitry. For this purpose, ex vivo striatal slices of SHR and Wistar (control strain) on the 35th postnatal day were dissected and incubated with [3H]-GABA to quantify the basal levels of uptake and release. SHR exhibited a reduced [3H]-GABA uptake and release, suggesting a defective striatal GABAergic transport system. GAT-1 appears to be the primary transporter for [3H]-GABA uptake in SHR striatum, as GAT-1 selective blocker, NO-711, completely abolished it. We also verified that acute exposure of striatal slices to caffeine improved [3H]-GABA uptake and release in SHR, whereas Wistar rats were not affected. GABA-uptake increase and cAMP accumulation promoted by caffeine was reverted by A1R activation with N6-cyclohexyl adenosine (CHA). As expected, the pharmacological blockade of cAMP-PKA signaling by H-89 also prevented caffeine-mediated [3H]-GABA uptake increment. Interestingly, a single caffeine exposure did not affect GAT-1 or A1R protein density in SHR, which was not different from Wistar protein levels, suggesting that the GAT-1-dependent transport in SHR has a defective functional activity rather than lower protein expression. The current data support that caffeine regulates GAT-1 function and improves striatal GABA transport via A1R-cAMP-PKA signaling, specifically in SHR. These results reinforce that caffeine may have therapeutic use in disorders where the GABA transport system is impaired.
Collapse
Affiliation(s)
| | | | | | - Pâmella de Moura
- Laboratório de Neurofarmacologia, Instituto Biomédico, Niterói, RJ, Brazil
| | | | - Robertta Silva Martins
- Laboratório de Neurofarmacologia, Instituto Biomédico, Niterói, RJ, Brazil
- Laboratório de Neurobiologia Celular E Molecular, Instituto de Biofísica Carlos Chagas Filho, Universidade Federal Do Rio de Janeiro, Rio de Janeiro, Brazil
| | | | | | - Ricardo Augusto de Melo Reis
- Laboratório de Neuroquímica, Instituto de Biofísica Carlos Chagas Filho, Universidade Federal Do Rio de Janeiro, Rio de Janeiro, Brazil
| | - Gustavo Costa Ferreira
- Laboratório de Neuroenergética E Erros Inatos Do Metabolismo, Instituto de Bioquímica Médica Leopoldo de Meis, Universidade Federal Do Rio de Janeiro, Rio de Janeiro, Brazil
| | - Alex Christian Manhães
- Laboratório de Neurofisiologia, Instituto de Biologia, Universidade Do Estado Do Rio de Janeiro, Rio de Janeiro, Brazil
| | - Maurício Dos Santos Pereira
- Laboratório de Neurofarmacologia, Instituto Biomédico, Niterói, RJ, Brazil.
- Laboratório de Neurofisiologia Molecular, Departamento de Biologia Básica E Oral, Faculdade de Odontologia de Ribeirão Preto, Universidade de São Paulo, Ribeirão Preto, Brazil.
| |
Collapse
|
6
|
Ryan RM, Ingram SL, Scimemi A. Regulation of Glutamate, GABA and Dopamine Transporter Uptake, Surface Mobility and Expression. Front Cell Neurosci 2021; 15:670346. [PMID: 33927596 PMCID: PMC8076567 DOI: 10.3389/fncel.2021.670346] [Citation(s) in RCA: 21] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/21/2021] [Accepted: 03/15/2021] [Indexed: 01/31/2023] Open
Abstract
Neurotransmitter transporters limit spillover between synapses and maintain the extracellular neurotransmitter concentration at low yet physiologically meaningful levels. They also exert a key role in providing precursors for neurotransmitter biosynthesis. In many cases, neurons and astrocytes contain a large intracellular pool of transporters that can be redistributed and stabilized in the plasma membrane following activation of different signaling pathways. This means that the uptake capacity of the brain neuropil for different neurotransmitters can be dynamically regulated over the course of minutes, as an indirect consequence of changes in neuronal activity, blood flow, cell-to-cell interactions, etc. Here we discuss recent advances in the mechanisms that control the cell membrane trafficking and biophysical properties of transporters for the excitatory, inhibitory and modulatory neurotransmitters glutamate, GABA, and dopamine.
Collapse
Affiliation(s)
- Renae M. Ryan
- School of Medical Sciences, Faculty of Medicine and Health, University of Sydney, Sydney, NSW, Australia
| | - Susan L. Ingram
- Department of Neurological Surgery, Oregon Health & Science University, Portland, OR, United States
| | | |
Collapse
|
7
|
Bae M, Roh JD, Kim Y, Kim SS, Han HM, Yang E, Kang H, Lee S, Kim JY, Kang R, Jung H, Yoo T, Kim H, Kim D, Oh H, Han S, Kim D, Han J, Bae YC, Kim H, Ahn S, Chan AM, Lee D, Kim JW, Kim E. SLC6A20 transporter: a novel regulator of brain glycine homeostasis and NMDAR function. EMBO Mol Med 2021; 13:e12632. [PMID: 33428810 PMCID: PMC7863395 DOI: 10.15252/emmm.202012632] [Citation(s) in RCA: 31] [Impact Index Per Article: 7.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/29/2020] [Revised: 10/22/2020] [Accepted: 11/19/2020] [Indexed: 12/25/2022] Open
Abstract
Glycine transporters (GlyT1 and GlyT2) that regulate levels of brain glycine, an inhibitory neurotransmitter with co-agonist activity for NMDA receptors (NMDARs), have been considered to be important targets for the treatment of brain disorders with suppressed NMDAR function such as schizophrenia. However, it remains unclear whether other amino acid transporters expressed in the brain can also regulate brain glycine levels and NMDAR function. Here, we report that SLC6A20A, an amino acid transporter known to transport proline based on in vitro data but is understudied in the brain, regulates proline and glycine levels and NMDAR function in the mouse brain. SLC6A20A transcript and protein levels were abnormally increased in mice carrying a mutant PTEN protein lacking the C terminus through enhanced β-catenin binding to the Slc6a20a gene. These mice displayed reduced extracellular levels of brain proline and glycine and decreased NMDAR currents. Elevating glycine levels back to normal ranges by antisense oligonucleotide-induced SLC6A20 knockdown, or the competitive GlyT1 antagonist sarcosine, normalized NMDAR currents and repetitive climbing behavior observed in these mice. Conversely, mice lacking SLC6A20A displayed increased extracellular glycine levels and NMDAR currents. Lastly, both mouse and human SLC6A20 proteins mediated proline and glycine transports, and SLC6A20 proteins could be detected in human neurons. These results suggest that SLC6A20 regulates proline and glycine homeostasis in the brain and that SLC6A20 inhibition has therapeutic potential for brain disorders involving NMDAR hypofunction.
Collapse
Affiliation(s)
- Mihyun Bae
- Center for Synaptic Brain DysfunctionsInstitute for Basic Science (IBS)DaejeonKorea
| | - Junyeop Daniel Roh
- Department of Biological SciencesKorea Advanced Institute for Science and Technology (KAIST)DaejeonKorea
| | - Youjoung Kim
- Department of Biological SciencesKorea Advanced Institute for Science and Technology (KAIST)DaejeonKorea
| | - Seong Soon Kim
- Therapeutics and Biotechnology DivisionKorea Research Institute of Chemical Technology (KRICT)DaejeonKorea
| | - Hye Min Han
- Department of Anatomy and NeurobiologySchool of DentistryKyungpook National UniversityDaeguKorea
| | - Esther Yang
- Department of Anatomy and Division of Brain Korea 21Biomedical ScienceCollege of MedicineKorea UniversitySeoulKorea
| | - Hyojin Kang
- Division of National SupercomputingKISTIDaejeonKorea
| | - Suho Lee
- Center for Synaptic Brain DysfunctionsInstitute for Basic Science (IBS)DaejeonKorea
| | - Jin Yong Kim
- Department of Anatomy and Division of Brain Korea 21Biomedical ScienceCollege of MedicineKorea UniversitySeoulKorea
| | - Ryeonghwa Kang
- Department of Biological SciencesKorea Advanced Institute for Science and Technology (KAIST)DaejeonKorea
| | - Hwajin Jung
- Center for Synaptic Brain DysfunctionsInstitute for Basic Science (IBS)DaejeonKorea
| | - Taesun Yoo
- Center for Synaptic Brain DysfunctionsInstitute for Basic Science (IBS)DaejeonKorea
| | - Hyosang Kim
- Department of Biological SciencesKorea Advanced Institute for Science and Technology (KAIST)DaejeonKorea
| | - Doyoun Kim
- Center for Synaptic Brain DysfunctionsInstitute for Basic Science (IBS)DaejeonKorea
| | - Heejeong Oh
- Department of Biological SciencesKorea Advanced Institute for Science and Technology (KAIST)DaejeonKorea
| | - Sungwook Han
- Department of Biological SciencesKorea Advanced Institute for Science and Technology (KAIST)DaejeonKorea
| | - Dayeon Kim
- Graduate School of Medical Science and EngineeringKAISTDaejeonKorea
| | - Jinju Han
- Graduate School of Medical Science and EngineeringKAISTDaejeonKorea
| | - Yong Chul Bae
- Department of Anatomy and NeurobiologySchool of DentistryKyungpook National UniversityDaeguKorea
| | - Hyun Kim
- Department of Anatomy and Division of Brain Korea 21Biomedical ScienceCollege of MedicineKorea UniversitySeoulKorea
| | - Sunjoo Ahn
- Therapeutics and Biotechnology DivisionKorea Research Institute of Chemical Technology (KRICT)DaejeonKorea
| | - Andrew M Chan
- School of Biomedical SciencesThe Chinese University of Hong KongHong KongHong Kong SARChina
| | - Daeyoup Lee
- Department of Biological SciencesKorea Advanced Institute for Science and Technology (KAIST)DaejeonKorea
| | - Jin Woo Kim
- Department of Biological SciencesKorea Advanced Institute for Science and Technology (KAIST)DaejeonKorea
| | - Eunjoon Kim
- Center for Synaptic Brain DysfunctionsInstitute for Basic Science (IBS)DaejeonKorea
- Department of Biological SciencesKorea Advanced Institute for Science and Technology (KAIST)DaejeonKorea
| |
Collapse
|
8
|
Mechanisms of action of clozapine in the treatment of neuroleptic-resistant and neuroleptic-intolerant schizophrenia. Eur Psychiatry 2020; 10 Suppl 1:39s-46s. [DOI: 10.1016/0767-399x(96)80083-6] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/30/2022] Open
Abstract
SummaryThe mechanisms of action which account for the effectiveness of clozapine as a pharmacotherapy for the treatment of neuroleptic non-responders and neuroleptic intolerant schizophrenic subjects remain elusive. We review recent data concerning the actions of clozapine in laboratory animals, and discuss the likely sites of action of clozapine and the receptors through which clozapine acts. We suggest that actions at dopamine D2 receptors in the caudate nucleus and putamen underlie the extrapyramidal side effects of conventional neuroleptics. In contrast, we propose that clozapine acts in the prefrontal cortex, specifically targeting an as yet unidentified DA receptor of the D2 family, to exert therapeutic actions in neuroleptic non-responders. We suggest that the ability of clozapine to augment extracellular dopamine levels in the prefrontal cortex may represent a key mechanism contributing to the therapeutic effects of this drug, and suggest some alternative approaches which might be expected to result in effects similar to those of clozapine.
Collapse
|
9
|
Fattorini G, Melone M, Conti F. A Reappraisal of GAT-1 Localization in Neocortex. Front Cell Neurosci 2020; 14:9. [PMID: 32116556 PMCID: PMC7031676 DOI: 10.3389/fncel.2020.00009] [Citation(s) in RCA: 17] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/29/2019] [Accepted: 01/13/2020] [Indexed: 12/31/2022] Open
Abstract
γ-Aminobutyric acid (GABA) transporter (GAT)-1, the major GABA transporter in the brain, plays a key role in modulating GABA signaling and is involved in the pathophysiology of several neuropsychiatric diseases, including epilepsy. The original description of GAT-1 as a neuronal transporter has guided the interpretation of the findings of all physiological, pharmacological, genetic, or clinical studies. However, evidence published in the past few years, some of which is briefly reviewed herein, does not seem to be consistent with a neurocentric view of GAT-1 function and calls for more detailed analysis of its localization. We therefore performed a thorough systematic assessment of GAT-1 localization in neocortex and subcortical white matter. In line with earlier work, we found that GAT-1 was robustly expressed in axon terminals forming symmetric synapses and in astrocytic processes, whereas its astrocytic expression was more diffuse than expected and, even more surprisingly, immature and mature oligodendrocytes and microglial cells also expressed the transporter. These data indicate that the era of “neuronal” and “glial” GABA transporters has finally come to a close and provide a wider perspective from which to view GABA-mediated physiological phenomena. In addition, given the well-known involvement of astrocytes, oligodendrocytes, and microglial cells in physiological as well as pathological conditions, the demonstration of functional GAT-1 in these cells is expected to provide greater insight into the phenomena occurring in the diseased brain as well as to prompt a reassessment of earlier findings.
Collapse
Affiliation(s)
- Giorgia Fattorini
- Department of Experimental and Clinical Medicine, Faculty of Medicine and Surgery, Università Politecnica delle Marche, Ancona, Italy.,Center for Neurobiology of Aging, IRCCS INRCA, Ancona, Italy
| | - Marcello Melone
- Department of Experimental and Clinical Medicine, Faculty of Medicine and Surgery, Università Politecnica delle Marche, Ancona, Italy.,Center for Neurobiology of Aging, IRCCS INRCA, Ancona, Italy
| | - Fiorenzo Conti
- Department of Experimental and Clinical Medicine, Faculty of Medicine and Surgery, Università Politecnica delle Marche, Ancona, Italy.,Center for Neurobiology of Aging, IRCCS INRCA, Ancona, Italy.,Fondazione di Medicina Molecolare, Università Politecnica delle Marche, Ancona, Italy
| |
Collapse
|
10
|
Γ-Aminobutyric acid in adult brain: an update. Behav Brain Res 2019; 376:112224. [DOI: 10.1016/j.bbr.2019.112224] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/20/2019] [Revised: 09/09/2019] [Accepted: 09/09/2019] [Indexed: 01/21/2023]
|
11
|
Fattorini G, Catalano M, Melone M, Serpe C, Bassi S, Limatola C, Conti F. Microglial expression of GAT-1 in the cerebral cortex. Glia 2019; 68:646-655. [PMID: 31692106 DOI: 10.1002/glia.23745] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/27/2019] [Revised: 10/12/2019] [Accepted: 10/18/2019] [Indexed: 01/08/2023]
Abstract
Microglial cells are the immune cells of the brain that, by sensing the microenvironment, permit a correct brain development and function. They communicate with other glial cells and with neurons, releasing and responding to a number of molecules that exert effects on surrounding cells. Among these, neurotransmitters and, in particular, gamma-aminobutyric acid (GABA) has recently gained interest in this context. We demonstrated the expression of GABA transporter 1 (GAT-1) in microglial cells both in soma and cell processes. We show that microglial cell treatment with 1,2,5,6-tetrahydro-1-[2-[[(diphenylmethylene)amino]oxy]ethyl]-3-pyridinecarboxylic acid hydrochloride (NNC-711), a potent and selective GAT-1 inhibitor, significantly reduced Na+ -dependent GABA uptake. On the other hand, GABA uptake was significantly increased by cell treatment with (S)-1-[2-[tris(4-methoxyphenyl)methoxy]ethyl]-3-piperidinecarboxylic acid (SNAP-5114), a GAT-2/3 inhibitor, and this effect was completely blocked by the botulinum toxin BoNT/C1, that specifically cleaves and inactives syntaxin 1A (STX1A). Overall, these findings show that microglial cells express GAT-1 and indicate that STX1A plays an important role in the regulation of GAT-1-dependent GABA uptake in microglia.
Collapse
Affiliation(s)
- Giorgia Fattorini
- Department of Experimental and Clinical Medicine, Section of Neuroscience and Cell Biology, Università Politecnica delle Marche, Ancona, Italy.,Center for Neurobiology of Aging, IRCCS INRCA, Ancona, Italy
| | - Myriam Catalano
- Department of Physiology and Pharmacology, Sapienza University of Rome, Laboratory Affiliated to Istituto Pasteur Italia, Rome, Italy
| | - Marcello Melone
- Department of Experimental and Clinical Medicine, Section of Neuroscience and Cell Biology, Università Politecnica delle Marche, Ancona, Italy.,Center for Neurobiology of Aging, IRCCS INRCA, Ancona, Italy
| | - Carmela Serpe
- Department of Physiology and Pharmacology, Sapienza University of Rome, Laboratory Affiliated to Istituto Pasteur Italia, Rome, Italy
| | - Silvia Bassi
- Department of Experimental and Clinical Medicine, Section of Neuroscience and Cell Biology, Università Politecnica delle Marche, Ancona, Italy
| | - Cristina Limatola
- Department of Physiology and Pharmacology, Sapienza University of Rome, Laboratory Affiliated to Istituto Pasteur Italia, Rome, Italy.,IRCCS Neuromed, Via Atinense, Pozzilli, Italy
| | - Fiorenzo Conti
- Department of Experimental and Clinical Medicine, Section of Neuroscience and Cell Biology, Università Politecnica delle Marche, Ancona, Italy.,Center for Neurobiology of Aging, IRCCS INRCA, Ancona, Italy
| |
Collapse
|
12
|
Kubo Y, Akanuma SI, Hosoya KI. Impact of SLC6A Transporters in Physiological Taurine Transport at the Blood-Retinal Barrier and in the Liver. Biol Pharm Bull 2017; 39:1903-1911. [PMID: 27904033 DOI: 10.1248/bpb.b16-00597] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022]
Abstract
Cumulative studies showed that taurine (2-aminoethanesulfonic acid) contributes to a variety of physiological events. Transport study suggested the cellular taurine transport in an Na+- and Cl--dependent manner, and the several members of SLC6A family have been shown as taurine transporter. At the inner blood-retinal barrier (BRB), taurine transporter (TauT/SLC6A) is involved in the transport of taurine to the retina from the circulating blood. The involvement of TauT is also suggested in γ-aminobutyric acid (GABA) transport at the inner BRB, and its role is assumed in the elimination of GABA from the retinal interstitial fluid. In the retina, taurine is thought to be a major organic osmolyte, and its influx and efflux through TauT and volume-sensitive organic osmolyte and anion channel (VSOAC) in Müller cells regulate the osmolarity in the retinal microenvironment to maintain a healthy retina. In the liver, hepatocytes take up taurine via GABA transporter 2 (GAT2/SLC6A13, the orthologue of mouse GAT3) expressed at the sinusoidal membrane of periportal hepatocytes, contributing to the metabolism of bile acid. Site-directed mutagenesis study suggests amino acid residues that are crucial in the recognition of substrates by GATs and TauT. The evidence suggests the physiological impact of taurine transporters in tissues.
Collapse
Affiliation(s)
- Yoshiyuki Kubo
- Department of Pharmaceutics, Graduate School of Medicine and Pharmaceutical Sciences, University of Toyama
| | | | | |
Collapse
|
13
|
Fattorini G, Melone M, Sánchez-Gómez MV, Arellano RO, Bassi S, Matute C, Conti F. GAT-1 mediated GABA uptake in rat oligodendrocytes. Glia 2017; 65:514-522. [PMID: 28071826 DOI: 10.1002/glia.23108] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/29/2015] [Revised: 11/30/2016] [Accepted: 12/05/2016] [Indexed: 01/18/2023]
Abstract
Stimulated by the results of a recent paper on the effects of tiagabine, a selective inhibitor of the main GABA transporter GAT-1, on oligodendrogenesis, we verified the possibility that GAT-1 may be expressed in oligodendrocytes using immunocytochemical methods and functional assays. Light microscopic analysis of the subcortical white matter of all animals revealed the presence of numerous GAT-1+ cells of different size (from 3 to 29 µm) and morphology. An electron microscope analysis revealed that, besides fibrous astrocytes and interstitial neurons, GAT-1 immunoreactivity was present in immature and mature oligodendrocytes. Co-localization studies between GAT-1 and markers specific for oligodendrocytes (NG2 and RIP) showed that about 12% of GAT-1 positive cells in the white matter were immature oligodendrocytes, while about 15% were mature oligodendrocytes. In vitro functional assays showed that oligodendrocytes exhibit tiagabine-sensitive Na+ -dependent GABA uptake. Although relationships between GABA and oligodendrocytes have been known for many years, this is the first demonstration that GAT-1 is expressed in oligodendrocytes. The present results on the one hand definitely closes the era of "neuronal" and "glial" GABA transporters, on the other they suggest that oligodendrocytes may contribute to pathophysiology of the several diseases in which GAT-1 have been implicated to date. GLIA 2017;65:514-522.
Collapse
Affiliation(s)
- Giorgia Fattorini
- Department of Experimental and Clinical Medicine, Section of Neuroscience and Cell Biology, Università Politecnica delle Marche, Ancona, 60026, Italy.,Center for Neurobiology of Aging, INRCA IRCCS, Ancona, 60121, Italy
| | - Marcello Melone
- Department of Experimental and Clinical Medicine, Section of Neuroscience and Cell Biology, Università Politecnica delle Marche, Ancona, 60026, Italy.,Center for Neurobiology of Aging, INRCA IRCCS, Ancona, 60121, Italy
| | - María Victoria Sánchez-Gómez
- Achucarro Basque Center for Neuroscience, CIBERNED, and Departamento de Neurociencias, Universidad del País Vasco, Leioa, 48940, Spain
| | - Rogelio O Arellano
- Instituto de Neurobiología, Universidad Nacional Autónoma de México, Querétaro, México
| | - Silvia Bassi
- Department of Experimental and Clinical Medicine, Section of Neuroscience and Cell Biology, Università Politecnica delle Marche, Ancona, 60026, Italy
| | - Carlos Matute
- Achucarro Basque Center for Neuroscience, CIBERNED, and Departamento de Neurociencias, Universidad del País Vasco, Leioa, 48940, Spain
| | - Fiorenzo Conti
- Department of Experimental and Clinical Medicine, Section of Neuroscience and Cell Biology, Università Politecnica delle Marche, Ancona, 60026, Italy.,Center for Neurobiology of Aging, INRCA IRCCS, Ancona, 60121, Italy.,Fondazione di Medicina Molecolare, Università Politecnica delle Marche, Ancona, 60026, Italy
| |
Collapse
|
14
|
Eskandari S, Willford SL, Anderson CM. Revised Ion/Substrate Coupling Stoichiometry of GABA Transporters. ADVANCES IN NEUROBIOLOGY 2017; 16:85-116. [PMID: 28828607 DOI: 10.1007/978-3-319-55769-4_5] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/26/2023]
Abstract
The purpose of this review is to highlight recent evidence in support of a 3 Na+: 1 Cl-: 1 GABA coupling stoichiometry for plasma membrane GABA transporters (SLC6A1 , SLC6A11 , SLC6A12 , SLC6A13 ) and how the revised stoichiometry impacts our understanding of the contribution of GABA transporters to GABA homeostasis in synaptic and extrasynaptic regions in the brain under physiological and pathophysiological states. Recently, our laboratory probed the GABA transporter stoichiometry by analyzing the results of six independent measurements, which included the shifts in the thermodynamic transporter reversal potential caused by changes in the extracellular Na+, Cl-, and GABA concentrations, as well as the ratio of charge flux to substrate flux for Na+, Cl-, and GABA under voltage-clamp conditions. The shifts in the transporter reversal potential for a tenfold change in the external concentration of Na+, Cl-, and GABA were 84 ± 4, 30 ± 1, and 29 ± 1 mV, respectively. Charge flux to substrate flux ratios were 0.7 ± 0.1 charges/Na+, 2.0 ± 0.2 charges/Cl-, and 2.1 ± 0.1 charges/GABA. We then compared these experimental results with the predictions of 150 different transporter stoichiometry models, which included 1-5 Na+, 0-5 Cl-, and 1-5 GABA per transport cycle. Only the 3 Na+: 1 Cl-: 1 GABA stoichiometry model correctly predicts the results of all six experimental measurements. Using the revised 3 Na+: 1 Cl-: 1 GABA stoichiometry, we propose that the GABA transporters mediate GABA uptake under most physiological conditions. Transporter-mediated GABA release likely takes place under pathophysiological or extreme physiological conditions.
Collapse
Affiliation(s)
- Sepehr Eskandari
- Biological Sciences Department, California State Polytechnic University, Pomona, CA, 91768, USA.
| | - Samantha L Willford
- Biological Sciences Department, California State Polytechnic University, Pomona, CA, 91768, USA
| | - Cynthia M Anderson
- Biological Sciences Department, California State Polytechnic University, Pomona, CA, 91768, USA
| |
Collapse
|
15
|
Albers HE, Walton JC, Gamble KL, McNeill JK, Hummer DL. The dynamics of GABA signaling: Revelations from the circadian pacemaker in the suprachiasmatic nucleus. Front Neuroendocrinol 2017; 44:35-82. [PMID: 27894927 PMCID: PMC5225159 DOI: 10.1016/j.yfrne.2016.11.003] [Citation(s) in RCA: 72] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/30/2016] [Revised: 10/16/2016] [Accepted: 11/22/2016] [Indexed: 12/31/2022]
Abstract
Virtually every neuron within the suprachiasmatic nucleus (SCN) communicates via GABAergic signaling. The extracellular levels of GABA within the SCN are determined by a complex interaction of synthesis and transport, as well as synaptic and non-synaptic release. The response to GABA is mediated by GABAA receptors that respond to both phasic and tonic GABA release and that can produce excitatory as well as inhibitory cellular responses. GABA also influences circadian control through the exclusively inhibitory effects of GABAB receptors. Both GABA and neuropeptide signaling occur within the SCN, although the functional consequences of the interactions of these signals are not well understood. This review considers the role of GABA in the circadian pacemaker, in the mechanisms responsible for the generation of circadian rhythms, in the ability of non-photic stimuli to reset the phase of the pacemaker, and in the ability of the day-night cycle to entrain the pacemaker.
Collapse
Affiliation(s)
- H Elliott Albers
- Center for Behavioral Neuroscience, Atlanta, GA 30302, United States; Neuroscience Institute, Georgia State University, Atlanta, GA 30302, United States.
| | - James C Walton
- Center for Behavioral Neuroscience, Atlanta, GA 30302, United States; Neuroscience Institute, Georgia State University, Atlanta, GA 30302, United States
| | - Karen L Gamble
- Department of Psychiatry and Behavioral Neurobiology, University of Alabama at Birmingham, Birmingham, AL 35294, United States
| | - John K McNeill
- Center for Behavioral Neuroscience, Atlanta, GA 30302, United States; Neuroscience Institute, Georgia State University, Atlanta, GA 30302, United States
| | - Daniel L Hummer
- Center for Behavioral Neuroscience, Atlanta, GA 30302, United States; Department of Psychology, Morehouse College, Atlanta, GA 30314, United States
| |
Collapse
|
16
|
Abstract
Neurotransmitters are normally released from neurons via calcium-dependent exocytosis of synaptic vesicles. However, after blockade of vesicular release by removal of calcium, or treatment with tetanus toxin, neurotransmitter release can still occur. In the case of GABA, nonvesicular release results from reversal of its uptake transporter, found on both neurons and glia. These GABA transporters are sodium-dependent and electrogenic, and therefore can be induced to operate in reverse by cell depolarization or by breakdown of the sodium gradient. Although demonstrated biochemically, less is known about whether this form of release occurs in vivo or whether it results in electrophysiological effects. Because conditions that favor reversal of the GABA transporter occur during high-frequency firing, nonvesicular GABA release may occur with excessive neuronal activity, such as during seizures. NEUROSCIENTIST 3:151-157, 1997
Collapse
|
17
|
Evidence for a Revised Ion/Substrate Coupling Stoichiometry of GABA Transporters. J Membr Biol 2015; 248:795-810. [DOI: 10.1007/s00232-015-9797-6] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/17/2015] [Accepted: 03/19/2015] [Indexed: 10/23/2022]
|
18
|
Yamashita A, Hamada A, Suhara Y, Kawabe R, Yanase M, Kuzumaki N, Narita M, Matsui R, Okano H, Narita M. Astrocytic activation in the anterior cingulate cortex is critical for sleep disorder under neuropathic pain. Synapse 2014; 68:235-47. [DOI: 10.1002/syn.21733] [Citation(s) in RCA: 66] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/07/2013] [Accepted: 01/09/2014] [Indexed: 01/21/2023]
Affiliation(s)
- Akira Yamashita
- Department of Pharmacology; Hoshi University School of Pharmacy and Pharmaceutical Sciences; 2-4-41 Ebara Shinagawa-ku Tokyo 142-8501 Japan
| | - Asami Hamada
- Department of Pharmacology; Hoshi University School of Pharmacy and Pharmaceutical Sciences; 2-4-41 Ebara Shinagawa-ku Tokyo 142-8501 Japan
| | - Yuki Suhara
- Department of Pharmacology; Hoshi University School of Pharmacy and Pharmaceutical Sciences; 2-4-41 Ebara Shinagawa-ku Tokyo 142-8501 Japan
| | - Rui Kawabe
- Department of Pharmacology; Hoshi University School of Pharmacy and Pharmaceutical Sciences; 2-4-41 Ebara Shinagawa-ku Tokyo 142-8501 Japan
| | - Makoto Yanase
- Department of Pharmacology; Hoshi University School of Pharmacy and Pharmaceutical Sciences; 2-4-41 Ebara Shinagawa-ku Tokyo 142-8501 Japan
| | - Naoko Kuzumaki
- Department of Physiology; Keio University School of Medicine; 35 Shinanomachi Shinjuku-ku Tokyo 160-8582 Japan
| | - Michiko Narita
- Department of Pharmacology; Hoshi University School of Pharmacy and Pharmaceutical Sciences; 2-4-41 Ebara Shinagawa-ku Tokyo 142-8501 Japan
| | - Ryosuke Matsui
- Department of Molecular and Systems Biology; Graduate School of Biostudies, Kyoto University; Yoshida Sakyo-ku Kyoto 606-8501 Japan
| | - Hideyuki Okano
- Department of Physiology; Keio University School of Medicine; 35 Shinanomachi Shinjuku-ku Tokyo 160-8582 Japan
| | - Minoru Narita
- Department of Pharmacology; Hoshi University School of Pharmacy and Pharmaceutical Sciences; 2-4-41 Ebara Shinagawa-ku Tokyo 142-8501 Japan
| |
Collapse
|
19
|
Melone M, Ciappelloni S, Conti F. A quantitative analysis of cellular and synaptic localization of GAT-1 and GAT-3 in rat neocortex. Brain Struct Funct 2013; 220:885-97. [PMID: 24368619 DOI: 10.1007/s00429-013-0690-8] [Citation(s) in RCA: 48] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/01/2013] [Accepted: 12/10/2013] [Indexed: 11/24/2022]
Abstract
High-affinity plasma membrane GABA transporters GAT-1 and GAT-3 contribute to the modulation of GABA-mediated inhibition in adult mammalian cerebral cortex. How GATs regulate inhibition in neocortical circuits remains however poorly understood for the lack of information on key localizational features. In this study, we used quantitative pre- and post-embedding electron microscopy to define the distribution of GAT-1 and GAT-3 in elements contributing to synapses and to unveil their ultrastructural organization at adult cortical GABAergic synapses. GAT-1 and GAT-3 were found in both neuronal and astrocytic processes: GAT-1 was prevalently segregated in neuronal elements and in profiles contributing to synapses, whereas GAT-3 was mostly expressed in astrocytes and did not exhibit a preferential distribution in elements contributing to synapses. Analysis of the ultrastructural distribution of GAT-1 and GAT-3 in the plasma membrane of axon terminals and perisynaptic astrocytic processes of symmetric synapses in relation to the active zone revealed that GAT-1 was more concentrated in restricted perisynaptic and extrasynaptic regions, whereas GAT-3 was prominent in extrasynaptic areas. These studies provide a basis for understanding the role GAT-1 and GAT-3 play in the modulation of GABA-mediated phasic and tonic inhibition in cerebral cortex.
Collapse
Affiliation(s)
- Marcello Melone
- Department of Experimental and Clinical Medicine, Section of Neuroscience and Cell Biology, Università Politecnica delle Marche, 60026, Ancona, Italy,
| | | | | |
Collapse
|
20
|
Wang T, Rusu SI, Hruskova B, Turecek R, Borst JGG. Modulation of synaptic depression of the calyx of Held synapse by GABA(B) receptors and spontaneous activity. J Physiol 2013; 591:4877-94. [PMID: 23940376 DOI: 10.1113/jphysiol.2013.256875] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/10/2023] Open
Abstract
The calyx of Held synapse of the medial nucleus of the trapezoid body is a giant axosomatic synapse in the auditory brainstem, which acts as a relay synapse showing little dependence of its synaptic strength on firing frequency. The main mechanism that is responsible for its resistance to synaptic depression is its large number of release sites with low release probability. Here, we investigated the contribution of presynaptic GABA(B) receptors and spontaneous activity to release probability both in vivo and in vitro in young-adult mice. Maximal activation of presynaptic GABA(B) receptors by baclofen reduced synaptic output by about 45% in whole-cell voltage clamp slice recordings, which was accompanied by a reduction in short-term depression. A similar reduction in transmission was observed when baclofen was applied in vivo by microiontophoresis during juxtacellular recordings using piggyback electrodes. No significant change in synaptic transmission was observed during application of the GABA(B) receptor antagonist CGP54626 both during in vivo and slice recordings, suggesting a low ambient GABA concentration. Interestingly, we observed that synapses with a high spontaneous frequency showed almost no synaptic depression during auditory stimulation, whereas synapses with a low spontaneous frequency did depress during noise bursts. Our data thus suggest that spontaneous firing can tonically reduce release probability in vivo. In addition, our data show that the ambient GABA concentration in the auditory brainstem is too low to activate the GABA(B) receptor at the calyx of Held significantly, but that activation of GABA(B) receptors can reduce sound-evoked synaptic depression.
Collapse
Affiliation(s)
- Tiantian Wang
- J. G. G. Borst: Department of Neuroscience, Erasmus MC, University Medical Center Rotterdam, Dr. Molewaterplein 50, 3015 GE Rotterdam, The Netherlands.
| | | | | | | | | |
Collapse
|
21
|
Usui T, Kubo Y, Akanuma SI, Hosoya KI. β-Alanine and l-histidine transport across the inner blood-retinal barrier: Potential involvement in l-carnosine supply. Exp Eye Res 2013; 113:135-42. [DOI: 10.1016/j.exer.2013.06.002] [Citation(s) in RCA: 24] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/25/2013] [Revised: 05/18/2013] [Accepted: 06/03/2013] [Indexed: 11/25/2022]
|
22
|
Kataoka K, Hara K, Haranishi Y, Terada T, Sata T. The antinociceptive effect of SNAP5114, a gamma-aminobutyric acid transporter-3 inhibitor, in rat experimental pain models. Anesth Analg 2013; 116:1162-1169. [PMID: 23456665 DOI: 10.1213/ane.0b013e318282dda7] [Citation(s) in RCA: 21] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/05/2022]
Abstract
BACKGROUND Gamma-aminobutyric acid (GABA) is the primary inhibitory neurotransmitter in the mammalian central nervous system. GABAergic transmission has an important role in regulating nociception at the spinal dorsal horn. It is terminated by rapid uptake of the neurotransmitter from the synaptic cleft into neurons and glial cells, via specific GABA transporters (GATs). Among the 4 GATs, GAT-3 has the greatest expression in central nervous system regions closely associated with nociceptive transmission, including the spinal cord. In this study, we examined the antinociceptive effect of intrathecal administration of a selective GAT-3 inhibitor, SNAP5114, on acute, inflammatory, and neuropathic pain in experimental models. METHODS Male Sprague-Dawley rats were used to assess thermal, mechanical, and chemical nociception in the tail flick and hotplate tests, the paw pressure test, and the formalin test. A rotarod test was performed to assess motor function. Chronic constriction injury to the sciatic nerve was induced in the rats. The electronic von Frey test and the plantar test were then performed to assess mechanical allodynia and thermal hyperalgesia. SNAP5114 (10, 50, 100, or 200 μg) was administered intrathecally to examine antinociceptive activity. To confirm whether the action of SNAP5114 was mediated by GABAergic transmission, the GABAA receptor antagonist bicuculline (0.3 μg) or the GABAB receptor antagonist CGP35348 (30 μg) was administered intrathecally before 200 μg of SNAP5114 in the tail flick test, the formalin test, and the electronic von Frey test. RESULTS Spinally applied SNAP5114 in normal rats dose-dependently prolonged withdrawal latencies in the tail flick test and suppressed the late-phase response in the formalin test. SNAP5114 did not affect motor performance. In the chronic constriction injury rats, SNAP5114 inhibited mechanical allodynia dose-dependently. The antinociceptive action of SNAP5114 was partially reversed by bicuculline or CGP35348 at doses at which the antagonist alone did not affect baseline behavioral responses. CONCLUSIONS These results suggest that SNAP5114 exerts antinociceptive effects by activating GABAA and GABAB receptors in the spinal cord. The GAT-3 inhibitor may prove useful in treatment of various painful conditions.
Collapse
Affiliation(s)
- Kazunori Kataoka
- From the Department of Anesthesiology, University of Occupational and Environmental Health, School of Medicine, Kitakyushu, Japan
| | | | | | | | | |
Collapse
|
23
|
Abstract
The solute carrier 6 (SLC6) family of the human genome comprises transporters for neurotransmitters, amino acids, osmolytes and energy metabolites. Members of this family play critical roles in neurotransmission, cellular and whole body homeostasis. Malfunction or altered expression of these transporters is associated with a variety of diseases. Pharmacological inhibition of the neurotransmitter transporters in this family is an important strategy in the management of neurological and psychiatric disorders. This review provides an overview of the biochemical and pharmacological properties of the SLC6 family transporters.
Collapse
Affiliation(s)
- Stefan Bröer
- Research School of Biology, Australian National University, Canberra, ACT, Australia.
| | | |
Collapse
|
24
|
Abstract
The mammalian genome contains four genes encoding GABA transporters (GAT1, slc6a1; GAT2, slc6a13; GAT3, slc6a11; BGT1, slc6a12) and five glutamate transporter genes (EAAT1, slc1a3; EAAT2, slc1a2; EAAT3, slc1a1; EAAT4, slc1a6; EAAT5, slc1a7). These transporters keep the extracellular levels of GABA and excitatory amino acids low and provide amino acids for metabolic purposes. The various transporters have different properties both with respect to their transport functions and with respect to their ability to act as ion channels. Further, they are differentially regulated. To understand the physiological roles of the individual transporter subtypes, it is necessary to obtain information on their distributions and expression levels. Quantitative data are important as the functional capacity is limited by the number of transporter molecules. The most important and most abundant transporters for removal of transmitter glutamate in the brain are EAAT2 (GLT-1) and EAAT1 (GLAST), while GAT1 and GAT3 are the major GABA transporters in the brain. EAAT3 (EAAC1) does not appear to play a role in signal transduction, but plays other roles. Due to their high uncoupled anion conductance, EAAT4 and EAAT5 seem to be acting more like inhibitory glutamate receptors than as glutamate transporters. GAT2 and BGT1 are primarily expressed in the liver and kidney, but are also found in the leptomeninges, while the levels in brain tissue proper are too low to have any impact on GABA removal, at least in normal young adult mice. The present review will provide summary of what is currently known and will also discuss some methodological pitfalls.
Collapse
Affiliation(s)
- Yun Zhou
- The Neurotransporter Group, Department of Anatomy, Institute of Basic Medical Sciences, University of Oslo, Oslo, Norway
| | - Niels Christian Danbolt
- The Neurotransporter Group, Department of Anatomy, Institute of Basic Medical Sciences, University of Oslo, Oslo, Norway
- *Correspondence: Niels Christian Danbolt, The Neurotransporter Group, Department of Anatomy, Institute of Basic Medical Sciences, University of Oslo, P.O. Box 1105 Blindern, Oslo N-0317, Norway e-mail:
| |
Collapse
|
25
|
Borycz J, Borycz JA, Edwards TN, Boulianne GL, Meinertzhagen IA. The metabolism of histamine in the Drosophila optic lobe involves an ommatidial pathway: β-alanine recycles through the retina. ACTA ACUST UNITED AC 2012; 215:1399-411. [PMID: 22442379 DOI: 10.1242/jeb.060699] [Citation(s) in RCA: 33] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022]
Abstract
Flies recycle the photoreceptor neurotransmitter histamine by conjugating it to β-alanine to form β-alanyl-histamine (carcinine). The conjugation is regulated by Ebony, while Tan hydrolyses carcinine, releasing histamine and β-alanine. In Drosophila, β-alanine synthesis occurs either from uracil or from the decarboxylation of aspartate but detailed roles for the enzymes responsible remain unclear. Immunohistochemically detected β-alanine is present throughout the fly's entire brain, and is enhanced in the retina especially in the pseudocone, pigment and photoreceptor cells of the ommatidia. HPLC determinations reveal 10.7 ng of β-alanine in the wild-type head, roughly five times more than histamine. When wild-type flies drink uracil their head β-alanine increases more than after drinking l-aspartic acid, indicating the effectiveness of the uracil pathway. Mutants of black, which lack aspartate decarboxylase, cannot synthesize β-alanine from l-aspartate but can still synthesize it efficiently from uracil. Our findings demonstrate a novel function for pigment cells, which not only screen ommatidia from stray light but also store and transport β-alanine and carcinine. This role is consistent with a β-alanine-dependent histamine recycling pathway occurring not only in the photoreceptor terminals in the lamina neuropile, where carcinine occurs in marginal glia, but vertically via a long pathway that involves the retina. The lamina's marginal glia are also a hub involved in the storage and/or disposal of carcinine and β-alanine.
Collapse
Affiliation(s)
- Janusz Borycz
- Department of Psychology and Neuroscience, Life Sciences Centre, Dalhousie University, Halifax, Canada, B3H 4J1
| | | | | | | | | |
Collapse
|
26
|
Ando D, Kubo Y, Akanuma SI, Yoneyama D, Tachikawa M, Hosoya KI. Function and regulation of taurine transport in Müller cells under osmotic stress. Neurochem Int 2012; 60:597-604. [DOI: 10.1016/j.neuint.2012.02.018] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/31/2011] [Revised: 02/16/2012] [Accepted: 02/18/2012] [Indexed: 11/26/2022]
|
27
|
Dellal SS, Luo R, Otis TS. GABAA receptors increase excitability and conduction velocity of cerebellar parallel fiber axons. J Neurophysiol 2012; 107:2958-70. [PMID: 22378171 DOI: 10.1152/jn.01028.2011] [Citation(s) in RCA: 26] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022] Open
Abstract
In the adult mammalian brain, GABA(A) receptors (GABA(A)Rs) are responsible for the predominant forms of synaptic inhibition, but these receptors can excite neurons when the chloride equilibrium potential (E(Cl)) is depolarized. In many mature neurons, GABA(A)Rs are found on presynaptic terminals where they exert depolarizing effects. To understand whether excitatory GABA action affects axonal function, we used transverse cerebellar slices to measure the effects of photolysis of caged GABA on the initiation and propagation of compound parallel fiber (PF) action potentials (APs). Photolysis of caged GABA increased the amplitude and conduction velocity of PF APs; GABA reuptake blockers and a positive modulator of GABA(A)Rs enhanced these effects. In contrast, a modulator selective for δ-subunit-containing GABA(A)Rs did not enhance these effects and responsiveness remained in δ(-/-) mice, arguing that δ-subunit-containing GABA(A)Rs are not required. Synaptically released GABA also increased PF excitability, indicating that the mechanism is engaged by physiological signals. A Hodgkin-Huxley-style compartmental model of the PF axon and granule cell body was constructed, and this model recapitulated the GABA-dependent decrease in AP threshold and the increase in conduction velocity, features that were sensitive to E(Cl) and to the voltage dependence of sodium channel inactivation. The model also predicts that axonal GABA(A)Rs could affect orthodromic spike initiation. We conclude that GABA acting on cerebellar PFs facilitates both spike generation and propagation, allowing axons of granule cells to passively integrate signals from inhibitory interneurons and influence information flow in the input layer to the cerebellar cortex.
Collapse
Affiliation(s)
- Shlomo S Dellal
- Dept. of Neurobiology, David Geffen School of Medicine, UCLA, Los Angeles, CA 90095, USA
| | | | | |
Collapse
|
28
|
Jin XT, Galvan A, Wichmann T, Smith Y. Localization and Function of GABA Transporters GAT-1 and GAT-3 in the Basal Ganglia. Front Syst Neurosci 2011; 5:63. [PMID: 21847373 PMCID: PMC3148782 DOI: 10.3389/fnsys.2011.00063] [Citation(s) in RCA: 58] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/26/2011] [Accepted: 07/13/2011] [Indexed: 02/04/2023] Open
Abstract
GABA transporter type 1 and 3 (GAT-1 and GAT-3, respectively) are the two main subtypes of GATs responsible for the regulation of extracellular GABA levels in the central nervous system. These transporters are widely expressed in neuronal (mainly GAT-1) and glial (mainly GAT-3) elements throughout the brain, but most data obtained so far relate to their role in the regulation of GABA(A) receptor-mediated postsynaptic tonic and phasic inhibition in the hippocampus, cerebral cortex and cerebellum. Taking into consideration the key role of GABAergic transmission within basal ganglia networks, and the importance for these systems to be properly balanced to mediate normal basal ganglia function, we analyzed in detail the localization and function of GAT-1 and GAT-3 in the globus pallidus of normal and Parkinsonian animals, in order to further understand the substrate and possible mechanisms by which GABA transporters may regulate basal ganglia outflow, and may become relevant targets for new therapeutic approaches for the treatment of basal ganglia-related disorders. In this review, we describe the general features of GATs in the basal ganglia, and give a detailed account of recent evidence that GAT-1 and GAT-3 regulation can have a major impact on the firing rate and pattern of basal ganglia neurons through pre- and post-synaptic GABA(A)- and GABA(B)-receptor-mediated effects.
Collapse
Affiliation(s)
- Xiao-Tao Jin
- Division of Neuroscience, Yerkes National Primate Research Center and Department of Neurology, Emory UniversityAtlanta, GA, USA
| | - Adriana Galvan
- Division of Neuroscience, Yerkes National Primate Research Center and Department of Neurology, Emory UniversityAtlanta, GA, USA
| | - Thomas Wichmann
- Division of Neuroscience, Yerkes National Primate Research Center and Department of Neurology, Emory UniversityAtlanta, GA, USA
| | - Yoland Smith
- Division of Neuroscience, Yerkes National Primate Research Center and Department of Neurology, Emory UniversityAtlanta, GA, USA
| |
Collapse
|
29
|
Kristensen AS, Andersen J, Jørgensen TN, Sørensen L, Eriksen J, Loland CJ, Strømgaard K, Gether U. SLC6 neurotransmitter transporters: structure, function, and regulation. Pharmacol Rev 2011; 63:585-640. [PMID: 21752877 DOI: 10.1124/pr.108.000869] [Citation(s) in RCA: 617] [Impact Index Per Article: 44.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/16/2022] Open
Abstract
The neurotransmitter transporters (NTTs) belonging to the solute carrier 6 (SLC6) gene family (also referred to as the neurotransmitter-sodium-symporter family or Na(+)/Cl(-)-dependent transporters) comprise a group of nine sodium- and chloride-dependent plasma membrane transporters for the monoamine neurotransmitters serotonin (5-hydroxytryptamine), dopamine, and norepinephrine, and the amino acid neurotransmitters GABA and glycine. The SLC6 NTTs are widely expressed in the mammalian brain and play an essential role in regulating neurotransmitter signaling and homeostasis by mediating uptake of released neurotransmitters from the extracellular space into neurons and glial cells. The transporters are targets for a wide range of therapeutic drugs used in treatment of psychiatric diseases, including major depression, anxiety disorders, attention deficit hyperactivity disorder and epilepsy. Furthermore, psychostimulants such as cocaine and amphetamines have the SLC6 NTTs as primary targets. Beginning with the determination of a high-resolution structure of a prokaryotic homolog of the mammalian SLC6 transporters in 2005, the understanding of the molecular structure, function, and pharmacology of these proteins has advanced rapidly. Furthermore, intensive efforts have been directed toward understanding the molecular and cellular mechanisms involved in regulation of the activity of this important class of transporters, leading to new methodological developments and important insights. This review provides an update of these advances and their implications for the current understanding of the SLC6 NTTs.
Collapse
Affiliation(s)
- Anders S Kristensen
- Department of Medicinal Chemistry, Faculty of Pharmaceutical Sciences, University of Copenhagen, Copenhagen, Denmark.
| | | | | | | | | | | | | | | |
Collapse
|
30
|
Narita M, Niikura K, Nanjo-Niikura K, Narita M, Furuya M, Yamashita A, Saeki M, Matsushima Y, Imai S, Shimizu T, Asato M, Kuzumaki N, Okutsu D, Miyoshi K, Suzuki M, Tsukiyama Y, Konno M, Yomiya K, Matoba M, Suzuki T. Sleep disturbances in a neuropathic pain-like condition in the mouse are associated with altered GABAergic transmission in the cingulate cortex. Pain 2011; 152:1358-1372. [DOI: 10.1016/j.pain.2011.02.016] [Citation(s) in RCA: 58] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/08/2010] [Revised: 02/04/2011] [Accepted: 02/07/2011] [Indexed: 11/17/2022]
|
31
|
Anderson CM, Kidd PD, Eskandari S. GATMD: γ-aminobutyric acid transporter mutagenesis database. DATABASE-THE JOURNAL OF BIOLOGICAL DATABASES AND CURATION 2010; 2010:baq028. [PMID: 21131297 PMCID: PMC2997607 DOI: 10.1093/database/baq028] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 11/14/2022]
Abstract
Since the cloning of the first γ-aminobutyric acid (GABA) transporter (GAT1; SLC6A1) from rat brain in 1990, more than 50 published studies have provided structure-function information on investigator-designed rat and mouse GAT1 mutants. To date, more than 200 of 599 GAT1 residues have been subjected to mutagenesis experiments by substitution with different amino acids, and the resulting transporter functional properties have significantly advanced our understanding of the mechanism of Na+- and Cl⁻-coupled GABA transport by this important member of the neurotransmitter:sodium symporter family. Moreover, many studies have addressed the functional consequences of amino acid deletion or insertion at various positions along the primary sequence. The enormity of this growing body of structure-function information has prompted us to develop GABA Transporter Mutagenesis Database (GATMD), a web-accessible, relational database of manually annotated biochemical, functional and pharmacological data reported on GAT1-the most intensely studied GABA transporter isoform. As of the last update of GATMD, 52 GAT1 mutagenesis papers have yielded 3360 experimental records, which collectively contain a total of ∼100 000 annotated parameters. Database URL: http://physiology.sci.csupomona.edu/GATMD/
Collapse
Affiliation(s)
- Cynthia M Anderson
- Biological Sciences Department, California State Polytechnic University, Pomona, CA 91768-4032, USA
| | | | | |
Collapse
|
32
|
Expression of GABAergic receptors in mouse taste receptor cells. PLoS One 2010; 5:e13639. [PMID: 21049022 PMCID: PMC2964312 DOI: 10.1371/journal.pone.0013639] [Citation(s) in RCA: 20] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/10/2010] [Accepted: 10/04/2010] [Indexed: 12/05/2022] Open
Abstract
Background Multiple excitatory neurotransmitters have been identified in the mammalian taste transduction, with few studies focused on inhibitory neurotransmitters. Since the synthetic enzyme glutamate decarboxylase (GAD) for gamma-aminobutyric acid (GABA) is expressed in a subset of mouse taste cells, we hypothesized that other components of the GABA signaling pathway are likely expressed in this system. GABA signaling is initiated by the activation of either ionotropic receptors (GABAA and GABAC) or metabotropic receptors (GABAB) while it is terminated by the re-uptake of GABA through transporters (GATs). Methodology/Principal Findings Using reverse transcriptase-PCR (RT-PCR) analysis, we investigated the expression of different GABA signaling molecules in the mouse taste system. Taste receptor cells (TRCs) in the circumvallate papillae express multiple subunits of the GABAA and GABAB receptors as well as multiple GATs. Immunocytochemical analyses examined the distribution of the GABA machinery in the circumvallate papillae. Both GABAA-and GABAB- immunoreactivity were detected in the peripheral taste receptor cells. We also used transgenic mice that express green fluorescent protein (GFP) in either the Type II taste cells, which can respond to bitter, sweet or umami taste stimuli, or in the Type III GAD67 expressing taste cells. Thus, we were able to identify that GABAergic receptors are expressed in some Type II and Type III taste cells. Mouse GAT4 labeling was concentrated in the cells surrounding the taste buds with a few positively labeled TRCs at the margins of the taste buds. Conclusions/Significance The presence of GABAergic receptors localized on Type II and Type III taste cells suggests that GABA is likely modulating evoked taste responses in the mouse taste bud.
Collapse
|
33
|
Guo C, Hirano AA, Stella SL, Bitzer M, Brecha NC. Guinea pig horizontal cells express GABA, the GABA-synthesizing enzyme GAD 65, and the GABA vesicular transporter. J Comp Neurol 2010; 518:1647-69. [PMID: 20235161 DOI: 10.1002/cne.22294] [Citation(s) in RCA: 37] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
Abstract
Gamma-aminobutyric acid (GABA) is likely expressed in horizontal cells of all species, although conflicting physiological findings have led to considerable controversy regarding its role as a transmitter in the outer retina. This study has evaluated key components of the GABA system in the outer retina of guinea pig, an emerging retinal model system. The presence of GABA, its rate-limiting synthetic enzyme glutamic acid decarboxylase (GAD(65) and GAD(67) isoforms), the plasma membrane GABA transporters (GAT-1 and GAT-3), and the vesicular GABA transporter (VGAT) was evaluated by using immunohistochemistry with well-characterized antibodies. The presence of GAD(65) mRNA was also evaluated by using laser capture microdissection and reverse transcriptase-polymerase chain reaction. Specific GABA, GAD(65), and VGAT immunostaining was localized to horizontal cell bodies, as well as to their processes and tips in the outer plexiform layer. Furthermore, immunostaining of retinal whole mounts and acutely dissociated retinas showed GAD(65) and VGAT immunoreactivity in both A-type and B-type horizontal cells. However, these cells did not contain GAD(67), GAT-1, or GAT-3 immunoreactivity. GAD(65) mRNA was detected in horizontal cells, and sequencing of the amplified GAD(65) fragment showed approximately 85% identity with other mammalian GAD(65) mRNAs. These studies demonstrate the presence of GABA, GAD(65), and VGAT in horizontal cells of the guinea pig retina, and support the idea that GABA is synthesized from GAD(65), taken up into synaptic vesicles by VGAT, and likely released by a vesicular mechanism from horizontal cells.
Collapse
Affiliation(s)
- Chenying Guo
- Department of Neurobiology, David Geffen School of Medicine at UCLA, University of California, Los Angeles, California 90095, USA
| | | | | | | | | |
Collapse
|
34
|
|
35
|
Kirmse K, Kirischuk S, Grantyn R. Role of GABA transporter 3 in GABAergic synaptic transmission at striatal output neurons. Synapse 2009; 63:921-9. [DOI: 10.1002/syn.20675] [Citation(s) in RCA: 27] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/09/2022]
|
36
|
Inhibitors of the gamma-aminobutyric acid transporter 1 (GAT1) do not reveal a channel mode of conduction. Neurochem Int 2009; 55:732-40. [PMID: 19622377 DOI: 10.1016/j.neuint.2009.07.005] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/08/2009] [Revised: 06/24/2009] [Accepted: 07/14/2009] [Indexed: 11/21/2022]
Abstract
We expressed the gamma-aminobutyric acid (GABA) transporter GAT1 (SLC6A1) in Xenopus laevis oocytes and performed GABA uptake experiments under voltage clamp at different membrane potentials as well as in the presence of the specific GAT1 inhibitors SKF-89976A and NO-711. In the absence of the inhibitors, GAT1 mediated the inward translocation of 2 net positive charges across the plasma membrane for every GABA molecule transported into the cell. This 2:1 charge flux/GABA flux ratio was the same over a wide range of membrane potentials from -110 mV to +10 mV. Moreover, when GABA-evoked (500 microM) currents were measured at -50 and -90 mV, neither SKF-89976A (5 and 25 microM) nor NO-711 (2 microM) altered the 2:1 charge flux/GABA flux ratio. The results are not consistent with previous hypotheses that (i) GABA evokes an uncoupled channel-mediated current in GAT1, and (ii) GAT1 inhibitors block the putative uncoupled current gated by GABA. Rather, the results suggest tight coupling of GAT1-mediated charge flux and GABA flux.
Collapse
|
37
|
Role of glutamate and GABA transporters in development of pentylenetetrazol-kindling. Neurochem Res 2009; 34:1324-31. [PMID: 19169815 DOI: 10.1007/s11064-009-9912-0] [Citation(s) in RCA: 31] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 01/06/2009] [Indexed: 12/15/2022]
Abstract
Kindling is a form of epileptogenesis that can be induced with pentylenetetrazol (PTZ). We undertook this study to evaluate the contribution of glutamate and GABA transporters to the process of PTZ kindling. Rats were injected i.p. three times per week with PTZ (40 mg/kg) until they were fully kindled. In rats who achieved full kindling, measurement of hippocampal glutamate and GABA transporters within 24 h by western blot showed that GLAST, GLT-1, and EAAC1 were elevated significantly. However, fully kindled rats at 30 days after their last seizure had no change in either glutamate or GABA transporters proteins. These sequential observations suggest that glutamate transporters may contribute to the occurrence of seizures, but were not associated with maintenance of epileptogenesis. During this experiment, we collected data from animals that had kindled easily and animals who were resistant to kindling. Easily-kindled rats reached full kindling with less than five injections of PTZ. Kindling resistant animals failed to achieve full kindling even after administration of 12 consecutive injections of PTZ. Levels of EAAC1 and GAT-1 in easily-kindled rats were decreased by 30% when compared to kindling resistant animals at 30 days after the last PTZ injection. Since decreased EAAC1 and GAT-1 would diminish GABA function, less quantity of these proteins would appear to be associated with the convulsive threshold at the beginning of kindling development. We wonder if glutamate and GABA transporters might be operant in a convulsion threshold set factor or as a pace factor for kindling.
Collapse
|
38
|
Ellefsen S, Stensløkken KO, Fagernes CE, Kristensen TA, Nilsson GE. Expression of genes involved in GABAergic neurotransmission in anoxic crucian carp brain (Carassius carassius). Physiol Genomics 2009; 36:61-8. [DOI: 10.1152/physiolgenomics.90301.2008] [Citation(s) in RCA: 26] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022] Open
Abstract
The crucian carp, Carassius carassius, survives days to months without oxygen, depending on temperature. In the anoxic crucian carp brain, increased GABAergic inhibition, mediated by increased extracellular levels of GABA, has been shown to suppress electric activity and ATP consumption. To investigate an involvement of gene expression in this response, we utilized real-time RT-PCR to test the effect of 1 and 7 days anoxia (8°C) on the expression of 22 genes, including nine GABAAreceptor subunits (α1–6, β2, δ, and γ2), three GABABreceptor subunits (GB1a-1b and GB2), three enzymes involved in GABA metabolism (GAD65 and GAD67, GABAT), four GABA transporters (GAT1, 2a-b and 3), two GABAAreceptor-associated proteins (GABARAP 1 and 2), and the K+/Cl−cotransporter KCC2. While the expression of GABAAreceptor subunits was dominated by α4-, α6-, and δ-subunits, all of which are located to extrasynaptic sites in mammalian brains and respond to elevations in extracellular levels of GABA by showing tonic activity patterns, the expression of GABA transporters was dominated by GAT2 (a and b) and GAT3, which also show extrasynaptic location in mammals. These expression patterns differ from those observed in mammals and may be a prerequisite for GABAergic inhibition of anoxic metabolic rate in crucian carp. Furthermore, while the expression of the majority of the genes was largely unaltered by anoxia, the expression of GAT2 and GAT3 decreased to 20%. This suggests impairment of GABA transport, which could be a mechanism behind the accumulation of extracellular GABA and the increased GABAergic inhibition.
Collapse
Affiliation(s)
- Stian Ellefsen
- Physiology Programme, Department of Molecular Biosciences, University of Oslo, Oslo
- Lillehammer University College, Lillehammer
| | | | - Cathrine E. Fagernes
- Physiology Programme, Department of Molecular Biosciences, University of Oslo, Oslo
| | - Tom A. Kristensen
- Gene Programme, Department of Molecular Biosciences, University of Oslo, Oslo, Norway
| | - Göran E. Nilsson
- Physiology Programme, Department of Molecular Biosciences, University of Oslo, Oslo
| |
Collapse
|
39
|
Tomi M, Tajima A, Tachikawa M, Hosoya KI. Function of taurine transporter (Slc6a6/TauT) as a GABA transporting protein and its relevance to GABA transport in rat retinal capillary endothelial cells. BIOCHIMICA ET BIOPHYSICA ACTA-BIOMEMBRANES 2008; 1778:2138-42. [DOI: 10.1016/j.bbamem.2008.04.012] [Citation(s) in RCA: 51] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/04/2008] [Revised: 04/08/2008] [Accepted: 04/28/2008] [Indexed: 10/22/2022]
|
40
|
Bragina L, Marchionni I, Omrani A, Cozzi A, Pellegrini-Giampietro DE, Cherubini E, Conti F. GAT-1 regulates both tonic and phasic GABAAreceptor-mediated inhibition in the cerebral cortex. J Neurochem 2008; 105:1781-93. [DOI: 10.1111/j.1471-4159.2008.05273.x] [Citation(s) in RCA: 62] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/29/2022]
|
41
|
Livesay DR, Kidd PD, Eskandari S, Roshan U. Assessing the ability of sequence-based methods to provide functional insight within membrane integral proteins: a case study analyzing the neurotransmitter/Na+ symporter family. BMC Bioinformatics 2007; 8:397. [PMID: 17941992 PMCID: PMC2194793 DOI: 10.1186/1471-2105-8-397] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/27/2007] [Accepted: 10/17/2007] [Indexed: 01/09/2023] Open
Abstract
Background Efforts to predict functional sites from globular proteins is increasingly common; however, the most successful of these methods generally require structural insight. Unfortunately, despite several recent technological advances, structural coverage of membrane integral proteins continues to be sparse. ConSequently, sequence-based methods represent an important alternative to illuminate functional roles. In this report, we critically examine the ability of several computational methods to provide functional insight within two specific areas. First, can phylogenomic methods accurately describe the functional diversity across a membrane integral protein family? And second, can sequence-based strategies accurately predict key functional sites? Due to the presence of a recently solved structure and a vast amount of experimental mutagenesis data, the neurotransmitter/Na+ symporter (NSS) family is an ideal model system to assess the quality of our predictions. Results The raw NSS sequence dataset contains 181 sequences, which have been aligned by various methods. The resultant phylogenetic trees always contain six major subfamilies are consistent with the functional diversity across the family. Moreover, in well-represented subfamilies, phylogenetic clustering recapitulates several nuanced functional distinctions. Functional sites are predicted using six different methods (phylogenetic motifs, two methods that identify subfamily-specific positions, and three different conservation scores). A canonical set of 34 functional sites identified by Yamashita et al. within the recently solved LeuTAa structure is used to assess the quality of the predictions, most of which are predicted by the bioinformatic methods. Remarkably, the importance of these sites is largely confirmed by experimental mutagenesis. Furthermore, the collective set of functional site predictions qualitatively clusters along the proposed transport pathway, further demonstrating their utility. Interestingly, the various prediction schemes provide results that are predominantly orthogonal to each other. However, when the methods do provide overlapping results, specificity is shown to increase dramatically (e.g., sites predicted by any three methods have both accuracy and coverage greater than 50%). Conclusion The results presented herein clearly establish the viability of sequence-based bioinformatic strategies to provide functional insight within the NSS family. As such, we expect similar bioinformatic investigations will streamline functional investigations within membrane integral families in the absence of structure.
Collapse
Affiliation(s)
- Dennis R Livesay
- Department of Computer Science and Bioinformatics Research Center, University of North Carolina at Charlotte, Charlotte, NC 28262, USA.
| | | | | | | |
Collapse
|
42
|
Ueda Y, Doi T, Nagatomo K, Willmore LJ, Nakajima A. Functional role for redox in the epileptogenesis: molecular regulation of glutamate in the hippocampus of FeCl3-induced limbic epilepsy model. Exp Brain Res 2007; 181:571-7. [PMID: 17486325 DOI: 10.1007/s00221-007-0954-8] [Citation(s) in RCA: 13] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/16/2007] [Accepted: 03/31/2007] [Indexed: 12/01/2022]
Abstract
We used western blotting to measure the quantity of glutamate and gamma-aminobutyric acid (GABA) transporters proteins within hippocampal tissue obtained from rats who had undergone epileptogenesis. Chronic seizures were induced by amygdalar injection of FeCl(3). We found that the glial glutamate transporters GLAST and GLT-1 were down-regulated at 60 days after initiation of chronic and recurrent seizures. However, the neuronal glutamate transporter EAAC-1 and the GABA transporter GAT-3 were increased. We performed in vivo microdialysis in freely moving animals to estimate in vivo redox state. We found that the hippocampal tissues were oxidized, resulting in even further impairment of glutamate transport. Our data show that epileptogenesis in rats resulting in chronic and recurrent seizures is associated with collapse of glutamate regulation caused by both the molecular down-regulation of glial glutamate transporters combined with the functional failure due to oxidation.
Collapse
Affiliation(s)
- Yuto Ueda
- Section of Psychiatry, Department of Clinical Neuroscience, Faculty of Medicine, University of Miyazaki, 5200 Kihara, Kiyotake, Miyazaki 889-1692, Japan.
| | | | | | | | | |
Collapse
|
43
|
Nagatomo K, Ueda Y, Doi T, Takaki M, Tsuru N. Functional role of GABA transporters for kindling development in GLAST KO mice. Neurosci Res 2006; 57:319-21. [PMID: 17156876 DOI: 10.1016/j.neures.2006.10.009] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/31/2006] [Revised: 10/04/2006] [Accepted: 10/24/2006] [Indexed: 10/23/2022]
Abstract
Kindling-induced after discharge in electroencephalograms depends on the protein associated with glutamatergic and/or GABAergic neuronal transmission. In glutamate transporter knockout (GLAST KO) mice, the kindling phenomena in GLAST KO developed more slowly while the after discharge duration (ADD) was briefer than that of the control C57BL-6J mice. These findings indicate that either the excitatory function was suppressed or the inhibitory function was enhanced in GLAST KO kindling. To explain these phenomena, we used Western blotting to evaluate the alterations in the expression of hippocampal GABA transporter proteins, and the estimation of the effect on the process of epileptogenesis. Although no alterations were observed in the GAT-3 expression, the hippocampal GAT-1 expression was significantly suppressed in comparison to that of C57BL-6J mice. A decreased GAT-1 level in the hippocampus, which might be associated with the increased extracellular GABA level, may therefore inhibit both ADD and seizure propagation as shown by the amygdaloid kindling phenomenon observed in GLAST KO mice.
Collapse
Affiliation(s)
- Keiko Nagatomo
- Section of Psychiatry, Department of Clinical Neuroscience, Faculty of Medicine, University of Miyazaki, 5200 Kihara, Kiyotake, Miyazaki, Japan
| | | | | | | | | |
Collapse
|
44
|
Barbaresi P. GABA-immunoreactive neurons and terminals in the cat periaqueductal gray matter: a light and electron microscopic study. ACTA ACUST UNITED AC 2006; 34:471-87. [PMID: 16902767 DOI: 10.1007/s11068-006-9440-7] [Citation(s) in RCA: 22] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/29/2005] [Revised: 02/13/2006] [Accepted: 06/08/2006] [Indexed: 10/24/2022]
Abstract
Immunocytochemical and electron microscopic methods were used to study the GABAergic innervation in adult cat periaqueductal gray matter (PAG). A mouse monoclonal antibody against gamma -aminobutyric acid (GABA) was used to visualize the inhibitory neuronal system of PAG. At light microscopy, GABA-immunopositive (GABA(IP)) neurons formed two longitudinally oriented columns in the dorsolateral and ventrolateral PAG that accounted for 36% of the neuronal population of both PAG columns; their perikaryal cross-sectional area was smaller than that of unlabeled (UNL) neurons found in the same PAG subdivisions. At electron microscopic level, patches of GABA immunoreactivity were readily detected in neuronal cell bodies, proximal and distal dendrites, axons and axon terminals. Approximately 35-36% of all terminals were GABA(IP); they established symmetric synapses with dendrites (84.72% of the sample in the dorsolateral PAG and 86.09% of the sample in the ventrolateral PAG) or with cell bodies (7-10% of the sample). Moreover, 49.15% of GABA(IP) axon terminals in the dorsolateral and 52.16% in the ventrolateral PAG established symmetric synapses with GABA(IP) dendrites. Immunopositive axon terminals and unlabeled terminals were also involved in the formation of a complex synaptic arrangment, i.e. clusters of synaptic terminals in close contact between them that were often observed in the PAG neuropil. Moreover, a fair number of axo-axonic synapses between GABA(IP) and/or UNL axon terminals were present in both PAG subdivisions. Several dendro-dendritic synapses between labeled and unlabeled dendrites were also observed in both PAG subdivisions. These results suggest that in the cat PAG there exist at least two classes of GABArgic neurons. The first class could exert a tonic control on PAG projecting neurons, the second could act on those GABAergic neurons that in turn keep PAG projecting neurons under tonic inhibition. The functional implications of this type of GABAergic synapse organization are discussed in relation to the dishinibitory processes that take place in the PAG.
Collapse
Affiliation(s)
- Paolo Barbaresi
- Department of Neurosciences, Section of Human Physiology, Marche Polytechnic University, Via Tronto 10/A-Torrette di Ancona, I-60020, Ancona, Italy
| |
Collapse
|
45
|
Casini G, Rickman DW, Brecha NC. Expression of the gamma-aminobutyric acid (GABA) plasma membrane transporter-1 in monkey and human retina. Invest Ophthalmol Vis Sci 2006; 47:1682-90. [PMID: 16565409 PMCID: PMC3696021 DOI: 10.1167/iovs.05-1117] [Citation(s) in RCA: 20] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/24/2022] Open
Abstract
PURPOSE To determine the expression pattern of the predominant gamma-aminobutyric acid (GABA) plasma membrane transporter GAT-1 in Old World monkey (Macaca mulatta) and human retina. METHODS GAT-1 was localized in retinal sections by using immunohistochemical techniques with fluorescence and confocal microscopy. Double-labeling studies were performed with the GAT-1 antibody using antibodies to GABA, vasoactive intestinal polypeptide (VIP), tyrosine hydroxylase (TH), and the bipolar cell marker Mab115A10. RESULTS The pattern of GAT-1 immunostaining was similar in human and monkey retinas. Numerous small immunoreactive somata were in the inner nuclear layer (INL) and were present rarely in the inner plexiform layer (IPL) of all retinal regions. Medium GAT-1 somata were in the ganglion cell layer in the parafoveal and peripheral retinal regions. GAT-1 fibers were densely distributed throughout the IPL. Varicose processes, originating from both the IPL and somata in the INL, arborized in the outer plexiform layer (OPL), forming a sparse network in all retinal regions, except the fovea. Sparsely occurring GAT-1 processes were in the nerve fiber layer in parafoveal regions and near the optic nerve head but not in the optic nerve. In the INL, 99% of the GAT-1 somata contained GABA, and 66% of the GABA immunoreactive somata expressed GAT-1. GAT-1 immunoreactivity was in all VIP-containing cells, but it was absent in TH-immunoreactive amacrine cells and in Mab115A10 immunoreactive bipolar cells. CONCLUSIONS GAT-1 in primate retinas is expressed by amacrine and displaced amacrine cells. The predominant expression of GAT-1 in the inner retina is consistent with the idea that GABA transporters influence neurotransmission and thus participate in visual information processing in the retina.
Collapse
Affiliation(s)
- Giovanni Casini
- Dipartimento di Scienze Ambientali, Università della Tuscia, Viterbo, Italy.
| | | | | |
Collapse
|
46
|
Wu Q, Wada M, Shimada A, Yamamoto A, Fujita T. Functional characterization of Zn2(+)-sensitive GABA transporter expressed in primary cultures of astrocytes from rat cerebral cortex. Brain Res 2006; 1075:100-9. [PMID: 16466645 DOI: 10.1016/j.brainres.2005.12.109] [Citation(s) in RCA: 13] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/18/2005] [Revised: 12/26/2005] [Accepted: 12/27/2005] [Indexed: 11/26/2022]
Abstract
The extracellular levels of gamma-aminobutyric acid (GABA), the main inhibitory neurotransmitter in the mammalian cerebral cortex, are regulated by specific high-affinity Na(+)/Cl(-) dependent transporters (GATs). GAT1 mainly expressed in cerebrocortical neurons is thought to play an important role for clearance of GABA in the extracellular fluid, whereas there is a little information available for pharmacological importance for astrocytic GABA transporters. In the present study, we therefore described the functional characterization of GABA transport in primary cultures of astrocytes from rat cerebral cortex and the identification of GABA transporter subtype(s). GABA transport was Na(+) and Cl(-) dependent and saturable with a Michaelis constant (K(t)) of 9.3+/-2.8 microM. Na(+)- and Cl(-)- activation kinetics revealed that the Na(+)-Cl(-)-to-GABA stoichiometry was 2:1:1 and concentrations of Na(+) and Cl(-) necessary for half-maximal transport (K(0.5)(Na) and K(0.5)(Cl)) were 78+/-28 mM and 9.6+/-2.6 mM, respectively. Na(+)-dependent GABA transport was competitively inhibited by various GABA transport inhibitors, especially GAT2- or GAT3-selective inhibitor. In addition, Zn(2+), which has been reported to be a potent inhibitor of GAT3, was found to have a significantly but partially inhibitory effect on the Na(+)-dependent GABA transport in a concentration-dependent manner. Furthermore, reverse transcription-PCR and Western blot analyses revealed that GAT2 and GAT3 are expressed in primary cultures of astrocytes. These results clearly showed that zinc is a useful reagent for separating GAT3 activity from GAT1- and GAT2-activities in CNS. To our knowledge, the present study represents the first report on the inhibitory effect of zinc on the Na(+)-dependent GABA transport in rat cerebrocortical astrocytes.
Collapse
Affiliation(s)
- Qiang Wu
- Department of Biopharmaceutics, Kyoto Pharmaceutical University, Yamashina, Kyoto 607-8414, Japan
| | | | | | | | | |
Collapse
|
47
|
Sulzer D, Sonders MS, Poulsen NW, Galli A. Mechanisms of neurotransmitter release by amphetamines: a review. Prog Neurobiol 2005; 75:406-33. [PMID: 15955613 DOI: 10.1016/j.pneurobio.2005.04.003] [Citation(s) in RCA: 861] [Impact Index Per Article: 43.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/22/2004] [Revised: 04/15/2005] [Accepted: 04/18/2005] [Indexed: 01/11/2023]
Abstract
Amphetamine and substituted amphetamines, including methamphetamine, methylphenidate (Ritalin), methylenedioxymethamphetamine (ecstasy), and the herbs khat and ephedra, encompass the only widely administered class of drugs that predominantly release neurotransmitter, in this case principally catecholamines, by a non-exocytic mechanism. These drugs play important medicinal and social roles in many cultures, exert profound effects on mental function and behavior, and can produce neurodegeneration and addiction. Numerous questions remain regarding the unusual molecular mechanisms by which these compounds induce catecholamine release. We review current issues on the two apparent primary mechanisms--the redistribution of catecholamines from synaptic vesicles to the cytosol, and induction of reverse transport of transmitter through plasma membrane uptake carriers--and on additional drug effects that affect extracellular catecholamine levels, including uptake inhibition, effects on exocytosis, neurotransmitter synthesis, and metabolism.
Collapse
Affiliation(s)
- David Sulzer
- Department of Psychiatry, Neurology and Pharmacology, New York State Psychiatric Institute, Columbia University, 650 W. 168th Street, Black Building Room 309, New York, NY 10032, USA.
| | | | | | | |
Collapse
|
48
|
Karakossian M, Spencer S, Gomez A, Padilla O, Sacher A, Loo D, Nelson N, Eskandari S. Novel properties of a mouse gamma-aminobutyric acid transporter (GAT4). J Membr Biol 2005; 203:65-82. [PMID: 15981712 PMCID: PMC3009668 DOI: 10.1007/s00232-004-0732-5] [Citation(s) in RCA: 36] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/31/2022]
Abstract
We expressed the mouse gamma-aminobutyric acid (GABA) transporter GAT4 (homologous to rat/ human GAT-3) in Xenopus laevis oocytes and examined its functional and pharmacological properties by using electrophysiological and tracer uptake methods. In the coupled mode of transport (Na+/ Cl-/GABA cotransport), there was tight coupling between charge flux and GABA flux across the plasma membrane (2 charges/GABA). Transport was highly temperature-dependent with a temperature coefficient (Q10) of 4.3. The GAT4 turnover rate (1.5 s(-l); -50 mV, 21 degrees C) and temperature dependence suggest physiological turnover rates of 15-20 s(-1). No uncoupled current was observed in the presence of Na+. In the absence of external Na+, GAT4 exhibited two distinct uncoupled currents. (i) A Cl- leak current (ICl(leak)) was observed when Na+ was replaced with choline or tetraethylammonium. The reversal potential of (ICl(leak)) followed the Cl- Nernst potential. (ii) A Li+ leak current (ILi(leak)) was observed when Na+ was replaced with Li+. Both leak currents were inhibited by Na+, and both were temperature-independent (Q10 approximately 1). The two leak modes appeared not to coexist, as Li+ inhibited (ICl(leak)). The results suggest the existence of cation- and anion-selective channel-like pathways in GAT4. Flufenamic acid inhibited GAT4 Na+/Cl-/GABA cotransport, ILi(leak), and ICl(leak), (Ki approximately 30 microM), and the voltage-induced presteady-state charge movements (Ki approximately 440 microM). Flufenamic acid exhibited little or no selectivity for GAT1, GAT2, or GAT3. Sodium and GABA concentration jicroumps revealed that slow Na+ binding to the transporter is followed by rapid GABA-induced translocation of the ligands across the plasma membrane. Thus, Na+ binding and associated conformational changes constitute the rate-limiting steps in the transport cycle.
Collapse
Affiliation(s)
- M.H. Karakossian
- Biological Sciences Department, California State Polytechnic University, Pomona, CA 91768–4032, USA
| | - S.R. Spencer
- Biological Sciences Department, California State Polytechnic University, Pomona, CA 91768–4032, USA
| | - A.Q. Gomez
- Biological Sciences Department, California State Polytechnic University, Pomona, CA 91768–4032, USA
| | - O.R. Padilla
- Biological Sciences Department, California State Polytechnic University, Pomona, CA 91768–4032, USA
| | - A. Sacher
- Department of Biochemistry, The George S. Wise Faculty of Life Sciences, Tel Aviv University, Tel Aviv 69978, Israel
| | - D.D.F. Loo
- Department of Physiology, David Geffen School of Medicine at UCLA, University of California, Los Angeles, CA 90095–1751, USA
| | - N. Nelson
- Department of Biochemistry, The George S. Wise Faculty of Life Sciences, Tel Aviv University, Tel Aviv 69978, Israel
| | - S. Eskandari
- Biological Sciences Department, California State Polytechnic University, Pomona, CA 91768–4032, USA
| |
Collapse
|
49
|
Melone M, Barbaresi P, Fattorini G, Conti F. Neuronal localization of the GABA transporter GAT-3 in human cerebral cortex: A procedural artifact? J Chem Neuroanat 2005; 30:45-54. [PMID: 15923108 DOI: 10.1016/j.jchemneu.2005.04.002] [Citation(s) in RCA: 24] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/31/2005] [Revised: 04/08/2005] [Accepted: 04/08/2005] [Indexed: 12/26/2022]
Abstract
Gamma-amino butyric acid (GABA) plasma membrane transporters (GATs) contribute to the modulation of GABA's actions and are implicated in neuropsychiatric diseases. In this study, the localization of GAT-3, the major glial GAT, was investigated in human cortex using immunocytochemical techniques. In prefrontal and temporal cortices, GAT-3 immunoreactivity (ir) was present throughout the depth of the cortex, both in puncta and in neurons. GAT-3-positive puncta were dispersed in the neuropil or closely related to cell bodies; neuronal staining was in perikarya, especially of pyramidal cells, and proximal dendrites. Electron microscopic studies showed that GAT-3 ir was in astrocytic processes as well as in neuronal elements. All GAT-3-positive neurons co-expressed heat shock protein 70. To test the possibility that the collection procedure of human samples induced the expression of GAT-3 in neurons which normally do not express it, we analyzed rat cortical tissue resected using the same procedure and found that numerous neurons are GAT-3-positive and that they co-express heat shock protein 70. Results show that in human cortex GAT-3 is expressed in astrocytic processes and in neurons and suggest that neuronal expression is related to the procedure used for collecting human samples.
Collapse
Affiliation(s)
- Marcello Melone
- Department of Neurosciences, Section of Physiology, Università Politecnica delle Marche, Via Tronto 10/A, Torrette di Ancona, I-60020 Ancona, Italy
| | | | | | | |
Collapse
|
50
|
Clausen RP, Moltzen EK, Perregaard J, Lenz SM, Sanchez C, Falch E, Frølund B, Bolvig T, Sarup A, Larsson OM, Schousboe A, Krogsgaard-Larsen P. Selective inhibitors of GABA uptake: synthesis and molecular pharmacology of 4-N-methylamino-4,5,6,7-tetrahydrobenzo[d]isoxazol-3-ol analogues. Bioorg Med Chem 2005; 13:895-908. [PMID: 15653355 DOI: 10.1016/j.bmc.2004.10.029] [Citation(s) in RCA: 63] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/04/2004] [Accepted: 10/12/2004] [Indexed: 11/23/2022]
Abstract
A series of lipophilic diaromatic derivatives of the glia-selective GABA uptake inhibitor (R)-4-amino-4,5,6,7-tetrahydrobenzo[d]isoxazol-3-ol [(R)-exo-THPO, 4] were synthesized via reductive amination of 3-ethoxy-4,5,6,7-tetrahydrobenzo[d]isoxazol-4-one (9) or via N-alkylation of O-alkylatedracemic 4. The effects of the target compounds on GABA uptake mechanisms in vitro were measured using a rat brain synaptosomal preparation or primary cultures of mouse cortical neurons and glia cells (astrocytes), as well as HEK cells transfected with cloned mouse GABA transporter subtypes (GAT1-4). The activity against isoniazid-induced convulsions in mice after subcutaneous administration of the compounds was determined. All of the compounds were potent inhibitors of synaptosomal uptake the most potent compound being (RS)-4-[N-(1,1-diphenylbut-1-en-4-yl)amino]-4,5,6,7-tetrahydrobenzo[d]isoxazol-3-ol (17a, IC50 = 0.14 microM). The majority of the compounds showed a weak preference for glial, as compared to neuronal, GABA uptake. The highest degree of selectivity was 10-fold corresponding to the glia selectivity of (R)-N-methyl-exo-THPO (5). All derivatives showed a preference for the GAT1 transporter, as compared with GAT2-4, with the exception of (RS)-4-[N-[1,1-bis(3-methyl-2-thienyl)but-1-en-4-yl]-N-methylamino]-4,5,6,7-tetrahydrobenzo[d]isoxazol-3-ol (28d), which quite surprisingly turned out to be more potent than GABA at both GAT1 and GAT2 subtypes. The GAT1 activity was shown to reside in (R)-28d whereas (R)-28d and (S)-28d contributed equally to GAT2 activity. This makes (S)-28d a GAT2 selective compound, and (R)-28d equally effective in inhibition of GAT1 and GAT2 mediated GABA transport. All compounds tested were effective as anticonvulsant reflecting that these compounds have blood-brain barrier permeating ability.
Collapse
Affiliation(s)
- Rasmus P Clausen
- Department of Medicinal Chemistry, The Danish University of Pharmaceutical Sciences, 2 Universitetsparken, DK-2100 Copenhagen, Denmark
| | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|