101
|
Shalaly ND, Aneiros E, Blank M, Mueller J, Nyman E, Blind M, Dabrowski MA, Andersson CV, Sandberg K. Positive Modulation of the Glycine Receptor by Means of Glycine Receptor-Binding Aptamers. ACTA ACUST UNITED AC 2015; 20:1112-23. [PMID: 26071243 PMCID: PMC4576506 DOI: 10.1177/1087057115590575] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/05/2015] [Accepted: 05/11/2015] [Indexed: 11/29/2022]
Abstract
According to the gate control theory of pain, the glycine receptors (GlyRs) are putative targets for development of therapeutic analgesics. A possible approach for novel analgesics is to develop a positive modulator of the glycine-activated Cl− channels. Unfortunately, there has been limited success in developing drug-like small molecules to study the impact of agonists or positive modulators on GlyRs. Eight RNA aptamers with low nanomolar affinity to GlyRα1 were generated, and their pharmacological properties analyzed. Cytochemistry using fluorescein-labeled aptamers demonstrated GlyRα1-dependent binding to the plasma membrane but also intracellular binding. Using a fluorescent membrane potential assay, we could identify five aptamers to be positive modulators. The positive modulation of one of the aptamers was confirmed by patch-clamp electrophysiology on L(tk) cells expressing GlyRα1 and/or GlyRα1β. This aptamer potentiated whole-cell Cl− currents in the presence of low concentrations of glycine. To our knowledge, this is the first demonstration ever of RNA aptamers acting as positive modulators for an ion channel. We believe that these aptamers are unique and valuable tools for further studies of GlyR biology and possibly also as tools for assay development in identifying small-molecule agonists and positive modulators.
Collapse
Affiliation(s)
- Nancy Dekki Shalaly
- Department of Neuroscience, AstraZeneca R&D, Södertälje, Sweden and Division of Protein Technology, School of Biotechnology, Royal Institute of Technology, Stockholm, Sweden
| | - Eduardo Aneiros
- Department of Neuroscience, AstraZeneca R&D, Södertälje, Sweden and Lead Discovery Technologies, Merck Serono SA, Geneva, Switzerland
| | | | - Johan Mueller
- Department of Structural Chemistry Laboratory, AstraZeneca R&D, Mölndal, Sweden
| | - Eva Nyman
- Department of Neuroscience, AstraZeneca R&D, Södertälje, Sweden
| | | | | | | | - Kristian Sandberg
- Department of Translational Science, Respiratory, AstraZeneca R&D, Mölndal, Sweden and Department of Medicinal Chemistry, Science for Life Laboratory, Biomedical Center, Uppsala University, Uppsala, Sweden
| |
Collapse
|
102
|
Sánchez A, Yévenes GE, San Martin L, Burgos CF, Moraga-Cid G, Harvey RJ, Aguayo LG. Control of ethanol sensitivity of the glycine receptor α3 subunit by transmembrane 2, the intracellular splice cassette and C-terminal domains. J Pharmacol Exp Ther 2015; 353:80-90. [PMID: 25589412 PMCID: PMC4366752 DOI: 10.1124/jpet.114.221143] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/29/2014] [Accepted: 01/13/2015] [Indexed: 01/06/2023] Open
Abstract
Previous studies have shown that the effect of ethanol onglycine receptors (GlyRs) containing the a1 subunit is affected by interaction with heterotrimeric G proteins (Gβγ). GlyRs containing the α3 subunit are involved in inflammatory pain sensitization and rhythmic breathing and have received much recent attention. For example, it is unknown whether ethanol affects the function of this important GlyR subtype. Electrophysiologic experiments showed that GlyR α3 subunits were not potentiated by pharmacologic concentrations of ethanol or by Gβγ. Thus, we studied GlyR α1–α3 chimeras and mutants to determine the molecular properties that confer ethanol insensitivity. Mutation of corresponding glycine 254 in transmembrane domain 2 (TM2) found in a1 in the α3(A254G) –α1 chimera induced a glycine-evoked current that displayed potentiation during application of ethanol (46 ± 5%, 100 mM) and Gβγ activation (80 ± 17%). Interestingly,insertion of the intracellular α3L splice cassette into GlyR α1 abolished the enhancement of the glycine-activated current by ethanol (5 ± 6%) and activation by Gβγ (21 6 7%). In corporation of the GlyR α1 C terminus into the ethanol-resistant α3S(A254G) mutant produced a construct that displayed potentiation of the glycine-activated current with 100 mM ethanol (40 ± 6%)together with a current enhancement after G protein activation (68 ± 25%). Taken together, these data demonstrate that GlyRα3 subunits are not modulated by ethanol. Residue A254 in TM2, the α3L splice cassette, and the C-terminal domain of α3GlyRs are determinants for low ethanol sensitivity and form the molecular basis of subtype-selective modulation of GlyRs by alcohol.
Collapse
Affiliation(s)
- Andrea Sánchez
- Laboratory of Neurophysiology, Department of Physiology, University of Concepción, Concepción, Chile
| | | | | | | | | | | | | |
Collapse
|
103
|
Disturbed neuronal ER-Golgi sorting of unassembled glycine receptors suggests altered subcellular processing is a cause of human hyperekplexia. J Neurosci 2015; 35:422-37. [PMID: 25568133 DOI: 10.1523/jneurosci.1509-14.2015] [Citation(s) in RCA: 25] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/25/2023] Open
Abstract
Recent studies on the pathogenic mechanisms of recessive hyperekplexia indicate disturbances in glycine receptor (GlyR) α1 biogenesis. Here, we examine the properties of a range of novel glycine receptor mutants identified in human hyperekplexia patients using expression in transfected cell lines and primary neurons. All of the novel mutants localized in the large extracellular domain of the GlyR α1 have reduced cell surface expression with a high proportion of receptors being retained in the ER, although there is forward trafficking of glycosylated subpopulations into the ER-Golgi intermediate compartment and cis-Golgi compartment. CD spectroscopy revealed that the mutant receptors have proportions of secondary structural elements similar to wild-type receptors. Two mutants in loop B (G160R, T162M) were functional, but none of those in loop D/β2-3 were. One nonfunctional truncated mutant (R316X) could be rescued by coexpression with the lacking C-terminal domain. We conclude that a proportion of GlyR α1 mutants can be transported to the plasma membrane but do not necessarily form functional ion channels. We suggest that loop D/β2-3 is an important determinant for GlyR trafficking and functionality, whereas alterations to loop B alter agonist potencies, indicating that residues here are critical elements in ligand binding.
Collapse
|
104
|
Lee SCS, Meyer A, Schubert T, Hüser L, Dedek K, Haverkamp S. Morphology and connectivity of the small bistratified A8 amacrine cell in the mouse retina. J Comp Neurol 2015; 523:1529-47. [PMID: 25630271 DOI: 10.1002/cne.23752] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/13/2014] [Revised: 01/22/2015] [Accepted: 01/24/2015] [Indexed: 01/23/2023]
Abstract
Amacrine cells comprise ∼ 30 morphological types in the mammalian retina. The synaptic connectivity and function of a few γ-aminobutyric acid (GABA)ergic wide-field amacrine cells have recently been studied; however, with the exception of the rod pathway-specific AII amacrine cell, the connectivity of glycinergic small-field amacrine cells has not been investigated in the mouse retina. Here, we studied the morphology and connectivity pattern of the small-field A8 amacrine cell. A8 cells in mouse retina are bistratified with lobular processes in the ON sublamina and arboreal dendrites in the OFF sublamina of the inner plexiform layer. The distinct bistratified morphology was first visible at postnatal day 8, reaching the adult shape at P13, around eye opening. The connectivity of A8 cells to bipolar cells and ganglion cells was studied by double and triple immunolabeling experiments by using various cell markers combined with synaptic markers. Our data suggest that A8 amacrine cells receive glutamatergic input from both OFF and ON cone bipolar cells. Furthermore, A8 cells are coupled to ON cone bipolar cells by gap junctions, and provide inhibitory input via glycine receptor (GlyR) subunit α1 to OFF cone bipolar cells and to ON A-type ganglion cells. Measurements of spontaneous glycinergic postsynaptic currents and GlyR immunolabeling revealed that A8 cells express GlyRs containing the α2 subunit. The results show that the bistratified A8 cell makes very similar synaptic contacts with cone bipolar cells as the rod pathway-specific AII amacrine cell. However, unlike AII cells, A8 amacrine cells provide glycinergic input to ON A-type ganglion cells.
Collapse
Affiliation(s)
- Sammy C S Lee
- Max Planck Institute for Brain Research, 60438, Frankfurt am Main, Germany.,University of Sydney-Save Sight Institute, Sydney, New South Wales, 2000, Australia
| | - Arndt Meyer
- Department of Neurobiology, University of Oldenburg, 26129, Oldenburg, Germany
| | - Timm Schubert
- Werner Reichardt Center for Integrative Neuroscience (CIN)/Institute for Ophthalmic Research, University of Tübingen, 72076, Tübingen, Germany
| | - Laura Hüser
- Max Planck Institute for Brain Research, 60438, Frankfurt am Main, Germany
| | - Karin Dedek
- Department of Neurobiology, University of Oldenburg, 26129, Oldenburg, Germany.,Research Center for Neurosensory Science, University of Oldenburg, 26129, Oldenburg, Germany
| | - Silke Haverkamp
- Max Planck Institute for Brain Research, 60438, Frankfurt am Main, Germany
| |
Collapse
|
105
|
Bicakci Z. Growth retardation, general hypotonia, and loss of acquired neuromotor skills in the infants of mothers with cobalamin deficiency and the possible role of succinyl-CoA and glycine in the pathogenesis. Medicine (Baltimore) 2015; 94:e584. [PMID: 25738478 PMCID: PMC4553967 DOI: 10.1097/md.0000000000000584] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/26/2022] Open
Abstract
Vitamin B12 (cobalamin, Cbl) deficiency can cause metabolic, hematological, and neurological abnormalities. Adequate levels of succinyl-coenzyme A (CoA) cannot be synthesized from methylmalonyl-CoA because of the decreased activity of the methylmalonyl-CoA mutase enzyme that uses Cbl as the cofactor. Succinyl-CoA synthesis deficiency leads to decreased heme synthesis and gluconeogenesis. The reason of growth retardation can be gluconeogenesis deficiency together with heme synthesis deficiency whereas the reason of the neurological abnormalities can be glycine increase in the tissue due to decreased heme synthesis. We present 7 infants diagnosed with severe nutritional Cbl deficiency and discuss the role of succinyl-CoA and glycine in the possible pathogenesis in this article. Patients brought to our clinic with a complaint of growth retardation and diagnosed with nutritional Cbl deficiency were included in the study. There were 5 females and 2 males. The mean age was 11 ± 2.30 (range 6-13) months. All patients had general muscular hypotonia and 4 had growth retardation. Neuromotor growth retardation was found in 4 of the children who had previously shown normal neuromotor development for age. The mean Cbl level was 83.8 ± 27.6 (45.6-114) pg/mL. The mean Cbl level of the mothers was 155 ± 56.6 (88-258) pg/mL. Six of the patients had anemia and 1 had thrombocytopenia. Mean corpuscular volume value was 91.5 ± 12.2 fL. Following treatment, the muscle tonus of the patients improved, the anemia and growth retardation decreased, and the lost neuromotor abilities were recovered. Severe nutritional Cbl deficiency is an important nutritional disease where complications can be prevented with early treatment. When evaluating the pathogenesis, it should be noted that nutritional Cbl deficiency is a succinyl-CoA synthesis deficiency.
Collapse
Affiliation(s)
- Zafer Bicakci
- From the Department of Pediatric Hematology, Kafkas University Faculty of Medicine, Paşa Çayırı, Kars, Turkey
| |
Collapse
|
106
|
Molecular basis for convergent evolution of glutamate recognition by pentameric ligand-gated ion channels. Sci Rep 2015; 5:8558. [PMID: 25708000 PMCID: PMC4338433 DOI: 10.1038/srep08558] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/23/2014] [Accepted: 01/27/2015] [Indexed: 12/31/2022] Open
Abstract
Glutamate is an indispensable neurotransmitter, triggering postsynaptic signals upon recognition by postsynaptic receptors. We questioned the phylogenetic position and the molecular details of when and where glutamate recognition arose in the glutamate-gated chloride channels. Experiments revealed that glutamate recognition requires an arginine residue in the base of the binding site, which originated at least three distinct times according to phylogenetic analysis. Most remarkably, the arginine emerged on the principal face of the binding site in the Lophotrochozoan lineage, but 65 amino acids upstream, on the complementary face, in the Ecdysozoan lineage. This combined experimental and computational approach throws new light on the evolution of synaptic signalling.
Collapse
|
107
|
Zhang Y, Dixon CL, Keramidas A, Lynch JW. Functional reconstitution of glycinergic synapses incorporating defined glycine receptor subunit combinations. Neuropharmacology 2015; 89:391-7. [DOI: 10.1016/j.neuropharm.2014.10.026] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/26/2014] [Revised: 10/16/2014] [Accepted: 10/26/2014] [Indexed: 10/24/2022]
|
108
|
Scott S, Lynch JW, Keramidas A. Correlating structural and energetic changes in glycine receptor activation. J Biol Chem 2015; 290:5621-34. [PMID: 25572390 DOI: 10.1074/jbc.m114.616573] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/06/2022] Open
Abstract
Pentameric ligand-gated ion channels (pLGICs) mediate fast chemoelectrical transduction in the nervous system. The mechanism by which the energy of ligand binding leads to current-conducting receptors is poorly understood and may vary among family members. We addressed these questions by correlating the structural and energetic mechanisms by which a naturally occurring M1 domain mutation (α1(Q-26'E)) enhances receptor activation in homo- and heteromeric glycine receptors. We systematically altered the charge of spatially clustered residues at positions 19' and 24', in the M2 and M2-M3 linker domains, respectively, which are known to be critical to efficient receptor activation, on a background of α1(Q-26'E). Changes in the durations of single receptor activations (clusters) and conductance were used to determine interaction coupling energies, which we correlated with conformational displacements as measured in pLGIC crystal structures. Presence of the α1(Q-26'E) enhanced cluster durations and reduced channel conductance in homo- and heteromeric receptors. Strong coupling between α1(-26') and α1(19') across the subunit interface suggests an important role in receptor activation. A lack of coupling between α1(-26') and α1(24') implies that 24' mutations disrupt activation via other interactions. A similar lack of energetic coupling between α1(-26') and reciprocal mutations in the β subunit suggests that this subunit remains relatively static during receptor activation. However, the channel effects of α1(Q-26'E) on α1β receptors suggests at least one α1-α1 interface per pentamer. The coupling-energy change between α1(-26') and α1(19') correlates with a local structural rearrangement essential for pLGIC activation, implying it comprises a key energetic pathway in activating glycine receptors and other pLGICs.
Collapse
Affiliation(s)
| | - Joseph W Lynch
- From the Queensland Brain Institute and the School of Biomedical Sciences, University of Queensland, Brisbane, Queensland, Australia 4072
| | | |
Collapse
|
109
|
Zhang JY, Wang YX. Gelsemium analgesia and the spinal glycine receptor/allopregnanolone pathway. Fitoterapia 2014; 100:35-43. [PMID: 25447163 DOI: 10.1016/j.fitote.2014.11.002] [Citation(s) in RCA: 40] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/07/2014] [Revised: 10/30/2014] [Accepted: 11/01/2014] [Indexed: 12/21/2022]
Abstract
Gelsemium, a small genus of flowering plant from the family Loganiaceae, comprises five species including the popular Gelsemium sempervirens Ait. and Gelsemium elegans Benth., which are indigenous to North America and China/East Asia, respectively. Approximately 120 alkaloids have been isolated and identified from Gelsemium, with the predominant indole alkaloids including gelsemine, koumine, gelsemicine, gelsenicine, gelsedine, sempervirine, koumidine, koumicine and humantenine. Gelsemine is the principal active alkaloid in G. sempervirens Ait., and koumine and gelsemine are the most and second-most dominant alkaloids in G. elegans Benth. Gelsemium extract and its active alkaloids serve a variety of biological functions, including neurobiological, immunosuppressive and antitumor effects, and have traditionally been used to treat pain, neuralgia, anxiety, insomnia, asthma, respiratory ailments and cancers. This review focuses on animal-based studies of Gelsemium as a pain treatment and its mechanism of action. In contrast to morphine, when administered intrathecally and systemically, koumine, gelsemine and gelsenicine have marked antinociception in inflammatory, neuropathic and bone cancer pains without inducing antinociceptive tolerance. Gelsemium and its active alkaloids may produce antinociception by activating the spinal α3 glycine/allopregnanolone pathway. The results of this review support the clinical use of Gelsemium and suggest that its active alkaloids may be developed to treat intractable and other types of pain, preferably after chemical modification. However, Gelsemium is a known toxic plant, and its toxicity limits its appropriate dosage and clinical use. To avoid or decrease the side/toxic effects of Gelsemium, an individual monomer of highly potent alkaloids must be selected, or alkaloids that exhibit greater α3 glycine receptor selectivity may be discovered or modified.
Collapse
Affiliation(s)
- Jing-Yang Zhang
- King's Lab, Shanghai Jiao Tong University School of Pharmacy, No 6 Biomedicine Building, 800 Dongchuan Road, Shanghai 200240, China
| | - Yong-Xiang Wang
- King's Lab, Shanghai Jiao Tong University School of Pharmacy, No 6 Biomedicine Building, 800 Dongchuan Road, Shanghai 200240, China.
| |
Collapse
|
110
|
Alexander SPH, Benson HE, Faccenda E, Pawson AJ, Sharman JL, Spedding M, Peters JA, Harmar AJ. The Concise Guide to PHARMACOLOGY 2013/14: ligand-gated ion channels. Br J Pharmacol 2014; 170:1582-606. [PMID: 24528238 PMCID: PMC3892288 DOI: 10.1111/bph.12446] [Citation(s) in RCA: 113] [Impact Index Per Article: 10.3] [Reference Citation Analysis] [Abstract] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/16/2022] Open
Abstract
The Concise Guide to PHARMACOLOGY 2013/14 provides concise overviews of the key properties of over 2000 human drug targets with their pharmacology, plus links to an open access knowledgebase of drug targets and their ligands (www.guidetopharmacology.org), which provides more detailed views of target and ligand properties. The full contents can be found at http://onlinelibrary.wiley.com/doi/10.1111/bph.12444/full. Ligand-gated ion channels are one of the seven major pharmacological targets into which the Guide is divided, with the others being G protein-coupled receptors, ion channels, catalytic receptors, nuclear hormone receptors, transporters and enzymes. These are presented with nomenclature guidance and summary information on the best available pharmacological tools, alongside key references and suggestions for further reading. A new landscape format has easy to use tables comparing related targets. It is a condensed version of material contemporary to late 2013, which is presented in greater detail and constantly updated on the website www.guidetopharmacology.org, superseding data presented in previous Guides to Receptors and Channels. It is produced in conjunction with NC-IUPHAR and provides the official IUPHAR classification and nomenclature for human drug targets, where appropriate. It consolidates information previously curated and displayed separately in IUPHAR-DB and the Guide to Receptors and Channels, providing a permanent, citable, point-in-time record that will survive database updates.
Collapse
Affiliation(s)
- Stephen P H Alexander
- School of Life Sciences, University of Nottingham Medical School, Nottingham, NG7 2UH, UK
| | | | | | | | | | | | | | | | | |
Collapse
|
111
|
Molecular determinants of agonist selectivity in glutamate-gated chloride channels which likely explain the agonist selectivity of the vertebrate glycine and GABAA-ρ receptors. PLoS One 2014; 9:e108458. [PMID: 25259865 PMCID: PMC4178172 DOI: 10.1371/journal.pone.0108458] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/01/2014] [Accepted: 08/11/2014] [Indexed: 11/29/2022] Open
Abstract
Orthologous Cys-loop glutamate-gated chloride channels (GluClR’s) have been cloned and described electrophysiologically and pharmacologically in arthropods and nematodes (both members of the invertebrate ecdysozoan superphylum). Recently, GluClR’s from Aplysia californica (a mollusc from the lophotrochozoan superphylum) have been cloned and similarly studied. In spite of sharing a common function, the ecdysozoan and lophotrochozoan receptors have been shown by phylogenetic analyses to have evolved independently. The recent crystallization of the GluClR from C. elegans revealed the binding pocket of the nematode receptor. An alignment of the protein sequences of the nematode and molluscan GluClRs showed that the Aplysia receptor does not contain all of the residues defining the binding mode of the ecdysozoan receptor. That the two receptors have slightly different binding modes is not surprising since earlier electrophysiological and pharmacological experiments had suggested that they were differentially responsive to certain agonists. Knowledge of the structure of the C. elegans GluClR has permitted us to generate a homology model of the binding pocket of the Aplysia receptor. We have analyzed the differences between the two binding modes and evaluated the relative significance of their non-common residues. We have compared the GluClRs electrophysiologically and pharmacologically and we have used site-directed mutagenesis on both receptor types to test predictions made from the model. Finally, we propose an explanation derived from the model for why the nematode receptors are gated only by glutamate, whereas the molluscan receptors can also be activated by β-alanine, GABA and taurine. Like the Aplysia receptor, the vertebrate glycine and GABAA-ρ receptors also respond to these other agonists. An alignment of the sequences of the molluscan and vertebrate receptors shows that the reasons we have given for the ability of the other agonists to activate the Aplysia receptor also explain the agonist profile seen in the glycine and GABAA-ρ receptors.
Collapse
|
112
|
Yu R, Hurdiss E, Greiner T, Lape R, Sivilotti L, Biggin PC. Agonist and antagonist binding in human glycine receptors. Biochemistry 2014; 53:6041-51. [PMID: 25184435 DOI: 10.1021/bi500815f] [Citation(s) in RCA: 35] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/30/2022]
Abstract
The human glycine receptor (hGlyR) is an anion-permeable ligand-gated channel that is part of a larger superfamily of receptors called the Cys-loop family. hGlyRs are particularly amenable to single-channel recordings, thus making them a model experimental system for understanding the Cys-loop receptor family in general. Understanding the relationship between agonist binding and efficacy in Cys-loop receptors should improve our future prospects for making specific agonists or antagonists. However, at present, there is no high-resolution structure for the complete hGlyR, and thus, modeling is needed to provide a physical framework on which to interpret single-channel data. The structure of the glutamate-gated chloride channel from Caenorhabditis elegans shows a much higher level of sequence identity to human hGlyR than previous templates such as AChBP or the bacterial channels, GLIC and ELIC. Thus, we constructed a model of the hGlyR and validated it against previously reported mutagenesis data. We used molecular dynamics to refine the model and to explore binding of both an agonist (glycine) and an antagonist (strychnine). The model shows excellent agreement with previous data but also suggests some unique features: (i) a water molecule that forms part of the binding site and allows us to account for some previous results that were difficult to reconcile, (ii) an interaction of the glycine agonist with S129, and (iii) an effect from E211, both of which we confirmed with new site-directed mutagenesis and patch clamp recordings. Finally, examination of the simulations suggests that strychnine binding induces movement to a conformational state distinct from the glycine-bound or apo state, not only within the ligand-binding domain but also in the transmembrane domain.
Collapse
Affiliation(s)
- Rilei Yu
- Structural Bioinformatics and Computational Biochemistry, Department of Biochemistry, University of Oxford , South Parks Road, Oxford OX1 3QU, United Kingdom
| | | | | | | | | | | |
Collapse
|
113
|
Meiselbach H, Vogel N, Langlhofer G, Stangl S, Schleyer B, Bahnassawy L, Sticht H, Breitinger HG, Becker CM, Villmann C. Single expressed glycine receptor domains reconstitute functional ion channels without subunit-specific desensitization behavior. J Biol Chem 2014; 289:29135-47. [PMID: 25143388 DOI: 10.1074/jbc.m114.559138] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/06/2022] Open
Abstract
Cys loop receptors are pentameric arrangements of independent subunits that assemble into functional ion channels. Each subunit shows a domain architecture. Functional ion channels can be reconstituted even from independent, nonfunctional subunit domains, as shown previously for GlyRα1 receptors. Here, we demonstrate that this reconstitution is not restricted to α1 but can be transferred to other members of the Cys loop receptor family. A nonfunctional GlyR subunit, truncated at the intracellular TM3-4 loop by a premature stop codon, can be complemented by co-expression of the missing tail portion of the receptor. Compared with α1 subunits, rescue by domain complementation was less efficient when GlyRα3 or the GABAA/C subunit ρ1 was used. If truncation disrupted an alternative splicing cassette within the intracellular TM3-4 loop of α3 subunits, which also regulates receptor desensitization, functional rescue was not possible. When α3 receptors were restored by complementation using domains with and without the spliced insert, no difference in desensitization was found. In contrast, desensitization properties could even be transferred between α1/α3 receptor chimeras harboring or lacking the α3 splice cassette proving that functional rescue depends on the integrity of the alternative splicing cassette in α3. Thus, an intact α3 splicing cassette in the TM3-4 loop environment is indispensable for functional rescue, and the quality of receptor restoration can be assessed from desensitization properties.
Collapse
Affiliation(s)
| | - Nico Vogel
- Institute of Biochemistry, Emil Fischer Center, Friedrich-Alexander University Erlangen-Nürnberg, Fahrstrasse 17, 91054 Erlangen, Germany
| | - Georg Langlhofer
- the Institute for Clinical Neurobiology, University of Würzburg, Versbacherstrasse 5, 97078 Würzburg, Germany, and
| | - Sabine Stangl
- Institute of Biochemistry, Emil Fischer Center, Friedrich-Alexander University Erlangen-Nürnberg, Fahrstrasse 17, 91054 Erlangen, Germany
| | - Barbara Schleyer
- the Institute for Clinical Neurobiology, University of Würzburg, Versbacherstrasse 5, 97078 Würzburg, Germany, and
| | - Lamia'a Bahnassawy
- the Biochemistry Department, German University Cairo, New Cairo City, Cairo 11835, Egypt
| | | | - Hans-Georg Breitinger
- the Biochemistry Department, German University Cairo, New Cairo City, Cairo 11835, Egypt
| | - Cord-Michael Becker
- Institute of Biochemistry, Emil Fischer Center, Friedrich-Alexander University Erlangen-Nürnberg, Fahrstrasse 17, 91054 Erlangen, Germany
| | - Carmen Villmann
- Institute of Biochemistry, Emil Fischer Center, Friedrich-Alexander University Erlangen-Nürnberg, Fahrstrasse 17, 91054 Erlangen, Germany, the Institute for Clinical Neurobiology, University of Würzburg, Versbacherstrasse 5, 97078 Würzburg, Germany, and
| |
Collapse
|
114
|
Schaefer N, Langlhofer G, Kluck CJ, Villmann C. Glycine receptor mouse mutants: model systems for human hyperekplexia. Br J Pharmacol 2014; 170:933-52. [PMID: 23941355 DOI: 10.1111/bph.12335] [Citation(s) in RCA: 30] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/03/2013] [Revised: 07/19/2013] [Accepted: 08/02/2013] [Indexed: 11/30/2022] Open
Abstract
Human hyperekplexia is a neuromotor disorder caused by disturbances in inhibitory glycine-mediated neurotransmission. Mutations in genes encoding for glycine receptor subunits or associated proteins, such as GLRA1, GLRB, GPHN and ARHGEF9, have been detected in patients suffering from hyperekplexia. Classical symptoms are exaggerated startle attacks upon unexpected acoustic or tactile stimuli, massive tremor, loss of postural control during startle and apnoea. Usually patients are treated with clonazepam, this helps to dampen the severe symptoms most probably by up-regulating GABAergic responses. However, the mechanism is not completely understood. Similar neuromotor phenotypes have been observed in mouse models that carry glycine receptor mutations. These mouse models serve as excellent tools for analysing the underlying pathomechanisms. Yet, studies in mutant mice looking for postsynaptic compensation of glycinergic dysfunction via an up-regulation in GABAA receptor numbers have failed, as expression levels were similar to those in wild-type mice. However, presynaptic adaptation mechanisms with an unusual switch from mixed GABA/glycinergic to GABAergic presynaptic terminals have been observed. Whether this presynaptic adaptation explains the improvement in symptoms or other compensation mechanisms exist is still under investigation. With the help of spontaneous glycine receptor mouse mutants, knock-in and knock-out studies, it is possible to associate behavioural changes with pharmacological differences in glycinergic inhibition. This review focuses on the structural and functional characteristics of the various mouse models used to elucidate the underlying signal transduction pathways and adaptation processes and describes a novel route that uses gene-therapeutic modulation of mutated receptors to overcome loss of function mutations.
Collapse
Affiliation(s)
- Natascha Schaefer
- Institute for Clinical Neurobiology, Julius-Maximilians-University of Würzburg, Würzburg, Germany
| | | | | | | |
Collapse
|
115
|
Abstract
The neurotransmitters GABA and glycine mediate fast synaptic inhibition by activating ligand-gated chloride channels--namely, type A GABA (GABA(A)) and glycine receptors. Both types of receptors are anchored postsynaptically by gephyrin, which self-assembles into a scaffold and interacts with the cytoskeleton. Current research indicates that postsynaptic gephyrin clusters are dynamic assemblies that are held together and regulated by multiple protein-protein interactions. Moreover, post-translational modifications of gephyrin regulate the formation and plasticity of GABAergic synapses by altering the clustering properties of postsynaptic scaffolds and thereby the availability and function of receptors and other signalling molecules. Here, we discuss the formation and regulation of the gephyrin scaffold, its role in GABAergic and glycinergic synaptic function and the implications for the pathophysiology of brain disorders caused by abnormal inhibitory neurotransmission.
Collapse
|
116
|
Lynagh T, Pless SA. Principles of agonist recognition in Cys-loop receptors. Front Physiol 2014; 5:160. [PMID: 24795655 PMCID: PMC4006026 DOI: 10.3389/fphys.2014.00160] [Citation(s) in RCA: 35] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/11/2014] [Accepted: 04/04/2014] [Indexed: 12/22/2022] Open
Abstract
Cys-loop receptors are ligand-gated ion channels that are activated by a structurally diverse array of neurotransmitters, including acetylcholine, serotonin, glycine, and GABA. After the term "chemoreceptor" emerged over 100 years ago, there was some wait until affinity labeling, molecular cloning, functional studies, and X-ray crystallography experiments identified the extracellular interface of adjacent subunits as the principal site of agonist binding. The question of how subtle differences at and around agonist-binding sites of different Cys-loop receptors can accommodate transmitters as chemically diverse as glycine and serotonin has been subject to intense research over the last three decades. This review outlines the functional diversity and current structural understanding of agonist-binding sites, including those of invertebrate Cys-loop receptors. Together, this provides a framework to understand the atomic determinants involved in how these valuable therapeutic targets recognize and bind their ligands.
Collapse
Affiliation(s)
| | - Stephan A. Pless
- Department of Drug Design and Pharmacology, Center for Biopharmaceuticals, University of CopenhagenCopenhagen, Denmark
| |
Collapse
|
117
|
The involvement of accumbal glycine receptors in the dopamine-elevating effects of addictive drugs. Neuropharmacology 2014; 82:69-75. [PMID: 24686030 DOI: 10.1016/j.neuropharm.2014.03.010] [Citation(s) in RCA: 32] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/12/2013] [Revised: 03/14/2014] [Accepted: 03/18/2014] [Indexed: 10/25/2022]
Abstract
The ability of drugs of abuse to increase mesolimbic levels of dopamine is a characteristic associated with their rewarding effects. Exactly how these effects are produced by different substances is not as well characterised. Our previous work in rats has demonstrated that accumbal glycine receptors (GlyRs) are involved in mediating the dopamine-activating effects of ethanol, and in modulating ethanol intake. In this study the investigation of GlyR involvement was extended to include several different drugs of abuse. By using microdialysis and electrophysiology we compared effects of addictive drugs, with and without the GlyR antagonist strychnine, on dopamine levels and neurotransmission in nucleus accumbens. The dopamine-increasing effect of systemic ethanol and the drug-induced change in neurotransmission in vitro, as measured by microdialysis and field potential recordings, were dependent on GlyRs in nAc. Accumbal GlyRs were also involved in the actions of tetrahydrocannabinol and nicotine, but not in those of cocaine or morphine. These data indicate that accumbal GlyRs play a key role in ethanol-induced dopamine activation and contribute also to that of cannabinoids and nicotine.
Collapse
|
118
|
Mariqueo TA, Agurto A, Muñoz B, San Martin L, Coronado C, Fernández-Pérez EJ, Murath P, Sánchez A, Homanics GE, Aguayo LG. Effects of ethanol on glycinergic synaptic currents in mouse spinal cord neurons. J Neurophysiol 2014; 111:1940-8. [PMID: 24572089 DOI: 10.1152/jn.00789.2013] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022] Open
Abstract
Ethanol increased the frequency of miniature glycinergic currents [miniature inhibitory postsynaptic currents (mIPSCs)] in cultured spinal neurons. This effect was dependent on intracellular calcium augmentation, since preincubation with BAPTA (an intracellular calcium chelator) or thapsigargin [a sarco(endo)plasmic reticulum Ca(2+)-ATPase (SERCA) pump inhibitor] significantly attenuated this effect. Similarly, U73122 (a phospholipase C inhibitor) or 2-aminoethoxydiphenyl borate [2-APB, an inositol 1,4,5-trisphosphate (IP₃) receptor (IP3R) inhibitor] reduced this effect. Block of ethanol action was also achieved after preincubation with Rp-cAMPS, inhibitor of the adenylate cyclase (AC)/PKA signaling pathway. These data suggest that there is a convergence at the level of IP₃R that accounts for presynaptic ethanol effects. At the postsynaptic level, ethanol increased the decay time constant of mIPSCs in a group of neurons (30 ± 10% above control, n = 13/26 cells). On the other hand, the currents activated by exogenously applied glycine were consistently potentiated (55 ± 10% above control, n = 11/12 cells), which suggests that ethanol modulates synaptic and nonsynaptic glycine receptors (GlyRs) in a different fashion. Supporting the role of G protein modulation on ethanol responses, we found that a nonhydrolyzable GTP analog [guanosine 5'-O-(3-thiotriphosphate) (GTPγS)] increased the decay time constant in ∼50% of the neurons (28 ± 12%, n = 11/19 cells) but potentiated the glycine-activated Cl(-) current in most of the neurons examined (83 ± 29%, n = 7/9 cells). In addition, confocal microscopy showed that α1-containing GlyRs colocalized with Gβ and Piccolo (a presynaptic cytomatrix protein) in ∼40% of synaptic receptor clusters, suggesting that colocalization of Gβγ and GlyRs might account for the difference in ethanol sensitivity at the postsynaptic level.
Collapse
Affiliation(s)
- Trinidad A Mariqueo
- Department of Physiology-Laboratory of Neurophysiology, University of Concepción, Concepción, Chile; PhD Program in Pharmacology, University of Chile, Santiago, Chile; and
| | - Adolfo Agurto
- Department of Physiology-Laboratory of Neurophysiology, University of Concepción, Concepción, Chile
| | - Braulio Muñoz
- Department of Physiology-Laboratory of Neurophysiology, University of Concepción, Concepción, Chile
| | - Loreto San Martin
- Department of Physiology-Laboratory of Neurophysiology, University of Concepción, Concepción, Chile
| | - Cesar Coronado
- Department of Physiology-Laboratory of Neurophysiology, University of Concepción, Concepción, Chile
| | | | - Pablo Murath
- Department of Physiology-Laboratory of Neurophysiology, University of Concepción, Concepción, Chile
| | - Andrea Sánchez
- Department of Physiology-Laboratory of Neurophysiology, University of Concepción, Concepción, Chile
| | - Gregg E Homanics
- Department of Anesthesiology and Department of Pharmacology and Chemical Biology, University of Pittsburgh, Pittsburgh, Pennsylvania
| | - Luis G Aguayo
- Department of Physiology-Laboratory of Neurophysiology, University of Concepción, Concepción, Chile;
| |
Collapse
|
119
|
Del Pino I, Koch D, Schemm R, Qualmann B, Betz H, Paarmann I. Proteomic analysis of glycine receptor β subunit (GlyRβ)-interacting proteins: evidence for syndapin I regulating synaptic glycine receptors. J Biol Chem 2014; 289:11396-11409. [PMID: 24509844 DOI: 10.1074/jbc.m113.504860] [Citation(s) in RCA: 20] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022] Open
Abstract
Glycine receptors (GlyRs) mediate inhibitory neurotransmission in spinal cord and brainstem. They are clustered at inhibitory postsynapses via a tight interaction of their β subunits (GlyRβ) with the scaffolding protein gephyrin. In an attempt to isolate additional proteins interacting with GlyRβ, we performed pulldown experiments with rat brain extracts using a glutathione S-transferase fusion protein encompassing amino acids 378-455 of the large intracellular loop of GlyRβ as bait. This identified syndapin I (SdpI) as a novel interaction partner of GlyRβ that coimmunoprecipitates with native GlyRs from brainstem extracts. Both SdpI and SdpII bound efficiently to the intracellular loop of GlyRβ in vitro and colocalized with GlyRβ upon coexpression in COS-7 cells. The SdpI-binding site was mapped to a proline-rich sequence of 22 amino acids within the intracellular loop of GlyRβ. Deletion and point mutation analysis disclosed that SdpI binding to GlyRβ is Src homology 3 domain-dependent. In cultured rat spinal cord neurons, SdpI immunoreactivity was found to partially colocalize with marker proteins of inhibitory and excitatory synapses. When SdpI was acutely knocked down in cultured spinal cord neurons by viral miRNA expression, postsynaptic GlyR clusters were significantly reduced in both size and number. Similar changes in GlyR cluster properties were found in spinal cultures from SdpI-deficient mice. Our results are consistent with a role of SdpI in the trafficking and/or cytoskeletal anchoring of synaptic GlyRs.
Collapse
Affiliation(s)
- Isabel Del Pino
- Department of Neurochemistry, Max-Planck-Institute for Brain Research, D-60438 Frankfurt/Main
| | - Dennis Koch
- Institute for Biochemistry I, Jena University Hospital, Friedrich Schiller University Jena, D-07743 Jena
| | - Rudolf Schemm
- Department for Theoretical and Computational Biophysics, Max-Planck-Institute for Biophysical Chemistry, D-37077 Göttingen, and
| | - Britta Qualmann
- Institute for Biochemistry I, Jena University Hospital, Friedrich Schiller University Jena, D-07743 Jena
| | - Heinrich Betz
- Department of Neurochemistry, Max-Planck-Institute for Brain Research, D-60438 Frankfurt/Main,; Max-Planck Institute for Medical Research, 69120 Heidelberg, Germany.
| | - Ingo Paarmann
- Department of Neurochemistry, Max-Planck-Institute for Brain Research, D-60438 Frankfurt/Main,.
| |
Collapse
|
120
|
Bode A, Lynch JW. The impact of human hyperekplexia mutations on glycine receptor structure and function. Mol Brain 2014; 7:2. [PMID: 24405574 PMCID: PMC3895786 DOI: 10.1186/1756-6606-7-2] [Citation(s) in RCA: 101] [Impact Index Per Article: 9.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/24/2013] [Accepted: 01/07/2014] [Indexed: 01/11/2023] Open
Abstract
Hyperekplexia is a rare neurological disorder characterized by neonatal hypertonia, exaggerated startle responses to unexpected stimuli and a variable incidence of apnoea, intellectual disability and delays in speech acquisition. The majority of motor defects are successfully treated by clonazepam. Hyperekplexia is caused by hereditary mutations that disrupt the functioning of inhibitory glycinergic synapses in neuromotor pathways of the spinal cord and brainstem. The human glycine receptor α1 and β subunits, which predominate at these synapses, are the major targets of mutations. International genetic screening programs, that together have analysed several hundred probands, have recently generated a clear picture of genotype-phenotype correlations and the prevalence of different categories of hyperekplexia mutations. Focusing largely on this new information, this review seeks to summarise the effects of mutations on glycine receptor structure and function and how these functional alterations lead to hyperekplexia.
Collapse
Affiliation(s)
| | - Joseph W Lynch
- Queensland Brain Institute and School of Biomedical Sciences, The University of Queensland, Queensland 4072, Australia.
| |
Collapse
|
121
|
Presynaptic glycine receptors as a potential therapeutic target for hyperekplexia disease. Nat Neurosci 2014; 17:232-9. [PMID: 24390226 DOI: 10.1038/nn.3615] [Citation(s) in RCA: 52] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/05/2013] [Accepted: 12/03/2013] [Indexed: 11/08/2022]
Abstract
Although postsynaptic glycine receptors (GlyRs) as αβ heteromers attract considerable research attention, little is known about the role of presynaptic GlyRs, likely α homomers, in diseases. Here, we demonstrate that dehydroxylcannabidiol (DH-CBD), a nonpsychoactive cannabinoid, can rescue GlyR functional deficiency and exaggerated acoustic and tactile startle responses in mice bearing point mutations in α1 GlyRs that are responsible for a hereditary startle-hyperekplexia disease. The GlyRs expressed as α1 homomers either in HEK-293 cells or at presynaptic terminals of the calyceal synapses in the auditory brainstem are more vulnerable than heteromers to hyperekplexia mutation-induced impairment. Homomeric mutants are more sensitive to DH-CBD than are heteromers, suggesting presynaptic GlyRs as a primary target. Consistent with this idea, DH-CBD selectively rescues impaired presynaptic GlyR activity and diminished glycine release in the brainstem and spinal cord of hyperekplexic mutant mice. Thus, presynaptic α1 GlyRs emerge as a potential therapeutic target for dominant hyperekplexia disease and other diseases with GlyR deficiency.
Collapse
|
122
|
Förstera B, a Dzaye OD, Winkelmann A, Semtner M, Benedetti B, Markovic DS, Synowitz M, Wend P, Fähling M, Junier MP, Glass R, Kettenmann H, Meier JC. Intracellular glycine receptor function facilitates glioma formation in vivo. J Cell Sci 2014; 127:3687-98. [DOI: 10.1242/jcs.146662] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022] Open
Abstract
The neuronal function of Cys-loop neurotransmitter receptors is established; however, their role in non-neuronal cells is poorly defined. As brain tumors accumulate the neurotransmitter glycine, we studied expression and function of glycine receptors (GlyR) in glioma cells. Human brain tumor biopsies selectively expressed GlyR subunits with nuclear import signal (NLS, α1 and α3). The mouse glioma cell line GL261 expressed GlyR α1, and knock-down of α1 protein expression impaired self-renewal capacity and tumorigenicity of GL261 glioma cells as evidenced by the neurosphere assay and GL261 cell inoculation in vivo, respectively. We furthermore show that the pronounced tumorigenic effect of GlyR α1 relies on a new intracellular signaling function that depends on the NLS region in the large cytosolic loop and impacts on GL261 glioma cell gene regulation. Stable expression of GlyR α1 and α3 loops rescued self-renewal capacity of GlyR α1 knock-down cells, which demonstrates their functional equivalence. The new intracellular signaling function identified here goes beyond the well-established role of GlyRs as neuronal ligand-gated ion channels and defines NLS-containing GlyRs as novel potential targets for brain tumor therapies.
Collapse
|
123
|
Bonin RP, De Koninck Y. Restoring ionotropic inhibition as an analgesic strategy. Neurosci Lett 2013; 557 Pt A:43-51. [PMID: 24080373 DOI: 10.1016/j.neulet.2013.09.047] [Citation(s) in RCA: 23] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/27/2013] [Revised: 09/11/2013] [Accepted: 09/16/2013] [Indexed: 12/31/2022]
Abstract
Neuronal inhibition in nociceptive relays of the spinal cord is essential for the proper processing of nociceptive information. In the spinal cord dorsal horn, the activity of synaptic and extrasynaptic GABAA and glycine receptors generates rapid, Cl(-)-dependent neuronal inhibition. A loss of this ionotropic inhibition, particularly through the collapse of the inhibitory Cl(-)-gradient, is a key mechanism by which pathological pain conditions develop. This review summarizes the roles of ionotropic inhibition in the regulation of nociception, and explores recent evidence that the potentiation of GABAA or glycine receptor activity or the enhancement of inhibitory drive can reverse pathological pain.
Collapse
Affiliation(s)
- Robert P Bonin
- Unité de neurosciences cellulaires et moléculaire, Centre de recherche de l'institut universitaire en santé mentale de Québec, Québec, Canada
| | | |
Collapse
|
124
|
Defective escape behavior in DEAH-box RNA helicase mutants improved by restoring glycine receptor expression. J Neurosci 2013; 33:14638-44. [PMID: 24027265 DOI: 10.1523/jneurosci.1157-13.2013] [Citation(s) in RCA: 22] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2022] Open
Abstract
RNA helicases regulate RNA metabolism, but their substrate specificity and in vivo function remain largely unknown. We isolated spontaneous mutant zebrafish that exhibit an abnormal dorsal bend at the beginning of tactile-evoked escape swimming. Similar behavioral defects were observed in zebrafish embryos treated with strychnine, which blocks glycine receptors (GlyRs), suggesting that the abnormal motor response in mutants may be attributable to a deficit in glycinergic synaptic transmission. We identified a missense mutation in the gene encoding RNA helicase Dhx37. In Dhx37 mutants, ribosomal RNA levels were unchanged, whereas GlyR α1, α3, and α4a subunit mRNA levels were decreased due to a splicing defect. We found that Dhx37 can interact with GlyR α1, α3, and α4a transcripts but not with the GlyR α2 subunit mRNA. Overexpression of GlyR α1, α3, or α4a subunits in Dhx37-deficient embryos restored normal behavior. Conversely, antisense-mediated knockdown of multiple GlyR α subunits in wild-type embryos was required to recapitulate the Dhx37 mutant phenotype. These results indicate that Dhx37 is specifically required for the biogenesis of a subset of GlyR α subunit mRNAs, thereby regulating glycinergic synaptic transmission and associated motor behaviors. To our knowledge, this is the first identification of pathologically relevant substrates for an RNA helicase.
Collapse
|
125
|
Activation of glycine and extrasynaptic GABA(A) receptors by taurine on the substantia gelatinosa neurons of the trigeminal subnucleus caudalis. Neural Plast 2013; 2013:740581. [PMID: 24379976 PMCID: PMC3863572 DOI: 10.1155/2013/740581] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/17/2013] [Revised: 11/05/2013] [Accepted: 11/12/2013] [Indexed: 11/23/2022] Open
Abstract
The substantia gelatinosa (SG) of the trigeminal subnucleus caudalis (Vc) has been known for the processing and transmission of orofacial nociceptive information. Taurine, one of the most plentiful free amino-acids in humans, has proved to be involved in pain modulation. In this study, using whole-cell patch clamp technique, we investigated the direct membrane effects of taurine and the action mechanism behind taurine-mediated responses on the SG neurons of the Vc. Taurine showed non-desensitizing and repeatable membrane depolarizations and inward currents which remained in the presence of amino-acid receptors blocking cocktail (AARBC) with tetrodotoxin, indicating that taurine acts directly on the postsynaptic SG neurons. Further, application of taurine at different doses (10 μM to 3 mM) showed a concentration dependent depolarizations and inward currents with the EC50 of 84.3 μM and 723 μM, respectively. Taurine-mediated responses were partially blocked by picrotoxin (50 μM) and almost completely blocked by strychnine (2 μM), suggesting that taurine-mediated responses are via glycine receptor (GlyR) activation. In addition, taurine (1 mM) activated extrasynaptic GABAA receptor (GABAAR)-mediated currents. Taken together, our results indicate that taurine can be a target molecule for orofacial pain modulation through the activation of GlyRs and/or extrasynaptic GABAARs on the SG neurons.
Collapse
|
126
|
Bode A, Wood SE, Mullins JGL, Keramidas A, Cushion TD, Thomas RH, Pickrell WO, Drew CJG, Masri A, Jones EA, Vassallo G, Born AP, Alehan F, Aharoni S, Bannasch G, Bartsch M, Kara B, Krause A, Karam EG, Matta S, Jain V, Mandel H, Freilinger M, Graham GE, Hobson E, Chatfield S, Vincent-Delorme C, Rahme JE, Afawi Z, Berkovic SF, Howell OW, Vanbellinghen JF, Rees MI, Chung SK, Lynch JW. New hyperekplexia mutations provide insight into glycine receptor assembly, trafficking, and activation mechanisms. J Biol Chem 2013; 288:33745-33759. [PMID: 24108130 DOI: 10.1074/jbc.m113.509240] [Citation(s) in RCA: 33] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/06/2022] Open
Abstract
Hyperekplexia is a syndrome of readily provoked startle responses, alongside episodic and generalized hypertonia, that presents within the first month of life. Inhibitory glycine receptors are pentameric ligand-gated ion channels with a definitive and clinically well stratified linkage to hyperekplexia. Most hyperekplexia cases are caused by mutations in the α1 subunit of the human glycine receptor (hGlyR) gene (GLRA1). Here we analyzed 68 new unrelated hyperekplexia probands for GLRA1 mutations and identified 19 mutations, of which 9 were novel. Electrophysiological analysis demonstrated that the dominant mutations p.Q226E, p.V280M, and p.R414H induced spontaneous channel activity, indicating that this is a recurring mechanism in hGlyR pathophysiology. p.Q226E, at the top of TM1, most likely induced tonic activation via an enhanced electrostatic attraction to p.R271 at the top of TM2, suggesting a structural mechanism for channel activation. Receptors incorporating p.P230S (which is heterozygous with p.R65W) desensitized much faster than wild type receptors and represent a new TM1 site capable of modulating desensitization. The recessive mutations p.R72C, p.R218W, p.L291P, p.D388A, and p.E375X precluded cell surface expression unless co-expressed with α1 wild type subunits. The recessive p.E375X mutation resulted in subunit truncation upstream of the TM4 domain. Surprisingly, on the basis of three independent assays, we were able to infer that p.E375X truncated subunits are incorporated into functional hGlyRs together with unmutated α1 or α1 plus β subunits. These aberrant receptors exhibit significantly reduced glycine sensitivity. To our knowledge, this is the first suggestion that subunits lacking TM4 domains might be incorporated into functional pentameric ligand-gated ion channel receptors.
Collapse
Affiliation(s)
- Anna Bode
- University of Queensland, Queensland Brain Institute and School of Biomedical Sciences, Queensland 4072, Australia
| | - Sian-Elin Wood
- Department of Neurology Research and Molecular Neuroscience, Institute of Life Science, College of Medicine, Swansea University Swansea SA2 8PP, United Kingdom
| | - Jonathan G L Mullins
- Department of Neurology Research and Molecular Neuroscience, Institute of Life Science, College of Medicine, Swansea University Swansea SA2 8PP, United Kingdom
| | - Angelo Keramidas
- University of Queensland, Queensland Brain Institute and School of Biomedical Sciences, Queensland 4072, Australia
| | - Thomas D Cushion
- Department of Neurology Research and Molecular Neuroscience, Institute of Life Science, College of Medicine, Swansea University Swansea SA2 8PP, United Kingdom
| | - Rhys H Thomas
- Department of Neurology Research and Molecular Neuroscience, Institute of Life Science, College of Medicine, Swansea University Swansea SA2 8PP, United Kingdom; Wales Epilepsy Research Network, College of Medicine, Swansea University Swansea SA2 8PP, United Kingdom
| | - William O Pickrell
- Department of Neurology Research and Molecular Neuroscience, Institute of Life Science, College of Medicine, Swansea University Swansea SA2 8PP, United Kingdom; Wales Epilepsy Research Network, College of Medicine, Swansea University Swansea SA2 8PP, United Kingdom
| | - Cheney J G Drew
- Department of Neurology Research and Molecular Neuroscience, Institute of Life Science, College of Medicine, Swansea University Swansea SA2 8PP, United Kingdom; Wales Epilepsy Research Network, College of Medicine, Swansea University Swansea SA2 8PP, United Kingdom
| | - Amira Masri
- Department of Paediatrics, Division of Child Neurology, Faculty of Medicine, University of Jordan, Amman 11942, Jordan
| | - Elizabeth A Jones
- Manchester Centre for Genomic Medicine, Central Manchester University Hospitals National Health Service Foundation Trust, Manchester Academic Health Sciences Centre, Manchester M13 9WL, United Kingdom; Manchester Centre for Genomic Medicine, Institute of Human Development, Faculty of Medical and Human Sciences, University of Manchester, Manchester Academic Health Sciences Centre, Manchester M13 9WL, United Kingdom
| | - Grace Vassallo
- Royal Manchester Children's Hospital, Central Manchester University Hospitals National Health Service Foundation Trust, Manchester Academic Health Sciences Centre, Manchester M13 9WL, United Kingdom
| | - Alfred P Born
- Department of Pediatrics, Copenhagen University Hospital, Rigshospitalet, 2100 Copenhagen, Denmark
| | - Fusun Alehan
- Department of Pediatrics, Division of Child Neurology, Faculty of Medicine, Basşkent University, 06990 Ankara, Turkey
| | - Sharon Aharoni
- Institute of Pediatric Neurology, Schneider Children's Medical Center of Israel, Petah Tikva 49202, Israel; Sackler Faculty of Medicine, Tel Aviv University, Tel Aviv 69987, Israel
| | - Gerald Bannasch
- Neurology Department, Affinity Medical Group, Menasha, Wisconsin 54952
| | - Marius Bartsch
- Department of Neonatology, University Medical Center of the Johannes Gutenberg University Mainz, D-55099 Mainz, Germany
| | - Bulent Kara
- Kocaeli University Medical Faculty, Department of Pediatrics, Division of Child Neurology, 41380 Kocaeli, Turkey
| | - Amanda Krause
- Division of Human Genetics, National Health Laboratory Service, and School of Pathology, Faculty of Health Sciences, University of the Witwatersrand, 2000 Johannesburg, South Africa
| | - Elie G Karam
- Department of Psychiatry and Clinical Psychology, Saint George Hospital University Medical Center, Balamand University, Faculty of Medicine, Beirut 1100 2807, Lebanon
| | - Stephanie Matta
- Department of Psychiatry and Clinical Psychology, Saint George Hospital University Medical Center, Balamand University, Faculty of Medicine, Beirut 1100 2807, Lebanon
| | - Vivek Jain
- Royal Children's Hospital Melbourne, Children's Neuroscience Centre, Royal Children's Hospital, Victoria 3052, Australia
| | - Hanna Mandel
- Metabolic Unit, Meyer Children's Hospital, Rambam Medical Center, Technion Faculty of Medicine, Haifa 31096, Israel
| | - Michael Freilinger
- Department of Pediatrics and Adolescent Medicine, Medical University of Vienna, 1090 Vienna, Austria
| | - Gail E Graham
- Department of Genetics, Children's Hospital of Eastern Ontario, Ottawa, Ontario K1H 8L1, Canada
| | - Emma Hobson
- Yorkshire Regional Genetic Service, Chapel Allerton Hospital, Leeds, West Yorkshire LS9 7TF, United Kingdom
| | - Sue Chatfield
- Neonatal Unit, Bradford Royal Infirmary, Bradford, West Yorkshire BD9 6RJ, United Kingdom
| | | | | | - Zaid Afawi
- Zlotowski Center for Neuroscience, Ben-Gurion University of the Negev, Beer-Sheva 8410501, Israel
| | - Samuel F Berkovic
- Epilepsy Research Centre, Melbourne Brain Centre, Austin Health, Heidelberg 3084, Victoria, Australia
| | - Owain W Howell
- Department of Neurology Research and Molecular Neuroscience, Institute of Life Science, College of Medicine, Swansea University Swansea SA2 8PP, United Kingdom; Wales Epilepsy Research Network, College of Medicine, Swansea University Swansea SA2 8PP, United Kingdom
| | | | - Mark I Rees
- Department of Neurology Research and Molecular Neuroscience, Institute of Life Science, College of Medicine, Swansea University Swansea SA2 8PP, United Kingdom; Wales Epilepsy Research Network, College of Medicine, Swansea University Swansea SA2 8PP, United Kingdom
| | - Seo-Kyung Chung
- Department of Neurology Research and Molecular Neuroscience, Institute of Life Science, College of Medicine, Swansea University Swansea SA2 8PP, United Kingdom; Wales Epilepsy Research Network, College of Medicine, Swansea University Swansea SA2 8PP, United Kingdom
| | - Joseph W Lynch
- University of Queensland, Queensland Brain Institute and School of Biomedical Sciences, Queensland 4072, Australia.
| |
Collapse
|
127
|
Liu Q, Wong-Riley MTT. Postnatal development of glycine receptor subunits α1, α2, α3, and β immunoreactivity in multiple brain stem respiratory-related nuclear groups of the rat. Brain Res 2013; 1538:1-16. [PMID: 24080401 DOI: 10.1016/j.brainres.2013.09.028] [Citation(s) in RCA: 28] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/16/2013] [Revised: 09/16/2013] [Accepted: 09/20/2013] [Indexed: 01/01/2023]
Abstract
The respiratory system is immature at birth and significant development occurs postnatally. A critical period of respiratory development occurs in rats around postnatal days 12-13, when enhanced inhibition dominates over suppressed excitation. The mechanisms underlying the heightened inhibition are not fully understood. The present study tested our hypothesis that the inhibition is marked by a switch in glycine receptor subunits from neonatal to adult form around the critical period. An in-depth immunohistochemical and single neuron optical densitometric study was undertaken on four respiratory-related nuclear groups (the pre-Bötzinger complex, nucleus ambiguus, hypoglossal nucleus, and ventrolateral subnucleus of solitary tract nucleus), and a non-respiratory cuneate nucleus in P2-21 rats. Our data revealed that in the respiratory-related nuclear groups: (1) the expressions of GlyRα2 and GlyRα3 were relatively high at P2, but declined after 1-1½ weeks to their lowest levels at P21; (2) the expression of GlyRα1 increased with age and reached significance at P12; and (3) the expression of GlyRβ rose from P2 to P12 followed by a slight decline until P21. No distinct increase in GlyRα1 at P12 was noted in the cuneate nucleus. Thus, there is a switch in dominance of expression from neonatal GlyRα2/α3 to the adult GlyRα1 and a heightened expression of GlyRα1 around the critical period in all respiratory-related nuclear groups, thereby supporting enhanced inhibition at that time. The rise in the expression of GlyRβ around P12 indicates that it plays an important role in forming the mature heteropentameric glycine receptors in these brain stem nuclear groups.
Collapse
Affiliation(s)
- Qiuli Liu
- Department of Cell Biology, Neurobiology and Anatomy, Medical College of Wisconsin, 8701 Watertown Plank Road, Milwaukee, WI 53226, USA
| | | |
Collapse
|
128
|
Thomas RH, Chung SK, Wood SE, Cushion TD, Drew CJG, Hammond CL, Vanbellinghen JF, Mullins JGL, Rees MI. Genotype-phenotype correlations in hyperekplexia: apnoeas, learning difficulties and speech delay. Brain 2013; 136:3085-95. [DOI: 10.1093/brain/awt207] [Citation(s) in RCA: 58] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/14/2022] Open
|
129
|
Gelsemine, a principal alkaloid from Gelsemium sempervirens Ait., exhibits potent and specific antinociception in chronic pain by acting at spinal α3 glycine receptors. Pain 2013; 154:2452-2462. [PMID: 23886522 DOI: 10.1016/j.pain.2013.07.027] [Citation(s) in RCA: 76] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/30/2013] [Revised: 07/14/2013] [Accepted: 07/17/2013] [Indexed: 11/22/2022]
Abstract
The present study examined the antinociceptive effects of gelsemine, the principal alkaloid in Gelsemium sempervirens Ait. A single intrathecal injection of gelsemine produced potent and specific antinociception in formalin-induced tonic pain, bone cancer-induced mechanical allodynia, and spinal nerve ligation-induced painful neuropathy. The antinociception was dose-dependent, with maximal inhibition of 50% to 60% and ED50 values of 0.5 to 0.6 μg. Multiple daily intrathecal injections of gelsemine for 7 days induced no tolerance to antinociception in the rat model of bone cancer pain. Spinal gelsemine was not effective in altering contralateral paw withdrawal thresholds, and had only a slight inhibitory effect on formalin-induced acute nociception. The specific antinociception of gelsemine in chronic pain was blocked dose-dependently by the glycine receptor (GlyR) antagonist strychnine with an apparent ID50 value of 3.8 μg. Gelsemine concentration-dependently displaced H(3)-strychnine binding to the membrane fraction of rat spinal cord homogenates, with a 100% displacement and a Ki of 21.9μM. Gene ablation of the GlyR α3 subunit (α3 GlyR) but not α1 GlyR, by a 7-day intrathecal injection of small interfering RNA (siRNA) targeting α3 GlyR or α1 GlyR, nearly completely prevented gelsemine-induced antinociception in neuropathic pain. Our results demonstrate that gelsemine produces potent and specific antinociception in chronic pain states without induction of apparent tolerance. The results also suggest that gelsemine produces antinociception by activation of spinal α3 glycine receptors, and support the notion that spinal α3 glycine receptors are a potential therapeutic target molecule for the management of chronic pain.
Collapse
|
130
|
Weltzien F, Puller C, O'Sullivan GA, Paarmann I, Betz H. Distribution of the glycine receptor β-subunit in the mouse CNS as revealed by a novel monoclonal antibody. J Comp Neurol 2013; 520:3962-81. [PMID: 22592841 DOI: 10.1002/cne.23139] [Citation(s) in RCA: 40] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/05/2022]
Abstract
Inhibitory glycine receptors (GlyRs) are composed of homologous α- (α1-4) and β-subunits. The β-subunits (GlyRβ) interact via their large cytosolic loops with the postsynaptic scaffolding protein gephyrin and are therefore considered essential for synaptic localization. In situ hybridization studies indicate a widespread distribution of GlyRβ transcripts throughout the mammalian central nervous system (CNS), whereas GlyRα mRNAs and proteins display more restricted expression patterns. Here we report the generation of a monoclonal antibody that specifically recognizes rodent GlyRβ (mAb-GlyRβ) and does not exhibit crossreactivity with any of the GlyRα1-4 subunits. Immunostaining with this antibody revealed high densities of punctate GlyRβ immunoreactivity at inhibitory synapses in mouse spinal cord, brainstem, midbrain, and olfactory bulb but not in the neocortex, cerebellum, or hippocampus. This contrasts the abundance of GlyRβ transcripts in all major regions of the rodent brain and suggests that GlyRβ protein levels are regulated posttranscriptionally. When mAb-GlyRβ was used in double-labeling experiments with GlyRα1-, α2-, α3-, or α4-specific antibodies to examine the colocalization of GlyRβ with these GlyR subunits in the mouse retina, >90% of the GlyRα1-3 clusters detected were found to be GlyRβ-immunoreactive. A subset (about 50%) of the GlyRα4 puncta in the inner plexiform layer, however, was found to lack GlyRβ and gephyrin immunostaining. These GlyRα4-only clusters were apposed to bassoon immunoreactivity and hence synaptically localized. Their existence points to a gephyrin-independent synaptic localization mechanism for a minor subset of GlyRs.
Collapse
Affiliation(s)
- Felix Weltzien
- Department of Neurochemistry, Max-Planck Institute for Brain Research, 60528 Frankfurt, Germany
| | | | | | | | | |
Collapse
|
131
|
New insights in endogenous modulation of ligand-gated ion channels: histamine is an inverse agonist at strychnine sensitive glycine receptors. Eur J Pharmacol 2013; 710:59-66. [PMID: 23603522 DOI: 10.1016/j.ejphar.2013.04.002] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/09/2012] [Revised: 03/28/2013] [Accepted: 04/03/2013] [Indexed: 01/18/2023]
Abstract
Histamine is involved in many physiological functions in the periphery and is an important neurotransmitter in the brain. It acts on metabotropic H1-H4 receptors mediating vasodilatation, bronchoconstriction and stimulation of gastric acid secretion. In the brain histamine is produced by neurons in the tuberomamillary nucleus (TMN), which controls arousal. Histamine is also a positive modulator of the inhibitory Cys-loop ligand-gated ion channel GABAA. We investigated now its effect on the second member of inhibitory Cys-loop ligand-gated ion channels, the strychnine sensitive glycine receptor. We expressed different human and rat glycine receptor subunits in Xenopus laevis oocytes and characterized the effect of histamine using the two electrode voltage clamp technique. Furthermore we investigated native glycine receptors in hypothalamic neurons using the patch-clamp technique. Histamine inhibited α1β glycine receptors with an IC50 of 5.2±0.3 mM. In presence of 10 mM histamine the glycine dose-response curve was shifted, increasing the EC50 for glycine from 25.5±1.4 μM to 42.4±2.3 μM. In addition, histamine blocked the spontaneous activity of RNA-edited α3β glycine receptors. Histamine inhibited glycine receptors expressed in hypothalamic TMN neurons with an IC50 of 4.6±0.3 mM. Our results give strong evidence that histamine is acting on the same binding site as glycine, being an inverse agonist that competitively antagonizes glycine receptors. Thus, we revealed histamine as an endogenous modulator of glycine receptors.
Collapse
|
132
|
Han L, Talwar S, Lynch JW. The relative orientation of the TM3 and TM4 domains varies between α1 and α3 glycine receptors. ACS Chem Neurosci 2013; 4:248-54. [PMID: 23421675 DOI: 10.1021/cn300177g] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/28/2022] Open
Abstract
Glycine receptors (GlyRs) are anion-conducting members of the pentameric ligand-gated ion channel family. We previously showed that the dramatic difference in glycine efficacies of α1 and α3 GlyRs is largely attributable to their nonconserved TM4 domains. Because mutation of individual nonconserved TM4 residues had little effect, we concluded that the efficacy difference was a distributed effect of all nonconserved TM4 residues. We therefore hypothesized that the TM4 domains of α1 and α3 GlyRs differ in structure, membrane orientation, and/or molecular dynamic properties. Here we employed voltage-clamp fluorometry to test whether their TM4 domains interact differently with their respective TM3 domains. We found a rhodamine fluorophore covalently attached to a homologous TM4 residue in each receptor interacts differentially with a conserved TM3 residue. We conclude that the α1 and α3 GlyR TM4 domains are orientated differently relative to their TM3 domains. This may underlie their differential ability to influence glycine efficacy.
Collapse
Affiliation(s)
- Lu Han
- Queensland Brain Institute and ‡School of Biomedical
Sciences, The University of Queensland, Brisbane QLD 4072, Australia
| | - Sahil Talwar
- Queensland Brain Institute and ‡School of Biomedical
Sciences, The University of Queensland, Brisbane QLD 4072, Australia
| | - Joseph W. Lynch
- Queensland Brain Institute and ‡School of Biomedical
Sciences, The University of Queensland, Brisbane QLD 4072, Australia
| |
Collapse
|
133
|
Differential distribution of glycine receptor subtypes at the rat calyx of Held synapse. J Neurosci 2013; 32:17012-24. [PMID: 23175852 DOI: 10.1523/jneurosci.1547-12.2012] [Citation(s) in RCA: 22] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2022] Open
Abstract
The properties of glycine receptors (GlyRs) depend upon their subunit composition. While the prevalent adult forms of GlyRs are heteromers, previous reports suggested functional α homomeric receptors in mature nervous tissues. Here we show two functionally different GlyRs populations in the rat medial nucleus of trapezoid body (MNTB). Postsynaptic receptors formed α1/β-containing clusters on somatodendritic domains of MNTB principal neurons, colocalizing with glycinergic nerve endings to mediate fast, phasic IPSCs. In contrast, presynaptic receptors on glutamatergic calyx of Held terminals were composed of dispersed, homomeric α1 receptors. Interestingly, the parent cell bodies of the calyces of Held, the globular bushy cells of the cochlear nucleus, expressed somatodendritic receptors (α1/β heteromers) and showed similar clustering and pharmacological profile as GlyRs on MNTB principal cells. These results suggest that specific targeting of GlyR β-subunit produces segregation of GlyR subtypes involved in two different mechanisms of modulation of synaptic strength.
Collapse
|
134
|
Lee CG, Kwon MJ, Yu HJ, Nam SH, Lee J, Ki CS, Lee M. Clinical features and genetic analysis of children with hyperekplexia in Korea. J Child Neurol 2013; 28:90-4. [PMID: 22532536 DOI: 10.1177/0883073812441058] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/15/2022]
Abstract
Hyperekplexia is a rare inherited neurologic disorder that is characterized by hypertonia and an exaggerated startle response to sudden external stimuli. Until now, 5 genes are known to be associated with hyperekplexia: GLRA1, SLC6A5, GLRB, GPHN, and ARHGEF9. In this report, we performed a clinical and genetic analysis of 4 Korean children with hyperekplexia. Two patients had typical clinical manifestations of hyperekplexia that initially were misdiagnosed as epilepsy. Direct sequencing of the GLRB and GLRA1 genes revealed 2 novel mutations, GLRB c.298-1G>A and c.1028C>T (p.S343F), in patient 1 and 1 novel mutation, GLRA1 c.895C>T (p.R299X), in patient 2. The other 2 familial cases, patients 3 and 4, exhibited startle responses, which appeared at the age of 1 year, and had global developmental delay. Those patients showed negative results for the 5 genes.
Collapse
Affiliation(s)
- Cha Gon Lee
- Department of Pediatrics, Samsung Medical Center, Sungkyunkwan University School of Medicine, Seoul, Korea
| | | | | | | | | | | | | |
Collapse
|
135
|
James VM, Bode A, Chung SK, Gill JL, Nielsen M, Cowan FM, Vujic M, Thomas RH, Rees MI, Harvey K, Keramidas A, Topf M, Ginjaar I, Lynch JW, Harvey RJ. Novel missense mutations in the glycine receptor β subunit gene (GLRB) in startle disease. Neurobiol Dis 2012; 52:137-49. [PMID: 23238346 PMCID: PMC3581774 DOI: 10.1016/j.nbd.2012.12.001] [Citation(s) in RCA: 45] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/11/2012] [Revised: 11/26/2012] [Accepted: 12/03/2012] [Indexed: 02/03/2023] Open
Abstract
Startle disease is a rare, potentially fatal neuromotor disorder characterized by exaggerated startle reflexes and hypertonia in response to sudden unexpected auditory, visual or tactile stimuli. Mutations in the GlyR α(1) subunit gene (GLRA1) are the major cause of this disorder, since remarkably few individuals with mutations in the GlyR β subunit gene (GLRB) have been found to date. Systematic DNA sequencing of GLRB in individuals with hyperekplexia revealed new missense mutations in GLRB, resulting in M177R, L285R and W310C substitutions. The recessive mutation M177R results in the insertion of a positively-charged residue into a hydrophobic pocket in the extracellular domain, resulting in an increased EC(50) and decreased maximal responses of α(1)β GlyRs. The de novo mutation L285R results in the insertion of a positively-charged side chain into the pore-lining 9' position. Mutations at this site are known to destabilize the channel closed state and produce spontaneously active channels. Consistent with this, we identified a leak conductance associated with spontaneous GlyR activity in cells expressing α(1)β(L285R) GlyRs. Peak currents were also reduced for α(1)β(L285R) GlyRs although glycine sensitivity was normal. W310C was predicted to interfere with hydrophobic side-chain stacking between M1, M2 and M3. We found that W310C had no effect on glycine sensitivity, but reduced maximal currents in α(1)β GlyRs in both homozygous (α(1)β(W310C)) and heterozygous (α(1)ββ(W310C)) stoichiometries. Since mild startle symptoms were reported in W310C carriers, this may represent an example of incomplete dominance in startle disease, providing a potential genetic explanation for the 'minor' form of hyperekplexia.
Collapse
Affiliation(s)
- Victoria M James
- Department of Pharmacology, UCL School of Pharmacy, 29-39 Brunswick Square, London WC1N 1AX, United Kingdom
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
136
|
Abstract
The subunit stoichiometry of heteromeric glycine-gated channels determines fundamental properties of these key inhibitory neurotransmitter receptors; however, the ratio of α1- to β-subunits per receptor remains controversial. We used single-molecule imaging and stepwise photobleaching in Xenopus oocytes to directly determine the subunit stoichiometry of a glycine receptor to be 3α1:2β. This approach allowed us to determine the receptor stoichiometry in mixed populations consisting of both heteromeric and homomeric channels, additionally revealing the quantitative proportions for the two populations.
Collapse
|
137
|
Chung SK, Bode A, Cushion TD, Thomas RH, Hunt C, Wood SE, Pickrell WO, Drew CJG, Yamashita S, Shiang R, Leiz S, Longardt AC, Longhardt AC, Raile V, Weschke B, Puri RD, Verma IC, Harvey RJ, Ratnasinghe DD, Parker M, Rittey C, Masri A, Lingappa L, Howell OW, Vanbellinghen JF, Mullins JG, Lynch JW, Rees MI. GLRB is the third major gene of effect in hyperekplexia. Hum Mol Genet 2012. [PMID: 23184146 DOI: 10.1093/hmg/dds498] [Citation(s) in RCA: 43] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/13/2022] Open
Abstract
Glycinergic neurotransmission is a major inhibitory influence in the CNS and its disruption triggers a paediatric and adult startle disorder, hyperekplexia. The postsynaptic α(1)-subunit (GLRA1) of the inhibitory glycine receptor (GlyR) and the cognate presynaptic glycine transporter (SLC6A5/GlyT2) are well-established genes of effect in hyperekplexia. Nevertheless, 52% of cases (117 from 232) remain gene negative and unexplained. Ligand-gated heteropentameric GlyRs form chloride ion channels that contain the α(1) and β-subunits (GLRB) in a 2α(1):3β configuration and they form the predominant population of GlyRs in the postnatal and adult human brain, brainstem and spinal cord. We screened GLRB through 117 GLRA1- and SLC6A5-negative hyperekplexia patients using a multiplex-polymerase chain reaction and Sanger sequencing approach. The screening identified recessive and dominant GLRB variants in 12 unrelated hyperekplexia probands. This primarily yielded homozygous null mutations, with nonsense (n = 3), small indel (n = 1), a large 95 kb deletion (n = 1), frameshifts (n = 1) and one recurrent splicing variant found in four cases. A further three cases were found with two homozygous and one dominant GLRB missense mutations. We provide strong evidence for the pathogenicity of GLRB mutations using splicing assays, deletion mapping, cell-surface biotinylation, expression studies and molecular modelling. This study describes the definitive assignment of GLRB as the third major gene for hyperekplexia and impacts on the genetic stratification and biological causation of this neonatal/paediatric disorder. Driven principally by consanguineous homozygosity of GLRB mutations, the study reveals long-term additive phenotypic outcomes for affected cases such as severe apnoea attacks, learning difficulties and developmental delay.
Collapse
Affiliation(s)
- Seo-Kyung Chung
- Neurology Research and Molecular Neuroscience, Institute of Life Science, Swansea University, Swansea SA2 8PP, UK.
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
138
|
Abstract
Strychnine-sensitive glycine receptors (GlyRs) mediate synaptic inhibition in the spinal cord, brainstem, and other regions of the mammalian central nervous system. In this minireview, we summarize our current view of the structure, ligand-binding sites, and chloride channel of these receptors and discuss recently emerging functions of distinct GlyR isoforms. GlyRs not only regulate the excitability of motor and afferent sensory neurons, including pain fibers, but also are involved in the processing of visual and auditory signals. Hence, GlyRs constitute promising targets for the development of therapeutically useful compounds.
Collapse
Affiliation(s)
- Sébastien Dutertre
- From the Institute for Molecular Bioscience, University of Queensland, Brisbane, Queensland 4072, Australia
| | - Cord-Michael Becker
- the Institute of Biochemistry, University of Erlangen-Nürnberg, 91054 Erlangen, Germany
| | - Heinrich Betz
- the Max-Planck-Institute for Medical Research, 69120 Heidelberg, Germany, and
- the Department of Molecular Neurobiology, Max-Planck-Institute for Experimental Medicine, 37075 Göttingen, Germany
| |
Collapse
|
139
|
Wang L, Li WG, Huang C, Zhu MX, Xu TL, Wu DZ, Li Y. Subunit-specific inhibition of glycine receptors by curcumol. J Pharmacol Exp Ther 2012; 343:371-9. [PMID: 22892339 DOI: 10.1124/jpet.112.195669] [Citation(s) in RCA: 11] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/15/2023] Open
Abstract
Emerging evidence has suggested that inhibitory glycine receptors (GlyRs) are an important molecular target in the treatment of numerous neurological disorders. Rhizoma curcumae is a medicinal plant with positive neurological effects. In this study, we showed that curcumol, a major bioactive component of R. curcumae, reversibly and concentration-dependently inhibited the glycine-activated current (IGly) in cultured rat hippocampal neurons. The inhibitory effect was neither voltage- nor agonist concentration-dependent. Moreover, curcumol selectively inhibited homomeric α2-containing, but not α1- or α3-containing, GlyRs. The addition of β subunit conferred the curcumol sensitivity of α3-containing, but not α1-containing, GlyRs. Site-directed mutagenesis analysis revealed that a threonine at position 59 of the α2 subunit is critical for the susceptibility of GlyRs to curcumol-mediated inhibition. Furthermore, paralleling a decline of α2 subunit expression during spinal cord development, the degree of IGly inhibition by curcumol decreased with prolonged culture of rat spinal dorsal horn neurons. Taken together, our results suggest that the GlyRs are novel molecular targets of curcumol, which may underlie its pharmaceutical effects in the central nervous system.
Collapse
MESH Headings
- Amino Acid Sequence
- Animals
- Cells, Cultured
- Central Nervous System/drug effects
- Curcuma/chemistry
- Data Interpretation, Statistical
- Electrophysiological Phenomena
- Female
- Hippocampus/cytology
- Hippocampus/drug effects
- Medicine, Chinese Traditional
- Molecular Sequence Data
- Mutagenesis, Site-Directed
- Patch-Clamp Techniques
- Pregnancy
- Rats
- Rats, Sprague-Dawley
- Receptors, GABA-A/drug effects
- Receptors, Glycine/antagonists & inhibitors
- Receptors, Glycine/chemistry
- Receptors, Glycine/drug effects
- Receptors, Glycine/genetics
- Recombinant Proteins
- Sesquiterpenes/pharmacology
- Threonine/chemistry
Collapse
Affiliation(s)
- Li Wang
- Institute of Chinese Materia Medica, Shanghai Key Laboratory of Complex Prescription, Shanghai University of Traditional Chinese Medicine, Shanghai, China
| | | | | | | | | | | | | |
Collapse
|
140
|
Schaefer N, Vogel N, Villmann C. Glycine receptor mutants of the mouse: what are possible routes of inhibitory compensation? Front Mol Neurosci 2012; 5:98. [PMID: 23118727 PMCID: PMC3484359 DOI: 10.3389/fnmol.2012.00098] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/07/2012] [Accepted: 10/11/2012] [Indexed: 12/02/2022] Open
Abstract
Defects in glycinergic inhibition result in a complex neuromotor disorder in humans known as hyperekplexia (OMIM 149400) with similar phenotypes in rodents characterized by an exaggerated startle reflex and hypertonia. Analogous to genetic defects in humans single point mutations, microdeletions, or insertions in the Glra1 gene but also in the Glrb gene underlie the pathology in mice. The mutations either localized in the α (spasmodic, oscillator, cincinnati, Nmf11) or the β (spastic) subunit of the glycine receptor (GlyR) are much less tolerated in mice than in humans, leaving the question for the existence of different regulatory elements of the pathomechanisms in humans and rodents. In addition to the spontaneous mutations, new insights into understanding of the regulatory pathways in hyperekplexia or glycine encephalopathy arose from the constantly increasing number of knock-out as well as knock-in mutants of GlyRs. Over the last five years, various efforts using in vivo whole cell recordings provided a detailed analysis of the kinetic parameters underlying glycinergic dysfunction. Presynaptic compensation as well as postsynaptic compensatory mechanisms in these mice by other GlyR subunits or GABAA receptors, and the role of extra-synaptic GlyRs is still a matter of debate. A recent study on the mouse mutant oscillator displayed a novel aspect for compensation of functionality by complementation of receptor domains that fold independently. This review focuses on defects in glycinergic neurotransmission in mice discussed with the background of human hyperekplexia en route to strategies of compensation.
Collapse
Affiliation(s)
- Natascha Schaefer
- Emil Fischer Center, Institute of Biochemistry, University Erlangen-Nuernberg Erlangen, Germany ; Institute for Clinical Neurobiology, University of Wuerzburg Wuerzburg, Germany
| | | | | |
Collapse
|
141
|
Lu Y, Zhang J, Ma B, Li K, Li X, Bai H, Yang Q, Zhu X, Ben J, Chen Q. Glycine attenuates cerebral ischemia/reperfusion injury by inhibiting neuronal apoptosis in mice. Neurochem Int 2012; 61:649-58. [PMID: 22796215 DOI: 10.1016/j.neuint.2012.07.005] [Citation(s) in RCA: 30] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/05/2012] [Revised: 06/18/2012] [Accepted: 07/03/2012] [Indexed: 10/28/2022]
|
142
|
de la Roche J, Leuwer M, Krampfl K, Haeseler G, Dengler R, Buchholz V, Ahrens J. 4-Chloropropofol enhances chloride currents in human hyperekplexic and artificial mutated glycine receptors. BMC Neurol 2012; 12:104. [PMID: 23006332 PMCID: PMC3517478 DOI: 10.1186/1471-2377-12-104] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/12/2011] [Accepted: 09/19/2012] [Indexed: 01/23/2023] Open
Abstract
BACKGROUND The mammalian neurological disorder hereditary hyperekplexia can be attributed to various mutations of strychnine sensitive glycine receptors. The clinical symptoms of "startle disease" predominantly occur in the newborn leading to convulsive hypertonia and an exaggerated startle response to unexpected mild stimuli. Amongst others, point mutations R271Q and R271L in the α1-subunit of strychnine sensitive glycine receptors show reduced glycine sensitivity and cause the clinical symptoms of hyperekplexia.Halogenation has been shown to be a crucial structural determinant for the potency of a phenolic compound to positively modulate glycine receptor function.The aim of this in vitro study was to characterize the effects of 4-chloropropofol (4-chloro-2,6-dimethylphenol) at four glycine receptor mutations. METHODS Glycine receptor subunits were expressed in HEK 293 cells and experiments were performed using the whole-cell patch-clamp technique. RESULTS 4-chloropropofol exerted a positive allosteric modulatory effect in a low sub-nanomolar concentration range at the wild type receptor (EC50 value of 0.08 ± 0.02 nM) and in a micromolar concentration range at the mutations (1.3 ± 0.6 μM, 0.1 ± 0.2 μM, 6.0 ± 2.3 μM and 55 ± 28 μM for R271Q, L, K and S267I, respectively). CONCLUSIONS 4-chloropropofol might be an effective compound for the activation of mutated glycine receptors in experimental models of startle disease.
Collapse
Affiliation(s)
- Jeanne de la Roche
- Clinic for Anesthesia and Critical Care Medicine, OE 8050, Hannover Medical School, Carl-Neuberg-Str, 1, 30625, Hannover, Germany.
| | | | | | | | | | | | | |
Collapse
|
143
|
Unterer B, Becker CM, Villmann C. The importance of TM3-4 loop subdomains for functional reconstitution of glycine receptors by independent domains. J Biol Chem 2012; 287:39205-15. [PMID: 22995908 DOI: 10.1074/jbc.m112.376053] [Citation(s) in RCA: 11] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/06/2022] Open
Abstract
Truncated glycine receptors that have been found in human patients suffering from the neuromotor disorder hyperekplexia or in spontaneous mouse models resulted in non-functional ion channels. Rescue of function experiments with the lacking protein portion expressed as a separate independent domain demonstrated restoration of glycine receptor functionality in vitro. This construct harbored most of the TM3-4 loop, TM4, and the C terminus and was required for concomitant transport of the truncated α1 and the complementation domain from the endoplasmic reticulum toward the cell surface, thereby enabling complex formation of functional glycine receptors. Here, the complementation domain was stepwise truncated from its N terminus in the TM3-4 loop. Truncation of more than 49 amino acids led again to loss of functionality in the receptor complex expressed from two independent domain constructs. We identified residues 357-418 in the intracellular TM3-4 loop as being required for reconstitution of functional glycine-gated channels. All complementation constructs showed cell surface protein expression and correct orientation according to glycine receptor topology. Moreover, we demonstrated that the truncations did not result in a decreased protein-protein interaction between both glycine receptor domains. Rather, deletions of more than 49 amino acids abolished conformational changes necessary for ion channel opening. When the TM3-4 loop subdomain harboring residues 357-418 was expressed as a third independent construct together with the truncated N-terminal and C-terminal glycine receptor domains, functionality of the glycine receptor was again restored. Thus, residues 357-418 represent an important determinant in the process of conformational rearrangements following ligand binding resulting in channel opening.
Collapse
Affiliation(s)
- Bea Unterer
- Institute of Biochemistry, Emil Fischer Center, Friedrich-Alexander University Erlangen-Nuernberg, Fahrstrasse 17, 91054 Erlangen, Germany
| | | | | |
Collapse
|
144
|
Shan Q, Han L, Lynch JW. Function of hyperekplexia-causing α1R271Q/L glycine receptors is restored by shifting the affected residue out of the allosteric signalling pathway. Br J Pharmacol 2012; 165:2113-23. [PMID: 21955162 DOI: 10.1111/j.1476-5381.2011.01701.x] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/27/2022] Open
Abstract
BACKGROUND AND PURPOSE Glycine receptor α1 subunit R271Q and R271L (α1R271Q/L) mutations cause the neuromotor disorder, hereditary hyperekplexia. Studies suggest that the 271 residue is located within the allosteric signalling pathway linking the agonist binding site to the channel gate. The present study aimed to investigate a possible mechanism for restoring the function of the α1R271Q/L glycine receptor. EXPERIMENTAL APPROACH A 12-amino-acid segment incorporating the 271 residue on the glycine receptor α1271Q/L subunit was replaced by the homologous segment from the glycine receptor β subunit (α1(Ch) 271Q/L). The function of the α1(Ch) 271Q/L glycine receptor was examined by whole-cell patch-clamp recording and voltage-clamp fluorometry techniques. KEY RESULTS The function of the α1(Ch) 271Q/L glycine receptor was restored to the level of the wild-type (WT) α1 glycine receptor. Moreover, in the α1(Ch) glycine receptor, in contrast to the α1 glycine receptor, the channel function was not sensitive to various substitutions of the 271 residue, and the conformational change in the vicinity of the 271 residue was uncoupled from the channel gating. CONCLUSIONS AND IMPLICATIONS The 271 residue is shifted out of the allosteric signalling pathway in the α1(Ch) glycine receptor. We propose that this mechanism provides a novel drug design strategy not only for glycine receptor α1R271Q/L-caused hereditary hyperekplexia, but also for any pathological condition that is caused by missense mutation- or covalent modification-induced disorders involving residues in allosteric signalling pathways. Such a strategy makes it possible to design an ideal drug, which only corrects the function of the mutant or modified protein without affecting the WT or naive protein.
Collapse
Affiliation(s)
- Qiang Shan
- School of Biomedical Sciences, University of Queensland, Brisbane, Queensland, Australia.
| | | | | |
Collapse
|
145
|
Becker K, Braune M, Benderska N, Buratti E, Baralle F, Villmann C, Stamm S, Eulenburg V, Becker CM. A retroelement modifies pre-mRNA splicing: the murine Glrb(spa) allele is a splicing signal polymorphism amplified by long interspersed nuclear element insertion. J Biol Chem 2012; 287:31185-94. [PMID: 22782896 DOI: 10.1074/jbc.m112.375691] [Citation(s) in RCA: 11] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/06/2022] Open
Abstract
The glycine receptor-deficient mutant mouse spastic carries a full-length long interspersed nuclear element (LINE1) retrotransposon in intron 6 of the glycine receptor β subunit gene, Glrb(spa). The mutation arose in the C57BL/6J strain and is associated with skipping of exon 6 or a combination of the exons 5 and 6, thus resulting in a translational frameshift within the coding regions of the GlyR β subunit. The effect of the Glrb(spa) LINE1 insertion on pre-mRNA splicing was studied using a minigene approach. Sequence comparison as well as motif prediction and mutational analysis revealed that in addition to the LINE1 insertion the inactivation of an exonic splicing enhancer (ESE) within exon 6 is required for skipping of exon 6. Reconstitution of the ESE by substitution of a single residue was sufficient to prevent exon skipping. In addition to the ESE, two regions within the 5' and 3' UTR of the LINE1 were shown to be critical determinants for exon skipping, indicating that LINE1 acts as efficient modifier of subtle endogenous splicing phenotypes. Thus, the spastic allele of the murine glycine receptor β subunit gene is a two-hit mutation, where the hypomorphic alteration in an ESE is amplified by the insertion of a LINE1 element in the adjacent intron. Conversely, the LINE1 effect on splicing may be modulated by individual polymorphisms, depending on the insertional environment within the host genome.
Collapse
Affiliation(s)
- Kristina Becker
- Institut für Biochemie, Emil Fischer Zentrum, Universität Erlangen-Nürnberg, 91054 Erlangen, Germany
| | | | | | | | | | | | | | | | | |
Collapse
|
146
|
Zhong X, Li X, Qian L, Xu Y, Lu Y, Zhang J, Li N, Zhu X, Ben J, Yang Q, Chen Q. Glycine attenuates myocardial ischemia-reperfusion injury by inhibiting myocardial apoptosis in rats. J Biomed Res 2012; 26:346-54. [PMID: 23554770 PMCID: PMC3613731 DOI: 10.7555/jbr.26.20110124] [Citation(s) in RCA: 25] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/03/2011] [Revised: 11/23/2011] [Accepted: 02/21/2012] [Indexed: 11/03/2022] Open
Abstract
Glycine is a well-documented cytoprotective agent. However, whether it has a protective effect against myocardial ischemia-reperfusion injury in vivo is still unknown. By using an open-chest anesthetized rat model, we found that glycine reduced the infarct size by 21% in ischemia-reperfusion injury rats compared with that in the vehicle-treated MI/R rats. The left ventricular ejection fraction and fractional shortening were increased by 19.11% and 30.98%, respectively, in glycine-treated rats. The plasma creatine kinase levels in ischemia-reperfusion injury rats decreased following glycine treatment. Importantly, administration of glycine significantly inhibited apoptosis in post-ischemia-reperfusion myocardium, which was accompanied by suppression of phosphorylated p38 mitogen-activated protein kinase and c-Jun NH2-terminal kinase, as well as the Fas ligand. These results suggest that glycine attenuates myocardial ischemia-reperfusion injury in vivo by inhibiting cardiomyocytes apoptosis.
Collapse
Affiliation(s)
- Xiaozheng Zhong
- Atherosclerosis Research Center, Key Laboratory of Cardiovascular Disease and Molecular Intervention, Nanjing Medical University, Nanjing, Jiangsu 210029, China
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|
147
|
Wöhri AB, Hillertz P, Eriksson PO, Meuller J, Dekker N, Snijder A. Thermodynamic studies of ligand binding to the human homopentameric glycine receptor using isothermal titration calorimetry. Mol Membr Biol 2012; 30:169-83. [DOI: 10.3109/09687688.2012.696733] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/13/2022]
|
148
|
Yang Z, Taran E, Webb TI, Lynch JW. Stoichiometry and subunit arrangement of α1β glycine receptors as determined by atomic force microscopy. Biochemistry 2012; 51:5229-31. [PMID: 22715885 DOI: 10.1021/bi300063m] [Citation(s) in RCA: 46] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/29/2022]
Abstract
The glycine receptor is an anion-permeable member of the Cys-loop ion channel receptor family. Synaptic glycine receptors predominantly comprise pentameric α1β subunit heteromers. To date, attempts to define the subunit stoichiometry and arrangement of these receptors have not yielded consistent results. Here we introduced FLAG and six-His epitopes into α1 and β subunits, respectively, and imaged single antibody-bound α1β receptors using atomic force microscopy. This permitted us to infer the number and relative locations of the respective subunits in functional pentamers. Our results indicate an invariant 2α1:3β stoichiometry with a β-α-β-α-β subunit arrangement.
Collapse
Affiliation(s)
- Zhe Yang
- Queensland Brain Institute, University of Queensland, Brisbane, QLD 4072, Australia
| | | | | | | |
Collapse
|
149
|
Lynagh T, Lynch JW. Ivermectin binding sites in human and invertebrate Cys-loop receptors. Trends Pharmacol Sci 2012; 33:432-41. [PMID: 22677714 DOI: 10.1016/j.tips.2012.05.002] [Citation(s) in RCA: 74] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/18/2012] [Revised: 04/27/2012] [Accepted: 05/07/2012] [Indexed: 10/28/2022]
Abstract
Ivermectin is a gold standard antiparasitic drug that has been used successfully to treat billions of humans, livestock and pets. Until recently, the binding site on its Cys-loop receptor target had been a mystery. Recent protein crystal structures, site-directed mutagenesis data and molecular modelling now explain how ivermectin binds to these receptors and reveal why it is selective for invertebrate members of the Cys-loop receptor family. Combining this with emerging genomic information, we are now in a position to predict species sensitivity to ivermectin and better understand the molecular basis of ivermectin resistance. An understanding of the molecular structure of the ivermectin binding site, which is formed at the interface of two adjacent subunits in the transmembrane domain of the receptor, should also aid the development of new lead compounds both as anthelmintics and as therapies for a wide variety of human neurological disorders.
Collapse
Affiliation(s)
- Timothy Lynagh
- Neurosensory Systems Group, Technical University of Darmstadt, Schnittspahnstrasse 3, 64287 Darmstadt, Germany
| | | |
Collapse
|
150
|
Molecular actions of smoking cessation drugs at α4β2 nicotinic receptors defined in crystal structures of a homologous binding protein. Proc Natl Acad Sci U S A 2012; 109:9173-8. [PMID: 22619328 DOI: 10.1073/pnas.1116397109] [Citation(s) in RCA: 59] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022] Open
Abstract
Partial agonists of the α4β2 nicotinic acetylcholine receptor (nAChR), such as varenicline, are therapeutically used in smoking cessation treatment. These drugs derive their therapeutic effect from fundamental molecular actions, which are to desensitize α4β2 nAChRs and induce channel opening with higher affinity, but lower efficacy than a full agonist at equal receptor occupancy. Here, we report X-ray crystal structures of a unique acetylcholine binding protein (AChBP) from the annelid Capitella teleta, Ct-AChBP, in complex with varenicline or lobeline, which are both partial agonists. These structures highlight the architecture for molecular recognition of these ligands, indicating the contact residues that potentially mediate their molecular actions in α4β2 nAChRs. We then used structure-guided mutagenesis and electrophysiological recordings to pinpoint crucial interactions of varenicline with residues on the complementary face of the binding site in α4β2 nAChRs. We observe that residues in loops D and E are molecular determinants of desensitization and channel opening with limited efficacy by the partial agonist varenicline. Together, this study analyzes molecular recognition of smoking cessation drugs by nAChRs in a structural context.
Collapse
|