1
|
Eulenburg V, Hülsmann S. Synergistic Control of Transmitter Turnover at Glycinergic Synapses by GlyT1, GlyT2, and ASC-1. Int J Mol Sci 2022; 23:ijms23052561. [PMID: 35269698 PMCID: PMC8909939 DOI: 10.3390/ijms23052561] [Citation(s) in RCA: 12] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/04/2022] [Revised: 02/21/2022] [Accepted: 02/23/2022] [Indexed: 01/25/2023] Open
Abstract
In addition to being involved in protein biosynthesis and metabolism, the amino acid glycine is the most important inhibitory neurotransmitter in caudal regions of the brain. These functions require a tight regulation of glycine concentration not only in the synaptic cleft, but also in various intracellular and extracellular compartments. This is achieved not only by confining the synthesis and degradation of glycine predominantly to the mitochondria, but also by the action of high-affinity large-capacity glycine transporters that mediate the transport of glycine across the membranes of presynaptic terminals or glial cells surrounding the synapses. Although most cells at glycine-dependent synapses express more than one transporter with high affinity for glycine, their synergistic functional interaction is only poorly understood. In this review, we summarize our current knowledge of the two high-affinity transporters for glycine, the sodium-dependent glycine transporters 1 (GlyT1; SLC6A9) and 2 (GlyT2; SLC6A5) and the alanine–serine–cysteine-1 transporter (Asc-1; SLC7A10).
Collapse
Affiliation(s)
- Volker Eulenburg
- Department for Anesthesiology and Intensive Care, Faculty of Medicine, University of Leipzig, Liebigstraße 20, D-04103 Leipzig, Germany
- Correspondence: (V.E.); (S.H.)
| | - Swen Hülsmann
- Department for Anesthesiology, University Medical Center, Georg-August University, Humboldtallee 23, D-37073 Göttingen, Germany
- Correspondence: (V.E.); (S.H.)
| |
Collapse
|
2
|
Inhibition of Glycine Re-Uptake: A Potential Approach for Treating Pain by Augmenting Glycine-Mediated Spinal Neurotransmission and Blunting Central Nociceptive Signaling. Biomolecules 2021; 11:biom11060864. [PMID: 34200954 PMCID: PMC8230656 DOI: 10.3390/biom11060864] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/24/2021] [Revised: 06/07/2021] [Accepted: 06/08/2021] [Indexed: 12/23/2022] Open
Abstract
Among the myriad of cellular and molecular processes identified as contributing to pathological pain, disinhibition of spinal cord nociceptive signaling to higher cortical centers plays a critical role. Importantly, evidence suggests that impaired glycinergic neurotransmission develops in the dorsal horn of the spinal cord in inflammatory and neuropathic pain models and is a key maladaptive mechanism causing mechanical hyperalgesia and allodynia. Thus, it has been hypothesized that pharmacological agents capable of augmenting glycinergic tone within the dorsal horn may be able to blunt or block aberrant nociceptor signaling to the brain and serve as a novel class of analgesics for various pathological pain states. Indeed, drugs that enhance dysfunctional glycinergic transmission, and in particular inhibitors of the glycine transporters (GlyT1 and GlyT2), are generating widespread interest as a potential class of novel analgesics. The GlyTs are Na+/Cl−-dependent transporters of the solute carrier 6 (SLC6) family and it has been proposed that the inhibition of them presents a possible mechanism by which to increase spinal extracellular glycine concentrations and enhance GlyR-mediated inhibitory neurotransmission in the dorsal horn. Various inhibitors of both GlyT1 and GlyT2 have demonstrated broad analgesic efficacy in several preclinical models of acute and chronic pain, providing promise for the approach to deliver a first-in-class non-opioid analgesic with a mechanism of action differentiated from current standard of care. This review will highlight the therapeutic potential of GlyT inhibitors as a novel class of analgesics, present recent advances reported for the field, and discuss the key challenges associated with the development of a GlyT inhibitor into a safe and effective agent to treat pain.
Collapse
|
3
|
Bhat S, El-Kasaby A, Freissmuth M, Sucic S. Functional and Biochemical Consequences of Disease Variants in Neurotransmitter Transporters: A Special Emphasis on Folding and Trafficking Deficits. Pharmacol Ther 2020; 222:107785. [PMID: 33310157 PMCID: PMC7612411 DOI: 10.1016/j.pharmthera.2020.107785] [Citation(s) in RCA: 27] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/31/2020] [Accepted: 12/02/2020] [Indexed: 01/30/2023]
Abstract
Neurotransmitters, such as γ-aminobutyric acid, glutamate, acetyl choline, glycine and the monoamines, facilitate the crosstalk within the central nervous system. The designated neurotransmitter transporters (NTTs) both release and take up neurotransmitters to and from the synaptic cleft. NTT dysfunction can lead to severe pathophysiological consequences, e.g. epilepsy, intellectual disability, or Parkinson’s disease. Genetic point mutations in NTTs have recently been associated with the onset of various neurological disorders. Some of these mutations trigger folding defects in the NTT proteins. Correct folding is a prerequisite for the export of NTTs from the endoplasmic reticulum (ER) and the subsequent trafficking to their pertinent site of action, typically at the plasma membrane. Recent studies have uncovered some of the key features in the molecular machinery responsible for transporter protein folding, e.g., the role of heat shock proteins in fine-tuning the ER quality control mechanisms in cells. The therapeutic significance of understanding these events is apparent from the rising number of reports, which directly link different pathological conditions to NTT misfolding. For instance, folding-deficient variants of the human transporters for dopamine or GABA lead to infantile parkinsonism/dystonia and epilepsy, respectively. From a therapeutic point of view, some folding-deficient NTTs are amenable to functional rescue by small molecules, known as chemical and pharmacological chaperones.
Collapse
Affiliation(s)
- Shreyas Bhat
- Institute of Pharmacology and the Gaston H. Glock Research Laboratories for Exploratory Drug Development, Center of Physiology and Pharmacology, Medical University of Vienna, A-1090 Vienna, Austria
| | - Ali El-Kasaby
- Institute of Pharmacology and the Gaston H. Glock Research Laboratories for Exploratory Drug Development, Center of Physiology and Pharmacology, Medical University of Vienna, A-1090 Vienna, Austria
| | - Michael Freissmuth
- Institute of Pharmacology and the Gaston H. Glock Research Laboratories for Exploratory Drug Development, Center of Physiology and Pharmacology, Medical University of Vienna, A-1090 Vienna, Austria
| | - Sonja Sucic
- Institute of Pharmacology and the Gaston H. Glock Research Laboratories for Exploratory Drug Development, Center of Physiology and Pharmacology, Medical University of Vienna, A-1090 Vienna, Austria.
| |
Collapse
|
4
|
Sheipouri D, Gallagher CI, Shimmon S, Rawling T, Vandenberg RJ. A System for Assessing Dual Action Modulators of Glycine Transporters and Glycine Receptors. Biomolecules 2020; 10:E1618. [PMID: 33266066 PMCID: PMC7760315 DOI: 10.3390/biom10121618] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/23/2020] [Revised: 11/23/2020] [Accepted: 11/25/2020] [Indexed: 12/13/2022] Open
Abstract
Reduced inhibitory glycinergic neurotransmission is implicated in a number of neurological conditions such as neuropathic pain, schizophrenia, epilepsy and hyperekplexia. Restoring glycinergic signalling may be an effective method of treating these pathologies. Glycine transporters (GlyTs) control synaptic and extra-synaptic glycine concentrations and slowing the reuptake of glycine using specific GlyT inhibitors will increase glycine extracellular concentrations and increase glycine receptor (GlyR) activation. Glycinergic neurotransmission can also be improved through positive allosteric modulation (PAM) of GlyRs. Despite efforts to manipulate this synapse, no therapeutics currently target it. We propose that dual action modulators of both GlyTs and GlyRs may show greater therapeutic potential than those targeting individual proteins. To show this, we have characterized a co-expression system in Xenopus laevis oocytes consisting of GlyT1 or GlyT2 co-expressed with GlyRα1. We use two electrode voltage clamp recording techniques to measure the impact of GlyTs on GlyRs and the effects of modulators of these proteins. We show that increases in GlyT density in close proximity to GlyRs diminish receptor currents. Reductions in GlyR mediated currents are not observed when non-transportable GlyR agonists are applied or when Na+ is not available. GlyTs reduce glycine concentrations across different concentration ranges, corresponding with their ion-coupling stoichiometry, and full receptor currents can be restored when GlyTs are blocked with selective inhibitors. We show that partial inhibition of GlyT2 and modest GlyRα1 potentiation using a dual action compound, is as useful in restoring GlyR currents as a full and potent single target GlyT2 inhibitor or single target GlyRα1 PAM. The co-expression system developed in this study will provide a robust means for assessing the likely impact of GlyR PAMs and GlyT inhibitors on glycine neurotransmission.
Collapse
Affiliation(s)
- Diba Sheipouri
- School of Medical Sciences, University of Sydney, Sydney, NSW 2006, Australia; (D.S.); (C.I.G.)
| | - Casey I. Gallagher
- School of Medical Sciences, University of Sydney, Sydney, NSW 2006, Australia; (D.S.); (C.I.G.)
| | - Susan Shimmon
- School of Mathematical and Physical Sciences, University of Technology Sydney, Sydney, NSW 2007, Australia; (S.S.); (T.R.)
| | - Tristan Rawling
- School of Mathematical and Physical Sciences, University of Technology Sydney, Sydney, NSW 2007, Australia; (S.S.); (T.R.)
| | - Robert J. Vandenberg
- School of Medical Sciences, University of Sydney, Sydney, NSW 2006, Australia; (D.S.); (C.I.G.)
| |
Collapse
|
5
|
Farr CV, El-Kasaby A, Freissmuth M, Sucic S. The Creatine Transporter Unfolded: A Knotty Premise in the Cerebral Creatine Deficiency Syndrome. Front Synaptic Neurosci 2020; 12:588954. [PMID: 33192443 PMCID: PMC7644880 DOI: 10.3389/fnsyn.2020.588954] [Citation(s) in RCA: 27] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/30/2020] [Accepted: 09/18/2020] [Indexed: 12/16/2022] Open
Abstract
Creatine provides cells with high-energy phosphates for the rapid reconstitution of hydrolyzed adenosine triphosphate. The eponymous creatine transporter (CRT1/SLC6A8) belongs to a family of solute carrier 6 (SLC6) proteins. The key role of CRT1 is to translocate creatine across tissue barriers and into target cells, such as neurons and myocytes. Individuals harboring mutations in the coding sequence of the human CRT1 gene develop creatine transporter deficiency (CTD), one of the pivotal underlying causes of cerebral creatine deficiency syndrome. CTD encompasses an array of clinical manifestations, including severe intellectual disability, epilepsy, autism, development delay, and motor dysfunction. CTD is characterized by the absence of cerebral creatine, which implies an indispensable role for CRT1 in supplying the brain cells with creatine. CTD-associated variants dramatically reduce or abolish creatine transport activity by CRT1. Many of these are point mutations that are known to trigger folding defects, leading to the retention of encoded CRT1 proteins in the endoplasmic reticulum and precluding their delivery to the cell surface. Misfolding of several related SLC6 transporters also gives rise to detrimental pathologic conditions in people; e.g., mutations in the dopamine transporter induce infantile parkinsonism/dystonia, while mutations in the GABA transporter 1 cause treatment-resistant epilepsy. In some cases, folding defects are amenable to rescue by small molecules, known as pharmacological and chemical chaperones, which restore the cell surface expression and transport activity of the previously non-functional proteins. Insights from the recent molecular, animal and human case studies of CTD add toward our understanding of this complex disorder and reveal the wide-ranging effects elicited upon CRT1 dysfunction. This grants novel therapeutic prospects for the treatment of patients afflicted with CTD, e.g., modifying the creatine molecule to facilitate CRT1-independent entry into brain cells, or correcting folding-deficient and loss-of-function CTD variants using pharmacochaperones and/or allosteric modulators. The latter justifies a search for additional compounds with a capacity to correct mutation-specific defects.
Collapse
Affiliation(s)
| | | | | | - Sonja Sucic
- Institute of Pharmacology, Center of Physiology and Pharmacology, Medical University of Vienna, Vienna, Austria
| |
Collapse
|
6
|
Hauf K, Barsch L, Bauer D, Buchert R, Armbruster A, Frauenfeld L, Grasshoff U, Eulenburg V. GlyT1 encephalopathy: Characterization of presumably disease causing GlyT1 mutations. Neurochem Int 2020; 139:104813. [PMID: 32712301 DOI: 10.1016/j.neuint.2020.104813] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/02/2020] [Revised: 06/26/2020] [Accepted: 07/09/2020] [Indexed: 12/22/2022]
Abstract
Glycine constitutes a major inhibitory neurotransmitter predominantly in caudal regions of the CNS. The extracellular glycine concentration is regulated synergistically by two high affinity, large capacity transporters GlyT1 and GlyT2. Both proteins are encoded by single genes SLC6A9 and SLC6A5, respectively. Mutations within the SLC6A5 gene encoding for GlyT2 have been demonstrated to be causative for hyperekplexia (OMIM #614618), a complex neuromuscular disease, in humans. In contrast, mutations within the SLC6A9 gene encoding for GlyT1 have been associated with GlyT1 encephalopathy (OMIM #601019), a disease causing severe postnatal respiratory deficiency, muscular hypotonia and arthrogryposis. The consequences of the respective GlyT1 mutations on the function of the transporter protein, however, have not yet been analysed. In this study we present the functional characterisation of three previously published GlyT1 mutations, two mutations predicted to cause truncation of GlyT1 (GlyT1Q573* and GlyT1K310F+fs*31) and one predicted to cause an amino acid exchange within transmembrane domain 7 of the transporter (GlyT1S407G), that are associated with GlyT1 encephalopathy. Additionally, the characterization of a novel mutation predicted to cause an amino acid exchange within transmembrane domain 1 (GlyT1V118M) identified in two fetuses showing increased nuchal translucency and arthrogryposis in routine ultrasound scans is demonstrated. We show that in recombinant systems the two presumably truncating mutations resulted in an intracellular retained GlyT1 protein lacking the intracellular C-terminal domain. In both cases this truncated protein did not show any residual transport activity. The point mutations, hGlyT1S407G and hGlyT1V118M, were processed correctly, but showed severely diminished activity, thus constituting a functional knock-out in-vivo. Taken together our data demonstrate that all analysed mutations of GlyT1 that have been identified in GlyT1 encephalopathy patients cause severe impairment of transporter function. This is consistent with the idea that loss of GlyT1 function is indeed causal for the disease phenotype.
Collapse
Affiliation(s)
- K Hauf
- Department of Anaesthesiology and Intensive Care, University of Leipzig, Leipzig, Germany
| | - L Barsch
- Department of Anaesthesiology and Intensive Care, University of Leipzig, Leipzig, Germany
| | - D Bauer
- Department of Biology, TU Darmstadt, Darmstadt, Germany
| | - R Buchert
- Institute of Medical Genetics and Applied Genomics, University of Tübingen, Tübingen, Germany
| | - A Armbruster
- Department of Biochemistry, University of Erlangen-Nuremberg, Erlangen, Germany
| | - L Frauenfeld
- Institute of Pathology and Neuropathology, University of Tübingen, Tübingen, Germany
| | - U Grasshoff
- Institute of Medical Genetics and Applied Genomics, University of Tübingen, Tübingen, Germany
| | - V Eulenburg
- Department of Anaesthesiology and Intensive Care, University of Leipzig, Leipzig, Germany; Department of Biochemistry, University of Erlangen-Nuremberg, Erlangen, Germany.
| |
Collapse
|
7
|
Mostyn SN, Sarker S, Muthuraman P, Raja A, Shimmon S, Rawling T, Cioffi CL, Vandenberg RJ. Photoswitchable ORG25543 Congener Enables Optical Control of Glycine Transporter 2. ACS Chem Neurosci 2020; 11:1250-1258. [PMID: 32191428 PMCID: PMC7206614 DOI: 10.1021/acschemneuro.9b00655] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/28/2022] Open
Abstract
![]()
Glycine
neurotransmission in the dorsal horn of the spinal cord
plays a key role in regulating nociceptive signaling, but in chronic
pain states reduced glycine neurotransmission is associated with the
development of allodynia and hypersensitivity to painful stimuli.
This suggests that restoration of glycine neurotransmission may be
therapeutic for the treatment of chronic pain. Glycine transporter
2 inhibitors have been demonstrated to enhance glycine neurotransmission
and provide relief from allodynia in rodent models of chronic pain.
In recent years, photoswitchable compounds have been developed to
provide the possibility of controlling the activity of target proteins
using light. In this study we have developed a photoswitchable noncompetitive
inhibitor of glycine transporter 2 that has different affinities for
the transporter at 365 nm compared to 470 nm light.
Collapse
Affiliation(s)
- Shannon N. Mostyn
- Discipline of Pharmacology, School of Medical Sciences, Faculty of Medicine and Health, University of Sydney, Camperdown, New South Wales 2006, Australia
| | - Subhodeep Sarker
- Discipline of Pharmacology, School of Medical Sciences, Faculty of Medicine and Health, University of Sydney, Camperdown, New South Wales 2006, Australia
| | - Parthasarathy Muthuraman
- Basic and Clinical Sciences and Pharmaceutical Sciences, Albany College of Pharmacy and Health Sciences, Albany, New York 12208, United States
| | - Arun Raja
- Basic and Clinical Sciences and Pharmaceutical Sciences, Albany College of Pharmacy and Health Sciences, Albany, New York 12208, United States
| | - Susan Shimmon
- School of Mathematical and Physical Sciences, University of Technology Sydney, Ultimo, New South Wales 2007, Australia
| | - Tristan Rawling
- School of Mathematical and Physical Sciences, University of Technology Sydney, Ultimo, New South Wales 2007, Australia
| | - Christopher L. Cioffi
- Basic and Clinical Sciences and Pharmaceutical Sciences, Albany College of Pharmacy and Health Sciences, Albany, New York 12208, United States
| | - Robert J. Vandenberg
- Discipline of Pharmacology, School of Medical Sciences, Faculty of Medicine and Health, University of Sydney, Camperdown, New South Wales 2006, Australia
| |
Collapse
|
8
|
Kitzenmaier A, Schaefer N, Kasaragod VB, Polster T, Hantschmann R, Schindelin H, Villmann C. The P429L loss of function mutation of the human glycine transporter 2 associated with hyperekplexia. Eur J Neurosci 2019; 50:3906-3920. [DOI: 10.1111/ejn.14533] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/12/2019] [Revised: 07/23/2019] [Accepted: 07/25/2019] [Indexed: 12/20/2022]
Affiliation(s)
- Alexandra Kitzenmaier
- Institute for Clinical Neurobiology Julius‐Maximilians‐University of Würzburg Würzburg Germany
| | - Natascha Schaefer
- Institute for Clinical Neurobiology Julius‐Maximilians‐University of Würzburg Würzburg Germany
| | - Vikram Babu Kasaragod
- Rudolf Virchow Centre for Experimental Biomedicine Julius‐Maximilians‐University of Würzburg Würzburg Germany
| | - Tilman Polster
- Pediatric Epileptology Mara Hospital Bethel Epilepsy Centre Bielefeld Germany
| | - Ralph Hantschmann
- Center for Developmental Pediatrics and Pediatric Neurology Hagen Germany
| | - Hermann Schindelin
- Rudolf Virchow Centre for Experimental Biomedicine Julius‐Maximilians‐University of Würzburg Würzburg Germany
| | - Carmen Villmann
- Institute for Clinical Neurobiology Julius‐Maximilians‐University of Würzburg Würzburg Germany
| |
Collapse
|
9
|
Vuilleumier PH, Fritsche R, Schliessbach J, Schmitt B, Arendt-Nielsen L, Zeilhofer HU, Curatolo M. Mutations affecting glycinergic neurotransmission in hyperekplexia increase pain sensitivity. Brain 2019; 141:63-71. [PMID: 29149236 DOI: 10.1093/brain/awx289] [Citation(s) in RCA: 20] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/10/2017] [Accepted: 09/19/2017] [Indexed: 11/14/2022] Open
Abstract
See Dickenson (doi:10.1093/brain/awx334) for a scientific commentary on this article.Inhibitory interneurons in the spinal cord use glycine and GABA for fast inhibitory neurotransmission. While there is abundant research on these inhibitory pain pathways in animal models, their relevance in humans remains unclear, largely due to the limited possibility to manipulate selectively these pathways in humans. Hyperekplexia is a rare human disease that is caused by loss-of-function mutations in genes encoding for glycine receptors and glycine transporters. In the present study, we tested whether hyperekplexia patients display altered pain perception or central pain modulation compared with healthy subjects. Seven patients with genetically and clinically confirmed hyperekplexia were compared to 14 healthy age- and sex-matched controls. The following quantitative sensory tests were performed: pressure pain detection threshold (primary outcome), ice water tolerance, single and repeated electrical pain detection thresholds, nociceptive withdrawal reflex threshold, and conditioned pain modulation. Statistical analysis was performed using linear mixed models. Hyperekplexia patients displayed lower pain thresholds than healthy controls for all of the quantitative sensory tests [mean (standard deviation)]: pressure pain detection threshold [273 (170) versus 475 (115) kPa, P = 0.003], ice water tolerance [49.2 (36.5) versus 85.7 (35.0) s, P = 0.015], electrical single pain detection threshold [5.42 (2.64) versus 7.47 (2.62) mA, P = 0.012], electrical repeated pain detection threshold [3.76 (1.41) versus 5.8 (1.73) mA, P = 0.003], and nociceptive withdrawal reflex [7.42 (3.63) versus 14.1 (6.9) mA, P = 0.015]. Conditioned pain modulation was significantly reduced in hyperekplexia [increase to baseline: 53.2 (63.7) versus 105 (57) kPa, P = 0.030]. Our data demonstrate increased pain sensitivity and impaired central pain modulation in hyperekplexia patients, supporting the importance of glycinergic neurotransmission for central pain modulation in humans.
Collapse
Affiliation(s)
- Pascal Henri Vuilleumier
- Department of Anaesthesiology and Pain Medicine, Bern University Hospital, University of Bern, Switzerland
| | - Raphael Fritsche
- Department of Ophthalmology, Canton Hospital of Lucerne, Switzerland
| | - Jürg Schliessbach
- Department of Anaesthesiology and Pain Medicine, Bern University Hospital, University of Bern, Switzerland
| | - Bernhard Schmitt
- Department of Child Neurology, Children's Hospital, University of Zurich, Switzerland
| | - Lars Arendt-Nielsen
- Center for Sensory-Motor Interaction, School of Medicine, University of Aalborg, Denmark
| | - Hanns Ulrich Zeilhofer
- Institute of Pharmacology and Toxicology, University of Zurich, and Institute of Pharmaceutical Sciences, Swiss Federal Institute of Technology (ETH) Zurich, Switzerland
| | - Michele Curatolo
- Center for Sensory-Motor Interaction, School of Medicine, University of Aalborg, Denmark.,Department of Anaesthesiology and Pain Medicine, University of Washington, Seattle, USA
| |
Collapse
|
10
|
Clinical features and genetic analysis of two siblings with startle disease in an Italian family: a case report. BMC MEDICAL GENETICS 2019; 20:40. [PMID: 30866851 PMCID: PMC6417078 DOI: 10.1186/s12881-019-0779-x] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 01/02/2019] [Accepted: 03/06/2019] [Indexed: 12/03/2022]
Abstract
Background Hyperekplexia also known as Startle disease is a rare neuromotor hereditary disorder characterized by exaggerated startle responses to unexpected auditory, tactile, and visual stimuli and generalized muscle stiffness, which both gradually subside during the first months of life. Although the diagnosis of Hyperekplexia is based on clinical findings, pathogenic variants in five genes have been reported to cause Hyperekplexia, of which GLRA1 accounts for about 80% of cases. Dominant and recessive mutations have been identified in GLRA1 gene as pathogenic variants in many individuals with the familial form of Hyperekplexia and occasionally in simplex cases. Case presentation In the present study, we describe clinical and genetic features of two Italian siblings, one with the major and one with the minor form of the disease. DNA samples from the probands and their parents were performed by NGS approach and validated by Sanger sequencing. The analysis of the GLRA1 gene revealed, in both probands, compound heterozygous mutations: c.895C > T or p.R299X inherited from the mother and c.587C > A or p.D98E inherited from the father. Conclusions Until now, these two identified mutations in GLRA1 have not been reported before as compound mutations. What clearly emerges within our study is the clinical heterogeneity in the same family. In fact, even though in the same pedigree, the affected mother showed only mild startle responses to unexpected noise stimuli, which might be explained by variable expressivity, while the father, showed no clear signs of symptomatology, which might be explained by non-penetrance. Finally, the two brothers have different form of the disease, even if the compound heterozygous mutations in GLRA1 are the same, showing that the same mutation in GLRA1 could have different phenotypic expressions and suggesting an underling mechanism of variable expressivity.
Collapse
|
11
|
Murphy SC, Recio A, de la Fuente C, Guo LT, Shelton GD, Clark LA. A glycine transporter SLC6A5 frameshift mutation causes startle disease in Spanish greyhounds. Hum Genet 2019; 138:509-513. [PMID: 30847549 DOI: 10.1007/s00439-019-01986-x] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/17/2018] [Accepted: 02/19/2019] [Indexed: 11/29/2022]
Abstract
Startle disease, or hyperekplexia, is a glycinergic disorder characterized by hypertonia and apnea that is triggered by noise and/or touch. Mutations in five genes have been associated with startle disease in humans, dogs, cattle, and mice. We identified a novel recessive startle disease in a family of Spanish greyhounds. Whole genome resequencing of an affected dog revealed a homozygous two base pair deletion in the ninth exon of SLC6A5, encoding the presynaptic glycine transporter. The deletion is predicted to cause a frameshift, p.S460FfsX47, leading to a premature stop codon that truncates over a third of the protein. Family members were genotyped for the deletion, and findings were consistent with an autosomal recessive inheritance pattern. The pathogenic variant was absent from 34 unrelated greyhounds, 659 domestic dogs of pure and mixed breeds, and 54 wild canids, suggesting it occurred recently and may be private to the family. The findings of this study can be used to inform future breeding decisions and prevent dissemination of the deleterious allele in greyhounds.
Collapse
Affiliation(s)
- Sarah C Murphy
- Department of Genetics and Biochemistry, Clemson University, Clemson, SC, 29634, USA
| | - Alfredo Recio
- Fundació Hospital Clínic Veterinari, Universitat Autònoma de Barcelona, Bellaterra, Spain
- Clinica Veterinaria Levante, San Javier, Murcia, Spain
| | - Cristian de la Fuente
- Fundació Hospital Clínic Veterinari, Universitat Autònoma de Barcelona, Bellaterra, Spain
| | - Ling T Guo
- Department of Pathology, School of Medicine, University of California San Diego, La Jolla, CA, 92093, USA
| | - G Diane Shelton
- Department of Pathology, School of Medicine, University of California San Diego, La Jolla, CA, 92093, USA.
| | - Leigh Anne Clark
- Department of Genetics and Biochemistry, Clemson University, Clemson, SC, 29634, USA.
| |
Collapse
|
12
|
López-Corcuera B, Arribas-González E, Aragón C. Hyperekplexia-associated mutations in the neuronal glycine transporter 2. Neurochem Int 2018; 123:95-100. [PMID: 29859229 DOI: 10.1016/j.neuint.2018.05.014] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/03/2018] [Revised: 05/26/2018] [Accepted: 05/29/2018] [Indexed: 12/20/2022]
Abstract
Hyperekplexia or startle disease is a dysfunction of inhibitory glycinergic neurotransmission characterized by an exaggerated startle in response to trivial tactile or acoustic stimuli. Although rare, this disorder can have serious consequences, including sudden infant death. One of the most frequent causes of hyperekplexia are mutations in the SLC6A5 gene, encoding the neuronal glycine transporter 2 (GlyT2), a key component of inhibitory glycinergic presynapses involved in synaptic glycine recycling though sodium and chloride-dependent co-transport. Most GlyT2 mutations detected so far are recessive, but two dominant missense mutations have been described. The detailed analysis of these mutations has revealed structural cues on the quaternary structure of GlyT2, and opens the possibility that novel selective pharmacochaperones have potential therapeutic effects in hyperekplexia.
Collapse
Affiliation(s)
- Beatriz López-Corcuera
- Centro de Biología Molecular ''Severo Ochoa'', Universidad Autónoma de Madrid-Consejo Superior de Investigaciones Científicas, Madrid, Spain; IdiPAZ-Hospital Universitario La Paz, Universidad Autónoma de Madrid, Madrid, Spain.
| | - Esther Arribas-González
- Centro de Biología Molecular ''Severo Ochoa'', Universidad Autónoma de Madrid-Consejo Superior de Investigaciones Científicas, Madrid, Spain; IdiPAZ-Hospital Universitario La Paz, Universidad Autónoma de Madrid, Madrid, Spain
| | - Carmen Aragón
- Centro de Biología Molecular ''Severo Ochoa'', Universidad Autónoma de Madrid-Consejo Superior de Investigaciones Científicas, Madrid, Spain; IdiPAZ-Hospital Universitario La Paz, Universidad Autónoma de Madrid, Madrid, Spain
| |
Collapse
|
13
|
Eulenburg V, Knop G, Sedmak T, Schuster S, Hauf K, Schneider J, Feigenspan A, Joachimsthaler A, Brandstätter JH. GlyT1 determines the glycinergic phenotype of amacrine cells in the mouse retina. Brain Struct Funct 2018; 223:3251-3266. [PMID: 29808289 DOI: 10.1007/s00429-018-1684-3] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/22/2016] [Accepted: 03/04/2018] [Indexed: 11/26/2022]
Abstract
The amino acid glycine acts as a neurotransmitter at both inhibitory glycinergic and excitatory glutamatergic synapses predominantly in caudal regions of the central nervous system but also in frontal brain regions and the retina. After its presynaptic release and binding to postsynaptic receptors at caudal glycinergic synapses, two high-affinity glycine transporters GlyT1 and GlyT2 remove glycine from the extracellular space. Glycinergic neurons express GlyT2, which is essential for the presynaptic replenishment of the transmitter, while glial-expressed GlyT1 was shown to control the extracellular glycine concentration. Here we show that GlyT1 expressed by glycinergic amacrine cells of the retina does not only contribute to the control of the extracellular glycine concentration in the retina but is also essential for the maintenance of the glycinergic transmitter phenotype of this cell population. Specifically, loss of GlyT1 from the glycinergic AII amacrine cells impairs AII-mediated glycinergic neurotransmission and alters regulation of the extracellular glycine concentration, without changes in the overall distribution and/or size of glycinergic synapses. Taken together, our results suggest that GlyT1 expressed by amacrine cells in the retina combines functions covered by neuronal GlyT2 and glial GlyT1 at caudal glycinergic synapses.
Collapse
Affiliation(s)
- Volker Eulenburg
- Department of Biochemistry and Molecular Medicine, Institute of Biochemistry, Friedrich-Alexander University of Erlangen-Nürnberg, Erlangen, Germany.
- Department of Anesthesiology and Intensive Care Medicine, University of Leipzig, Liebigstrasse 20, 04103, Leipzig, Germany.
| | - Gabriel Knop
- Department of Biology, Animal Physiology, Friedrich-Alexander-University of Erlangen-Nürnberg, Erlangen, Germany
| | - Tina Sedmak
- Department of Biology, Animal Physiology, Friedrich-Alexander-University of Erlangen-Nürnberg, Erlangen, Germany
| | - Stefanie Schuster
- Department of Biochemistry and Molecular Medicine, Institute of Biochemistry, Friedrich-Alexander University of Erlangen-Nürnberg, Erlangen, Germany
| | - Katharina Hauf
- Department of Biochemistry and Molecular Medicine, Institute of Biochemistry, Friedrich-Alexander University of Erlangen-Nürnberg, Erlangen, Germany
- Department of Anesthesiology and Intensive Care Medicine, University of Leipzig, Liebigstrasse 20, 04103, Leipzig, Germany
| | - Julia Schneider
- Department of Biochemistry and Molecular Medicine, Institute of Biochemistry, Friedrich-Alexander University of Erlangen-Nürnberg, Erlangen, Germany
| | - Andreas Feigenspan
- Department of Biology, Animal Physiology, Friedrich-Alexander-University of Erlangen-Nürnberg, Erlangen, Germany
| | - Anneka Joachimsthaler
- Department of Biology, Animal Physiology, Friedrich-Alexander-University of Erlangen-Nürnberg, Erlangen, Germany
- Department of Ophthalmology, University Hospital Erlangen, 91054, Erlangen, Germany
| | - Johann Helmut Brandstätter
- Department of Biology, Animal Physiology, Friedrich-Alexander-University of Erlangen-Nürnberg, Erlangen, Germany
| |
Collapse
|
14
|
Yang Z, Sun G, Yao F, Tao D, Zhu B. A novel compound mutation in GLRA1 cause hyperekplexia in a Chinese boy- a case report and review of the literature. BMC MEDICAL GENETICS 2017; 18:110. [PMID: 28985719 PMCID: PMC5631533 DOI: 10.1186/s12881-017-0476-6] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 06/20/2017] [Accepted: 10/03/2017] [Indexed: 11/10/2022]
Abstract
Background The pathogenesis of hereditary hyperekplexia is thought to involve abnormalities in the glycinergic neurotransmission system, the most of mutations reported in GLRA1. This gene encodes the glycine receptor α1 subunit, which has an extracellular domain (ECD) and a transmembrane domain (TMD) with 4 α-helices (TM1–TM4). Case presentation We investigated the genetic cause of hyperekplexia in a Chinese family with one affected member. Whole-exome sequencing of the 5 candidate genes was performed on the proband patient, and direct sequencing was performed to validate and confirm the detected mutation in other family members. We also review and analyse all reported GLRA1 mutations. The proband had a compound heterozygous GLRA1 mutation that comprised 2 novel GLRA1 missense mutations, C.569C > T (p.T190 M) from the mother and C.1270G > A (p.D424N) from the father. SIFT, Polyphen-2 and MutationTaster analysis identified the mutations as disease-causing, but the parents had no signs of hyperekplexia. The p.T190 M mutation is located in the ECD, while p.D424N is located in TM4. Conclusions Our findings contribute to a growing list GLRA1 mutations associated with hyperekplexia and provide new insights into correlations between phenotype and GLRA1 mutations. Some recessive mutations can induce hyperekplexia in combination with other recessive GLRA1 mutations. Mutations in the ECD, TM1, TM1-TM2 loop, TM3, TM3-TM4 loop and TM4 are more often recessive and part of a compound mutation, while those in TM2 and the TM2-TM3 loop are more likely to be dominant hereditary mutations. Electronic supplementary material The online version of this article (10.1186/s12881-017-0476-6) contains supplementary material, which is available to authorized users.
Collapse
Affiliation(s)
- Zhiliang Yang
- Department of Pediatrics, The First Hospital of China Medical University, No. 155 Nanjing North Street, Heping District, Shenyang, 110001, Liaoning Province, People's Republic of China.
| | - Guilian Sun
- Department of Pediatrics, The First Hospital of China Medical University, No. 155 Nanjing North Street, Heping District, Shenyang, 110001, Liaoning Province, People's Republic of China
| | - Fang Yao
- Department of Pediatrics, The First Hospital of China Medical University, No. 155 Nanjing North Street, Heping District, Shenyang, 110001, Liaoning Province, People's Republic of China
| | - Dongying Tao
- Department of Pediatrics, The First Hospital of China Medical University, No. 155 Nanjing North Street, Heping District, Shenyang, 110001, Liaoning Province, People's Republic of China
| | - Binlu Zhu
- Department of Pediatrics, The First Hospital of China Medical University, No. 155 Nanjing North Street, Heping District, Shenyang, 110001, Liaoning Province, People's Republic of China
| |
Collapse
|
15
|
Cioffi CL. Modulation of Glycine-Mediated Spinal Neurotransmission for the Treatment of Chronic Pain. J Med Chem 2017; 61:2652-2679. [PMID: 28876062 DOI: 10.1021/acs.jmedchem.7b00956] [Citation(s) in RCA: 25] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Abstract
Chronic pain constitutes a significant and expanding worldwide health crisis. Currently available analgesics poorly serve individuals suffering from chronic pain, and new therapeutic agents that are more effective, safer, and devoid of abuse liabilities are desperately needed. Among the myriad of cellular and molecular processes contributing to chronic pain, spinal disinhibition of pain signaling to higher cortical centers plays a critical role. Accumulating evidence shows that glycinergic inhibitory neurotransmission in the spinal cord dorsal horn gates nociceptive signaling, is essential in maintaining physiological pain sensitivity, and is diminished in pathological pain states. Thus, it is hypothesized that agents capable of enhancing glycinergic tone within the dorsal horn could obtund nociceptor signaling to the brain and serve as analgesics for persistent pain. This Perspective highlights the potential that pharmacotherapies capable of increasing inhibitory spinal glycinergic neurotransmission hold in providing new and transformative analgesic therapies for the treatment of chronic pain.
Collapse
Affiliation(s)
- Christopher L Cioffi
- Departments of Basic and Clinical Sciences and Pharmaceutical Sciences , Albany College of Pharmacy and Health Sciences , 106 New Scotland Avenue , Albany , New York 12208 United States
| |
Collapse
|
16
|
Lowrie M, Garosi L. Classification of Involuntary Movements in Dogs: Myoclonus and Myotonia. J Vet Intern Med 2017; 31:979-987. [PMID: 28557061 PMCID: PMC5508344 DOI: 10.1111/jvim.14771] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/24/2017] [Revised: 04/07/2017] [Accepted: 04/27/2017] [Indexed: 12/25/2022] Open
Abstract
Myoclonus is a sudden brief, involuntary muscle jerk. Of all the movement disorders, myoclonus is the most difficult to encapsulate into any simple framework. On the one hand, a classification system is required that is clinically useful to aid in guiding diagnosis and treatment. On the other hand, there is need for a system that organizes current knowledge regarding biological mechanisms to guide scientific research. These 2 needs are distinct, making it challenging to develop a robust classification system suitable for all purposes. We attempt to classify myoclonus as “epileptic” and “nonepileptic” based on its association with epileptic seizures. Myotonia in people may be divided into 2 clinically and molecularly defined forms: (1) nondystrophic myotonias and (2) myotonic dystrophies. The former are a group of skeletal muscle channelopathies characterized by delayed skeletal muscle relaxation. Many distinct clinical phenotypes are recognized in people, the majority relating to mutations in skeletal muscle voltage‐gated chloride (CLCN1) and sodium channel (SCN4A) genes. In dogs, myotonia is associated with mutations in CLCN1. The myotonic dystrophies are considered a multisystem clinical syndrome in people encompassing 2 clinically and molecularly defined forms designated myotonic dystrophy types 1 and 2. No mutation has been linked to veterinary muscular dystrophies. We detail veterinary examples of myotonia and attempt classification according to guidelines used in humans. This more precise categorization of myoclonus and myotonia aims to promote the search for molecular markers contributing to the phenotypic spectrum of disease. Our work aimed to assist recognition for these 2 enigmatic conditions.
Collapse
Affiliation(s)
- M Lowrie
- Dovecote Veterinary Hospital, Derby, UK
| | - L Garosi
- Davies Veterinary Specialists, Hitchin, UK
| |
Collapse
|
17
|
Freissmuth M, Stockner T, Sucic S. SLC6 Transporter Folding Diseases and Pharmacochaperoning. Handb Exp Pharmacol 2017; 245:249-270. [PMID: 29086036 DOI: 10.1007/164_2017_71] [Citation(s) in RCA: 21] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/24/2022]
Abstract
The human genome encodes 19 genes of the solute carrier 6 (SLC6) family; non-synonymous changes in the coding sequence give rise to mutated transporters, which are misfolded and thus cause diseases in the affected individuals. Prominent examples include mutations in the transporters for dopamine (DAT, SLC6A3), for creatine (CT1, SLC6A8), and for glycine (GlyT2, SLC6A5), which result in infantile dystonia, mental retardation, and hyperekplexia, respectively. Thus, there is an obvious unmet medical need to identify compounds, which can remedy the folding deficit. The pharmacological correction of folding defects was originally explored in mutants of the serotonin transporter (SERT, SLC6A4), which were created to study the COPII-dependent export from the endoplasmic reticulum. This led to the serendipitous discovery of the pharmacochaperoning action of ibogaine. Ibogaine and its metabolite noribogaine also rescue several disease-relevant mutants of DAT. Because the pharmacology of DAT and SERT is exceptionally rich, it is not surprising that additional compounds have been identified, which rescue folding-deficient mutants. These compounds are not only of interest for restoring DAT function in the affected children. They are also likely to serve as useful tools to interrogate the folding trajectory of the transporter. This is likely to initiate a virtuous cycle: if the principles underlying folding of SLC6 transporters are understood, the design of pharmacochaperones ought to be facilitated.
Collapse
Affiliation(s)
- Michael Freissmuth
- Institute of Pharmacology and the Gaston H. Glock Research Laboratories for Exploratory Drug Development, Center of Physiology and Pharmacology, Medical University of Vienna, Vienna, Austria.
| | - Thomas Stockner
- Institute of Pharmacology and the Gaston H. Glock Research Laboratories for Exploratory Drug Development, Center of Physiology and Pharmacology, Medical University of Vienna, Vienna, Austria
| | - Sonja Sucic
- Institute of Pharmacology and the Gaston H. Glock Research Laboratories for Exploratory Drug Development, Center of Physiology and Pharmacology, Medical University of Vienna, Vienna, Austria
| |
Collapse
|
18
|
Glycinergic transmission: glycine transporter GlyT2 in neuronal pathologies. Neuronal Signal 2016; 1:NS20160009. [PMID: 32714574 PMCID: PMC7377260 DOI: 10.1042/ns20160009] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/14/2016] [Revised: 11/04/2016] [Accepted: 11/09/2016] [Indexed: 11/17/2022] Open
Abstract
Glycinergic neurons are major contributors to the regulation of neuronal excitability, mainly in caudal areas of the nervous system. These neurons control fluxes of sensory information between the periphery and the CNS and diverse motor activities like locomotion, respiration or vocalization. The phenotype of a glycinergic neuron is determined by the expression of at least two proteins: GlyT2, a plasma membrane transporter of glycine, and VIAAT, a vesicular transporter shared by glycine and GABA. In this article, we review recent advances in understanding the role of GlyT2 in the pathophysiology of inhibitory glycinergic neurotransmission. GlyT2 mutations are associated to decreased glycinergic function that results in a rare movement disease termed hyperekplexia (HPX) or startle disease. In addition, glycinergic neurons control pain transmission in the dorsal spinal cord and their function is reduced in chronic pain states. A moderate inhibition of GlyT2 may potentiate glycinergic inhibition and constitutes an attractive target for pharmacological intervention against these devastating conditions.
Collapse
|
19
|
Sucic S, Kasture A, Mazhar Asjad HM, Kern C, El-Kasaby A, Freissmuth M. When transporters fail to be transported: how to rescue folding-deficient SLC6 transporters. ACTA ACUST UNITED AC 2016; 1:34-40. [PMID: 28405636 PMCID: PMC5386142 DOI: 10.29245/2572.942x/2016/9.1098] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/25/2022]
Abstract
The human dopamine transporter (hDAT) belongs to the solute carrier 6 (SLC6) gene family. Point mutations in hDAT (SLC6A3) have been linked to a syndrome of dopamine transporter deficiency or infantile dystonia/parkinsonism. The mutations impair DAT folding, causing retention of variant DATs in the endoplasmic reticulum and subsequently impair transport activity. The folding trajectory of DAT itself is not understood, though many insights have been gained from studies of folding-deficient mutants of the closely related serotonin transporter (SERT); i.e. their functional rescue by pharmacochaperoning with (nor)ibogaine or heat-shock protein inhibitors. We recently provided a proof-of-principle that folding-deficits in DAT are amenable to rescue in vitro and in vivo. As a model we used the Drosophila melanogaster DAT mutant dDAT-G108Q, which phenocopies the fumin/sleepless DAT-knockout. Treatment with noribogaine and/or HSP70 inhibitor pifithrin-μ restored folding of, and dopamine transport by, dDAT-G108Q, its axonal delivery and normal sleep time in mutant flies. The possibility of functional rescue of misfolded DATs in living flies by pharmacochaperoning grants new therapeutic prospects in the remedy of folding diseases, not only in hDAT, but also in other SLC6 transporters, in particular mutants of the creatine transporter-1, which give rise to X-linked mental retardation.
Collapse
Affiliation(s)
- Sonja Sucic
- Institute of Pharmacology, Center of Physiology and Pharmacology, Medical University of Vienna, A-1090 Vienna, Austria
| | - Ameya Kasture
- Institute of Pharmacology, Center of Physiology and Pharmacology, Medical University of Vienna, A-1090 Vienna, Austria
| | - H M Mazhar Asjad
- Institute of Pharmacology, Center of Physiology and Pharmacology, Medical University of Vienna, A-1090 Vienna, Austria
| | - Carina Kern
- Institute of Pharmacology, Center of Physiology and Pharmacology, Medical University of Vienna, A-1090 Vienna, Austria
| | - Ali El-Kasaby
- Institute of Pharmacology, Center of Physiology and Pharmacology, Medical University of Vienna, A-1090 Vienna, Austria
| | - Michael Freissmuth
- Institute of Pharmacology, Center of Physiology and Pharmacology, Medical University of Vienna, A-1090 Vienna, Austria
| |
Collapse
|
20
|
Kurolap A, Armbruster A, Hershkovitz T, Hauf K, Mory A, Paperna T, Hannappel E, Tal G, Nijem Y, Sella E, Mahajnah M, Ilivitzki A, Hershkovitz D, Ekhilevitch N, Mandel H, Eulenburg V, Baris HN. Loss of Glycine Transporter 1 Causes a Subtype of Glycine Encephalopathy with Arthrogryposis and Mildly Elevated Cerebrospinal Fluid Glycine. Am J Hum Genet 2016; 99:1172-1180. [PMID: 27773429 DOI: 10.1016/j.ajhg.2016.09.004] [Citation(s) in RCA: 27] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/15/2016] [Accepted: 09/07/2016] [Indexed: 01/02/2023] Open
Abstract
Glycine is a major neurotransmitter that activates inhibitory glycine receptors and is a co-agonist for excitatory glutamatergic N-methyl-D-aspartate (NMDA) receptors. Two transporters, GLYT1 and GLYT2, regulate extracellular glycine concentrations within the CNS. Dysregulation of the extracellular glycine has been associated with hyperekplexia and nonketotic hyperglycinemia. Here, we report four individuals from two families who presented at birth with facial dysmorphism, encephalopathy, arthrogryposis, hypotonia progressing to hypertonicity with startle-like clonus, and respiratory failure. Only one individual survived the respiratory failure and was weaned off ventilation but has significant global developmental delay. Mildly elevated cerebrospinal fluid (CSF) glycine and normal serum glycine were observed in two individuals. In both families, we identified truncating mutations in SLC6A9, encoding GLYT1. We demonstrate that pharmacologic or genetic abolishment of GlyT1 activity in mice leads to mildly elevated glycine in the CSF but not in blood. Additionally, previously reported slc6a9-null mice and zebrafish mutants also display phenotypes consistent with the affected individuals we examined. Our data suggest that truncating SLC6A9 mutations lead to a distinct human neurological syndrome hallmarked by mildly elevated CSF glycine and normal serum glycine.
Collapse
|
21
|
Advances in understanding the functions of native GlyT1 and GlyT2 neuronal glycine transporters. Neurochem Int 2016; 99:169-177. [DOI: 10.1016/j.neuint.2016.07.001] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/06/2016] [Revised: 07/05/2016] [Accepted: 07/05/2016] [Indexed: 11/20/2022]
|
22
|
The lidocaine metabolite N-ethylglycine has antinociceptive effects in experimental inflammatory and neuropathic pain. Pain 2016; 156:1647-1659. [PMID: 25932687 PMCID: PMC4617288 DOI: 10.1097/j.pain.0000000000000206] [Citation(s) in RCA: 33] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022]
Abstract
Supplemental Digital Content is Available in the Text. The lidocaine metabolite N-ethylglycine specifically reduces GlyT1-dependent glycine uptake and has antinociceptive effects in experimental inflammatory and neuropathic pain, while no adverse effects are observed. Glycine transporter 1 (GlyT1) plays a crucial role in regulating extracellular glycine concentrations and might thereby constitute a new drug target for the modulation of glycinergic inhibition in pain signaling. Consistent with this view, inhibition of GlyT1 has been found to induce antinociceptive effects in various animal pain models. We have shown previously that the lidocaine metabolite N-ethylglycine (EG) reduces GlyT1-dependent glycine uptake by functioning as an artificial substrate for this transporter. Here, we show that EG is specific for GlyT1 and that in rodent models of inflammatory and neuropathic pain, systemic treatment with EG results in an efficient amelioration of hyperalgesia and allodynia without affecting acute pain. There was no effect on motor coordination or the development of inflammatory edema. No adverse neurological effects were observed after repeated high-dose application of EG. EG concentrations both in blood and spinal fluid correlated with an increase of glycine concentration in spinal fluid. The time courses of the EG and glycine concentrations corresponded well with the antinociceptive effect. Additionally, we found that EG reduced the increase in neuronal firing of wide-dynamic-range neurons caused by inflammatory pain induction. These findings suggest that systemically applied lidocaine exerts antihyperalgesic effects through its metabolite EG in vivo, by enhancing spinal inhibition of pain processing through GlyT1 modulation and subsequent increase of glycine concentrations at glycinergic inhibitory synapses. EG and other substrates of GlyT1, therefore, may be a useful therapeutic agent in chronic pain states involving spinal disinhibition.
Collapse
|
23
|
Spillane J, Kullmann DM, Hanna MG. Genetic neurological channelopathies: molecular genetics and clinical phenotypes. J Neurol Neurosurg Psychiatry 2016; 87:37-48. [PMID: 26558925 PMCID: PMC4717447 DOI: 10.1136/jnnp-2015-311233] [Citation(s) in RCA: 43] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/11/2015] [Accepted: 09/13/2015] [Indexed: 01/08/2023]
Abstract
Evidence accumulated over recent years has shown that genetic neurological channelopathies can cause many different neurological diseases. Presentations relating to the brain, spinal cord, peripheral nerve or muscle mean that channelopathies can impact on almost any area of neurological practice. Typically, neurological channelopathies are inherited in an autosomal dominant fashion and cause paroxysmal disturbances of neurological function, although the impairment of function can become fixed with time. These disorders are individually rare, but an accurate diagnosis is important as it has genetic counselling and often treatment implications. Furthermore, the study of less common ion channel mutation-related diseases has increased our understanding of pathomechanisms that is relevant to common neurological diseases such as migraine and epilepsy. Here, we review the molecular genetic and clinical features of inherited neurological channelopathies.
Collapse
Affiliation(s)
- J Spillane
- Royal Free Hospital Foundation Trust London, London, UK MRC Centre for Neuromuscular Disease, UCL, London, UK
| | - D M Kullmann
- MRC Centre for Neuromuscular Disease, UCL, London, UK UCL, Institute of Neurology, London, UK
| | - M G Hanna
- MRC Centre for Neuromuscular Disease, UCL, London, UK UCL, Institute of Neurology, London, UK
| |
Collapse
|
24
|
Safory H, Neame S, Shulman Y, Zubedat S, Radzishevsky I, Rosenberg D, Sason H, Engelender S, Avital A, Hülsmann S, Schiller J, Wolosker H. The alanine-serine-cysteine-1 (Asc-1) transporter controls glycine levels in the brain and is required for glycinergic inhibitory transmission. EMBO Rep 2015; 16:590-8. [PMID: 25755256 DOI: 10.15252/embr.201439561] [Citation(s) in RCA: 37] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/10/2014] [Accepted: 02/13/2015] [Indexed: 11/09/2022] Open
Abstract
Asc-1 (SLC7A10) is an amino acid transporter whose deletion causes neurological abnormalities and early postnatal death in mice. Using metabolomics and behavioral and electrophysiological methods, we demonstrate that Asc-1 knockout mice display a marked decrease in glycine levels in the brain and spinal cord along with impairment of glycinergic inhibitory transmission, and a hyperekplexia-like phenotype that is rescued by replenishing brain glycine. Asc-1 works as a glycine and L-serine transporter, and its transport activity is required for the subsequent conversion of L-serine into glycine in vivo. Asc-1 is a novel regulator of glycine metabolism and a candidate for hyperekplexia disorders.
Collapse
Affiliation(s)
- Hazem Safory
- The Rappaport Faculty of Medicine and Research Institute, Technion-Israel Institute of Technology, Haifa, Israel
| | - Samah Neame
- The Rappaport Faculty of Medicine and Research Institute, Technion-Israel Institute of Technology, Haifa, Israel
| | - Yoav Shulman
- The Rappaport Faculty of Medicine and Research Institute, Technion-Israel Institute of Technology, Haifa, Israel
| | - Salman Zubedat
- The Rappaport Faculty of Medicine and Research Institute, Technion-Israel Institute of Technology, Haifa, Israel
| | - Inna Radzishevsky
- The Rappaport Faculty of Medicine and Research Institute, Technion-Israel Institute of Technology, Haifa, Israel
| | - Dina Rosenberg
- The Rappaport Faculty of Medicine and Research Institute, Technion-Israel Institute of Technology, Haifa, Israel
| | - Hagit Sason
- The Rappaport Faculty of Medicine and Research Institute, Technion-Israel Institute of Technology, Haifa, Israel
| | - Simone Engelender
- The Rappaport Faculty of Medicine and Research Institute, Technion-Israel Institute of Technology, Haifa, Israel
| | - Avi Avital
- The Rappaport Faculty of Medicine and Research Institute, Technion-Israel Institute of Technology, Haifa, Israel Emek Medical Center, Afula, Israel
| | - Swen Hülsmann
- Department of Anesthesiology, Emergency and Intensive Care Medicine and Center for Nanoscale Microscopy and Molecular Physiology of the Brain Georg-August-University, Göttingen, Germany
| | - Jackie Schiller
- The Rappaport Faculty of Medicine and Research Institute, Technion-Israel Institute of Technology, Haifa, Israel
| | - Herman Wolosker
- The Rappaport Faculty of Medicine and Research Institute, Technion-Israel Institute of Technology, Haifa, Israel
| |
Collapse
|
25
|
Arribas-González E, de Juan-Sanz J, Aragón C, López-Corcuera B. Molecular basis of the dominant negative effect of a glycine transporter 2 mutation associated with hyperekplexia. J Biol Chem 2014; 290:2150-65. [PMID: 25480793 DOI: 10.1074/jbc.m114.587055] [Citation(s) in RCA: 24] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/06/2022] Open
Abstract
Hyperekplexia or startle disease is a rare clinical syndrome characterized by an exaggerated startle in response to trivial tactile or acoustic stimuli. This neurological disorder can have serious consequences in neonates, provoking brain damage and/or sudden death due to apnea episodes and cardiorespiratory failure. Hyperekplexia is caused by defective inhibitory glycinergic neurotransmission. Mutations in the human SLC6A5 gene encoding the neuronal GlyT2 glycine transporter are responsible for the presynaptic form of the disease. GlyT2 mediates synaptic glycine recycling, which constitutes the main source of releasable transmitter at glycinergic synapses. Although the majority of GlyT2 mutations detected so far are recessive, a dominant negative mutant that affects GlyT2 trafficking does exist. In this study, we explore the properties and structural alterations of the S512R mutation in GlyT2. We analyze its dominant negative effect that retains wild-type GlyT2 in the endoplasmic reticulum (ER), preventing surface expression. We show that the presence of an arginine rather than serine 512 provoked transporter misfolding, enhanced association to the ER-chaperone calnexin, altered association with the coat-protein complex II component Sec24D, and thereby impeded ER exit. The S512R mutant formed oligomers with wild-type GlyT2 causing its retention in the ER. Overexpression of calnexin rescued wild-type GlyT2 from the dominant negative effect of the mutant, increasing the amount of transporter that reached the plasma membrane and dampening the interaction between the wild-type and mutant GlyT2. The ability of chemical chaperones to overcome the dominant negative effect of the disease mutation on the wild-type transporter was demonstrated in heterologous cells and primary neurons.
Collapse
Affiliation(s)
- Esther Arribas-González
- From the Departamento de Biología Molecular and Centro de Biología Molecular "Severo Ochoa," Consejo Superior de Investigaciones Científicas-Universidad Autónoma de Madrid, Madrid 28049, Spain, the IdiPAZ-Hospital Universitario La Paz, Universidad Autónoma de Madrid, Madrid 28046, Spain
| | - Jaime de Juan-Sanz
- From the Departamento de Biología Molecular and Centro de Biología Molecular "Severo Ochoa," Consejo Superior de Investigaciones Científicas-Universidad Autónoma de Madrid, Madrid 28049, Spain, the IdiPAZ-Hospital Universitario La Paz, Universidad Autónoma de Madrid, Madrid 28046, Spain the Centro de Investigación Biomédica en Red de Enfermedades Raras, Instituto de Salud Carlos III, Madrid 28029, Spain, and
| | - Carmen Aragón
- From the Departamento de Biología Molecular and Centro de Biología Molecular "Severo Ochoa," Consejo Superior de Investigaciones Científicas-Universidad Autónoma de Madrid, Madrid 28049, Spain, the IdiPAZ-Hospital Universitario La Paz, Universidad Autónoma de Madrid, Madrid 28046, Spain the Centro de Investigación Biomédica en Red de Enfermedades Raras, Instituto de Salud Carlos III, Madrid 28029, Spain, and
| | - Beatriz López-Corcuera
- From the Departamento de Biología Molecular and Centro de Biología Molecular "Severo Ochoa," Consejo Superior de Investigaciones Científicas-Universidad Autónoma de Madrid, Madrid 28049, Spain, the IdiPAZ-Hospital Universitario La Paz, Universidad Autónoma de Madrid, Madrid 28046, Spain the Centro de Investigación Biomédica en Red de Enfermedades Raras, Instituto de Salud Carlos III, Madrid 28029, Spain, and
| |
Collapse
|
26
|
Abstract
Hyperekplexia is a rare disorder caused by autosomal dominant or recessive modes of inheritance and characterized by episodes of exaggerated startle. Five causative genes have been identified to date. The syndrome has been recognized for decades and due to its rarity, the literature contains mostly descriptive reports, many early studies lacking molecular genetic diagnoses. A spectrum of clinical severity exists. Severe cases can lead to neonatal cardiac arrest and death during an episode, an outcome prevented by early diagnosis and clinical vigilance. Large treatment studies are not feasible, so therapeutic measures continue to be empiric. A marked response to clonazepam is often reported but refractory cases exist. Herein we report the clinical course and treatment response of a severely affected infant homozygous for an SLC6A5 nonsense mutation and review the literature summarizing the history and genetic understanding of the disease as well as the described comorbidities and treatment options.
Collapse
|
27
|
Jiménez E, Núñez E, Ibáñez I, Zafra F, Aragón C, Giménez C. Glycine transporters GlyT1 and GlyT2 are differentially modulated by glycogen synthase kinase 3β. Neuropharmacology 2014; 89:245-54. [PMID: 25301276 DOI: 10.1016/j.neuropharm.2014.09.023] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/25/2014] [Revised: 09/08/2014] [Accepted: 09/16/2014] [Indexed: 11/16/2022]
Abstract
Inhibitory glycinergic neurotransmission is terminated by the specific glycine transporters GlyT1 and GlyT2 which actively reuptake glycine from the synaptic cleft. GlyT1 is associated with both glycinergic and glutamatergic pathways, and is the main regulator of the glycine levels in the synapses. GlyT2 is the main supplier of glycine for vesicle refilling, a process that is vital to preserve the quantal glycine content in synaptic vesicles. Therefore, to control glycinergic neurotransmission efficiently, GlyT1 and GlyT2 activity must be regulated by diverse neuronal and glial signaling pathways. In this work, we have investigated the possible functional modulation of GlyT1 and GlyT2 by glycogen synthase kinase 3 (GSK3β). This kinase is involved in mood stabilization, neurodegeneration and plasticity at excitatory and inhibitory synapses. The co-expression of GSK3β with GlyT1 or GlyT2 in COS-7 cells and Xenopus laevis oocytes, leads to inhibition and stimulation of GlyT1 and GlyT2 activities, respectively, with a decrease of GlyT1, and an increase in GlyT2 levels at the plasma membrane. The specificity of these changes is supported by the antagonism exerted by a catalytically inactive form of the kinase and through inhibitors of GSK3β such as lithium chloride and TDZD-8. GSK3β also increases the incorporation of 32Pi into GlyT1 and decreases that of GlyT2. The pharmacological inhibition of the endogenous GSK3β in neuron cultures of brainstem and spinal cord leads to an opposite modulation of GlyT1 and GlyT2.Our results suggest that GSK3β is important for stabilizing and/or controlling the expression of functional GlyTs on the neural cell surface.
Collapse
Affiliation(s)
- Esperanza Jiménez
- Centro de Biología Molecular Severo Ochoa, Universidad Autónoma de Madrid, Consejo Superior de Investigaciones Científicas, 28049 Madrid, Spain; Centro de Investigación Biomédica en Red de Enfermedades Raras, ISCIII, Madrid, Spain; IdiPAZ-Hospital Universitario La Paz, Madrid, Spain
| | - Enrique Núñez
- Centro de Biología Molecular Severo Ochoa, Universidad Autónoma de Madrid, Consejo Superior de Investigaciones Científicas, 28049 Madrid, Spain; Centro de Investigación Biomédica en Red de Enfermedades Raras, ISCIII, Madrid, Spain; IdiPAZ-Hospital Universitario La Paz, Madrid, Spain
| | - Ignacio Ibáñez
- Centro de Biología Molecular Severo Ochoa, Universidad Autónoma de Madrid, Consejo Superior de Investigaciones Científicas, 28049 Madrid, Spain; Centro de Investigación Biomédica en Red de Enfermedades Raras, ISCIII, Madrid, Spain; IdiPAZ-Hospital Universitario La Paz, Madrid, Spain
| | - Francisco Zafra
- Centro de Biología Molecular Severo Ochoa, Universidad Autónoma de Madrid, Consejo Superior de Investigaciones Científicas, 28049 Madrid, Spain; Centro de Investigación Biomédica en Red de Enfermedades Raras, ISCIII, Madrid, Spain; IdiPAZ-Hospital Universitario La Paz, Madrid, Spain
| | - Carmen Aragón
- Centro de Biología Molecular Severo Ochoa, Universidad Autónoma de Madrid, Consejo Superior de Investigaciones Científicas, 28049 Madrid, Spain; Centro de Investigación Biomédica en Red de Enfermedades Raras, ISCIII, Madrid, Spain; IdiPAZ-Hospital Universitario La Paz, Madrid, Spain
| | - Cecilio Giménez
- Centro de Biología Molecular Severo Ochoa, Universidad Autónoma de Madrid, Consejo Superior de Investigaciones Científicas, 28049 Madrid, Spain; Centro de Investigación Biomédica en Red de Enfermedades Raras, ISCIII, Madrid, Spain; IdiPAZ-Hospital Universitario La Paz, Madrid, Spain.
| |
Collapse
|
28
|
Mihalikova A, Baliova M, Jursky F. Calcium dependent interaction of calmodulin with the GlyT1 C-terminus. Neurochem Res 2014; 39:2225-33. [PMID: 25168305 DOI: 10.1007/s11064-014-1424-x] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/15/2014] [Revised: 08/19/2014] [Accepted: 08/21/2014] [Indexed: 11/25/2022]
Abstract
The cytoplasmic regions of neurotransmitter transporters play an important role in their trafficking. This process is, to a high extent, tuned by calcium and calcium binding proteins, but the exact molecular connection are still not fully understood. In this work we found that the C-terminal region of the mouse glycine transporter GlyT1b is able to specifically interact with calmodulin in the presence of calcium. We found that several GlyT1 C-terminal mutations, including those in the ER retention signal, either eliminate or increase calmodulin interaction in vitro. In tissue-culture-expressed GlyT1 at least two of these mutations altered the sensitivity of GlyT1 surface expression and glycine uptake to calmodulin antagonists. These results suggest the possible involvement of calmodulin or calmodulin-like interactions in the regulation of GlyT1C-mediated transporter trafficking.
Collapse
Affiliation(s)
- Andrea Mihalikova
- Laboratory of Neurobiology, Institute of Molecular Biology, Slovak Academy of Sciences, Dubravska cesta 21, 845 51, Bratislava, Slovakia
| | | | | |
Collapse
|
29
|
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: 93] [Impact Index Per Article: 9.3] [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
|
30
|
Defining the blanks--pharmacochaperoning of SLC6 transporters and ABC transporters. Pharmacol Res 2013; 83:63-73. [PMID: 24316454 PMCID: PMC4059943 DOI: 10.1016/j.phrs.2013.11.009] [Citation(s) in RCA: 37] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/12/2013] [Revised: 11/27/2013] [Accepted: 11/27/2013] [Indexed: 02/06/2023]
Abstract
SLC6 family members and ABC transporters represent two extremes: SLC6 transporters are confined to the membrane proper and only expose small segments to the hydrophilic milieu. In ABC transporters the hydrophobic core is connected to a large intracellular (eponymous) ATP binding domain that is comprised of two discontiguous repeats. Accordingly, their folding problem is fundamentally different. This can be gauged from mutations that impair the folding of the encoded protein and give rise to clinically relevant disease phenotypes: in SLC6 transporters, these cluster at the protein–lipid interface on the membrane exposed surface. Mutations in ABC-transporters map to the interface between nucleotide binding domains and the coupling helices, which provide the connection to the hydrophobic core. Folding of these mutated ABC-transporters can be corrected with ligands/substrates that bind to the hydrophobic core. This highlights a pivotal role of the coupling helices in the folding trajectory. In contrast, insights into pharmacochaperoning of SLC6 transporters are limited to monoamine transporters – in particular the serotonin transporter (SERT) – because of their rich pharmacology. Only ligands that stabilize the inward facing conformation act as effective pharmacochaperones. This indicates that the folding trajectory of SERT proceeds via the inward facing conformation. Mutations that impair folding of SLC6 family members can be transmitted as dominant or recessive alleles. The dominant phenotype of the mutation can be rationalized, because SLC6 transporters are exported in oligomeric form from the endoplasmic reticulum (ER). Recessive transmission requires shielding of the unaffected gene product from the mutated transporter in the ER. This can be accounted for by a chaperone-COPII (coatomer protein II) exchange model, where proteinaceous ER-resident chaperones engage various intermediates prior to formation of the oligomeric state and subsequent export from the ER. It is likely that the action of pharmacochaperones is contingent on and modulated by these chaperones.
Collapse
|
31
|
de Juan-Sanz J, Núñez E, Villarejo-López L, Pérez-Hernández D, Rodriguez-Fraticelli AE, López-Corcuera B, Vázquez J, Aragón C. Na+/K+-ATPase is a new interacting partner for the neuronal glycine transporter GlyT2 that downregulates its expression in vitro and in vivo. J Neurosci 2013; 33:14269-81. [PMID: 23986260 PMCID: PMC6618510 DOI: 10.1523/jneurosci.1532-13.2013] [Citation(s) in RCA: 34] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/10/2013] [Revised: 07/09/2013] [Accepted: 07/18/2013] [Indexed: 01/16/2023] Open
Abstract
The neuronal glycine transporter GlyT2 plays a fundamental role in the glycinergic neurotransmission by recycling the neurotransmitter to the presynaptic terminal. GlyT2 is the main supplier of glycine for vesicle refilling, a process that is absolutely necessary to preserve quantal glycine content in synaptic vesicles. Alterations in GlyT2 activity modify glycinergic neurotransmission and may underlie several neuromuscular disorders, such as hyperekplexia, myoclonus, dystonia, and epilepsy. Indeed, mutations in the gene encoding GlyT2 are the main presynaptic cause of hyperekplexia in humans and produce congenital muscular dystonia type 2 (CMD2) in Belgian Blue cattle. GlyT2 function is strictly coupled to the sodium electrochemical gradient actively generated by the Na+/K+-ATPase (NKA). GlyT2 cotransports 3Na+/Cl-/glycine generating large rises of Na+ inside the presynaptic terminal that must be efficiently reduced by the NKA to preserve Na+ homeostasis. In this work, we have used high-throughput mass spectrometry to identify proteins interacting with GlyT2 in the CNS. NKA was detected as a putative candidate and through reciprocal coimmunoprecipitations and immunocytochemistry analyses the association between GlyT2 and NKA was confirmed. NKA mainly interacts with the raft-associated active pool of GlyT2, and low and high levels of the specific NKA ligand ouabain modulate the endocytosis and total expression of GlyT2 in neurons. The ouabain-mediated downregulation of GlyT2 also occurs in vivo in two different systems: zebrafish embryos and adult rats, indicating that this NKA-mediated regulatory mechanism is evolutionarily conserved and may play a relevant role in the physiological control of inhibitory glycinergic neurotransmission.
Collapse
Affiliation(s)
- Jaime de Juan-Sanz
- Centro de Biología Molecular Severo Ochoa, Universidad Autónoma de Madrid, Consejo Superior de Investigaciones Científicas, 28049 Madrid, Spain
- Centro de Investigación Biomédica en Red de Enfermedades Raras, ISCIII, 46010 Valencia, Spain
- IdiPAZ-Hospital Universitario La Paz, 28046 Madrid, Spain
| | - Enrique Núñez
- Centro de Biología Molecular Severo Ochoa, Universidad Autónoma de Madrid, Consejo Superior de Investigaciones Científicas, 28049 Madrid, Spain
- Centro de Investigación Biomédica en Red de Enfermedades Raras, ISCIII, 46010 Valencia, Spain
- IdiPAZ-Hospital Universitario La Paz, 28046 Madrid, Spain
| | - Lucía Villarejo-López
- Centro de Biología Molecular Severo Ochoa, Universidad Autónoma de Madrid, Consejo Superior de Investigaciones Científicas, 28049 Madrid, Spain
| | | | - Alejo E. Rodriguez-Fraticelli
- Centro de Biología Molecular Severo Ochoa, Universidad Autónoma de Madrid, Consejo Superior de Investigaciones Científicas, 28049 Madrid, Spain
| | - Beatriz López-Corcuera
- Centro de Biología Molecular Severo Ochoa, Universidad Autónoma de Madrid, Consejo Superior de Investigaciones Científicas, 28049 Madrid, Spain
- Centro de Investigación Biomédica en Red de Enfermedades Raras, ISCIII, 46010 Valencia, Spain
- IdiPAZ-Hospital Universitario La Paz, 28046 Madrid, Spain
| | - Jesús Vázquez
- Centro Nacional de Investigaciones Cardiovasculares, 28029 Madrid, Spain, and
| | - Carmen Aragón
- Centro de Biología Molecular Severo Ochoa, Universidad Autónoma de Madrid, Consejo Superior de Investigaciones Científicas, 28049 Madrid, Spain
- Centro de Investigación Biomédica en Red de Enfermedades Raras, ISCIII, 46010 Valencia, Spain
- IdiPAZ-Hospital Universitario La Paz, 28046 Madrid, Spain
| |
Collapse
|
32
|
Pariset L, Bongiorni S, Bueno S, Gruber CEM, Prosperini G, Chillemi G, Bicorgna S, Gentile A, Valentini A. Microarray gene expression profiling of neural tissues in bovine spastic paresis. BMC Vet Res 2013; 9:122. [PMID: 23782433 PMCID: PMC3693873 DOI: 10.1186/1746-6148-9-122] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/20/2012] [Accepted: 06/11/2013] [Indexed: 11/24/2022] Open
Abstract
Background Bovine Spastic Paresis (BSP) is a neuromuscular disorder which affects both male and female cattle. BSP is characterized by spastic contraction and overextension of the gastrocnemious muscle of one or both limbs and is associated with a scarce increase in body weight. This disease seems to be caused by an autosomal and recessive gene, with incomplete penetration, although no genes clearly involved with its onset have been so far identified. We employed cDNA microarrays to identify metabolic pathways affected by BSP in Romagnola cattle breed. Investigation of those pathways at the genome level can help to understand this disease. Results Microarray analysis of control and affected individuals resulted in 268 differentially expressed genes. These genes were subjected to KEGG pathway functional clustering analysis, revealing that they are predominantly involved in Cell Communication, Signalling Molecules and Interaction and Signal Transduction, Diseases and Nervous System classes. Significantly enriched KEGG pathway’s classes for the differentially expressed genes were calculated; interestingly, all those significantly under-expressed in the affected samples are included in Neurodegenerative Diseases. To identify genome locations possibly harbouring gene(s) involved in the disease, the chromosome distribution of the differentially expressed genes was also investigated. Conclusions The cDNA microarray we used in this study contains a brain library and, even if carrying an incomplete transcriptome representation, it has proven to be a valuable tool allowing us to add useful and new information to a poorly studied disease. By using this tool, we examined nearly 15000 transcripts and analysed gene pathways affected by the disease. Particularly, our data suggest also a defective glycinergic synaptic transmission in the development of the disease and an alteration of calcium signalling proteins. We provide data to acquire knowledge of a genetic disease for which literature still presents poor results and that could be further and specifically analysed in the next future. Moreover this study, performed in livestock, may also harbour molecular information useful for understanding human diseases.
Collapse
Affiliation(s)
- Lorraine Pariset
- Department for Innovation in Biological, Agro-food and Forest systems (DIBAF), University of Tuscia, via S, Camillo de Lellis snc, Viterbo, 01100, Italy.
| | | | | | | | | | | | | | | | | |
Collapse
|
33
|
Arribas-González E, Alonso-Torres P, Aragón C, López-Corcuera B. Calnexin-assisted biogenesis of the neuronal glycine transporter 2 (GlyT2). PLoS One 2013; 8:e63230. [PMID: 23650557 PMCID: PMC3641136 DOI: 10.1371/journal.pone.0063230] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/08/2012] [Accepted: 03/30/2013] [Indexed: 11/20/2022] Open
Abstract
The neuronal transporter GlyT2 is a polytopic, 12-transmembrane domain, plasma membrane glycoprotein involved in the removal and recycling of synaptic glycine from inhibitory synapses. Mutations in the human GlyT2 gene (SLC6A5) that cause deficient glycine transport or defective GlyT2 trafficking are the second most common cause of hyperekplexia or startle disease. In this study we examined several aspects of GlyT2 biogenesis that involve the endoplasmic reticulum chaperone calnexin (CNX). CNX binds transiently to an intermediate under-glycosylated transporter precursor and facilitates GlyT2 processing. In cells expressing GlyT2, transporter accumulation and transport activity were attenuated by siRNA-mediated CNX knockdown and enhanced by CNX overexpression. GlyT2 binding to CNX was mediated by glycan and polypeptide-based interactions as revealed by pharmacological approaches and the behavior of GlyT2 N-glycan-deficient mutants. Moreover, transporter folding appeared to be stabilized by N-glycans. Co-expression of CNX and a fully non-glycosylated mutant rescues glycine transport but not mutant surface expression. Hence, CNX discriminates between different conformational states of GlyT2 displaying a lectin-independent chaperone activity. GlyT2 wild-type and mutant transporters were finally degraded in the lysosome. Our findings provide further insight into GlyT2 biogenesis, and a useful framework for the study of newly synthesized GlyT2 transporters bearing hyperekplexia mutations.
Collapse
Affiliation(s)
- Esther Arribas-González
- Departamento de Biología Molecular and Centro de Biología Molecular “Severo Ochoa”, Consejo Superior de Investigaciones Científicas, Universidad Autónoma de Madrid, Madrid, Spain
- IdiPAZ-Hospital Universitario La Paz, Universidad Autónoma de Madrid, Madrid, Spain
| | - Pablo Alonso-Torres
- Departamento de Biología Molecular and Centro de Biología Molecular “Severo Ochoa”, Consejo Superior de Investigaciones Científicas, Universidad Autónoma de Madrid, Madrid, Spain
| | - Carmen Aragón
- Departamento de Biología Molecular and Centro de Biología Molecular “Severo Ochoa”, Consejo Superior de Investigaciones Científicas, Universidad Autónoma de Madrid, Madrid, Spain
- Centro de Investigación Biomédica en Red de Enfermedades Raras, Instituto de Salud Carlos III, Madrid, Spain
- IdiPAZ-Hospital Universitario La Paz, Universidad Autónoma de Madrid, Madrid, Spain
| | - Beatriz López-Corcuera
- Departamento de Biología Molecular and Centro de Biología Molecular “Severo Ochoa”, Consejo Superior de Investigaciones Científicas, Universidad Autónoma de Madrid, Madrid, Spain
- Centro de Investigación Biomédica en Red de Enfermedades Raras, Instituto de Salud Carlos III, Madrid, Spain
- IdiPAZ-Hospital Universitario La Paz, Universidad Autónoma de Madrid, Madrid, Spain
| |
Collapse
|
34
|
Jursky F, Baliova M. Expression and purification of recombinant calpain-derived N-terminal peptides from glycine transporter GlyT2. Protein Expr Purif 2013; 88:143-9. [DOI: 10.1016/j.pep.2012.12.007] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/03/2012] [Revised: 12/17/2012] [Accepted: 12/18/2012] [Indexed: 11/15/2022]
|
35
|
Giménez C, Pérez-Siles G, Martínez-Villarreal J, Arribas-González E, Jiménez E, Núñez E, de Juan-Sanz J, Fernández-Sánchez E, García-Tardón N, Ibáñez I, Romanelli V, Nevado J, James VM, Topf M, Chung SK, Thomas RH, Desviat LR, Aragón C, Zafra F, Rees MI, Lapunzina P, Harvey RJ, López-Corcuera B. A novel dominant hyperekplexia mutation Y705C alters trafficking and biochemical properties of the presynaptic glycine transporter GlyT2. J Biol Chem 2012; 287:28986-9002. [PMID: 22753417 PMCID: PMC3436537 DOI: 10.1074/jbc.m111.319244] [Citation(s) in RCA: 34] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/31/2011] [Revised: 06/18/2012] [Indexed: 11/06/2022] Open
Abstract
Hyperekplexia or startle disease is characterized by an exaggerated startle response, evoked by tactile or auditory stimuli, producing hypertonia and apnea episodes. Although rare, this orphan disorder can have serious consequences, including sudden infant death. Dominant and recessive mutations in the human glycine receptor (GlyR) α1 gene (GLRA1) are the major cause of this disorder. However, recessive mutations in the presynaptic Na(+)/Cl(-)-dependent glycine transporter GlyT2 gene (SLC6A5) are rapidly emerging as a second major cause of startle disease. In this study, systematic DNA sequencing of SLC6A5 revealed a new dominant GlyT2 mutation: pY705C (c.2114A→G) in transmembrane domain 11, in eight individuals from Spain and the United Kingdom. Curiously, individuals harboring this mutation show significant variation in clinical presentation. In addition to classical hyperekplexia symptoms, some individuals had abnormal respiration, facial dysmorphism, delayed motor development, or intellectual disability. We functionally characterized this mutation using molecular modeling, electrophysiology, [(3)H]glycine transport, cell surface expression, and cysteine labeling assays. We found that the introduced cysteine interacts with the cysteine pair Cys-311-Cys-320 in the second external loop of GlyT2. This interaction impairs transporter maturation through the secretory pathway, reduces surface expression, and inhibits transport function. Additionally, Y705C presents altered H(+) and Zn(2+) dependence of glycine transport that may affect the function of glycinergic neurotransmission in vivo.
Collapse
Affiliation(s)
- Cecilio Giménez
- From the Departamento de Biología Molecular and Centro de Biología Molecular “Severo Ochoa,” (Consejo Superior de Investigaciones Científicas-Universidad Autónoma de Madrid), Madrid 28049, Spain
- the Centro de Investigación Biomédica en Red de Enfermedades Raras, Instituto de Salud Carlos III, Madrid 28029, Spain
- the IdiPAZ-Hospital Universitario La Paz
| | - Gonzalo Pérez-Siles
- From the Departamento de Biología Molecular and Centro de Biología Molecular “Severo Ochoa,” (Consejo Superior de Investigaciones Científicas-Universidad Autónoma de Madrid), Madrid 28049, Spain
- the Centro de Investigación Biomédica en Red de Enfermedades Raras, Instituto de Salud Carlos III, Madrid 28029, Spain
| | - Jaime Martínez-Villarreal
- From the Departamento de Biología Molecular and Centro de Biología Molecular “Severo Ochoa,” (Consejo Superior de Investigaciones Científicas-Universidad Autónoma de Madrid), Madrid 28049, Spain
- the Centro de Investigación Biomédica en Red de Enfermedades Raras, Instituto de Salud Carlos III, Madrid 28029, Spain
- the IdiPAZ-Hospital Universitario La Paz
| | - Esther Arribas-González
- From the Departamento de Biología Molecular and Centro de Biología Molecular “Severo Ochoa,” (Consejo Superior de Investigaciones Científicas-Universidad Autónoma de Madrid), Madrid 28049, Spain
- the IdiPAZ-Hospital Universitario La Paz
| | - Esperanza Jiménez
- From the Departamento de Biología Molecular and Centro de Biología Molecular “Severo Ochoa,” (Consejo Superior de Investigaciones Científicas-Universidad Autónoma de Madrid), Madrid 28049, Spain
- the Centro de Investigación Biomédica en Red de Enfermedades Raras, Instituto de Salud Carlos III, Madrid 28029, Spain
- the IdiPAZ-Hospital Universitario La Paz
| | - Enrique Núñez
- From the Departamento de Biología Molecular and Centro de Biología Molecular “Severo Ochoa,” (Consejo Superior de Investigaciones Científicas-Universidad Autónoma de Madrid), Madrid 28049, Spain
- the Centro de Investigación Biomédica en Red de Enfermedades Raras, Instituto de Salud Carlos III, Madrid 28029, Spain
- the IdiPAZ-Hospital Universitario La Paz
| | - Jaime de Juan-Sanz
- From the Departamento de Biología Molecular and Centro de Biología Molecular “Severo Ochoa,” (Consejo Superior de Investigaciones Científicas-Universidad Autónoma de Madrid), Madrid 28049, Spain
- the Centro de Investigación Biomédica en Red de Enfermedades Raras, Instituto de Salud Carlos III, Madrid 28029, Spain
- the IdiPAZ-Hospital Universitario La Paz
| | - Enrique Fernández-Sánchez
- From the Departamento de Biología Molecular and Centro de Biología Molecular “Severo Ochoa,” (Consejo Superior de Investigaciones Científicas-Universidad Autónoma de Madrid), Madrid 28049, Spain
| | - Noemí García-Tardón
- From the Departamento de Biología Molecular and Centro de Biología Molecular “Severo Ochoa,” (Consejo Superior de Investigaciones Científicas-Universidad Autónoma de Madrid), Madrid 28049, Spain
- the Centro de Investigación Biomédica en Red de Enfermedades Raras, Instituto de Salud Carlos III, Madrid 28029, Spain
- the IdiPAZ-Hospital Universitario La Paz
| | - Ignacio Ibáñez
- From the Departamento de Biología Molecular and Centro de Biología Molecular “Severo Ochoa,” (Consejo Superior de Investigaciones Científicas-Universidad Autónoma de Madrid), Madrid 28049, Spain
| | - Valeria Romanelli
- the Centro de Investigación Biomédica en Red de Enfermedades Raras, Instituto de Salud Carlos III, Madrid 28029, Spain
- the Instituto de Genética Médica y Molecular, IdiPAZ-Hospital Universitario La Paz, Universidad Autónoma de Madrid, Madrid 28046, Spain
| | - Julián Nevado
- the Centro de Investigación Biomédica en Red de Enfermedades Raras, Instituto de Salud Carlos III, Madrid 28029, Spain
- the Instituto de Genética Médica y Molecular, IdiPAZ-Hospital Universitario La Paz, Universidad Autónoma de Madrid, Madrid 28046, Spain
| | - Victoria M. James
- the Department of Pharmacology, University College London School of Pharmacy, London WC1N 1AX, United Kingdom
| | - Maya Topf
- the Institute of Structural and Molecular Biology, Crystallography, Birkbeck College, London WC1E 7HX, United Kingdom, and
| | - Seo-Kyung Chung
- the Institute of Life Science, College of Medicine, Swansea University, Swansea SA2 8PP, United Kingdom
| | - Rhys H. Thomas
- the Institute of Life Science, College of Medicine, Swansea University, Swansea SA2 8PP, United Kingdom
| | - Lourdes R. Desviat
- From the Departamento de Biología Molecular and Centro de Biología Molecular “Severo Ochoa,” (Consejo Superior de Investigaciones Científicas-Universidad Autónoma de Madrid), Madrid 28049, Spain
| | - Carmen Aragón
- From the Departamento de Biología Molecular and Centro de Biología Molecular “Severo Ochoa,” (Consejo Superior de Investigaciones Científicas-Universidad Autónoma de Madrid), Madrid 28049, Spain
- the Centro de Investigación Biomédica en Red de Enfermedades Raras, Instituto de Salud Carlos III, Madrid 28029, Spain
- the IdiPAZ-Hospital Universitario La Paz
| | - Francisco Zafra
- From the Departamento de Biología Molecular and Centro de Biología Molecular “Severo Ochoa,” (Consejo Superior de Investigaciones Científicas-Universidad Autónoma de Madrid), Madrid 28049, Spain
- the Centro de Investigación Biomédica en Red de Enfermedades Raras, Instituto de Salud Carlos III, Madrid 28029, Spain
- the IdiPAZ-Hospital Universitario La Paz
| | - Mark I. Rees
- the Institute of Life Science, College of Medicine, Swansea University, Swansea SA2 8PP, United Kingdom
| | - Pablo Lapunzina
- the Centro de Investigación Biomédica en Red de Enfermedades Raras, Instituto de Salud Carlos III, Madrid 28029, Spain
- the Instituto de Genética Médica y Molecular, IdiPAZ-Hospital Universitario La Paz, Universidad Autónoma de Madrid, Madrid 28046, Spain
| | - Robert J. Harvey
- the Department of Pharmacology, University College London School of Pharmacy, London WC1N 1AX, United Kingdom
| | - Beatriz López-Corcuera
- From the Departamento de Biología Molecular and Centro de Biología Molecular “Severo Ochoa,” (Consejo Superior de Investigaciones Científicas-Universidad Autónoma de Madrid), Madrid 28049, Spain
- the Centro de Investigación Biomédica en Red de Enfermedades Raras, Instituto de Salud Carlos III, Madrid 28029, Spain
- the IdiPAZ-Hospital Universitario La Paz
| |
Collapse
|
36
|
Carta E, Chung SK, James VM, Robinson A, Gill JL, Remy N, Vanbellinghen JF, Drew CJG, Cagdas S, Cameron D, Cowan FM, Del Toro M, Graham GE, Manzur AY, Masri A, Rivera S, Scalais E, Shiang R, Sinclair K, Stuart CA, Tijssen MAJ, Wise G, Zuberi SM, Harvey K, Pearce BR, Topf M, Thomas RH, Supplisson S, Rees MI, Harvey RJ. Mutations in the GlyT2 gene (SLC6A5) are a second major cause of startle disease. J Biol Chem 2012; 287:28975-85. [PMID: 22700964 PMCID: PMC3436555 DOI: 10.1074/jbc.m112.372094] [Citation(s) in RCA: 68] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/19/2012] [Revised: 06/11/2012] [Indexed: 11/06/2022] Open
Abstract
Hereditary hyperekplexia or startle disease is characterized by an exaggerated startle response, evoked by tactile or auditory stimuli, leading to hypertonia and apnea episodes. Missense, nonsense, frameshift, splice site mutations, and large deletions in the human glycine receptor α1 subunit gene (GLRA1) are the major known cause of this disorder. However, mutations are also found in the genes encoding the glycine receptor β subunit (GLRB) and the presynaptic Na(+)/Cl(-)-dependent glycine transporter GlyT2 (SLC6A5). In this study, systematic DNA sequencing of SLC6A5 in 93 new unrelated human hyperekplexia patients revealed 20 sequence variants in 17 index cases presenting with homozygous or compound heterozygous recessive inheritance. Five apparently unrelated cases had the truncating mutation R439X. Genotype-phenotype analysis revealed a high rate of neonatal apneas and learning difficulties associated with SLC6A5 mutations. From the 20 SLC6A5 sequence variants, we investigated glycine uptake for 16 novel mutations, confirming that all were defective in glycine transport. Although the most common mechanism of disrupting GlyT2 function is protein truncation, new pathogenic mechanisms included splice site mutations and missense mutations affecting residues implicated in Cl(-) binding, conformational changes mediated by extracellular loop 4, and cation-π interactions. Detailed electrophysiology of mutation A275T revealed that this substitution results in a voltage-sensitive decrease in glycine transport caused by lower Na(+) affinity. This study firmly establishes the combination of missense, nonsense, frameshift, and splice site mutations in the GlyT2 gene as the second major cause of startle disease.
Collapse
Affiliation(s)
- Eloisa Carta
- Department of Pharmacology, UCL School of Pharmacy, London WC1N 1AX, United Kingdom
| | - Seo-Kyung Chung
- Institute of Life Science, College of Medicine, Swansea University, Swansea SA2 8PP, United Kingdom
| | - Victoria M. James
- Department of Pharmacology, UCL School of Pharmacy, London WC1N 1AX, United Kingdom
| | - Angela Robinson
- Institute of Life Science, College of Medicine, Swansea University, Swansea SA2 8PP, United Kingdom
| | - Jennifer L. Gill
- Department of Pharmacology, UCL School of Pharmacy, London WC1N 1AX, United Kingdom
| | - Nathalie Remy
- Human Genetics, Liège University Hospital, B-4000 Liège, Belgium
| | | | - Cheney J. G. Drew
- Institute of Life Science, College of Medicine, Swansea University, Swansea SA2 8PP, United Kingdom
| | - Sophie Cagdas
- Department of Child Neuropsychiatry, C Poma Hospital, 46100 Mantova, Italy
| | - Duncan Cameron
- Department of Paediatrics, Glan Clwyd Hospital, Rhyl LL18 5UJ, United Kingdom
| | - Frances M. Cowan
- Department of Paediatrics, Imperial College, London W12 0HS, United Kingdom
| | - Mireria Del Toro
- Servicio de Neurologia Pediatrica, Hospital General Universitari Vall d'Hebron, 08035 Barcelona, Spain
| | - Gail E. Graham
- Department of Genetics, Children's Hospital of Eastern Ontario, Ottawa, Ontario K1H 8L1, Canada
| | - Adnan Y. Manzur
- Department of Paediatric Neurology, Great Ormond Street Hospital for Children, London WC1N 3JH, United Kingdom
| | - Amira Masri
- Department of Paediatrics, University of Jordan, 11941 Amman, Jordan
| | - Serge Rivera
- Service de Pédiatrie, Centre Hospitalier de la Côte Basque, 64109 Bayonne, France
| | - Emmanuel Scalais
- Neurologie Pédiatrique, Centre Hospitalier de Luxembourg, L-1210 Luxembourg
| | - Rita Shiang
- Department of Human and Molecular Genetics, Virginia Commonwealth University, Richmond, Virginia 23298-0033
| | - Kate Sinclair
- Queensland Paediatric Rehabilitation Service, Royal Children's Hospital, Herston 4029, Australia
| | | | - Marina A. J. Tijssen
- Department of Neurology, University Medical Centre Groningen, 9713 GZ, Groningen, The Netherlands
| | - Grahame Wise
- Sydney Children's Hospital, Randwick NSW 2031, Australia
| | - Sameer M. Zuberi
- Paediatric Neurosciences Research Group, Royal Hospital for Sick Children, Glasgow G3 8SJ, United Kingdom
| | - Kirsten Harvey
- Department of Pharmacology, UCL School of Pharmacy, London WC1N 1AX, United Kingdom
| | - Brian R. Pearce
- Department of Pharmacology, UCL School of Pharmacy, London WC1N 1AX, United Kingdom
| | - Maya Topf
- Institute of Structural and Molecular Biology, Department of Biological Sciences, Birkbeck College, London WC1E 7HX, United Kingdom
| | - Rhys H. Thomas
- 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
| | - Stéphane Supplisson
- Institut de Biologie de l'Ecole Normale Supérieure, Paris 75000, France
- Institut National de la Santé et de la Recherche Médicale, U1024, Paris 75000, France, and
- CNRS, UMR 8197, Paris 75000, France
| | - Mark I. Rees
- 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
| | - Robert J. Harvey
- Department of Pharmacology, UCL School of Pharmacy, London WC1N 1AX, United Kingdom
| |
Collapse
|
37
|
A MusD retrotransposon insertion in the mouse Slc6a5 gene causes alterations in neuromuscular junction maturation and behavioral phenotypes. PLoS One 2012; 7:e30217. [PMID: 22272310 PMCID: PMC3260239 DOI: 10.1371/journal.pone.0030217] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/16/2011] [Accepted: 12/15/2011] [Indexed: 11/20/2022] Open
Abstract
Glycine is the major inhibitory neurotransmitter in the spinal cord and some brain regions. The presynaptic glycine transporter, GlyT2, is required for sustained glycinergic transmission through presynaptic reuptake and recycling of glycine. Mutations in SLC6A5, encoding GlyT2, cause hereditary hyperekplexia in humans, and similar phenotypes in knock-out mice, and variants are associated with schizophrenia. We identified a spontaneous mutation in mouse Slc6a5, caused by a MusD retrotransposon insertion. The GlyT2 protein is undetectable in homozygous mutants, indicating a null allele. Homozygous mutant mice are normal at birth, but develop handling-induced spasms at five days of age, and only survive for two weeks, but allow the study of early activity-regulated developmental processes. At the neuromuscular junction, synapse elimination and the switch from embryonic to adult acetylcholine receptor subunits are hastened, consistent with a presumed increase in motor neuron activity, and transcription of acetylcholine receptors is elevated. Heterozygous mice, which show no reduction in lifespan but nonetheless have reduced levels of GlyT2, have a normal thermal sensitivity with the hot-plate test, but differences in repetitive grooming and decreased sleep time with home-cage monitoring. Open-field and elevated plus-maze tests did not detect anxiety-like behaviors; however, the latter showed a hyperactivity phenotype. Importantly, grooming and hyperactivity are observed in mouse schizophrenia models. Thus, mutations in Slc6a5 show changes in neuromuscular junction development as homozygotes, and behavioral phenotypes as heterozygotes, indicating their usefulness for studies related to glycinergic dysfunction.
Collapse
|
38
|
Forman OP, Penderis J, Hartley C, Hayward LJ, Ricketts SL, Mellersh CS. Parallel mapping and simultaneous sequencing reveals deletions in BCAN and FAM83H associated with discrete inherited disorders in a domestic dog breed. PLoS Genet 2012; 8:e1002462. [PMID: 22253609 PMCID: PMC3257292 DOI: 10.1371/journal.pgen.1002462] [Citation(s) in RCA: 61] [Impact Index Per Article: 5.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/12/2011] [Accepted: 11/21/2011] [Indexed: 12/28/2022] Open
Abstract
The domestic dog (Canis familiaris) segregates more naturally-occurring diseases and phenotypic variation than any other species and has become established as an unparalled model with which to study the genetics of inherited traits. We used a genome-wide association study (GWAS) and targeted resequencing of DNA from just five dogs to simultaneously map and identify mutations for two distinct inherited disorders that both affect a single breed, the Cavalier King Charles Spaniel. We investigated episodic falling (EF), a paroxysmal exertion-induced dyskinesia, alongside the phenotypically distinct condition congenital keratoconjunctivitis sicca and ichthyosiform dermatosis (CKCSID), commonly known as dry eye curly coat syndrome. EF is characterised by episodes of exercise-induced muscular hypertonicity and abnormal posturing, usually occurring after exercise or periods of excitement. CKCSID is a congenital disorder that manifests as a rough coat present at birth, with keratoconjunctivitis sicca apparent on eyelid opening at 10–14 days, followed by hyperkeratinisation of footpads and distortion of nails that develops over the next few months. We undertook a GWAS with 31 EF cases, 23 CKCSID cases, and a common set of 38 controls and identified statistically associated signals for EF and CKCSID on chromosome 7 (Praw 1.9×10−14; Pgenome = 1.0×10−5) and chromosome 13 (Praw 1.2×10−17; Pgenome = 1.0×10−5), respectively. We resequenced both the EF and CKCSID disease-associated regions in just five dogs and identified a 15,724 bp deletion spanning three exons of BCAN associated with EF and a single base-pair exonic deletion in FAM83H associated with CKCSID. Neither BCAN or FAM83H have been associated with equivalent disease phenotypes in any other species, thus demonstrating the ability to use the domestic dog to study the genetic basis of more than one disease simultaneously in a single breed and to identify multiple novel candidate genes in parallel.
Collapse
Affiliation(s)
- Oliver P Forman
- Kennel Club Genetics Centre, Animal Health Trust, Kentford, United Kingdom.
| | | | | | | | | | | |
Collapse
|
39
|
Kristensen AS, Andersen J, Jørgensen TN, Sørensen L, Eriksen J, Loland CJ, Strømgaard K, Gether U. SLC6 neurotransmitter transporters: structure, function, and regulation. Pharmacol Rev 2011; 63:585-640. [PMID: 21752877 DOI: 10.1124/pr.108.000869] [Citation(s) in RCA: 608] [Impact Index Per Article: 46.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/16/2022] Open
Abstract
The neurotransmitter transporters (NTTs) belonging to the solute carrier 6 (SLC6) gene family (also referred to as the neurotransmitter-sodium-symporter family or Na(+)/Cl(-)-dependent transporters) comprise a group of nine sodium- and chloride-dependent plasma membrane transporters for the monoamine neurotransmitters serotonin (5-hydroxytryptamine), dopamine, and norepinephrine, and the amino acid neurotransmitters GABA and glycine. The SLC6 NTTs are widely expressed in the mammalian brain and play an essential role in regulating neurotransmitter signaling and homeostasis by mediating uptake of released neurotransmitters from the extracellular space into neurons and glial cells. The transporters are targets for a wide range of therapeutic drugs used in treatment of psychiatric diseases, including major depression, anxiety disorders, attention deficit hyperactivity disorder and epilepsy. Furthermore, psychostimulants such as cocaine and amphetamines have the SLC6 NTTs as primary targets. Beginning with the determination of a high-resolution structure of a prokaryotic homolog of the mammalian SLC6 transporters in 2005, the understanding of the molecular structure, function, and pharmacology of these proteins has advanced rapidly. Furthermore, intensive efforts have been directed toward understanding the molecular and cellular mechanisms involved in regulation of the activity of this important class of transporters, leading to new methodological developments and important insights. This review provides an update of these advances and their implications for the current understanding of the SLC6 NTTs.
Collapse
Affiliation(s)
- Anders S Kristensen
- Department of Medicinal Chemistry, Faculty of Pharmaceutical Sciences, University of Copenhagen, Copenhagen, Denmark.
| | | | | | | | | | | | | | | |
Collapse
|
40
|
Moly PK, Hatta K. Early glycinergic axon contact with the Mauthner neuron during zebrafish development. Neurosci Res 2011; 70:251-9. [PMID: 21397641 DOI: 10.1016/j.neures.2011.03.003] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/04/2011] [Revised: 03/04/2011] [Accepted: 03/04/2011] [Indexed: 10/18/2022]
Abstract
Glycinergic neurons are the major inhibitory neurons in the vertebrate central nervous system. In teleosts, they play important roles in the escape response by regulating the activity of the Mauthner (M-) cells. Here we studied the contact between glycinergic axons and the M-cells in early zebrafish embryos by double immunostaining with an anti-glycine antibody and the 3A10 antibody that labels M-cells. We also studied a transgenic line, Tg(GlyT2:GFP), in which GFP is expressed under the control of the promoter for the glycine transporter-2 gene. The initial contacts by ascending glycinergic axons on the M-soma were observed within 27h post-fertilization (hpf) on the lateral part of the ventral surface of the M-soma. Stochastic labeling of glycinergic neurons was then performed by injecting a GlyT2:GFP construct into early cleaving eggs. We identified the origin of the earliest glycinergic axons that contact the M-soma as commissural neurons, located in the anterior spinal cord, whose axons ascend along the lateral longitudinal fascicles with a short descending branch. We also found, in the fourth rhombomere, late-developed glycinergic commissural neurons whose axons contact anterior or posterior edge of both M-somas. This study provides the first example of the initial development of an inhibitory network on an identifiable neuron in vertebrates.
Collapse
Affiliation(s)
- Pricila Khan Moly
- Graduate School of Life Science, University of Hyogo, Hyogo 678-1297, Japan
| | | |
Collapse
|
41
|
Startle disease in Irish wolfhounds associated with a microdeletion in the glycine transporter GlyT2 gene. Neurobiol Dis 2011; 43:184-9. [PMID: 21420493 PMCID: PMC4068303 DOI: 10.1016/j.nbd.2011.03.010] [Citation(s) in RCA: 35] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/23/2010] [Revised: 03/01/2011] [Accepted: 03/11/2011] [Indexed: 11/21/2022] Open
Abstract
Defects in glycinergic synaptic transmission in humans, cattle, and rodents result in an exaggerated startle reflex and hypertonia in response to either acoustic or tactile stimuli. Molecular genetic studies have determined that mutations in the genes encoding the postsynaptic glycine receptor (GlyR) α1 and β subunits (GLRA1 and GLRB) and the presynaptic glycine transporter GlyT2 (SLC6A5) are the major cause of these disorders. Here, we report the first genetically confirmed canine cases of startle disease. A litter of seven Irish wolfhounds was identified in which two puppies developed muscle stiffness and tremor in response to handling. Although sequencing of GLRA1 and GLRB did not reveal any pathogenic mutations, analysis of SLC6A5 revealed a homozygous 4.2 kb microdeletion encompassing exons 2 and 3 in both affected animals. This results in the loss of part of the large cytoplasmic N-terminus and all subsequent transmembrane domains due to a frameshift. This genetic lesion was confirmed by defining the deletion breakpoint, Southern blotting, and multiplex ligation-dependent probe amplification (MLPA). This analysis enabled the development of a rapid genotyping test that revealed heterozygosity for the deletion in the dam and sire and three other siblings, confirming recessive inheritance. Wider testing of related animals has identified a total of 13 carriers of the SLC6A5 deletion as well as non-carrier animals. These findings will inform future breeding strategies and enable a rational pharmacotherapy of this new canine disorder.
Collapse
|
42
|
Núñez E, Alonso-Torres P, Fornés A, Aragón C, López-Corcuera B. The neuronal glycine transporter GLYT2 associates with membrane rafts: functional modulation by lipid environment. J Neurochem 2010; 105:2080-90. [PMID: 18266927 DOI: 10.1111/j.1471-4159.2008.05292.x] [Citation(s) in RCA: 27] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/10/2023]
Abstract
The neuronal glycine transporter GLYT2 is a plasma membrane protein that removes the neurotransmitter glycine from the synaptic cleft, thereby aiding the pre-synaptic terminal reloading and the termination of the glycinergic signal. Missense mutations in the gene encoding GLYT2 (SLC6A5) cause hyperekplexia in humans. The activity of GLYT2 seems to be highly regulated. In this report, we demonstrate that GLYT2 is associated with membrane rafts in the plasma membrane of brainstem terminals and neurons. The transporter is localized to Triton X-100-insoluble light synaptosomal membranes together with flotillin-1, a marker protein for membrane rafts, in a methyl-beta-cyclodextrin (MbetaCD)-sensitive manner. In brainstem primary neurons, the GLYT2 punctuate pattern visualized by confocal microscopy was modified by cholesterol depletion with MbetaCD, unlike other non-raft neuronal markers. GLYT2-associated gold particles were observed by electron microscopy on purified rafts from brainstem synaptosomes. Furthermore, either in brainstem terminals and cultured neurons, the pharmacological reduction of the levels of raft components, cholesterol and sphingomyelin, impairs both the association of GLYT2 with membrane rafts and its transport activity. Thus, GLYT2 may require membrane raft location for optimal function, and therefore the lipid environment may constitute a new mechanism to modulate GLYT2.
Collapse
Affiliation(s)
- Enrique Núñez
- Departamento de Biología Molecular, Consejo Superior de Investigaciones Científicas-Universidad Autónoma de Madrid, Madrid, Spain
| | | | | | | | | |
Collapse
|
43
|
Latal AT, Kremer T, Gomeza J, Eulenburg V, Hülsmann S. Development of synaptic inhibition in glycine transporter 2 deficient mice. Mol Cell Neurosci 2010; 44:342-52. [PMID: 20447457 DOI: 10.1016/j.mcn.2010.04.005] [Citation(s) in RCA: 21] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/09/2010] [Revised: 04/19/2010] [Accepted: 04/23/2010] [Indexed: 11/20/2022] Open
Abstract
Mice deficient for the neuronal glycine transporter subtype 2 (GlyT2) die during the second postnatal week after developing neuromotor deficiencies, which resembles severe forms of human hyperekplexia. This phenotype has been attributed to a dramatic reduction in glycinergic neurotransmission. In the present study we analyzed the development of GABAergic and glycinergic synaptic transmission in GlyT2-knockout mice during early postnatal life. Anti-glycine immunohistochemistry in spinal cord and brainstem slices and whole-cell voltage-clamp recordings of glycinergic inhibitory postsynaptic currents (IPSCs) from hypoglossal motoneurons revealed strikingly reduced levels of synaptic glycine already at birth. Since GABA and glycine use the same vesicular inhibitory amino acid transporter (VIAAT or VGAT) we also analysed GABAergic neurotransmission. No increase of GABA immunoreactivity was observed in the spinal cord and brainstem of GlyT2(-/-) mice at any stage of postnatal development. Correspondingly no up-regulation of GABAergic IPSCs was detected in GlyT2(-/-) hypoglossal motoneurons. These data suggest that in the first postnatal week, loss of the glycine transporter 2 is neither compensated by glycine de-novo synthesis nor by up-regulation of the GABAergic transmission in GlyT2(-/-) mice.
Collapse
Affiliation(s)
- A Tobias Latal
- Dept. Neuro- and Sensory Physiology, Center Physiology and Pathophysiology, Georg-August-University Göttingen, Humboldtallee 23, D-37073 Göttingen, Germany
| | | | | | | | | |
Collapse
|
44
|
Chalphin AV, Saha MS. The specification of glycinergic neurons and the role of glycinergic transmission in development. Front Mol Neurosci 2010; 3:11. [PMID: 20461146 PMCID: PMC2866564 DOI: 10.3389/fnmol.2010.00011] [Citation(s) in RCA: 13] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/01/2010] [Accepted: 03/23/2010] [Indexed: 12/16/2022] Open
Abstract
Glycine's role as an inhibitory neurotransmitter in the adult vertebrate nervous system has been well characterized in a number of different model organisms. However, a full understanding of glycinergic transmission requires a knowledge of how glycinergic synapses emerge and the role of glycinergic signaling during development. Recent literature has provided a detailed picture of the developmental expression of many of the molecular components that comprise the glycinergic phenotype, namely the glycine transporters and the glycine receptor subunits; the transcriptional networks leading to the expression of this important neurotransmitter phenotype are also being elucidated. An equally important focus of research has revealed the critical role of glycinergic signaling in sculpting many different aspects of neural development. This review examines the current literature detailing the expression patterns of the components of the glycinergic phenotype in various vertebrate model organisms over the course of development and the molecular mechanisms governing the expression of the glycinergic phenotype. The review then surveys the recent work on the role of glycinergic signaling in the developing nervous system and concludes with an overview of areas for further research.
Collapse
|
45
|
Davies JS, Chung SK, Thomas RH, Robinson A, Hammond CL, Mullins JGL, Carta E, Pearce BR, Harvey K, Harvey RJ, Rees MI. The glycinergic system in human startle disease: a genetic screening approach. Front Mol Neurosci 2010; 3:8. [PMID: 20407582 PMCID: PMC2854534 DOI: 10.3389/fnmol.2010.00008] [Citation(s) in RCA: 26] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/02/2009] [Accepted: 03/08/2010] [Indexed: 11/17/2022] Open
Abstract
Human startle disease, also known as hyperekplexia (OMIM 149400), is a paroxysmal neurological disorder caused by defects in glycinergic neurotransmission. Hyperekplexia is characterised by an exaggerated startle reflex in response to tactile or acoustic stimuli which first presents as neonatal hypertonia, followed in some with episodes of life-threatening infantile apnoea. Genetic screening studies have demonstrated that hyperekplexia is genetically heterogeneous with several missense and nonsense mutations in the postsynaptic glycine receptor (GlyR) alpha1 subunit gene (GLRA1) as the primary cause. More recently, missense, nonsense and frameshift mutations have also been identified in the glycine transporter GlyT2 gene, SLC6A5, demonstrating a presynaptic component to this disease. Further mutations, albeit rare, have been identified in the genes encoding the GlyR beta subunit (GLRB), collybistin (ARHGEF9) and gephyrin (GPHN) - all of which are postsynaptic proteins involved in orchestrating glycinergic neurotransmission. In this review, we describe the clinical ascertainment aspects, phenotypic considerations and the downstream molecular genetic tools utilised to analyse both presynaptic and postsynaptic components of this heterogeneous human neurological disorder. Moreover, we will describe how the ancient startle response is the preserve of glycinergic neurotransmission and how animal models and human hyperekplexia patients have provided synergistic evidence that implicates this inhibitory system in the control of startle reflexes.
Collapse
Affiliation(s)
- Jeff S Davies
- Institute of Life Science, School of Medicine, Swansea University Singleton Park, Swansea, UK
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|
46
|
Villmann C, Oertel J, Melzer N, Becker CM. Recessive hyperekplexia mutations of the glycine receptor alpha1 subunit affect cell surface integration and stability. J Neurochem 2009; 111:837-47. [PMID: 19732286 DOI: 10.1111/j.1471-4159.2009.06372.x] [Citation(s) in RCA: 29] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/02/2023]
Abstract
The human neurological disorder hyperekplexia is frequently caused by recessive and dominant mutations of the glycine receptor alpha1 subunit gene, GLRA1. Dominant forms are mostly attributed to amino acid substitutions within the ion pore or adjacent loops, resulting in altered channel properties. Here, the biogenesis of glycine receptor alpha1 subunit mutants underlying recessive forms of hyperekplexia was analyzed following recombinant expression in HEK293 cells. The alpha1 mutant S231R resulted in a decrease of surface integrated protein, consistent with reduced maximal current values. Decreased maximal currents shown for the recessive alpha1 mutant I244N were associated with protein instability, rather than decreased surface integration. The recessive mutants R252H and R392H encode exchanges of arginine residues delineating the intracellular faces of transmembrane domains. After expression, the mutant R252H was virtually absent from the cell surface, consistent with non-functionality and the importance of the positive charge for membrane integration. Surface expression of R392H was highly reduced, resulting in residual chloride conductance. Independent of the site of the mutation within the alpha1 polypeptide, metabolic radiolabelling and pulse chase studies revealed a shorter half-life of the full-length alpha1 protein for all recessive mutants as compared to the wild-type. Treatment with the proteasome blocker, lactacystin, significantly increased the accumulation of alpha1 mutants in intracellular membranes. These observations indicated that the recessive alpha1 mutants are recognized by the endoplasmatic reticulum control system, and degraded via the proteasome pathway. Thus, the lack of glycinergic inhibition associated with recessive hyperekplexia may be attributed to sequestration of mutant subunits within the endoplasmatic reticulum quality control system.
Collapse
Affiliation(s)
- Carmen Villmann
- Institut für Biochemie, Emil-Fischer-Zentrum, Universität Erlangen-Nürnberg, Erlangen, Germany
| | | | | | | |
Collapse
|
47
|
Núñez E, Pérez-Siles G, Rodenstein L, Alonso-Torres P, Zafra F, Jiménez E, Aragón C, López-Corcuera B. Subcellular localization of the neuronal glycine transporter GLYT2 in brainstem. Traffic 2009; 10:829-43. [PMID: 19374720 DOI: 10.1111/j.1600-0854.2009.00911.x] [Citation(s) in RCA: 28] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/30/2022]
Abstract
The neuronal glycine transporter GLYT2 belongs to the neurotransmitter:sodium:symporter (NSS) family and removes glycine from the synaptic cleft, thereby aiding the termination of the glycinergic signal and achieving the reloading of the presynaptic terminal. The task fulfilled by this transporter is fine tuned by regulating both transport activity and intracellular trafficking. Different stimuli such as neuronal activity or protein kinase C (PKC) activation can control GLYT2 surface levels although the intracellular compartments where GLYT2 resides are largely unknown. Here, by biochemical and immunological techniques in combination with electron and confocal microscopy, we have investigated the subcellular distribution of GLYT2 in rat brainstem tissue, and characterized the vesicles that contain the transporter. GLYT2 is shown to be present in small and larger vesicles that contain the synaptic vesicle protein synaptophysin, the recycling endosome small GTPase Rab11, and in the larger vesicle population, the vesicular inhibitory amino acid transporter VIAAT. Rab5A, the GABA transporter GAT1, synaptotagmin2 and synaptobrevin2 (VAMP2) were not present. Coexpression of a Rab11 dominant negative mutant with recombinant GLYT2 impaired transporter trafficking and glycine transport. Dual immunogold labeling of brainstem synaptosomes showed a very close proximity of GLYT2 and Rab11. Therefore, the intracellular GLYT2 resides in a subset of endosomal membranes and may traffic around several compartments, mainly Rab11-positive endosomes.
Collapse
Affiliation(s)
- Enrique Núñez
- Departamento de Biología Molecular and Centro de Biología Molecular "Severo Ochoa" (UAM-CSIC). Universidad Autónoma de Madrid, Spain
| | | | | | | | | | | | | | | |
Collapse
|
48
|
Functional complementation of Glra1(spd-ot), a glycine receptor subunit mutant, by independently expressed C-terminal domains. J Neurosci 2009; 29:2440-52. [PMID: 19244519 DOI: 10.1523/jneurosci.4400-08.2009] [Citation(s) in RCA: 25] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2022] Open
Abstract
The oscillator mouse (Glra1(spd-ot)) carries a 9 bp microdeletion plus a 2 bp microinsertion in the glycine receptor alpha1 subunit gene, resulting in the absence of functional alpha1 polypeptides from the CNS and lethality 3 weeks after birth. Depending on differential use of two splice acceptor sites in exon 9 of the Glra1 gene, the mutant allele encodes either a truncated alpha1 subunit (spd(ot)-trc) or a polypeptide with a C-terminal missense sequence (spd(ot)-elg). During recombinant expression, both splice variants fail to form ion channels. In complementation studies, a tail construct, encoding the deleted C-terminal sequence, was coexpressed with both mutants. Coexpression with spd(ot)-trc produced glycine-gated ion channels. Rescue efficiency was increased by inclusion of the wild-type motif RRKRRH. In cultured spinal cord neurons from oscillator homozygotes, viral infection with recombinant C-terminal tail constructs resulted in appearance of endogenous alpha1 antigen. The functional rescue of alpha1 mutants by the C-terminal tail polypeptides argues for a modular subunit architecture of members of the Cys-loop receptor family.
Collapse
|
49
|
|
50
|
Gilfillan R, Kerr J, Walker G, Wishart G. Glycine Transporters and Their Inhibitors. TOPICS IN MEDICINAL CHEMISTRY 2009. [DOI: 10.1007/7355_2009_030] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/09/2023]
|