1
|
Simard JR, Michelsen K, Wang Y, Yang C, Youngblood B, Grubinska B, Taborn K, Gillie DJ, Cook K, Chung K, Long AM, Hall BE, Shaffer PL, Foti RS, Gingras J. Modulation of Ligand-Gated Glycine Receptors Via Functional Monoclonal Antibodies. J Pharmacol Exp Ther 2022; 383:56-69. [PMID: 35926871 DOI: 10.1124/jpet.121.001026] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/17/2021] [Revised: 07/15/2022] [Accepted: 07/19/2022] [Indexed: 12/15/2022] Open
Abstract
Ion channels are targets of considerable therapeutic interest to address a wide variety of neurologic indications, including pain perception. Current pharmacological strategies have focused mostly on small molecule approaches that can be limited by selectivity requirements within members of a channel family or superfamily. Therapeutic antibodies have been proposed, designed, and characterized to alleviate this selectivity limitation; however, there are no Food and Drug Administration-approved therapeutic antibody-based drugs targeting ion channels on the market to date. Here, in an effort to identify novel classes of engineered ion channel modulators for potential neurologic therapeutic applications, we report the generation and characterization of six (EC50 < 25nM) Cys-loop receptor family monoclonal antibodies with modulatory function against rat and human glycine receptor alpha 1 (GlyRα1) and/or GlyRα3. These antibodies have activating (i.e., positive modulator) or inhibiting (i.e., negative modulator) profiles. Moreover, GlyRα3 selectivity was successfully achieved for two of the three positive modulators identified. When dosed intravenously, the antibodies achieved sufficient brain exposure to cover their calculated in vitro EC50 values. When compared head-to-head at identical exposures, the GlyRα3-selective antibody showed a more desirable safety profile over the nonselective antibody, thus demonstrating, for the first time, an advantage for GlyRα3-selectivity. Our data show that ligand-gated ion channels of the glycine receptor family within the central nervous system can be functionally modulated by engineered biologics in a dose-dependent manner and that, despite high protein homology between the alpha subunits, selectivity can be achieved within this receptor family, resulting in future therapeutic candidates with more desirable drug safety profiles. SIGNIFICANCE STATEMENT: This study presents immunization and multiplatform screening approaches to generate a diverse library of functional antibodies (agonist, potentiator, or inhibitory) raised against human glycine receptors (GlyRs). This study also demonstrates the feasibility of acquiring alpha subunit selectivity, a desirable therapeutic profile. When tested in vivo, these tool molecules demonstrated an increased safety profile in favor of GlyRα3-selectivity. These are the first reported functional GlyR antibodies that may open new avenues to treating central nervous system diseases with subunit selective biologics.
Collapse
Affiliation(s)
- Jeffrey R Simard
- Departments of Neuroscience (J.R.S., C.Y., B.Y. B.G., K.T., D.J.G., J.G.), Molecular Engineering (K.M., A.M.L., P.L.S.), Protein Technologies (Y.W., B.E.H.), and Pharmacokinetics and Drug Metabolism (R.S.F.), Amgen Research, Cambridge, Massachusetts; and Department of Pharmacokinetics and Drug Metabolism, Amgen Research, 1120 Veterans Boulevard, South San Francisco, California (K.Co., K.Ch.)
| | - Klaus Michelsen
- Departments of Neuroscience (J.R.S., C.Y., B.Y. B.G., K.T., D.J.G., J.G.), Molecular Engineering (K.M., A.M.L., P.L.S.), Protein Technologies (Y.W., B.E.H.), and Pharmacokinetics and Drug Metabolism (R.S.F.), Amgen Research, Cambridge, Massachusetts; and Department of Pharmacokinetics and Drug Metabolism, Amgen Research, 1120 Veterans Boulevard, South San Francisco, California (K.Co., K.Ch.)
| | - Yan Wang
- Departments of Neuroscience (J.R.S., C.Y., B.Y. B.G., K.T., D.J.G., J.G.), Molecular Engineering (K.M., A.M.L., P.L.S.), Protein Technologies (Y.W., B.E.H.), and Pharmacokinetics and Drug Metabolism (R.S.F.), Amgen Research, Cambridge, Massachusetts; and Department of Pharmacokinetics and Drug Metabolism, Amgen Research, 1120 Veterans Boulevard, South San Francisco, California (K.Co., K.Ch.)
| | - Chunhua Yang
- Departments of Neuroscience (J.R.S., C.Y., B.Y. B.G., K.T., D.J.G., J.G.), Molecular Engineering (K.M., A.M.L., P.L.S.), Protein Technologies (Y.W., B.E.H.), and Pharmacokinetics and Drug Metabolism (R.S.F.), Amgen Research, Cambridge, Massachusetts; and Department of Pharmacokinetics and Drug Metabolism, Amgen Research, 1120 Veterans Boulevard, South San Francisco, California (K.Co., K.Ch.)
| | - Beth Youngblood
- Departments of Neuroscience (J.R.S., C.Y., B.Y. B.G., K.T., D.J.G., J.G.), Molecular Engineering (K.M., A.M.L., P.L.S.), Protein Technologies (Y.W., B.E.H.), and Pharmacokinetics and Drug Metabolism (R.S.F.), Amgen Research, Cambridge, Massachusetts; and Department of Pharmacokinetics and Drug Metabolism, Amgen Research, 1120 Veterans Boulevard, South San Francisco, California (K.Co., K.Ch.)
| | - Barbara Grubinska
- Departments of Neuroscience (J.R.S., C.Y., B.Y. B.G., K.T., D.J.G., J.G.), Molecular Engineering (K.M., A.M.L., P.L.S.), Protein Technologies (Y.W., B.E.H.), and Pharmacokinetics and Drug Metabolism (R.S.F.), Amgen Research, Cambridge, Massachusetts; and Department of Pharmacokinetics and Drug Metabolism, Amgen Research, 1120 Veterans Boulevard, South San Francisco, California (K.Co., K.Ch.)
| | - Kristin Taborn
- Departments of Neuroscience (J.R.S., C.Y., B.Y. B.G., K.T., D.J.G., J.G.), Molecular Engineering (K.M., A.M.L., P.L.S.), Protein Technologies (Y.W., B.E.H.), and Pharmacokinetics and Drug Metabolism (R.S.F.), Amgen Research, Cambridge, Massachusetts; and Department of Pharmacokinetics and Drug Metabolism, Amgen Research, 1120 Veterans Boulevard, South San Francisco, California (K.Co., K.Ch.)
| | - Daniel J Gillie
- Departments of Neuroscience (J.R.S., C.Y., B.Y. B.G., K.T., D.J.G., J.G.), Molecular Engineering (K.M., A.M.L., P.L.S.), Protein Technologies (Y.W., B.E.H.), and Pharmacokinetics and Drug Metabolism (R.S.F.), Amgen Research, Cambridge, Massachusetts; and Department of Pharmacokinetics and Drug Metabolism, Amgen Research, 1120 Veterans Boulevard, South San Francisco, California (K.Co., K.Ch.)
| | - Kevin Cook
- Departments of Neuroscience (J.R.S., C.Y., B.Y. B.G., K.T., D.J.G., J.G.), Molecular Engineering (K.M., A.M.L., P.L.S.), Protein Technologies (Y.W., B.E.H.), and Pharmacokinetics and Drug Metabolism (R.S.F.), Amgen Research, Cambridge, Massachusetts; and Department of Pharmacokinetics and Drug Metabolism, Amgen Research, 1120 Veterans Boulevard, South San Francisco, California (K.Co., K.Ch.)
| | - Kyu Chung
- Departments of Neuroscience (J.R.S., C.Y., B.Y. B.G., K.T., D.J.G., J.G.), Molecular Engineering (K.M., A.M.L., P.L.S.), Protein Technologies (Y.W., B.E.H.), and Pharmacokinetics and Drug Metabolism (R.S.F.), Amgen Research, Cambridge, Massachusetts; and Department of Pharmacokinetics and Drug Metabolism, Amgen Research, 1120 Veterans Boulevard, South San Francisco, California (K.Co., K.Ch.)
| | - Alexander M Long
- Departments of Neuroscience (J.R.S., C.Y., B.Y. B.G., K.T., D.J.G., J.G.), Molecular Engineering (K.M., A.M.L., P.L.S.), Protein Technologies (Y.W., B.E.H.), and Pharmacokinetics and Drug Metabolism (R.S.F.), Amgen Research, Cambridge, Massachusetts; and Department of Pharmacokinetics and Drug Metabolism, Amgen Research, 1120 Veterans Boulevard, South San Francisco, California (K.Co., K.Ch.)
| | - Brian E Hall
- Departments of Neuroscience (J.R.S., C.Y., B.Y. B.G., K.T., D.J.G., J.G.), Molecular Engineering (K.M., A.M.L., P.L.S.), Protein Technologies (Y.W., B.E.H.), and Pharmacokinetics and Drug Metabolism (R.S.F.), Amgen Research, Cambridge, Massachusetts; and Department of Pharmacokinetics and Drug Metabolism, Amgen Research, 1120 Veterans Boulevard, South San Francisco, California (K.Co., K.Ch.)
| | - Paul L Shaffer
- Departments of Neuroscience (J.R.S., C.Y., B.Y. B.G., K.T., D.J.G., J.G.), Molecular Engineering (K.M., A.M.L., P.L.S.), Protein Technologies (Y.W., B.E.H.), and Pharmacokinetics and Drug Metabolism (R.S.F.), Amgen Research, Cambridge, Massachusetts; and Department of Pharmacokinetics and Drug Metabolism, Amgen Research, 1120 Veterans Boulevard, South San Francisco, California (K.Co., K.Ch.)
| | - Robert S Foti
- Departments of Neuroscience (J.R.S., C.Y., B.Y. B.G., K.T., D.J.G., J.G.), Molecular Engineering (K.M., A.M.L., P.L.S.), Protein Technologies (Y.W., B.E.H.), and Pharmacokinetics and Drug Metabolism (R.S.F.), Amgen Research, Cambridge, Massachusetts; and Department of Pharmacokinetics and Drug Metabolism, Amgen Research, 1120 Veterans Boulevard, South San Francisco, California (K.Co., K.Ch.)
| | - Jacinthe Gingras
- Departments of Neuroscience (J.R.S., C.Y., B.Y. B.G., K.T., D.J.G., J.G.), Molecular Engineering (K.M., A.M.L., P.L.S.), Protein Technologies (Y.W., B.E.H.), and Pharmacokinetics and Drug Metabolism (R.S.F.), Amgen Research, Cambridge, Massachusetts; and Department of Pharmacokinetics and Drug Metabolism, Amgen Research, 1120 Veterans Boulevard, South San Francisco, California (K.Co., K.Ch.)
| |
Collapse
|
2
|
Subunit-Specific Photocontrol of Glycine Receptors by Azobenzene-Nitrazepam Photoswitcher. eNeuro 2021; 8:ENEURO.0294-20.2020. [PMID: 33298457 PMCID: PMC7877471 DOI: 10.1523/eneuro.0294-20.2020] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/03/2020] [Revised: 11/04/2020] [Accepted: 11/23/2020] [Indexed: 12/24/2022] Open
Abstract
Photopharmacology is a unique approach that through a combination of photochemistry methods and advanced life science techniques allows the study and control of specific biological processes, ranging from intracellular pathways to brain circuits. Recently, a first photochromic channel blocker of anion-selective GABAA receptors, the azobenzene-nitrazepam-based photochromic compound (Azo-NZ1), has been described. In the present study, using patch-clamp technique in heterologous system and in mice brain slices, site-directed mutagenesis and molecular modeling we provide evidence of the interaction of Azo-NZ1 with glycine receptors (GlyRs) and determine the molecular basis of this interaction. Glycinergic synaptic neurotransmission determines an important inhibitory drive in the vertebrate nervous system and plays a crucial role in the control of neuronal circuits in the spinal cord and brain stem. GlyRs are involved in locomotion, pain sensation, breathing, and auditory function, as well as in the development of such disorders as hyperekplexia, epilepsy, and autism. Here, we demonstrate that Azo-NZ1 blocks in a UV-dependent manner the activity of α2 GlyRs (GlyR2), while being barely active on α1 GlyRs (GlyR1). The site of Azo-NZ1 action is in the chloride-selective pore of GlyR at the 2’ position of transmembrane helix 2 and amino acids forming this site determine the difference in Azo-NZ1 blocking activity between GlyR2 and GlyR1. This subunit-specific modulation is also shown on motoneurons of brainstem slices from neonatal mice that switch during development from expressing “fetal” GlyR2 to “adult” GlyR1 receptors.
Collapse
|
3
|
Gomila AMJ, Rustler K, Maleeva G, Nin-Hill A, Wutz D, Bautista-Barrufet A, Rovira X, Bosch M, Mukhametova E, Petukhova E, Ponomareva D, Mukhamedyarov M, Peiretti F, Alfonso-Prieto M, Rovira C, König B, Bregestovski P, Gorostiza P. Photocontrol of Endogenous Glycine Receptors In Vivo. Cell Chem Biol 2020; 27:1425-1433.e7. [PMID: 32846115 DOI: 10.1016/j.chembiol.2020.08.005] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/21/2019] [Revised: 04/14/2020] [Accepted: 08/04/2020] [Indexed: 12/15/2022]
Abstract
Glycine receptors (GlyRs) are indispensable for maintaining excitatory/inhibitory balance in neuronal circuits that control reflexes and rhythmic motor behaviors. Here we have developed Glyght, a GlyR ligand controlled with light. It is selective over other Cys-loop receptors, is active in vivo, and displays an allosteric mechanism of action. The photomanipulation of glycinergic neurotransmission opens new avenues to understanding inhibitory circuits in intact animals and to developing drug-based phototherapies.
Collapse
Affiliation(s)
- Alexandre M J Gomila
- Institute for Bioengineering of Catalonia (IBEC), The Barcelona Institute of Science and Technology (BIST), Barcelona 08028, Spain
| | - Karin Rustler
- University of Regensburg, Institute of Organic Chemistry, Regensburg 93053, Germany
| | - Galyna Maleeva
- Institute for Bioengineering of Catalonia (IBEC), The Barcelona Institute of Science and Technology (BIST), Barcelona 08028, Spain; Aix-Marseille Université, INSERM, INS, Institut de Neurosciences des Systèmes, Marseille 13005, France
| | - Alba Nin-Hill
- University of Barcelona, Department of Inorganic and Organic Chemistry, Institute of Theoretical Chemistry (IQTCUB), Barcelona 08028, Spain
| | - Daniel Wutz
- University of Regensburg, Institute of Organic Chemistry, Regensburg 93053, Germany
| | - Antoni Bautista-Barrufet
- Institute for Bioengineering of Catalonia (IBEC), The Barcelona Institute of Science and Technology (BIST), Barcelona 08028, Spain
| | - Xavier Rovira
- Institute for Bioengineering of Catalonia (IBEC), The Barcelona Institute of Science and Technology (BIST), Barcelona 08028, Spain
| | - Miquel Bosch
- Institute for Bioengineering of Catalonia (IBEC), The Barcelona Institute of Science and Technology (BIST), Barcelona 08028, Spain
| | - Elvira Mukhametova
- Aix-Marseille Université, INSERM, INS, Institut de Neurosciences des Systèmes, Marseille 13005, France; Kazan Federal University, Open Lab of Motor Neurorehabilitation, Kazan, Russia
| | - Elena Petukhova
- Institute of Neurosciences, Kazan State Medical University, Kazan, Russia
| | - Daria Ponomareva
- Institute of Neurosciences, Kazan State Medical University, Kazan, Russia
| | | | - Franck Peiretti
- Aix Marseille Université, INSERM 1263, INRA 1260, C2VN, Marseille, France
| | - Mercedes Alfonso-Prieto
- Institute for Advanced Simulation IAS-5 and Institute of Neuroscience and Medicine INM-9, Computational Biomedicine, Forschungszentrum Jülich, 52425 Jülich, Germany; Cécile and Oskar Vogt Institute for Brain Research, Medical Faculty, Heinrich Heine University Düsseldorf, 40225 Düsseldorf, Germany
| | - Carme Rovira
- University of Barcelona, Department of Inorganic and Organic Chemistry, Institute of Theoretical Chemistry (IQTCUB), Barcelona 08028, Spain; Catalan Institution for Research and Advanced Studies (ICREA), Barcelona 08003 Spain.
| | - Burkhard König
- University of Regensburg, Institute of Organic Chemistry, Regensburg 93053, Germany.
| | - Piotr Bregestovski
- Aix-Marseille Université, INSERM, INS, Institut de Neurosciences des Systèmes, Marseille 13005, France; Institute of Neurosciences, Kazan State Medical University, Kazan, Russia.
| | - Pau Gorostiza
- Institute for Bioengineering of Catalonia (IBEC), The Barcelona Institute of Science and Technology (BIST), Barcelona 08028, Spain; Catalan Institution for Research and Advanced Studies (ICREA), Barcelona 08003 Spain; CIBER-BBN, Madrid 28001 Spain.
| |
Collapse
|
4
|
Arias HR, Do Rego JL, Do Rego JC, Chen Z, Anouar Y, Scholze P, Gonzales EB, Huang R, Chagraoui A. Coronaridine congeners potentiate GABA A receptors and induce sedative activity in mice in a benzodiazepine-insensitive manner. Prog Neuropsychopharmacol Biol Psychiatry 2020; 101:109930. [PMID: 32194202 DOI: 10.1016/j.pnpbp.2020.109930] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/30/2019] [Revised: 03/13/2020] [Accepted: 03/15/2020] [Indexed: 12/23/2022]
Abstract
To determine whether (+)-catharanthine induces sedative- or anxiolytic/anxiogenic-like activity in male mice, proper animal paradigms were used. The results showed that (+)-catharanthine induces sedative-like activity in the 63-72 mg/Kg dose range in a flumazenil-insensitive manner, but neither this effect nor anxiolytic/anxiogenic-like activity was observed at lower doses. To determine the underlying molecular mechanism of the sedative-like activity, electrophysiological and radioligand binding experiments were performed with (+)-catharanthine and (±)-18-methoxycoronaridine [(±)-18-MC] on GABAA (GABAARs) and glycine receptors (GlyRs). Coronaridine congeners both activated and potentiated a variety of human (h) GABAARs, except hρ1. (+)-Catharanthine-induced potentiation followed this receptor selectivity (EC50's in μM): hα1β2 (4.6 ± 0.8) > hα2β2γ2 (12.6 ± 3.8) ~ hα1β2γ2 (14.4 ± 4.6) indicating that both α1 and α2 are equally important, whereas γ2 is not necessary. (+)-Catharanthine was >2-fold more potent and efficient than (±)-18-MC at hα1β2γ2. (+)-Catharanthine also potentiated, whereas (±)-18-MC inhibited, hα1 GlyRs with very low potency. Additional [3H]-flunitrazepam competition binding experiments using rat cerebellum membranes clearly demonstrated that these ligands do not bind to the benzodiazepine site. This is supported by the observed activity at hα1β2 (lacking the BDZ site) and similar effects between α1- and α2-containing GABAARs. Our study shows, for the first time, that (+)-catharanthine induced sedative-like effects in mice, and coronaridine congeners potentiated human α1β2γ2, α1β2, and hα2β2γ2, but not ρ1, GABAARs, both in a benzodiazepine-insensitive fashion, whereas only (+)-catharanthine slightly potentiated GlyRs.
Collapse
Affiliation(s)
- Hugo R Arias
- Department of Pharmacology and Physiology, Oklahoma State University College of Osteopathic Medicine, Tahlequah, OK, USA.
| | - Jean Luc Do Rego
- Behavioral Analysis Platform SCAC, Institute for Research and Innovation in Biomedicine (IRIB), University of Rouen Normandy, Rouen, France
| | - Jean Claude Do Rego
- Behavioral Analysis Platform SCAC, Institute for Research and Innovation in Biomedicine (IRIB), University of Rouen Normandy, Rouen, France
| | - Zhenglan Chen
- Department of Pharmacology and Neuroscience, Institute for Health Aging, University of North Texas Health Science Center at Fort Worth, TX, USA
| | - Youssef Anouar
- Laboratory of Neuronal and Neuroendocrine Differentiation and Communication, Normandie University, UNIROUEN, INSERM U1239, Institute for Research and Innovation in Biomedicine of Normandy (IRIB) Rouen, France
| | - Petra Scholze
- Department of Pathobiology of the Nervous System, Center for Brain Research, Medical University of Vienna, Vienna, Austria
| | - Eric B Gonzales
- Department of Pharmacology and Neuroscience, Institute for Health Aging, University of North Texas Health Science Center at Fort Worth, TX, USA
| | - Renqi Huang
- Department of Pharmacology and Neuroscience, Institute for Health Aging, University of North Texas Health Science Center at Fort Worth, TX, USA
| | - Abdeslam Chagraoui
- Laboratory of Neuronal and Neuroendocrine Differentiation and Communication, Normandie University, UNIROUEN, INSERM U1239, Institute for Research and Innovation in Biomedicine of Normandy (IRIB) Rouen, France; Department of Medical Biochemistry, Rouen University Hospital, CHU de Rouen, France.
| |
Collapse
|
5
|
Abstract
The inhibitory glycine receptor is a member of the Cys-loop superfamily of ligand-gated ion channels. It is the principal mediator of rapid synaptic inhibition in the spinal cord and brainstem and plays an important role in the modulation of higher brain functions including vision, hearing, and pain signaling. Glycine receptor function is controlled by only a few agonists, while the number of antagonists and positive or biphasic modulators is steadily increasing. These modulators are important for the study of receptor activation and regulation and have found clinical interest as potential analgesics and anticonvulsants. High-resolution structures of the receptor have become available recently, adding to our understanding of structure-function relationships and revealing agonistic, inhibitory, and modulatory sites on the receptor protein. This Review presents an overview of compounds that activate, inhibit, or modulate glycine receptor function in vitro and in vivo.
Collapse
Affiliation(s)
- Ulrike Breitinger
- Department of Biochemistry, German University in Cairo, New Cairo 11835, Egypt
| | | |
Collapse
|
6
|
Hegazy NH, Breitinger HG, Breitinger U. Kavalactones from Kava (Piper methysticum) root extract as modulators of recombinant human glycine receptors. Biol Chem 2020; 400:1205-1215. [PMID: 31141476 DOI: 10.1515/hsz-2019-0112] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/15/2019] [Accepted: 05/17/2019] [Indexed: 02/07/2023]
Abstract
Roots of kava (Piper methysticum) plant are used in almost all Pacific Ocean cultures to prepare a drink with sedative, anesthetic and euphoric properties. One of the main active ingredients of the extract are kava lactones. Here, kava root CO2 extract and three kavalactones, DL-kavain, dihydrokavain and yangonin (isolated from whole extract by column chromatography) were tested for their inhibitory action on recombinant homomeric human α1 glycine receptors expressed in HEK293 cells. Kava CO2 root extract, as well as the individual components DL-kavain, dihydrokavain and yangonin inhibited glycine receptor activity in a dose-dependent manner. DL-kavain was the most potent inhibitor (IC50 = 0.077 ± 0.002 mm), followed by yangonin (IC50 = 0.31 ± 0.04 mm) and dihydrokavain (IC50 = 3.23 ± 0.10 mm) which were 4- and 40-fold less active than DL-kavain, respectively. Application of kava root extract did not reduce maximum currents, but increased EC50 of glycine. Simultaneous application of kava extract and strychnine showed additive inhibition, suggesting that binding of kavalactones and strychnine on the receptor is mutually exclusive. Overall, kavalactones exert a moderate inhibitory effect on the human α1 glycine receptor with DL-kavain being the most potent constituent.
Collapse
Affiliation(s)
- Nada Hany Hegazy
- Department of Biochemistry, German University in Cairo, Main Entrance of Al Tagamoa Al Khames, New Cairo 11835, Egypt
| | - Hans-Georg Breitinger
- Department of Biochemistry, German University in Cairo, Main Entrance of Al Tagamoa Al Khames, New Cairo 11835, Egypt
| | - Ulrike Breitinger
- Department of Biochemistry, German University in Cairo, Main Entrance of Al Tagamoa Al Khames, New Cairo 11835, Egypt
| |
Collapse
|
7
|
Ginkgolic acid promotes autophagy-dependent clearance of intracellular alpha-synuclein aggregates. Mol Cell Neurosci 2019; 101:103416. [PMID: 31654699 DOI: 10.1016/j.mcn.2019.103416] [Citation(s) in RCA: 27] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/21/2019] [Revised: 10/18/2019] [Accepted: 10/21/2019] [Indexed: 12/31/2022] Open
Abstract
The accumulation of intracytoplasmic inclusion bodies (Lewy bodies) composed of aggregates of the alpha-synuclein (α-syn) protein is the principal pathological characteristic of Parkinson's disease (PD) and may lead to degeneration of dopaminergic neurons. To date there is no medication that can promote the efficient clearance of these pathological aggregates. In this study, the effect on α-syn aggregate clearance of ginkgolic acid (GA), a natural compound extracted from Ginkgo biloba leaves that inhibits SUMOylation amongst other pathways, was assessed in SH-SY5Y neuroblastoma cells and rat primary cortical neurons. Depolarization of SH-SY5Y neuroblastoma cells and rat primary cortical neurons with KCl was used to induce α-syn aggregate formation. Cells pre-treated with either GA or the related compound, anacardic acid, revealed a significant decrease in intracytoplasmic aggregates immunopositive for α-syn and SUMO-1. An increased frequency of autophagosomes was also detected with both compounds. GA post-treatment 24 h after depolarization also significantly diminished α-syn aggregate bearing cells, indicating the clearance of pre-formed aggregates. Autophagy inhibitors blocked GA-dependent clearance of α-syn aggregates, but not increased autophagosome frequency. Western analysis revealed that the reduction in α-syn aggregate frequency obtained with GA pre-treatment was accompanied by little change in the abundance of SUMO conjugates. The current findings show that GA can promote autophagy-dependent clearance of α-syn aggregates and may have potential in disease modifying therapy.
Collapse
|
8
|
Sparling BA, DiMauro EF. Progress in the discovery of small molecule modulators of the Cys-loop superfamily receptors. Bioorg Med Chem Lett 2017; 27:3207-3218. [DOI: 10.1016/j.bmcl.2017.04.073] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/18/2017] [Revised: 04/20/2017] [Accepted: 04/24/2017] [Indexed: 12/11/2022]
|
9
|
Maleeva G, Peiretti F, Zhorov BS, Bregestovski P. Voltage-Dependent Inhibition of Glycine Receptor Channels by Niflumic Acid. Front Mol Neurosci 2017; 10:125. [PMID: 28559795 PMCID: PMC5432571 DOI: 10.3389/fnmol.2017.00125] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/18/2017] [Accepted: 04/12/2017] [Indexed: 01/11/2023] Open
Abstract
Niflumic acid (NFA) is a member of the fenamate class of nonsteroidal anti-inflammatory drugs. This compound and its derivatives are used worldwide clinically for the relief of chronic and acute pain. NFA is also a commonly used blocker of voltage-gated chloride channels. Here we present evidence that NFA is an efficient blocker of chloride-permeable glycine receptors (GlyRs) with subunit heterogeneity of action. Using the whole-cell configuration of patch-clamp recordings and molecular modeling, we analyzed the action of NFA on homomeric α1ΔIns, α2B, α3L, and heteromeric α1β and α2β GlyRs expressed in CHO cells. NFA inhibited glycine-induced currents in a voltage-dependent manner and its blocking potency in α2 and α3 GlyRs was higher than that in α1 GlyR. The Woodhull analysis suggests that NFA blocks α1 and α2 GlyRs at the fractional electrical distances of 0.16 and 0.65 from the external membrane surface, respectively. Thus, NFA binding site in α1 GlyR is closer to the external part of the membrane, while in α2 GlyR it is significantly deeper in the pore. Mutation G254A at the cytoplasmic part of the α1 GlyR pore-lining TM2 helix (level 2') increased the NFA blocking potency, while incorporation of the β subunit did not have a significant effect. The Hill plot analysis suggests that α1 and α2 GlyRs are preferably blocked by two and one NFA molecules, respectively. Molecular modeling using Monte Carlo energy minimizations provides the structural rationale for the experimental data and proposes more than one interaction site along the pore where NFA can suppress the ion permeation.
Collapse
Affiliation(s)
- Galyna Maleeva
- INSERM, INS, Institut de Neurosciences des Systèmes, Aix-Marseille UniversityMarseille, France.,Department of Cytology, Bogomoletz Institute of PhysiologyKyiv, Ukraine
| | - Franck Peiretti
- INSERM 1062, INRA 1260, NORT, Aix-Marseille UniversityMarseille, France
| | - Boris S Zhorov
- Sechenov Institute of Evolutionary Physiology and Biochemistry, Russian Academy of SciencesSt. Petersburg, Russia.,Department of Biochemistry and Biomedical Sciences, McMaster UniversityHamilton, ON, Canada
| | - Piotr Bregestovski
- INSERM, INS, Institut de Neurosciences des Systèmes, Aix-Marseille UniversityMarseille, France.,Department of Physiology, Kazan State Medical UniversityKazan, Russia
| |
Collapse
|
10
|
Chakka N, Andrews KL, Berry LM, Bregman H, Gunaydin H, Huang L, Guzman-Perez A, Plant MH, Simard JR, Gingras J, DiMauro EF. Applications of parallel synthetic lead hopping and pharmacophore-based virtual screening in the discovery of efficient glycine receptor potentiators. Eur J Med Chem 2017; 137:63-75. [PMID: 28575722 DOI: 10.1016/j.ejmech.2017.05.036] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/23/2017] [Revised: 05/09/2017] [Accepted: 05/11/2017] [Indexed: 12/24/2022]
Abstract
Glycine receptors (GlyRs) are pentameric glycine-gated chloride ion channels that are enriched in the brainstem and spinal cord where they have been demonstrated to play a role in central nervous system (CNS) inhibition. Herein we describe two novel classes of glycine receptor potentiators that have been developed using similarity- and property-guided scaffold hopping enabled by parallel synthesis and pharmacophore-based virtual screening strategies. This effort resulted in the identification of novel, efficient and modular leads having favorable in vitro ADME profiles and high CNS multi-parameter optimization (MPO) scores, exemplified by azetidine sulfonamide 19 and aminothiazole sulfone (ent2)-20.
Collapse
Affiliation(s)
- Nagasree Chakka
- Department of Medicinal Chemistry, Amgen Inc., 360 Binney Street, Cambridge, MA 02142, USA
| | - Kristin L Andrews
- Department of Molecular Engineering, Amgen Inc., 360 Binney Street, Cambridge, MA 02142, USA
| | - Loren M Berry
- Department of Pharmacokinetics, Amgen Inc., 360 Binney Street, Cambridge, MA 02142, USA
| | - Howard Bregman
- Department of Medicinal Chemistry, Amgen Inc., 360 Binney Street, Cambridge, MA 02142, USA
| | - Hakan Gunaydin
- Department of Molecular Engineering, Amgen Inc., 360 Binney Street, Cambridge, MA 02142, USA
| | - Liyue Huang
- Department of Pharmacokinetics, Amgen Inc., 360 Binney Street, Cambridge, MA 02142, USA
| | - Angel Guzman-Perez
- Department of Medicinal Chemistry, Amgen Inc., 360 Binney Street, Cambridge, MA 02142, USA
| | - Matthew H Plant
- Department of Discovery Attribute Sciences, Amgen Inc., 360 Binney Street, Cambridge, MA 02142, USA
| | - Jeffrey R Simard
- Department of Neuroscience, Amgen Inc., 360 Binney Street, Cambridge, MA 02142, USA
| | - Jacinthe Gingras
- Department of Neuroscience, Amgen Inc., 360 Binney Street, Cambridge, MA 02142, USA
| | - Erin F DiMauro
- Department of Medicinal Chemistry, Amgen Inc., 360 Binney Street, Cambridge, MA 02142, USA.
| |
Collapse
|
11
|
Lynch JW, Zhang Y, Talwar S, Estrada-Mondragon A. Glycine Receptor Drug Discovery. ADVANCES IN PHARMACOLOGY 2017; 79:225-253. [DOI: 10.1016/bs.apha.2017.01.003] [Citation(s) in RCA: 49] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
|
12
|
Bregman H, Simard JR, Andrews KL, Ayube S, Chen H, Gunaydin H, Guzman-Perez A, Hu J, Huang L, Huang X, Krolikowski PH, Lehto SG, Lewis RT, Michelsen K, Pegman P, Plant MH, Shaffer PL, Teffera Y, Yi S, Zhang M, Gingras J, DiMauro EF. The Discovery and Hit-to-Lead Optimization of Tricyclic Sulfonamides as Potent and Efficacious Potentiators of Glycine Receptors. J Med Chem 2016; 60:1105-1125. [PMID: 28001399 DOI: 10.1021/acs.jmedchem.6b01496] [Citation(s) in RCA: 27] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
Current pain therapeutics suffer from undesirable psychotropic and sedative side effects, as well as abuse potential. Glycine receptors (GlyRs) are inhibitory ligand-gated ion channels expressed in nerves of the spinal dorsal horn, where their activation is believed to reduce transmission of painful stimuli. Herein, we describe the identification and hit-to-lead optimization of a novel class of tricyclic sulfonamides as allosteric GlyR potentiators. Initial optimization of high-throughput screening (HTS) hit 1 led to the identification of 3, which demonstrated ex vivo potentiation of glycine-activated current in mouse dorsal horn neurons from spinal cord slices. Further improvement of potency and pharmacokinetics produced in vivo proof-of-concept tool molecule 20 (AM-1488), which reversed tactile allodynia in a mouse spared-nerve injury (SNI) model. Additional structural optimization provided highly potent potentiator 32 (AM-3607), which was cocrystallized with human GlyRα3cryst to afford the first described potentiator-bound X-ray cocrystal structure within this class of ligand-gated ion channels (LGICs).
Collapse
Affiliation(s)
| | - Jeffrey R Simard
- Department of Neuroscience, Amgen Inc. , One Amgen Center Drive, Thousand Oaks, California 91320, United States
| | | | | | | | | | | | | | | | | | | | - Sonya G Lehto
- Department of Neuroscience, Amgen Inc. , One Amgen Center Drive, Thousand Oaks, California 91320, United States
| | | | | | | | | | | | | | | | - Maosheng Zhang
- Department of Neuroscience, Amgen Inc. , One Amgen Center Drive, Thousand Oaks, California 91320, United States
| | | | | |
Collapse
|
13
|
Crystal structures of human glycine receptor α3 bound to a novel class of analgesic potentiators. Nat Struct Mol Biol 2016; 24:108-113. [PMID: 27991902 DOI: 10.1038/nsmb.3329] [Citation(s) in RCA: 95] [Impact Index Per Article: 11.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/15/2016] [Accepted: 10/27/2016] [Indexed: 01/19/2023]
Abstract
Current therapies to treat persistent pain and neuropathic pain are limited by poor efficacy, side effects and risk of addiction. Here, we present a novel class of potent selective, central nervous system (CNS)-penetrant potentiators of glycine receptors (GlyRs), ligand-gated ion channels expressed in the CNS. AM-1488 increased the response to exogenous glycine in mouse spinal cord and significantly reversed mechanical allodynia induced by nerve injury in a mouse model of neuropathic pain. We obtained an X-ray crystal structure of human homopentameric GlyRα3 in complex with AM-3607, a potentiator of the same class with increased potency, and the agonist glycine, at 2.6-Å resolution. AM-3607 binds a novel allosteric site between subunits, which is adjacent to the orthosteric site where glycine binds. Our results provide new insights into the potentiation of cysteine-loop receptors by positive allosteric modulators and hold promise in structure-based design of GlyR modulators for the treatment of neuropathic pain.
Collapse
|
14
|
Ogino K, Hirata H. Defects of the Glycinergic Synapse in Zebrafish. Front Mol Neurosci 2016; 9:50. [PMID: 27445686 PMCID: PMC4925712 DOI: 10.3389/fnmol.2016.00050] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/31/2016] [Accepted: 06/13/2016] [Indexed: 12/26/2022] Open
Abstract
Glycine mediates fast inhibitory synaptic transmission. Physiological importance of the glycinergic synapse is well established in the brainstem and the spinal cord. In humans, the loss of glycinergic function in the spinal cord and brainstem leads to hyperekplexia, which is characterized by an excess startle reflex to sudden acoustic or tactile stimulation. In addition, glycinergic synapses in this region are also involved in the regulation of respiration and locomotion, and in the nociceptive processing. The importance of the glycinergic synapse is conserved across vertebrate species. A teleost fish, the zebrafish, offers several advantages as a vertebrate model for research of glycinergic synapse. Mutagenesis screens in zebrafish have isolated two motor defective mutants that have pathogenic mutations in glycinergic synaptic transmission: bandoneon (beo) and shocked (sho). Beo mutants have a loss-of-function mutation of glycine receptor (GlyR) β-subunit b, alternatively, sho mutant is a glycinergic transporter 1 (GlyT1) defective mutant. These mutants are useful animal models for understanding of glycinergic synaptic transmission and for identification of novel therapeutic agents for human diseases arising from defect in glycinergic transmission, such as hyperekplexia or glycine encephalopathy. Recent advances in techniques for genome editing and for imaging and manipulating of a molecule or a physiological process make zebrafish more attractive model. In this review, we describe the glycinergic defective zebrafish mutants and the technical advances in both forward and reverse genetic approaches as well as in vivo visualization and manipulation approaches for the study of the glycinergic synapse in zebrafish.
Collapse
Affiliation(s)
- Kazutoyo Ogino
- Department of Chemistry and Biological Science, College of Science and Engineering, Aoyama Gakuin University Sagamihara, Japan
| | - Hiromi Hirata
- Department of Chemistry and Biological Science, College of Science and Engineering, Aoyama Gakuin University Sagamihara, Japan
| |
Collapse
|