1
|
Jin F, Lin YY, Wang RC, Xie TX, Zhao Y, Shen C, Sheng D, Ichikawa M, Yu Y, Wang J, Hattori M. Cryo-EM structure of the zinc-activated channel (ZAC) in the Cys-loop receptor superfamily. Proc Natl Acad Sci U S A 2024; 121:e2405659121. [PMID: 39441630 DOI: 10.1073/pnas.2405659121] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/20/2024] [Accepted: 08/28/2024] [Indexed: 10/25/2024] Open
Abstract
Cys-loop receptors are a large superfamily of pentameric ligand-gated ion channels with various physiological roles, especially in neurotransmission in the central nervous system. Among them, zinc-activated channel (ZAC) is a Zn2+-activated ion channel that is widely expressed in the human body and is conserved among eukaryotes. Due to its gating by extracellular Zn2+, ZAC has been considered a Zn2+ sensor, but it has undergone minimal structural and functional characterization since its molecular cloning. Among the families in the Cys-loop receptor superfamily, only the structure of ZAC has yet to be determined. Here, we determined the cryo-EM structure of ZAC in the apo state and performed structure-based mutation analyses. We identified a few residues in the extracellular domain whose mutations had a mild impact on Zn2+ sensitivity. The constriction site in the ion-conducting pore differs from the one in other Cys-loop receptor structures, and further mutational analysis identified a key residue that is important for ion selectivity. In summary, our work provides a structural framework for understanding the ion-conducting mechanism of ZAC.
Collapse
Affiliation(s)
- Fei Jin
- State Key Laboratory of Genetic Engineering, Shanghai Key Laboratory of Bioactive Small Molecules, Collaborative Innovation Center of Genetics and Development, Department of Physiology and Neurobiology, School of Life Sciences, Fudan University, Shanghai 200438, China
| | - Yi-Yu Lin
- Department of Basic Medicine, School of Basic Medicine and Clinical Pharmacy, China Pharmaceutical University, Nanjing 200098, China
| | - Ru-Chun Wang
- Department of Basic Medicine, School of Basic Medicine and Clinical Pharmacy, China Pharmaceutical University, Nanjing 200098, China
| | - Tang-Xuan Xie
- Department of Basic Medicine, School of Basic Medicine and Clinical Pharmacy, China Pharmaceutical University, Nanjing 200098, China
| | - Yimeng Zhao
- State Key Laboratory of Genetic Engineering, Shanghai Key Laboratory of Bioactive Small Molecules, Collaborative Innovation Center of Genetics and Development, Department of Physiology and Neurobiology, School of Life Sciences, Fudan University, Shanghai 200438, China
- Human Phenome Institute, Fudan University, Shanghai 201203, China
| | - Cheng Shen
- State Key Laboratory of Genetic Engineering, Shanghai Key Laboratory of Bioactive Small Molecules, Collaborative Innovation Center of Genetics and Development, Department of Physiology and Neurobiology, School of Life Sciences, Fudan University, Shanghai 200438, China
| | - Danqi Sheng
- State Key Laboratory of Genetic Engineering, Shanghai Key Laboratory of Bioactive Small Molecules, Collaborative Innovation Center of Genetics and Development, Department of Physiology and Neurobiology, School of Life Sciences, Fudan University, Shanghai 200438, China
| | - Muneyoshi Ichikawa
- State Key Laboratory of Genetic Engineering, Department of Biochemistry and Biophysics, School of Life Sciences, Fudan University, Shanghai 200438, China
| | - Ye Yu
- Department of Basic Medicine, School of Basic Medicine and Clinical Pharmacy, China Pharmaceutical University, Nanjing 200098, China
| | - Jin Wang
- Department of Chemistry, School of Science, China Pharmaceutical University, Nanjing 200098, China
| | - Motoyuki Hattori
- State Key Laboratory of Genetic Engineering, Shanghai Key Laboratory of Bioactive Small Molecules, Collaborative Innovation Center of Genetics and Development, Department of Physiology and Neurobiology, School of Life Sciences, Fudan University, Shanghai 200438, China
| |
Collapse
|
2
|
Yu H, Wang W. Modulation of heteromeric glycine receptor function through high concentration clustering. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.10.17.618879. [PMID: 39464082 PMCID: PMC11507885 DOI: 10.1101/2024.10.17.618879] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 10/29/2024]
Abstract
Ion channels are targeted by many drugs for treating neurological, musculoskeletal, renal and other diseases. These drugs bind to and alter the function of individual channels to achieve desired therapeutic effects. However, many ion channels function in high concentration clusters in their native environment. It is unclear if and how clustering modulates ion channel function. Human heteromeric glycine receptors (GlyRs) are the major inhibitory neurotransmitter receptors in the spinal cord and are active targets for developing chronic pain medications. We show that the α2β heteromeric GlyR assembles with the master postsynaptic scaffolding gephyrin (GPHN) into micron-sized clustered at the plasma membrane after heterologous expression. The inhibitory trans- synaptic adhesion protein neuroligin-2 (NL2) further increases both the cluster sizes and GlyR concentration. The apparent glycine affinity increases monotonically as a function of GlyR concentration but not with cluster size. We also show that ligand re-binding to adjacent GlyRs alters kinetics but not chemical equilibrium. A positively charged N- terminus sequence of the GlyR β subunit was further identified essential for glycine affinity modulation through clustering. Taken together, we propose a mechanism where clustering enhances local electrostatic potential, which in turn concentrates ions and ligands, modulating the function of GlyR. This mechanism is likely universal across ion channel clusters found ubiquitously in biology and provides new perspectives in possible pharmaceutical development.
Collapse
|
3
|
Liu X, Wang W. Gating mechanism of the human α1β GlyR by glycine. Structure 2024; 32:1621-1631.e3. [PMID: 39146932 DOI: 10.1016/j.str.2024.07.012] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/02/2024] [Revised: 06/20/2024] [Accepted: 07/19/2024] [Indexed: 08/17/2024]
Abstract
Glycine receptors (GlyRs) are members of the Cys-loop receptors that constitute a major portion of mammalian neurotransmitter receptors. Recent resolution of heteromeric GlyR structures in multiple functional states raised fundamental questions regarding the gating mechanism of GlyR, and generally the Cys-loop family receptors. Here, we characterized in detail equilibrium properties as well as the transition kinetics between functional states. We show that, while all allosteric sites bind cooperatively to glycine, occupation of 2 sites at the α-α interfaces is sufficient for activation and necessary for high-efficacy gating. Differential glycine concentration dependence of desensitization rate, extent, and its recovery suggests separate but concerted roles of ligand-binding and ionophore reorganization. Based on these observations and available structural information, we developed a quantitative gating model that accurately predicts both equilibrium and kinetical properties throughout the glycine gating cycle. This model likely applies generally to the Cys-loop receptors and informs on pharmaceutical endeavors.
Collapse
Affiliation(s)
- Xiaofen Liu
- Departments of Biophysics, University of Texas Southwestern Medical Center, Dallas, TX, USA
| | - Weiwei Wang
- Departments of Biophysics, University of Texas Southwestern Medical Center, Dallas, TX, USA.
| |
Collapse
|
4
|
Yan J, Chen L, Warshel A, Bai C. Exploring the Activation Process of the Glycine Receptor. J Am Chem Soc 2024; 146:26297-26312. [PMID: 39279763 DOI: 10.1021/jacs.4c08489] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 09/18/2024]
Abstract
Glycine receptors (GlyR) conduct inhibitory glycinergic neurotransmission in the spinal cord and the brainstem. They play an important role in muscle tone, motor coordination, respiration, and pain perception. However, the mechanism underlying GlyR activation remains unclear. There are five potential glycine binding sites in α1 GlyR, and different binding patterns may cause distinct activation or desensitization behaviors. In this study, we investigated the coupling of protein conformational changes and glycine binding events to elucidate the influence of binding patterns on the activation and desensitization processes of α1 GlyRs. Subsequently, we explored the energetic distinctions between the apical and lateral pathways during α1 GlyR conduction to identify the pivotal factors in the ion conduction pathway preference. Moreover, we predicted the mutational effects of the key residues and verified our predictions using electrophysiological experiments. For the mutants that can be activated by glycine, the predictions of the mutational directions were all correct. The strength of the mutational effects was assessed using Pearson's correlation coefficient, yielding a value of -0.77 between the calculated highest energy barriers and experimental maximum current amplitudes. These findings contribute to our understanding of GlyR activation, identify the key residues of GlyRs, and provide guidance for mechanistic studies on other pLGICs.
Collapse
Affiliation(s)
- Junfang Yan
- School of Medicine, Warshel Institute for Computational Biology, The Chinese University of Hong Kong, Shenzhen 518172, China
| | - Luonan Chen
- Key Laboratory of Systems Biology, Institute of Biochemistry and Cell Biology, Center for Excellence in Molecular Cell Science, Chinese Academy of Sciences, Shanghai 200031, China
| | - Arieh Warshel
- Department of Chemistry, University of Southern California, Los Angeles, California 90089-1062, United States
| | - Chen Bai
- School of Medicine, Warshel Institute for Computational Biology, The Chinese University of Hong Kong, Shenzhen 518172, China
- College of Pharmaceutical Sciences, Zhejiang University, Hangzhou 310058, China
- Chenzhu (MoMeD) Biotechnology Co., Ltd., Hangzhou 310005, China
| |
Collapse
|
5
|
Liu F, Li T, Gong H, Tian F, Bai Y, Wang H, Yang C, Li Y, Guo F, Liu S, Chen Q. Structural insights into the molecular effects of the anthelmintics monepantel and betaine on the Caenorhabditis elegans acetylcholine receptor ACR-23. EMBO J 2024; 43:3787-3806. [PMID: 39009676 PMCID: PMC11377560 DOI: 10.1038/s44318-024-00165-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/05/2024] [Revised: 06/27/2024] [Accepted: 06/28/2024] [Indexed: 07/17/2024] Open
Abstract
Anthelmintics are drugs used for controlling pathogenic helminths in animals and plants. The natural compound betaine and the recently developed synthetic compound monepantel are both anthelmintics that target the acetylcholine receptor ACR-23 and its homologs in nematodes. Here, we present cryo-electron microscopy structures of ACR-23 in apo, betaine-bound, and betaine- and monepantel-bound states. We show that ACR-23 forms a homo-pentameric channel, similar to some other pentameric ligand-gated ion channels (pLGICs). While betaine molecules are bound to the classical neurotransmitter sites in the inter-subunit interfaces in the extracellular domain, monepantel molecules are bound to allosteric sites formed in the inter-subunit interfaces in the transmembrane domain of the receptor. Although the pore remains closed in betaine-bound state, monepantel binding results in an open channel by wedging into the cleft between the transmembrane domains of two neighboring subunits, which causes dilation of the ion conduction pore. By combining structural analyses with site-directed mutagenesis, electrophysiology and in vivo locomotion assays, we provide insights into the mechanism of action of the anthelmintics monepantel and betaine.
Collapse
Affiliation(s)
- Fenglian Liu
- Center for Life Sciences, Yunnan Key Laboratory of Cell Metabolism and Diseases, State Key Laboratory for Conservation and Utilization of Bio-Resources in Yunnan, School of Life Sciences, Yunnan University, Kunming, 650091, China
| | - Tianyu Li
- Shanghai Key Laboratory of Maternal Fetal Medicine, Shanghai Institute of Maternal-Fetal Medicine and Gynecologic Oncology, Department of Anesthesiology, Shanghai First Maternity and Infant Hospital, School of Medicine, Tongji University, Shanghai, 201204, China
- State Key Laboratory of Drug Research, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai, 201203, China
| | - Huihui Gong
- Center for Life Sciences, Yunnan Key Laboratory of Cell Metabolism and Diseases, State Key Laboratory for Conservation and Utilization of Bio-Resources in Yunnan, School of Life Sciences, Yunnan University, Kunming, 650091, China
| | - Fei Tian
- Center for Life Sciences, Yunnan Key Laboratory of Cell Metabolism and Diseases, State Key Laboratory for Conservation and Utilization of Bio-Resources in Yunnan, School of Life Sciences, Yunnan University, Kunming, 650091, China
| | - Yan Bai
- Center for Life Sciences, Yunnan Key Laboratory of Cell Metabolism and Diseases, State Key Laboratory for Conservation and Utilization of Bio-Resources in Yunnan, School of Life Sciences, Yunnan University, Kunming, 650091, China
| | - Haowei Wang
- Center for Life Sciences, Yunnan Key Laboratory of Cell Metabolism and Diseases, State Key Laboratory for Conservation and Utilization of Bio-Resources in Yunnan, School of Life Sciences, Yunnan University, Kunming, 650091, China
| | - Chonglin Yang
- Center for Life Sciences, Yunnan Key Laboratory of Cell Metabolism and Diseases, State Key Laboratory for Conservation and Utilization of Bio-Resources in Yunnan, School of Life Sciences, Yunnan University, Kunming, 650091, China
| | - Yang Li
- State Key Laboratory of Drug Research, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai, 201203, China
- University of Chinese Academy of Sciences, Beijing, 100049, China
- National Clinical Research Center for Aging and Medicine, Huashan Hospital, Fudan University, Shanghai, 200040, China
| | - Fei Guo
- State Key Laboratory of Drug Research, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai, 201203, China.
- University of Chinese Academy of Sciences, Beijing, 100049, China.
- National Clinical Research Center for Aging and Medicine, Huashan Hospital, Fudan University, Shanghai, 200040, China.
| | - Sheng Liu
- Institute of Pediatrics, Shenzhen Children's Hospital, Shenzhen, Guangdong Province, 518026, China.
- Department of Infectious Diseases, Shenzhen Children's Hospital, Shenzhen, Guangdong Province, 518038, China.
| | - Qingfeng Chen
- Center for Life Sciences, Yunnan Key Laboratory of Cell Metabolism and Diseases, State Key Laboratory for Conservation and Utilization of Bio-Resources in Yunnan, School of Life Sciences, Yunnan University, Kunming, 650091, China.
| |
Collapse
|
6
|
Lu Y, Lin Y, Wang J. Progress on functions of intracellular domain of trimeric ligand-gated ion channels. Zhejiang Da Xue Xue Bao Yi Xue Ban 2024; 53:221-230. [PMID: 38310082 PMCID: PMC11057991 DOI: 10.3724/zdxbyxb-2023-0472] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/30/2023] [Accepted: 11/11/2023] [Indexed: 02/05/2024]
Abstract
Ligand-gated ion channels are a large category of essential ion channels, modulating their state by binding to specific ligands to allow ions to pass through the cell membrane. Purinergic ligand-gated ion channel receptors (P2XRs) and acid-sensitive ion channels (ASICs) are representative members of trimeric ligand-gated ion channel. Recent studies have shown that structural differences in the intracellular domain of P2XRs may determine the desensitization process. The lateral fenestrations of P2XRs potentially serve as a pathway for ion conductance and play a decisive role in ion selectivity. Phosphorylation of numerous amino acid residues in the P2XRs are involved in regulating the activity of ion channels. Additionally, the P2XRs interact with other ligand-gated ion channels including N-methyl-D-aspartate receptors, γ-aminobutyric acid receptors, 5-hydroxytryptamin receptors and nicotinic acetylcholine receptors, mediating physiological processes such as synaptic plasticity. Conformational changes in the intracellular domain of the ASICs expose binding sites of intracellular signal partners, facilitating metabolic signal transduction. Amino acids such as Val16, Ser17, Ile18, Gln19 and Ala20 in the ASICs participate in channel opening and membrane expression. ASICs can also bind to intracellular proteins, such as CIPP and p11, to regulate channel function. Many phosphorylation sites at the C-terminus and N-terminus of ASICs are involved in the regulation of receptors. Furthermore, ASICs are involved in various physiological and pathophysiological processes, which include pain, ischemic stroke, psychiatric disorders, and neurodegenerative disease. In this article, we review the roles of the intracellular domains of these trimeric ligand-gated ion channels in channel gating as well as their physiological and pathological functions, in order to provide new insights into the discovery of related drugs.
Collapse
Affiliation(s)
- Yan Lu
- School of Basic Medicine and Clinical Pharmacy, China Pharmaceutical University, Nanjing 210009, China.
| | - Yiyu Lin
- School of Basic Medicine and Clinical Pharmacy, China Pharmaceutical University, Nanjing 210009, China
| | - Jin Wang
- School of Basic Medicine and Clinical Pharmacy, China Pharmaceutical University, Nanjing 210009, China.
| |
Collapse
|
7
|
Alvarez LD, Carina Alves NR. Structural Basis for Molecular Recognition of Cannabinoids by Inhibitory Cys-Loop Channels. J Med Chem 2024; 67:3274-3286. [PMID: 38428383 DOI: 10.1021/acs.jmedchem.3c02391] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 03/03/2024]
Abstract
Cannabis sativa has a long history of medicinal use, dating back to ancient times. This plant produces cannabinoids, which are now known to interact with several human proteins, including Cys-loop receptors for glycine (GlyR) and gamma-aminobutyric acid (GABAAR). As these channels are the primary mediators of inhibitory signals, they contribute to the diverse effects of cannabinoids on the nervous system. Evidence suggests that cannabinoid binding sites are located within the transmembrane domain, although their precise location has remained undetermined for over a decade. The process of identification of the binding site and the computational approaches employed are the main subjects of this Perspective, which includes an analysis of the most recently resolved cryo-EM structures of zebrafish GlyR bound to Δ9-tetrahydrocannabinol and the THC-GlyR complex obtained through molecular dynamics simulations. With this work, we aim to contribute to guiding future studies investigating the molecular basis of cannabinoid action on inhibitory channels.
Collapse
Affiliation(s)
- Lautaro D Alvarez
- Departamento de Química Biológica, Facultad de Ciencias Exactas y Naturales, Universidad de Buenos Aires, Ciudad Universitaria, Buenos Aires C1428EGA, Argentina
- UMYMFOR, CONICET-Universidad de Buenos Aires, Ciudad UniversitariaBuenos Aires C1428EGA, Argentina
| | - N R Carina Alves
- Departamento de Química Biológica, Facultad de Ciencias Exactas y Naturales, Universidad de Buenos Aires, Ciudad Universitaria, Buenos Aires C1428EGA, Argentina
- UMYMFOR, CONICET-Universidad de Buenos Aires, Ciudad UniversitariaBuenos Aires C1428EGA, Argentina
| |
Collapse
|
8
|
Wiessler AL, Hasenmüller AS, Fuhl I, Mille C, Cortes Campo O, Reinhard N, Schenk J, Heinze KG, Schaefer N, Specht CG, Villmann C. Role of the Glycine Receptor β Subunit in Synaptic Localization and Pathogenicity in Severe Startle Disease. J Neurosci 2024; 44:e0837232023. [PMID: 37963764 PMCID: PMC10860499 DOI: 10.1523/jneurosci.0837-23.2023] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/08/2023] [Revised: 09/20/2023] [Accepted: 10/16/2023] [Indexed: 11/16/2023] Open
Abstract
Startle disease is due to the disruption of recurrent inhibition in the spinal cord. Most common causes are genetic variants in genes (GLRA1, GLRB) encoding inhibitory glycine receptor (GlyR) subunits. The adult GlyR is a heteropentameric complex composed of α1 and β subunits that localizes at postsynaptic sites and replaces embryonically expressed GlyRα2 homomers. The human GlyR variants of GLRA1 and GLRB, dominant and recessive, have been intensively studied in vitro. However, the role of unaffected GlyRβ, essential for synaptic GlyR localization, in the presence of mutated GlyRα1 in vivo is not fully understood. Here, we used knock-in mice expressing endogenous mEos4b-tagged GlyRβ that were crossed with mouse Glra1 startle disease mutants. We explored the role of GlyRβ under disease conditions in mice carrying a missense mutation (shaky) or resulting from the loss of GlyRα1 (oscillator). Interestingly, synaptic targeting of GlyRβ was largely unaffected in both mouse mutants. While synaptic morphology appears unaltered in shaky animals, synapses were notably smaller in homozygous oscillator animals. Hence, GlyRβ enables transport of functionally impaired GlyRα1 missense variants to synaptic sites in shaky animals, which has an impact on the efficacy of possible compensatory mechanisms. The observed enhanced GlyRα2 expression in oscillator animals points to a compensation by other GlyRα subunits. However, trafficking of GlyRα2β complexes to synaptic sites remains functionally insufficient, and homozygous oscillator mice still die at 3 weeks after birth. Thus, both functional and structural deficits can affect glycinergic neurotransmission in severe startle disease, eliciting different compensatory mechanisms in vivo.
Collapse
Affiliation(s)
- Anna-Lena Wiessler
- Institute for Clinical Neurobiology, University Hospital, Julius-Maximilians-University of Würzburg, 97078 Würzburg, Germany
| | - Ann-Sofie Hasenmüller
- Institute for Clinical Neurobiology, University Hospital, Julius-Maximilians-University of Würzburg, 97078 Würzburg, Germany
| | - Isabell Fuhl
- Institute for Clinical Neurobiology, University Hospital, Julius-Maximilians-University of Würzburg, 97078 Würzburg, Germany
| | - Clémence Mille
- Institut National de la Santé et de la Recherche Médicale (Inserm U1195), Université Paris-Saclay, 94276 Le Kremlin-Bicêtre, France
| | - Orlando Cortes Campo
- Institute for Clinical Neurobiology, University Hospital, Julius-Maximilians-University of Würzburg, 97078 Würzburg, Germany
| | - Nicola Reinhard
- Institute for Clinical Neurobiology, University Hospital, Julius-Maximilians-University of Würzburg, 97078 Würzburg, Germany
| | - Joachim Schenk
- Rudolf Virchow Center for Integrative and Translational Bioimaging, Julius-Maximilians-University of Würzburg, 97080 Würzburg, Germany
| | - Katrin G Heinze
- Rudolf Virchow Center for Integrative and Translational Bioimaging, Julius-Maximilians-University of Würzburg, 97080 Würzburg, Germany
| | - Natascha Schaefer
- Institute for Clinical Neurobiology, University Hospital, Julius-Maximilians-University of Würzburg, 97078 Würzburg, Germany
| | - Christian G Specht
- Institut National de la Santé et de la Recherche Médicale (Inserm U1195), Université Paris-Saclay, 94276 Le Kremlin-Bicêtre, France
| | - Carmen Villmann
- Institute for Clinical Neurobiology, University Hospital, Julius-Maximilians-University of Würzburg, 97078 Würzburg, Germany
| |
Collapse
|
9
|
Thompson JRE, Beaudoin CA, Lummis SCR. Modelling and Molecular Dynamics Predict the Structure and Interactions of the Glycine Receptor Intracellular Domain. Biomolecules 2023; 13:1757. [PMID: 38136628 PMCID: PMC10741472 DOI: 10.3390/biom13121757] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/31/2023] [Revised: 11/28/2023] [Accepted: 12/01/2023] [Indexed: 12/24/2023] Open
Abstract
Glycine receptors (GlyRs) are glycine-gated inhibitory pentameric ligand-gated ion channels composed of α or α + β subunits. A number of structures of these proteins have been reported, but to date, these have only revealed details of the extracellular and transmembrane domains, with the intracellular domain (ICD) remaining uncharacterised due to its high flexibility. The ICD is a region that can modulate function in addition to being critical for receptor localisation and clustering via proteins such as gephyrin. Here, we use modelling and molecular dynamics (MD) to reveal details of the ICDs of both homomeric and heteromeric GlyR. At their N and C ends, both the α and β subunit ICDs have short helices, which are major sites of stabilising interactions; there is a large flexible loop between them capable of forming transient secondary structures. The α subunit can affect the β subunit ICD structure, which is more flexible in a 4α2:1β than in a 4α1:1β GlyR. We also explore the effects of gephyrin binding by creating GlyR models bound to the gephyrin E domain; MD simulations suggest these are more stable than the unbound forms, and again there are α subunit-dependent differences, despite the fact the gephyrin binds to the β subunit. The bound models also suggest that gephyrin causes compaction of the ICD. Overall, the data expand our knowledge of this important receptor protein and in particular clarify features of the underexplored ICD.
Collapse
Affiliation(s)
| | | | - Sarah C. R. Lummis
- Department of Biochemistry, University of Cambridge, Cambridge CB2 1QW, UK
| |
Collapse
|
10
|
Schaefer N, Harvey RJ, Villmann C. Startle Disease: New Molecular Insights into an Old Neurological Disorder. Neuroscientist 2023; 29:767-781. [PMID: 35754344 PMCID: PMC10623600 DOI: 10.1177/10738584221104724] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
Startle disease (SD) is characterized by enhanced startle responses, generalized muscle stiffness, unexpected falling, and fatal apnea episodes due to disturbed feedback inhibition in the spinal cord and brainstem of affected individuals. Mutations within the glycine receptor (GlyR) subunit and glycine transporter 2 (GlyT2) genes have been identified in individuals with SD. Impaired inhibitory neurotransmission in SD is due to pre- and/or postsynaptic GlyR or presynaptic GlyT2 dysfunctions. Previous research has focused on mutated GlyRs and GlyT2 that impair ion channel/transporter function or trafficking. With insights provided by recently solved cryo-electron microscopy and X-ray structures of GlyRs, a detailed picture of structural transitions important for receptor gating has emerged, allowing a deeper understanding of SD at the molecular level. Moreover, studies on novel SD mutations have demonstrated a higher complexity of SD, with identification of additional clinical signs and symptoms and interaction partners representing key players for fine-tuning synaptic processes. Although our knowledge has steadily improved during the last years, changes in synaptic localization and GlyR or GlyT2 homeostasis under disease conditions are not yet completely understood. Combined proteomics, interactomics, and high-resolution microscopy techniques are required to reveal alterations in receptor dynamics at the synaptic level under disease conditions.
Collapse
Affiliation(s)
- Natascha Schaefer
- Institute of Clinical Neurobiology, University Hospital, Julius-Maximilians-University of Würzburg, Würzburg, Germany
| | - Robert J. Harvey
- School of Health and Behavioural Sciences, University of the Sunshine Coast, Maroochydore DC, Australia
- Sunshine Coast Health Institute, Birtinya, Australia
| | - Carmen Villmann
- Institute of Clinical Neurobiology, University Hospital, Julius-Maximilians-University of Würzburg, Würzburg, Germany
| |
Collapse
|
11
|
Liu X, Wang W. Asymmetric gating of a human hetero-pentameric glycine receptor. Nat Commun 2023; 14:6377. [PMID: 37821459 PMCID: PMC10567788 DOI: 10.1038/s41467-023-42051-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/16/2022] [Accepted: 09/28/2023] [Indexed: 10/13/2023] Open
Abstract
Hetero-pentameric Cys-loop receptors constitute a major type of neurotransmitter receptors that enable signal transmission and processing in the nervous system. Despite intense investigations into their working mechanism and pharmaceutical potentials, how neurotransmitters activate these receptors remains unclear due to the lack of high-resolution structural information in the activated open state. Here we report near-atomic resolution structures resolved in digitonin consistent with all principle functional states of the human α1β GlyR, which is a major Cys-loop receptor that mediates inhibitory neurotransmission in the central nervous system of adults. Glycine binding induces cooperative and symmetric structural rearrangements in the neurotransmitter-binding extracellular domain but asymmetrical pore dilation in the transmembrane domain. Symmetric response in the extracellular domain is consistent with electrophysiological data showing cooperative glycine activation and contribution from both α1 and β subunits. A set of functionally essential but differentially charged amino acid residues in the transmembrane domain of the α1 and β subunits explains asymmetric activation. These findings provide a foundation for understanding how the gating of the Cys-loop receptor family members diverges to accommodate specific physiological environments.
Collapse
Affiliation(s)
- Xiaofen Liu
- Department of Biophysics, University of Texas Southwestern Medical Center, Dallas, TX, USA
| | - Weiwei Wang
- Department of Biophysics, University of Texas Southwestern Medical Center, Dallas, TX, USA.
| |
Collapse
|
12
|
Yang L, Hou A, Jiang Q, Cheng M, Liu Y. Methodological Development and Applications of Tryptamine-Ynamide Cyclizations in Synthesizing Core Skeletons of Indole Alkaloids. J Org Chem 2023; 88:11377-11391. [PMID: 37540141 DOI: 10.1021/acs.joc.3c01088] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 08/05/2023]
Abstract
Over the past two decades, synthetic strategies for synthesizing the skeletons of various indole alkaloids based on tryptamine-ynamide have been continuously developed and applied to the total syntheses or formal total syntheses of related molecules. In this synopsis, we summarized the cyclization pathways of tryptamine-ynamide under different catalytic conditions, emphasizing the reaction mechanism and applications in the syntheses of indole alkaloids.
Collapse
Affiliation(s)
- Lu Yang
- Key Laboratory of Structure-Based Drug Design and Discovery of Ministry of Education, Shenyang Pharmaceutical University, Shenyang 110016, P. R. China
- Wuya College of Innovation, Shenyang Pharmaceutical University, Shenyang 110016, P. R. China
- Institute of Drug Research in Medicine Capital of China, Benxi 117000, P. R. China
| | - Anbin Hou
- Key Laboratory of Structure-Based Drug Design and Discovery of Ministry of Education, Shenyang Pharmaceutical University, Shenyang 110016, P. R. China
- Wuya College of Innovation, Shenyang Pharmaceutical University, Shenyang 110016, P. R. China
- Institute of Drug Research in Medicine Capital of China, Benxi 117000, P. R. China
| | - Qing Jiang
- Key Laboratory of Structure-Based Drug Design and Discovery of Ministry of Education, Shenyang Pharmaceutical University, Shenyang 110016, P. R. China
- Wuya College of Innovation, Shenyang Pharmaceutical University, Shenyang 110016, P. R. China
- Institute of Drug Research in Medicine Capital of China, Benxi 117000, P. R. China
| | - Maosheng Cheng
- Key Laboratory of Structure-Based Drug Design and Discovery of Ministry of Education, Shenyang Pharmaceutical University, Shenyang 110016, P. R. China
- Institute of Drug Research in Medicine Capital of China, Benxi 117000, P. R. China
| | - Yongxiang Liu
- Key Laboratory of Structure-Based Drug Design and Discovery of Ministry of Education, Shenyang Pharmaceutical University, Shenyang 110016, P. R. China
- Wuya College of Innovation, Shenyang Pharmaceutical University, Shenyang 110016, P. R. China
- Institute of Drug Research in Medicine Capital of China, Benxi 117000, P. R. China
| |
Collapse
|
13
|
Liu X, Wang W. Gating mechanism of the human α1β GlyR by glycine. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2023:2023.08.08.552474. [PMID: 37609197 PMCID: PMC10441291 DOI: 10.1101/2023.08.08.552474] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 08/24/2023]
Abstract
Glycine receptors (GlyRs) are members of the Cys-loop receptors that constitute a major portion of neurotransmitter receptors in the human nervous system. GlyRs are found in the spinal cord and brain mediating locomotive, sensory and cognitive functions, and are targets for pharmaceutical development. GlyRs share a general gating scheme with Cys-loop receptor family members, but the underlying mechanism is unclear. Recent resolution of heteromeric GlyRs structures in multiple functional states identified an invariable 4:1 α:β subunit stoichiometry and provided snapshots in the gating cycle, challenging previous beliefs and raising the fundamental questions of how α and β subunit functions in glycine binding and channel activation. In addition, how a single glycine-bound extracellular domain conformation leads to structurally and functionally different open and desensitized states remained enigmatic. In this study, we characterized in detail equilibrium properties as well as the transition kinetics between functional states. We show that while all allosteric sites bind cooperatively to glycine, occupation of 2 sites at the α-α interfaces is necessary and sufficient for GlyR activation. We also demonstrate differential glycine concentration dependence of desensitization rate, extent, and its recovery, which suggests separate but concerted roles of ligand-binding and ionophore reorganization. Based on these observations and available structural information, we developed a comprehensive quantitative gating model that accurately predicts both equilibrium and kinetical properties throughout glycine gating cycle. This model likely applies generally to the Cys-loop receptor family and informs on pharmaceutical endeavors in function modulation of this receptor family.
Collapse
|
14
|
Elverson K, Freeman S, Manson F, Warwicker J. Computational Investigation of Mechanisms for pH Modulation of Human Chloride Channels. Molecules 2023; 28:5753. [PMID: 37570721 PMCID: PMC10420675 DOI: 10.3390/molecules28155753] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/29/2022] [Revised: 07/25/2023] [Accepted: 07/26/2023] [Indexed: 08/13/2023] Open
Abstract
Many transmembrane proteins are modulated by intracellular or extracellular pH. Investigation of pH dependence generally proceeds by mutagenesis of a wide set of amino acids, guided by properties such as amino-acid conservation and structure. Prediction of pKas can streamline this process, allowing rapid and effective identification of amino acids of interest with respect to pH dependence. Commencing with the calcium-activated chloride channel bestrophin 1, the carboxylate ligand structure around calcium sites relaxes in the absence of calcium, consistent with a measured lack of pH dependence. By contrast, less relaxation in the absence of calcium in TMEM16A, and maintenance of elevated carboxylate sidechain pKas, is suggested to give rise to pH-dependent chloride channel activity. This hypothesis, modulation of calcium/proton coupling and pH-dependent activity through the extent of structural relaxation, is shown to apply to the well-characterised cytosolic proteins calmodulin (pH-independent) and calbindin D9k (pH-dependent). Further application of destabilised, ionisable charge sites, or electrostatic frustration, is made to other human chloride channels (that are not calcium-activated), ClC-2, GABAA, and GlyR. Experimentally determined sites of pH modulation are readily identified. Structure-based tools for pKa prediction are freely available, allowing users to focus on mutagenesis studies, construct hypothetical proton pathways, and derive hypotheses such as the model for control of pH-dependent calcium activation through structural flexibility. Predicting altered pH dependence for mutations in ion channel disorders can support experimentation and, ultimately, clinical intervention.
Collapse
Affiliation(s)
- Kathleen Elverson
- Division of Evolution, Infection and Genomics, Faculty of Biology, Medicine and Health, The University of Manchester, Manchester M13 9PT, UK
| | - Sally Freeman
- Division of Pharmacy and Optometry, School of Health Sciences, Faculty of Biology, Medicine and Health, The University of Manchester, Manchester M13 9PT, UK
| | - Forbes Manson
- Division of Evolution, Infection and Genomics, Faculty of Biology, Medicine and Health, The University of Manchester, Manchester M13 9PT, UK
| | - Jim Warwicker
- Division of Molecular and Cellular Function, Faculty of Biology, Medicine and Health, Manchester Institute of Biotechnology, The University of Manchester, Manchester M1 7DN, UK
| |
Collapse
|
15
|
Negin B, Jander G. Convergent and divergent evolution of plant chemical defenses. CURRENT OPINION IN PLANT BIOLOGY 2023; 73:102368. [PMID: 37087925 DOI: 10.1016/j.pbi.2023.102368] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/17/2023] [Revised: 03/06/2023] [Accepted: 03/22/2023] [Indexed: 05/03/2023]
Abstract
The majority of the several hundred thousand specialized metabolites produced by plants function in defense against insects and other herbivores. Despite this diversity, identical metabolites or structurally distinct metabolites hitting the same targets in herbivorous animals have evolved repeatedly. This convergent evolution may reflect the constraints of plant primary metabolism in providing metabolic precursors, as well as the limited number of readily accessible targets in animals. These restrictions may make it uncommon for plants to develop completely novel toxic and deterrent metabolites, despite the ongoing evolution of resistance mechanisms in insect herbivores. Defensive compounds that are unique to individual genera or species often have long biosynthetic pathways that may complicate the repeated evolution of these metabolites in different plant species.
Collapse
Affiliation(s)
- Boaz Negin
- Boyce Thompson Institute, Ithaca, NY, 14853, USA
| | - Georg Jander
- Boyce Thompson Institute, Ithaca, NY, 14853, USA.
| |
Collapse
|
16
|
Gibbs E, Klemm E, Seiferth D, Kumar A, Ilca SL, Biggin PC, Chakrapani S. Conformational transitions and allosteric modulation in a heteromeric glycine receptor. Nat Commun 2023; 14:1363. [PMID: 36914669 PMCID: PMC10011588 DOI: 10.1038/s41467-023-37106-7] [Citation(s) in RCA: 9] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/22/2022] [Accepted: 03/02/2023] [Indexed: 03/16/2023] Open
Abstract
Glycine Receptors (GlyRs) provide inhibitory neuronal input in the spinal cord and brainstem, which is critical for muscle coordination and sensory perception. Synaptic GlyRs are a heteromeric assembly of α and β subunits. Here we present cryo-EM structures of full-length zebrafish α1βBGlyR in the presence of an antagonist (strychnine), agonist (glycine), or agonist with a positive allosteric modulator (glycine/ivermectin). Each structure shows a distinct pore conformation with varying degrees of asymmetry. Molecular dynamic simulations found the structures were in a closed (strychnine) and desensitized states (glycine and glycine/ivermectin). Ivermectin binds at all five interfaces, but in a distinct binding pose at the β-α interface. Subunit-specific features were sufficient to solve structures without a fiduciary marker and to confirm the 4α:1β stoichiometry recently observed. We also report features of the extracellular and intracellular domains. Together, our results show distinct compositional and conformational properties of α1βGlyR and provide a framework for further study of this physiologically important channel.
Collapse
Affiliation(s)
- Eric Gibbs
- Department of Physiology and Biophysics, Case Western Reserve University, Cleveland, OH, 44106-4970, USA
| | - Emily Klemm
- Department of Physiology and Biophysics, Case Western Reserve University, Cleveland, OH, 44106-4970, USA
| | - David Seiferth
- Department of Biochemistry, University of Oxford, Oxford, OX1 3QU, UK
| | - Arvind Kumar
- Department of Physiology and Biophysics, Case Western Reserve University, Cleveland, OH, 44106-4970, USA
| | - Serban L Ilca
- New York Structural Biology Center, New York, NY, 10027, USA
- Simons Electron Microscopy Center, New York, NY, 10027, USA
| | - Philip C Biggin
- Department of Biochemistry, University of Oxford, Oxford, OX1 3QU, UK
| | - Sudha Chakrapani
- Department of Physiology and Biophysics, Case Western Reserve University, Cleveland, OH, 44106-4970, USA.
- Department of Neuroscience, School of Medicine, Case Western Reserve University, Cleveland, OH, 44106-4970, USA.
| |
Collapse
|
17
|
Mi T, Mack JO, Koolmees W, Lyon Q, Yochimowitz L, Teng ZQ, Jiang P, Montell C, Zhang YV. Alkaline taste sensation through the alkaliphile chloride channel in Drosophila. Nat Metab 2023; 5:466-480. [PMID: 36941450 PMCID: PMC10665042 DOI: 10.1038/s42255-023-00765-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/15/2022] [Accepted: 02/09/2023] [Indexed: 03/23/2023]
Abstract
The sense of taste is an important sentinel governing what should or should not be ingested by an animal, with high pH sensation playing a critical role in food selection. Here we explore the molecular identities of taste receptors detecting the basic pH of food using Drosophila melanogaster as a model. We identify a chloride channel named alkaliphile (Alka), which is both necessary and sufficient for aversive taste responses to basic food. Alka forms a high-pH-gated chloride channel and is specifically expressed in a subset of gustatory receptor neurons (GRNs). Optogenetic activation of alka-expressing GRNs is sufficient to suppress attractive feeding responses to sucrose. Conversely, inactivation of these GRNs causes severe impairments in the aversion to high pH. Altogether, our discovery of Alka as an alkaline taste receptor lays the groundwork for future research on alkaline taste sensation in other animals.
Collapse
Affiliation(s)
- Tingwei Mi
- Monell Chemical Senses Center, Philadelphia, PA, USA
| | - John O Mack
- Monell Chemical Senses Center, Philadelphia, PA, USA
| | | | - Quinn Lyon
- Monell Chemical Senses Center, Philadelphia, PA, USA
| | | | - Zhao-Qian Teng
- State Key Laboratory of Stem Cell and Reproductive Biology, Institute of Zoology, Chinese Academy of Sciences, Beijing Institute for Stem Cell and Regenerative Medicine, Beijing, China
| | - Peihua Jiang
- Monell Chemical Senses Center, Philadelphia, PA, USA
| | - Craig Montell
- Department of Molecular, Cellular, and Developmental Biology, University of California, Santa Barbara, CA, USA
| | - Yali V Zhang
- Monell Chemical Senses Center, Philadelphia, PA, USA.
- Department of Physiology, The Diabetes Research Center, University of Pennsylvania Perelman School of Medicine, Philadelphia, PA, USA.
| |
Collapse
|
18
|
Alvarez LD, Alves NRC. Molecular determinants of tetrahydrocannabinol binding to the glycine receptor. Proteins 2023; 91:400-411. [PMID: 36271319 DOI: 10.1002/prot.26438] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/02/2022] [Revised: 10/04/2022] [Accepted: 10/18/2022] [Indexed: 02/03/2023]
Abstract
The recognition of Cannabis as a source of new compounds suitable for medical use has attracted strong interest from the scientific community in its research, and substantial progress has accumulated regarding cannabinoids' activity; however, a thorough description of their molecular mechanisms of action remains a task to complete. Highlighting their complex pharmacology, the list of cannabinoids' interactors has vastly expanded beyond the canonical cannabinoid receptors. Among those, we have focused our study on the glycine receptor (GlyR), an ion channel involved in the modulation of nervous system responses, including, to our interest, sensitivity to peripheral pain. Here, we report the use of computational methods to investigate possible binding modes between the GlyR and Δ9 -tetrahydrocannabinol (THC). After obtaining a first pose for the THC binding from a biased molecular docking simulation and subsequently evaluating it by molecular dynamic simulations, we found a dynamic system with an identifiable representative binding mode characterized by the specific interaction with two transmembrane residues (Phe293 and Ser296). Complementarily, we assessed the role of membrane cholesterol in this interaction and positively established its relevance for THC binding to GlyR. Lastly, the use of restrained molecular dynamics simulations allowed us to refine the description of the binding mode and of the cholesterol effect. Altogether, our findings contribute to the current knowledge about the GlyR-THC mode of binding and propose a new starting point for future research on how cannabinoids in general, and THC in particular, modulate pain perception in view of its possible clinical applications.
Collapse
Affiliation(s)
- Lautaro D Alvarez
- Facultad de Ciencias Exactas y Naturales, Departamento de Química Biológica, Universidad de Buenos Aires, Buenos Aires, Argentina.,CONICET-Universidad de Buenos Aires, UMYMFOR, Buenos Aires, Argentina
| | - N R Carina Alves
- Facultad de Ciencias Exactas y Naturales, Departamento de Química Biológica, Universidad de Buenos Aires, Buenos Aires, Argentina.,CONICET-Universidad de Buenos Aires, IFIBYNE, Buenos Aires, Argentina
| |
Collapse
|
19
|
Wang HC, Cheng KI, Tseng KY, Kwan AL, Chang LL. AAV-glycine receptor α3 alleviates CFA-induced inflammatory pain by downregulating ERK phosphorylation and proinflammatory cytokine expression in SD rats. Mol Med 2023; 29:22. [PMID: 36792984 PMCID: PMC9933394 DOI: 10.1186/s10020-023-00606-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/21/2022] [Accepted: 01/10/2023] [Indexed: 02/17/2023] Open
Abstract
BACKGROUND Glycine receptors (GlyRs) play key roles in the processing of inflammatory pain. The use of adeno-associated virus (AAV) vectors for gene therapy in human clinical trials has shown promise, as AAV generally causes a very mild immune response and long-term gene transfer, and there have been no reports of disease. Therefore, we used AAV for GlyRα1/3 gene transfer in F11 neuron cells and into Sprague-Dawley (SD) rats to investigate the effects and roles of AAV-GlyRα1/3 on cell cytotoxicity and inflammatory response. METHODS In vitro experiments were performed using plasmid adeno-associated virus (pAAV)-GlyRα1/3-transfected F11 neurons to investigate the effects of pAAV-GlyRα1/3 on cell cytotoxicity and the prostaglandin E2 (PGE2)-mediated inflammatory response. In vivo experiment, the association between GlyRα3 and inflammatory pain was analyzed in normal rats after AAV-GlyRα3 intrathecal injection and after complete Freund's adjuvant (CFA) intraplantar administration. Intrathecal AAV-GlyRα3 delivery into SD rats was evaluated in terms of its potential for alleviating CFA-induced inflammatory pain. RESULTS The activation of mitogen-activated protein kinase (MAPK) inflammatory signaling and neuronal injury marker activating transcription factor 3 (ATF-3) were evaluated by western blotting and immunofluorescence; the level of cytokine expression was measured by ELISA. The results showed that pAAV/pAAV-GlyRα1/3 transfection into F11 cells did not significantly reduce cell viability or induce extracellular signal-regulated kinase (ERK) phosphorylation or ATF-3 activation. PGE2-induced ERK phosphorylation in F11 cells was repressed by the expression of pAAV-GlyRα3 and administration of an EP2 inhibitor, GlyRαs antagonist (strychnine), and a protein kinase C inhibitor. Additionally, intrathecal AAV-GlyRα3 administration to SD rats significantly decreased CFA-induced inflammatory pain and suppressed CFA-induced ERK phosphorylation, did not induce obvious histopathological injury but increased ATF-3 activation in dorsal root ganglion (DRGs). CONCLUSIONS Antagonists of the prostaglandin EP2 receptor, PKC, and glycine receptor can inhibit PGE2-induced ERK phosphorylation. Intrathecal AAV-GlyRα3 administration to SD rats significantly decreased CFA-induced inflammatory pain and suppressed CFA-induced ERK phosphorylation, did not significantly induce gross histopathological injury but elicited ATF-3 activation. We suggest that PGE2-induced ERK phosphorylation can be modulated by GlyRα3, and AAV-GlyRα3 significantly downregulated CFA-induced cytokine activation.
Collapse
Affiliation(s)
- Hung-Chen Wang
- grid.145695.a0000 0004 1798 0922Department of Neurosurgery, Kaohsiung Chang Gung Memorial Hospital, Chang Gung University College of Medicine, Kaohsiung, Taiwan
| | - Kuang-I Cheng
- grid.412019.f0000 0000 9476 5696Department of Anesthesiology, Faculty of Medicine, College of Medicine, Kaohsiung Medical University, Kaohsiung, Taiwan ,grid.412019.f0000 0000 9476 5696Graduate Institute of Medicine, College of Medicine, Kaohsiung Medical University, Kaohsiung, Taiwan
| | - Kuang-Yi Tseng
- grid.412019.f0000 0000 9476 5696Department of Anesthesiology, Faculty of Medicine, College of Medicine, Kaohsiung Medical University, Kaohsiung, Taiwan ,grid.412019.f0000 0000 9476 5696Graduate Institute of Medicine, College of Medicine, Kaohsiung Medical University, Kaohsiung, Taiwan
| | - Aij-Lie Kwan
- grid.412019.f0000 0000 9476 5696Department of Neurosurgery, Faculty of Medicine, College of Medicine, Kaohsiung Medical University, Kaohsiung, Taiwan
| | - Lin-Li Chang
- Graduate Institute of Medicine, College of Medicine, Kaohsiung Medical University, Kaohsiung, Taiwan. .,Department of Microbiology and Immunology, Faculty of Medicine, College of Medicine, Kaohsiung Medical University, 100, Shih-Chuan 1st Road, Kaohsiung, 80708, Taiwan. .,Graduate Institute of Clinical Medicine, College of Medicine, Kaohsiung Medical University, Kaohsiung, Taiwan. .,Center for Infectious Disease and Cancer Research, Kaohsiung Medical University, Kaohsiung, Taiwan. .,Department of Medical Research, Kaohsiung Medical University Hospital, Kaohsiung, Taiwan.
| |
Collapse
|
20
|
Rauschenberger V, Piro I, Kasaragod VB, Hörlin V, Eckes AL, Kluck CJ, Schindelin H, Meinck HM, Wickel J, Geis C, Tüzün E, Doppler K, Sommer C, Villmann C. Glycine receptor autoantibody binding to the extracellular domain is independent from receptor glycosylation. Front Mol Neurosci 2023; 16:1089101. [PMID: 36860666 PMCID: PMC9969106 DOI: 10.3389/fnmol.2023.1089101] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/03/2022] [Accepted: 01/19/2023] [Indexed: 02/17/2023] Open
Abstract
Glycine receptor (GlyR) autoantibodies are associated with stiff-person syndrome and the life-threatening progressive encephalomyelitis with rigidity and myoclonus in children and adults. Patient histories show variability in symptoms and responses to therapeutic treatments. A better understanding of the autoantibody pathology is required to develop improved therapeutic strategies. So far, the underlying molecular pathomechanisms include enhanced receptor internalization and direct receptor blocking altering GlyR function. A common epitope of autoantibodies against the GlyRα1 has been previously defined to residues 1A-33G at the N-terminus of the mature GlyR extracellular domain. However, if other autoantibody binding sites exist or additional GlyR residues are involved in autoantibody binding is yet unknown. The present study investigates the importance of receptor glycosylation for binding of anti-GlyR autoantibodies. The glycine receptor α1 harbors only one glycosylation site at the amino acid residue asparagine 38 localized in close vicinity to the identified common autoantibody epitope. First, non-glycosylated GlyRs were characterized using protein biochemical approaches as well as electrophysiological recordings and molecular modeling. Molecular modeling of non-glycosylated GlyRα1 did not show major structural alterations. Moreover, non-glycosylation of the GlyRα1N38Q did not prevent the receptor from surface expression. At the functional level, the non-glycosylated GlyR demonstrated reduced glycine potency, but patient GlyR autoantibodies still bound to the surface-expressed non-glycosylated receptor protein in living cells. Efficient adsorption of GlyR autoantibodies from patient samples was possible by binding to native glycosylated and non-glycosylated GlyRα1 expressed in living not fixed transfected HEK293 cells. Binding of patient-derived GlyR autoantibodies to the non-glycosylated GlyRα1 offered the possibility to use purified non-glycosylated GlyR extracellular domain constructs coated on ELISA plates and use them as a fast screening readout for the presence of GlyR autoantibodies in patient serum samples. Following successful adsorption of patient autoantibodies by GlyR ECDs, binding to primary motoneurons and transfected cells was absent. Our results indicate that the glycine receptor autoantibody binding is independent of the receptor's glycosylation state. Purified non-glycosylated receptor domains harbouring the autoantibody epitope thus provide, an additional reliable experimental tool besides binding to native receptors in cell-based assays for detection of autoantibody presence in patient sera.
Collapse
Affiliation(s)
- Vera Rauschenberger
- Institute of Clinical Neurobiology, University Hospital, Julius-Maximilians-University of Würzburg, Würzburg, Germany
| | - Inken Piro
- Department of Neurology, University Hospital Würzburg, Würzburg, Germany
| | - Vikram Babu Kasaragod
- Rudolf Virchow Centre for Integrative and Translational Bioimaging, Julius-Maximilians-University of Würzburg, Würzburg, Germany
| | - Verena Hörlin
- Institute of Clinical Neurobiology, University Hospital, Julius-Maximilians-University of Würzburg, Würzburg, Germany
| | - Anna-Lena Eckes
- Institute of Clinical Neurobiology, University Hospital, Julius-Maximilians-University of Würzburg, Würzburg, Germany
| | - Christoph J. Kluck
- Institute of Biochemistry, Emil-Fischer-Center, FAU Erlangen-Nürnberg, Erlangen, Germany
| | - Hermann Schindelin
- Rudolf Virchow Centre for Integrative and Translational Bioimaging, Julius-Maximilians-University of Würzburg, Würzburg, Germany
| | - Hans-Michael Meinck
- Department of Neurology, University Hospital Heidelberg, Heidelberg, Germany
| | - Jonathan Wickel
- Section Translational Neuroimmunology, Department of Neurology, Jena University Hospital, Jena, Germany
| | - Christian Geis
- Section Translational Neuroimmunology, Department of Neurology, Jena University Hospital, Jena, Germany
| | - Erdem Tüzün
- Institute of Experimental Medicine, Istanbul University, Istanbul, Türkiye
| | - Kathrin Doppler
- Department of Neurology, University Hospital Würzburg, Würzburg, Germany
| | - Claudia Sommer
- Department of Neurology, University Hospital Würzburg, Würzburg, Germany
| | - Carmen Villmann
- Institute of Clinical Neurobiology, University Hospital, Julius-Maximilians-University of Würzburg, Würzburg, Germany,*Correspondence: Carmen Villmann, ✉
| |
Collapse
|
21
|
Tian Q, Tong P, Chen G, Deng M, Cai T, Tian R, Zhang Z, Xia K, Hu Z. GLRA2 gene mutations cause high myopia in humans and mice. J Med Genet 2023; 60:193-203. [PMID: 35396272 PMCID: PMC9887403 DOI: 10.1136/jmedgenet-2022-108425] [Citation(s) in RCA: 7] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/04/2022] [Accepted: 03/16/2022] [Indexed: 02/03/2023]
Abstract
BACKGROUND High myopia (HM) is a leading cause of blindness that has a strong genetic predisposition. However, its genetic and pathogenic mechanisms remain largely unknown. Thus, this study aims to determine the genetic profile of individuals from two large Chinese families with HM and 200 patients with familial/sporadic HM. We also explored the pathogenic mechanism of HM using HEK293 cells and a mouse model. METHODS The participants underwent genome-wide linkage analysis and exome sequencing. Visual acuity, electroretinogram response, refractive error, optical parameters and retinal rod cell genesis were measured in knockout mice. Immunofluorescent staining, biotin-labelled membrane protein isolation and electrophysiological characterisation were conducted in cells transfected with overexpression plasmids. RESULTS A novel HM locus on Xp22.2-p11.4 was identified. Variant c.539C>T (p.Pro180Leu) in GLRA2 gene was co-segregated with HM in the two families. Another variant, c.458G>A (p.Arg153Gln), was identified in a sporadic sample. The Glra2 knockout mice showed myopia-related phenotypes, decreased electroretinogram responses and impaired retinal rod cell genesis. Variants c.458G>A and c.539C>T altered the localisation of GlyRα2 on the cell membrane and decreased agonist sensitivity. CONCLUSION GLRA2 was identified as a novel HM-causing gene. Its variants would cause HM through altered visual experience by impairing photoperception and visual transmission.
Collapse
Affiliation(s)
- Qi Tian
- Center for Medical Genetics, School of Life Sciences, Central South University, Changsha, Hunan, People's Republic of China,Hunan Key Laboratory of Medical Genetics, School of Life Sciences, Central South University, Changsha, Hunan, People's Republic of China,Hunan Key Laboratory of Animal Models for Human Disease, School of Life Sciences, Central South University, Changsha, Hunan, People's Republic of China
| | - Ping Tong
- Department of Ophthalmology, The Second Xiangya Hospital, Central South University, Changsha, Hunan, People's Republic of China
| | - Gong Chen
- Hunan Key Laboratory of Medical Genetics, School of Life Sciences, Central South University, Changsha, Hunan, People's Republic of China,Hunan Key Laboratory of Animal Models for Human Disease, School of Life Sciences, Central South University, Changsha, Hunan, People's Republic of China,Hunan Province Key Laboratory of Basic and Applied Hematology, School of Life Sciences, Central South University, Changsha, Hunan, People's Republic of China,Department of Biochemistry and Molecular Biology, School of Life Sciences, Central South University, Changsha, Hunan, People's Republic of China
| | - Meichun Deng
- Hunan Key Laboratory of Medical Genetics, School of Life Sciences, Central South University, Changsha, Hunan, People's Republic of China,Hunan Key Laboratory of Animal Models for Human Disease, School of Life Sciences, Central South University, Changsha, Hunan, People's Republic of China,Hunan Province Key Laboratory of Basic and Applied Hematology, School of Life Sciences, Central South University, Changsha, Hunan, People's Republic of China,Department of Biochemistry and Molecular Biology, School of Life Sciences, Central South University, Changsha, Hunan, People's Republic of China
| | - Tian'e Cai
- Reproductive Center, Sanya Central Hospital, Sanya, Hainan, People's Republic of China
| | - Runyi Tian
- Center for Medical Genetics, School of Life Sciences, Central South University, Changsha, Hunan, People's Republic of China,Hunan Key Laboratory of Medical Genetics, School of Life Sciences, Central South University, Changsha, Hunan, People's Republic of China,Hunan Key Laboratory of Animal Models for Human Disease, School of Life Sciences, Central South University, Changsha, Hunan, People's Republic of China
| | - Zimin Zhang
- Center for Medical Genetics, School of Life Sciences, Central South University, Changsha, Hunan, People's Republic of China,Hunan Key Laboratory of Medical Genetics, School of Life Sciences, Central South University, Changsha, Hunan, People's Republic of China,Hunan Key Laboratory of Animal Models for Human Disease, School of Life Sciences, Central South University, Changsha, Hunan, People's Republic of China
| | - Kun Xia
- Center for Medical Genetics, School of Life Sciences, Central South University, Changsha, Hunan, People's Republic of China .,Hunan Key Laboratory of Medical Genetics, School of Life Sciences, Central South University, Changsha, Hunan, People's Republic of China.,Hunan Key Laboratory of Animal Models for Human Disease, School of Life Sciences, Central South University, Changsha, Hunan, People's Republic of China
| | - Zhengmao Hu
- Center for Medical Genetics, School of Life Sciences, Central South University, Changsha, Hunan, People's Republic of China .,Hunan Key Laboratory of Medical Genetics, School of Life Sciences, Central South University, Changsha, Hunan, People's Republic of China.,Hunan Key Laboratory of Animal Models for Human Disease, School of Life Sciences, Central South University, Changsha, Hunan, People's Republic of China
| |
Collapse
|
22
|
Liu X, Wang W. Asymmetric gating of a human hetero-pentameric glycine receptor. RESEARCH SQUARE 2023:rs.3.rs-2386831. [PMID: 36711971 PMCID: PMC9882600 DOI: 10.21203/rs.3.rs-2386831/v1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Indexed: 01/11/2023]
Abstract
Hetero-pentameric Cys-loop receptors constitute a major type of neurotransmitter receptors that enable signal transmission and processing in the nervous system. Despite intense investigations in their working mechanism and pharmaceutical potentials, how neurotransmitters activate these receptors remain unclear due to the lack of high-resolution structural information in the activated open state. Here we report near-atomic resolution structures in all principle functional states of the human α1β GlyR, which is a major Cys-loop receptor that mediates inhibitory neurotransmission in the central nervous system of adults. Glycine binding induced cooperative and symmetric structural rearrangements in the neurotransmitter-binding extracellular domain, but asymmetrical pore dilation in the transmembrane domain. Symmetric response in the extracellular domain is consistent with electrophysiological data showing similar contribution to activation from all the α1 and β subunits. A set of functionally essential but differentially charged amino-acid residues in the transmembrane domain of the α1 and β subunits explains asymmetric activation. These findings point to a gating mechanism that is distinct from homomeric receptors but more compatible with heteromeric GlyRs being clustered at synapses through β subunit-scaffolding protein interactions. Such mechanism provides foundation for understanding how gating of the Cys-loop receptor members diverge to accommodate specific physiological environment.
Collapse
Affiliation(s)
- Xiaofen Liu
- University of Texas Southwestern Medical Center
| | - Weiwei Wang
- University of Texas Southwestern Medical Center
| |
Collapse
|
23
|
Lummis SCR, Dougherty DA. Expression of Mutant Glycine Receptors in Xenopus Oocytes Using Canonical and Non-Canonical Amino Acids Reveals Distinct Roles of Conserved Proline Residues. MEMBRANES 2022; 12:1012. [PMID: 36295771 PMCID: PMC9607081 DOI: 10.3390/membranes12101012] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 09/16/2022] [Revised: 10/05/2022] [Accepted: 10/08/2022] [Indexed: 06/16/2023]
Abstract
Pentameric ligand-gated ion channels (pLGIC) play important roles in fast neuronal signal transmission. Functional receptors are pentamers, with each subunit having an extracellular domain (ECD), a transmembrane domain (TMD) and an intracellular domain. The binding of the agonist to the ECD induces a structural change that is transduced to the TMD to open the channel. Molecular details of this process are emerging, but a comprehensive understanding is still lacking. Proline (Pro) is one amino acid that has attracted much interest; its unusual features generate bends in loops and kinks and bulges in helices, which can be essential for function in some pLGICs. Here, we explore the roles of four conserved Pros in the glycine receptor (GlyR), creating substitutions with canonical and noncanonical amino acids, characterizing them using two electrode voltage clamp electrophysiology in Xenopus oocytes, and interpreting changes in receptor parameters using structural data from the open and closed states of the receptor. The data reveal that for efficient function, the Pro in the α1β1 loop is needed to create a turn and to be the correct size and shape to interact with nearby residues; the peptide bond of the Pro in the Cys-loop requires the cis conformation; and the Pros in loop A and M1 allow efficient function because of their reduced hydrogen bonding capacity. These data are broadly consistent with data from other pLGICs, and therefore likely represent the important features of these Pros in all members of the family.
Collapse
Affiliation(s)
- Sarah C. R. Lummis
- Department of Biochemistry, University of Cambridge, Tennis Court Road, Cambridge CB2 1GA, UK
| | - Dennis A. Dougherty
- Division of Chemistry and Chemical Engineering, California Institute of Technology, Pasadena, CA 91125, USA
| |
Collapse
|
24
|
Cerdan AH, Peverini L, Changeux JP, Corringer PJ, Cecchini M. Lateral fenestrations in the extracellular domain of the glycine receptor contribute to the main chloride permeation pathway. SCIENCE ADVANCES 2022; 8:eadc9340. [PMID: 36240268 PMCID: PMC9565810 DOI: 10.1126/sciadv.adc9340] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 05/10/2022] [Accepted: 08/29/2022] [Indexed: 06/16/2023]
Abstract
Glycine receptors (GlyRs) are ligand-gated ion channels mediating signal transduction at chemical synapses. Since the early patch-clamp electrophysiology studies, the details of the ion permeation mechanism have remained elusive. Here, we combine molecular dynamics simulations of a zebrafish GlyR-α1 model devoid of the intracellular domain with mutagenesis and single-channel electrophysiology of the full-length human GlyR-α1. We show that lateral fenestrations between subunits in the extracellular domain provide the main translocation pathway for chloride ions to enter/exit a central water-filled vestibule at the entrance of the transmembrane channel. In addition, we provide evidence that these fenestrations are at the origin of current rectification in known anomalous mutants and design de novo two inward-rectifying channels by introducing mutations within them. These results demonstrate the central role of lateral fenestrations on synaptic neurotransmission.
Collapse
Affiliation(s)
- Adrien H. Cerdan
- Institut de Chimie de Strasbourg, UMR7177, CNRS, Université de Strasbourg, F-67083 Strasbourg Cedex, France
- Institut Pasteur, Université Paris Cité, CNRS UMR 3571, Channel-Receptors Unit, Paris, France
| | - Laurie Peverini
- Institut Pasteur, Université Paris Cité, CNRS UMR 3571, Channel-Receptors Unit, Paris, France
| | - Jean-Pierre Changeux
- Institut Pasteur, Université Paris Cité, CNRS UMR 3571, Channel-Receptors Unit, Paris, France
- Kavli Institute for Brain and Mind, University of California, San Diego, La Jolla, CA, USA
- Collège de France, Paris, France
| | - Pierre-Jean Corringer
- Institut Pasteur, Université Paris Cité, CNRS UMR 3571, Channel-Receptors Unit, Paris, France
| | - Marco Cecchini
- Institut de Chimie de Strasbourg, UMR7177, CNRS, Université de Strasbourg, F-67083 Strasbourg Cedex, France
| |
Collapse
|
25
|
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
|
26
|
Xu W, Wu L, Liu S, Liu X, Cao X, Zhou C, Zhang J, Fu Y, Guo Y, Wu Y, Tan Q, Wang L, Liu J, Jiang L, Fan Z, Pei Y, Yu J, Cheng J, Zhao S, Hao X, Liu ZJ, Hua T. Structural basis for strychnine activation of human bitter taste receptor TAS2R46. Science 2022; 377:1298-1304. [DOI: 10.1126/science.abo1633] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/02/2022]
Abstract
Taste sensing is a sophisticated chemosensory process, and bitter taste perception is mediated by type 2 taste receptors (TAS2Rs), or class T G protein–coupled receptors. Understanding the detailed molecular mechanisms behind taste sensation is hindered by a lack of experimental receptor structures. Here, we report the cryo–electron microscopy structures of human TAS2R46 complexed with chimeric mini–G protein gustducin, in both strychnine-bound and apo forms. Several features of TAS2R46 are disclosed, including distinct receptor structures that compare with known GPCRs, a new “toggle switch,” activation-related motifs, and precoupling with mini–G protein gustducin. Furthermore, the dynamic extracellular and more-static intracellular parts of TAS2R46 suggest possible diverse ligand-recognition and activation processes. This study provides a basis for further exploration of other bitter taste receptors and their therapeutic applications.
Collapse
Affiliation(s)
- Weixiu Xu
- iHuman Institute, ShanghaiTech University, Shanghai 201210, China
- School of Life Science and Technology, ShanghaiTech University, Shanghai 201210, China
| | - Lijie Wu
- iHuman Institute, ShanghaiTech University, Shanghai 201210, China
| | - Shenhui Liu
- iHuman Institute, ShanghaiTech University, Shanghai 201210, China
- School of Life Science and Technology, ShanghaiTech University, Shanghai 201210, China
| | - Xiao Liu
- iHuman Institute, ShanghaiTech University, Shanghai 201210, China
- School of Life Science and Technology, ShanghaiTech University, Shanghai 201210, China
| | - Xiaoling Cao
- iHuman Institute, ShanghaiTech University, Shanghai 201210, China
- School of Life Science and Technology, ShanghaiTech University, Shanghai 201210, China
| | - Cui Zhou
- iHuman Institute, ShanghaiTech University, Shanghai 201210, China
- School of Life Science and Technology, ShanghaiTech University, Shanghai 201210, China
| | - Jinyi Zhang
- iHuman Institute, ShanghaiTech University, Shanghai 201210, China
- School of Life Science and Technology, ShanghaiTech University, Shanghai 201210, China
| | - You Fu
- iHuman Institute, ShanghaiTech University, Shanghai 201210, China
- School of Life Science and Technology, ShanghaiTech University, Shanghai 201210, China
| | - Yu Guo
- iHuman Institute, ShanghaiTech University, Shanghai 201210, China
| | - Yiran Wu
- iHuman Institute, ShanghaiTech University, Shanghai 201210, China
| | - Qiwen Tan
- iHuman Institute, ShanghaiTech University, Shanghai 201210, China
| | - Ling Wang
- iHuman Institute, ShanghaiTech University, Shanghai 201210, China
| | - Junlin Liu
- iHuman Institute, ShanghaiTech University, Shanghai 201210, China
| | - Longquan Jiang
- iHuman Institute, ShanghaiTech University, Shanghai 201210, China
- School of Life Science and Technology, ShanghaiTech University, Shanghai 201210, China
| | - Zhongbo Fan
- iHuman Institute, ShanghaiTech University, Shanghai 201210, China
- School of Life Science and Technology, ShanghaiTech University, Shanghai 201210, China
| | - Yuan Pei
- iHuman Institute, ShanghaiTech University, Shanghai 201210, China
| | - Jingyi Yu
- School of Information Science and Technology, ShanghaiTech University, Shanghai 201210, China
| | - Jianjun Cheng
- iHuman Institute, ShanghaiTech University, Shanghai 201210, China
- School of Life Science and Technology, ShanghaiTech University, Shanghai 201210, China
| | - Suwen Zhao
- iHuman Institute, ShanghaiTech University, Shanghai 201210, China
- School of Life Science and Technology, ShanghaiTech University, Shanghai 201210, China
| | - Xiaojiang Hao
- State Key Laboratory of Phytochemistry and Plant Resource in West China, Kunming Institute of Botany, Chinese Academy of Sciences, Kunming 650210, China
| | - Zhi-Jie Liu
- iHuman Institute, ShanghaiTech University, Shanghai 201210, China
- School of Life Science and Technology, ShanghaiTech University, Shanghai 201210, China
| | - Tian Hua
- iHuman Institute, ShanghaiTech University, Shanghai 201210, China
- School of Life Science and Technology, ShanghaiTech University, Shanghai 201210, China
| |
Collapse
|
27
|
Kumar A, Kindig K, Rao S, Zaki AM, Basak S, Sansom MSP, Biggin PC, Chakrapani S. Structural basis for cannabinoid-induced potentiation of alpha1-glycine receptors in lipid nanodiscs. Nat Commun 2022; 13:4862. [PMID: 35982060 PMCID: PMC9388682 DOI: 10.1038/s41467-022-32594-5] [Citation(s) in RCA: 9] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/17/2022] [Accepted: 08/08/2022] [Indexed: 11/23/2022] Open
Abstract
Nociception and motor coordination are critically governed by glycine receptor (GlyR) function at inhibitory synapses. Consequentially, GlyRs are attractive targets in the management of chronic pain and in the treatment of several neurological disorders. High-resolution mechanistic details of GlyR function and its modulation are just emerging. While it has been known that cannabinoids such as Δ9-tetrahydrocannabinol (THC), the principal psychoactive constituent in marijuana, potentiate GlyR in the therapeutically relevant concentration range, the molecular mechanism underlying this effect is still not understood. Here, we present Cryo-EM structures of full-length GlyR reconstituted into lipid nanodisc in complex with THC under varying concentrations of glycine. The GlyR-THC complexes are captured in multiple conformational states that reveal the basis for THC-mediated potentiation, manifested as different extents of opening at the level of the channel pore. Taken together, these structural findings, combined with molecular dynamics simulations and functional analysis, provide insights into the potential THC binding site and the allosteric coupling to the channel pore.
Collapse
Affiliation(s)
- Arvind Kumar
- Department of Physiology and Biophysics, School of Medicine, Case Western Reserve University, Cleveland, OH, USA
| | - Kayla Kindig
- Department of Physiology and Biophysics, School of Medicine, Case Western Reserve University, Cleveland, OH, USA
| | - Shanlin Rao
- Department of Biochemistry, University of Oxford, Oxford, UK
| | | | - Sandip Basak
- Department of Physiology and Biophysics, School of Medicine, Case Western Reserve University, Cleveland, OH, USA
| | - Mark S P Sansom
- Department of Biochemistry, University of Oxford, Oxford, UK
| | - Philip C Biggin
- Department of Biochemistry, University of Oxford, Oxford, UK
| | - Sudha Chakrapani
- Department of Physiology and Biophysics, School of Medicine, Case Western Reserve University, Cleveland, OH, USA.
- Cleveland Center for Membrane and Structural Biology, School of Medicine, Case Western Reserve University, Cleveland, OH, USA.
- Department of Neuroscience, School of Medicine, Case Western Reserve University, Cleveland, OH, USA.
| |
Collapse
|
28
|
Zhu H. Structure and Mechanism of Glycine Receptor Elucidated by Cryo-Electron Microscopy. Front Pharmacol 2022; 13:925116. [PMID: 36016557 PMCID: PMC9395720 DOI: 10.3389/fphar.2022.925116] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/21/2022] [Accepted: 06/14/2022] [Indexed: 11/17/2022] Open
Abstract
Glycine receptors (GlyRs) are pentameric ion channels that mediate fast inhibitory neurotransmission. GlyRs are found in the central nervous system including the spinal cord, brain stem, and cerebellum, as well as in the retina, sperm, macrophages, hippocampus, cochlea, and liver. Due to their crucial roles in counter-balancing excitatory signals and pain signal transmission, GlyR dysfunction can lead to severe diseases, and as a result, compounds that modify GlyR activity may have tremendous therapeutic potential. Despite this potential, the development of GlyR-specific small-molecule ligands is lacking. Over the past few years, high-resolution structures of both homomeric and heteromeric GlyRs structures in various conformations have provided unprecedented details defining the pharmacology of ligand binding, subunit composition, and mechanisms of channel gating. These high-quality structures will undoubtedly help with the development of GlyR-targeted therapies.
Collapse
|
29
|
Hong B, Grzech D, Caputi L, Sonawane P, López CER, Kamileen MO, Hernández Lozada NJ, Grabe V, O'Connor SE. Biosynthesis of strychnine. Nature 2022; 607:617-622. [PMID: 35794473 PMCID: PMC9300463 DOI: 10.1038/s41586-022-04950-4] [Citation(s) in RCA: 53] [Impact Index Per Article: 26.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/01/2022] [Accepted: 06/08/2022] [Indexed: 11/08/2022]
Abstract
Strychnine is a natural product that, through isolation, structural elucidation and synthetic efforts, shaped the field of organic chemistry. Currently, strychnine is used as a pesticide to control rodents1 because of its potent neurotoxicity2,3. The polycyclic architecture of strychnine has inspired chemists to develop new synthetic transformations and strategies to access this molecular scaffold4, yet it is still unknown how plants create this complex structure. Here we report the biosynthetic pathway of strychnine, along with the related molecules brucine and diaboline. Moreover, we successfully recapitulate strychnine, brucine and diaboline biosynthesis in Nicotiana benthamiana from an upstream intermediate, thus demonstrating that this complex, pharmacologically active class of compounds can now be harnessed through metabolic engineering approaches.
Collapse
Affiliation(s)
- Benke Hong
- Department of Natural Product Biosynthesis, Max-Planck Institute for Chemical Ecology, Jena, Germany
| | - Dagny Grzech
- Department of Natural Product Biosynthesis, Max-Planck Institute for Chemical Ecology, Jena, Germany
| | - Lorenzo Caputi
- Department of Natural Product Biosynthesis, Max-Planck Institute for Chemical Ecology, Jena, Germany
| | - Prashant Sonawane
- Department of Natural Product Biosynthesis, Max-Planck Institute for Chemical Ecology, Jena, Germany
| | - Carlos E Rodríguez López
- Department of Natural Product Biosynthesis, Max-Planck Institute for Chemical Ecology, Jena, Germany
| | - Mohamed Omar Kamileen
- Department of Natural Product Biosynthesis, Max-Planck Institute for Chemical Ecology, Jena, Germany
| | - Néstor J Hernández Lozada
- Department of Natural Product Biosynthesis, Max-Planck Institute for Chemical Ecology, Jena, Germany
| | - Veit Grabe
- Microscopic Imaging Service Group, Max-Planck Institute for Chemical Ecology, Jena, Germany
| | - Sarah E O'Connor
- Department of Natural Product Biosynthesis, Max-Planck Institute for Chemical Ecology, Jena, Germany.
| |
Collapse
|
30
|
Mhashal AR, Yoluk O, Orellana L. Exploring the Conformational Impact of Glycine Receptor TM1-2 Mutations Through Coarse-Grained Analysis and Atomistic Simulations. Front Mol Biosci 2022; 9:890851. [PMID: 35836931 PMCID: PMC9275627 DOI: 10.3389/fmolb.2022.890851] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/06/2022] [Accepted: 05/05/2022] [Indexed: 11/13/2022] Open
Abstract
Pentameric ligand-gated ion channels (PLGICs) are a family of proteins that convert chemical signals into ion fluxes through cellular membranes. Their structures are highly conserved across all kingdoms from bacteria to eukaryotes. Beyond their classical roles in neurotransmission and neurological disorders, PLGICs have been recently related to cell proliferation and cancer. Here, we focus on the best characterized eukaryotic channel, the glycine receptor (GlyR), to investigate its mutational patterns in genomic-wide tumor screens and compare them with mutations linked to hyperekplexia (HPX), a Mendelian neuromotor disease that disrupts glycinergic currents. Our analysis highlights that cancer mutations significantly accumulate across TM1 and TM2, partially overlapping with HPX changes. Based on 3D-clustering, conservation, and phenotypic data, we select three mutations near the pore, expected to impact GlyR conformation, for further study by molecular dynamics (MD). Using principal components from experimental GlyR ensembles as framework, we explore the motions involved in transitions from the human closed and desensitized structures and how they are perturbed by mutations. Our MD simulations show that WT GlyR spontaneously explores opening and re-sensitization transitions that are significantly impaired by mutations, resulting in receptors with altered permeability and desensitization properties in agreement with HPX functional data.
Collapse
Affiliation(s)
| | | | - Laura Orellana
- Protein Dynamics and Cancer Lab, Department of Oncology-Pathology, Karolinska Institute, Solna, Sweden
| |
Collapse
|
31
|
Mesoy SM, Bridgland-Taylor M, Lummis SCR. Mutations of the nACh Receptor M4 Helix Reveal Different Phenotypes in Different Expression Systems: Could Lipids be Responsible? Front Physiol 2022; 13:850782. [PMID: 35600303 PMCID: PMC9116227 DOI: 10.3389/fphys.2022.850782] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/08/2022] [Accepted: 03/31/2022] [Indexed: 11/18/2022] Open
Abstract
The role of the outermost helix (M4) in the pentameric ligand-gated ion channel (pLGIC) family is currently not fully understood. It is known that M4 is important for receptor assembly, possibly via interactions with neighboring M1 and M3 helices. M4 can also transmit information on the lipid content of the membrane to the gating mechanism, and it may form a link to the extracellular domain via the Cys-loop. Our previous study examining the α4β2 nACh receptor M4 helix using HEK cells indicated M4 here is more sensitive to change than those of other pLGIC. Many of these other studies, however, were performed in Xenopus oocytes. Here we examine the nine previously identified nonfunctional α4β2 nACh receptor M4 mutant receptors using this system. The data reveal that seven of these mutant receptors do function when expressed in oocytes, with only 2, the conserved Asp at the intracellular end of M4 and a Phe in the center, having a similar phenotype (nonfunctional) in both HEK cells and oocytes. The oocyte data are more consistent with studies in other pLGIC and demonstrate the importance of the expression system used. Of the many differences between these two expression systems, we suggest that the different lipid content of the plasma membrane is a possible candidate for explaining these discrepancies.
Collapse
Affiliation(s)
- Susanne M. Mesoy
- Department of Biochemistry, University of Cambridge, University of Cambridge, Cambridge, United Kingdom
| | - Matthew Bridgland-Taylor
- Clinical Pharmacology and Safety Sciences, Biopharmaceuticals R&D, AstraZeneca, Cambridge, United Kingdom
| | - Sarah C. R. Lummis
- Department of Biochemistry, University of Cambridge, University of Cambridge, Cambridge, United Kingdom
- *Correspondence: Sarah C. R. Lummis,
| |
Collapse
|
32
|
Chen X, Wilson KA, Schaefer N, De Hayr L, Windsor M, Scalais E, van Rijckevorsel G, Stouffs K, Villmann C, O’Mara ML, Lynch JW, Harvey RJ. Loss, Gain and Altered Function of GlyR α2 Subunit Mutations in Neurodevelopmental Disorders. Front Mol Neurosci 2022; 15:886729. [PMID: 35571374 PMCID: PMC9103196 DOI: 10.3389/fnmol.2022.886729] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/28/2022] [Accepted: 04/07/2022] [Indexed: 12/11/2022] Open
Abstract
Glycine receptors (GlyRs) containing the α2 subunit govern cell fate, neuronal migration and synaptogenesis in the developing cortex and spinal cord. Rare missense variants and microdeletions in the X-linked GlyR α2 subunit gene (GLRA2) have been associated with human autism spectrum disorder (ASD), where they typically cause a loss-of-function via protein truncation, reduced cell-surface trafficking and/or reduced glycine sensitivity (e.g., GLRA2Δex8-9 and extracellular domain variants p.N109S and p.R126Q). However, the GlyR α2 missense variant p.R323L in the intracellular M3-M4 domain results in a gain-of-function characterized by slower synaptic decay times, longer duration active periods and increases in channel conductance. This study reports the functional characterization of four missense variants in GLRA2 associated with ASD or developmental disorders (p.V-22L, p.N38K, p.K213E, p.T269M) using a combination of bioinformatics, molecular dynamics simulations, cellular models of GlyR trafficking and electrophysiology in artificial synapses. The GlyR α2V–22L variant resulted in altered predicted signal peptide cleavage and a reduction in cell-surface expression, suggestive of a partial loss-of-function. Similarly, GlyR α2N38K homomers showed reduced cell-surface expression, a reduced affinity for glycine and a reduced magnitude of IPSCs in artificial synapses. By contrast, GlyR α2K213E homomers showed a slight reduction in cell-surface expression, but IPSCs were larger, with faster rise/decay times, suggesting a gain-of-function. Lastly, GlyR α2T269M homomers exhibited a high glycine sensitivity accompanied by a substantial leak current, suggestive of an altered function that could dramatically enhance glycinergic signaling. These results may explain the heterogeneity of clinical phenotypes associated with GLRA2 mutations and reveal that missense variants can result in a loss, gain or alteration of GlyR α2 function. In turn, these GlyR α2 missense variants are likely to either negatively or positively deregulate cortical progenitor homeostasis and neuronal migration in the developing brain, leading to changes in cognition, learning, and memory.
Collapse
Affiliation(s)
- Xiumin Chen
- Queensland Brain Institute, The University of Queensland, Brisbane, QLD, Australia
| | - Katie A. Wilson
- Research School of Chemistry, The Australian National University, Canberra, ACT, Australia
| | - Natascha Schaefer
- Institute of Clinical Neurobiology, University Hospital, Julius-Maximilians-University Würzburg, Würzburg, Germany
| | - Lachlan De Hayr
- School of Health and Behavioural Sciences, University of the Sunshine Coast, Maroochydore, QLD, Australia
- Sunshine Coast Health Institute, Birtinya, QLD, Australia
| | - Mark Windsor
- School of Health and Behavioural Sciences, University of the Sunshine Coast, Maroochydore, QLD, Australia
- Sunshine Coast Health Institute, Birtinya, QLD, Australia
| | - Emmanuel Scalais
- Neurologie Pédiatrique, Centre Hospitalier de Luxembourg, Luxembourg, Luxembourg
| | | | - Katrien Stouffs
- Center for Medical Genetics, Universitair Ziekenhuis Brussel, Vrije Universiteit Brussel, Brussels, Belgium
| | - Carmen Villmann
- Institute of Clinical Neurobiology, University Hospital, Julius-Maximilians-University Würzburg, Würzburg, Germany
| | - Megan L. O’Mara
- Research School of Chemistry, The Australian National University, Canberra, ACT, Australia
- Australian Institute for Bioengineering and Nanotechnology (AIBN), The University of Queensland, Brisbane, QLD, Australia
| | - Joseph W. Lynch
- Queensland Brain Institute, The University of Queensland, Brisbane, QLD, Australia
| | - Robert J. Harvey
- School of Health and Behavioural Sciences, University of the Sunshine Coast, Maroochydore, QLD, Australia
- Sunshine Coast Health Institute, Birtinya, QLD, Australia
- *Correspondence: Robert J. Harvey,
| |
Collapse
|
33
|
Zlotos DP, Mandour YM, Jensen AA. Strychnine and its mono- and dimeric analogues: a pharmaco-chemical perspective. Nat Prod Rep 2022; 39:1910-1937. [PMID: 35380133 DOI: 10.1039/d1np00079a] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2022]
Abstract
Covering: up to November 2021Since its isolation in 1818, strychnine has attracted the attention of a plethora of chemists and pharmacologists who have established its structure, developed total syntheses, and examined its complex pharmacology. While numerous reviews on structure elucidation and total synthesis of strychnine are available, reports on structure-activity relationships (SARs) of this fascinating alkaloid are rare. In this review, we present and discuss structures, synthetic approaches, metabolic transformations, and the diverse pharmacological actions of strychnine and its mono- and dimeric analogues. Particular attention is given to its SARs at glycine receptors (GlyRs) in light of recently published high-resolution structures of strychnine-GlyR complexes. Other pharmacological actions of strychnine and its derivatives, such as their antagonistic properties at nicotinic acetylcholine receptors (nAChRs), allosteric modulation of muscarinic acetylcholine receptors as well as anti-cancer and anti-plasmodial effects are also critically reviewed, and possible future developments in the field are discussed.
Collapse
Affiliation(s)
- Darius P Zlotos
- Department of Pharmaceutical Chemistry, Faculty of Pharmacy and Biotechnology, The German University in Cairo, New Cairo City, 11835 Cairo, Egypt.
| | - Yasmine M Mandour
- School of Life and Medical Sciences, University of Hertfordshire hosted by Global Academic Foundation, New Administrative Capital, Cairo, Egypt
| | - Anders A Jensen
- Department of Drug Design and Pharmacology, Faculty of Health and Medical Sciences, University of Copenhagen, DK-2100 Copenhagen, Denmark
| |
Collapse
|
34
|
Olsson Y, Lidö H, Ericson M, Söderpalm B. The glycine-containing dipeptide leucine-glycine raises accumbal dopamine levels in a subpopulation of rats presenting a lower endogenous dopamine tone. J Neural Transm (Vienna) 2022; 129:395-407. [PMID: 35322277 PMCID: PMC9007805 DOI: 10.1007/s00702-022-02487-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/28/2022] [Accepted: 03/09/2022] [Indexed: 11/26/2022]
Abstract
Interventions that elevate glycine levels and target the glycine receptor (GlyR) in the nucleus Accumbens (nAc) reduce ethanol intake in rats, supposedly by acting on the brain reward system via increased basal and attenuated ethanol-induced nAc dopamine release. Glycine transport across the blood brain barrier (BBB) appears inefficient, but glycine-containing dipeptides elevate whole brain tissue dopamine levels in mice. This study explores whether treatment with the glycine-containing dipeptides leucine-glycine (Leu-Gly) and glycine-leucine (Gly-Leu) by means of a hypothesized, facilitated BBB passage, alter nAc glycine and dopamine levels and locomotor activity in two rodent models. The acute effects of Leu-Gly and Gly-Leu (1–1000 mg/kg, i.p.) alone or Leu-Gly in combination with ethanol on locomotion in male NMRI mice were examined in locomotor activity boxes. Striatal and brainstem slices were obtained for ex vivo HPLC analyses of tissue levels of glycine and dopamine. Furthermore, the effects of Leu-Gly i.p. (1–1000 mg/kg) on glycine and dopamine output in the nAc were examined using in vivo microdialysis coupled to HPLC in freely moving male Wistar rats. Leu-Gly and Gly-Leu did not significantly alter locomotion, ethanol-induced hyperlocomotor activity or tissue levels of glycine or dopamine, apart from Gly-Leu 10 mg/kg that slightly raised nAc dopamine. Microdialysis revealed no significant alterations in nAc glycine or dopamine levels when regarding all rats as a homogenous group. In a subgroup of rats defined as dopamine responders, a significant elevation of nAc dopamine (20%) was seen following Leu-Gly 10–1000 mg/kg i.p, and this group of animals presented lower baseline dopamine levels compared to dopamine non-responders. To conclude, peripheral injection of glycine-containing dipeptides appears inefficient in elevating central glycine levels but raises accumbal dopamine levels in a subgroup of rats with a lower endogenous dopamine tone. The tentative relationship between dopamine baseline and ensuing response to glycinergic treatment and presumptive direct interactions between glycine-containing dipeptides and the GlyR bear insights for refinement of the glycinergic treatment concept for alcohol use disorder (AUD).
Collapse
Affiliation(s)
- Yasmin Olsson
- Addiction Biology Unit, Department of Psychiatry and Neurochemistry, Institute of Neuroscience and Physiology, Sahlgrenska Academy, University of Gothenburg, PO Box 410, 405 30, Gothenburg, Sweden.
- Beroendekliniken, Sahlgrenska University Hospital, Gothenburg, Sweden.
- Department of Neurology, Sahlgrenska University Hospital, Gothenburg, Sweden.
| | - Helga Lidö
- Addiction Biology Unit, Department of Psychiatry and Neurochemistry, Institute of Neuroscience and Physiology, Sahlgrenska Academy, University of Gothenburg, PO Box 410, 405 30, Gothenburg, Sweden
- Beroendekliniken, Sahlgrenska University Hospital, Gothenburg, Sweden
| | - Mia Ericson
- Addiction Biology Unit, Department of Psychiatry and Neurochemistry, Institute of Neuroscience and Physiology, Sahlgrenska Academy, University of Gothenburg, PO Box 410, 405 30, Gothenburg, Sweden
| | - Bo Söderpalm
- Addiction Biology Unit, Department of Psychiatry and Neurochemistry, Institute of Neuroscience and Physiology, Sahlgrenska Academy, University of Gothenburg, PO Box 410, 405 30, Gothenburg, Sweden
- Beroendekliniken, Sahlgrenska University Hospital, Gothenburg, Sweden
| |
Collapse
|
35
|
San Martín VP, Sazo A, Utreras E, Moraga-Cid G, Yévenes GE. Glycine Receptor Subtypes and Their Roles in Nociception and Chronic Pain. Front Mol Neurosci 2022; 15:848642. [PMID: 35401105 PMCID: PMC8984470 DOI: 10.3389/fnmol.2022.848642] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/04/2022] [Accepted: 01/28/2022] [Indexed: 01/23/2023] Open
Abstract
Disruption of the inhibitory control provided by the glycinergic system is one of the major mechanisms underlying chronic pain. In line with this concept, recent studies have provided robust proof that pharmacological intervention of glycine receptors (GlyRs) restores the inhibitory function and exerts anti-nociceptive effects on preclinical models of chronic pain. A targeted regulation of the glycinergic system requires the identification of the GlyR subtypes involved in chronic pain states. Nevertheless, the roles of individual GlyR subunits in nociception and in chronic pain are yet not well defined. This review aims to provide a systematic outline on the contribution of GlyR subtypes in chronic pain mechanisms, with a particular focus on molecular pathways of spinal glycinergic dis-inhibition mediated by post-translational modifications at the receptor level. The current experimental evidence has shown that phosphorylation of synaptic α1β and α3β GlyRs are involved in processes of spinal glycinergic dis-inhibition triggered by chronic inflammatory pain. On the other hand, the participation of α2-containing GlyRs and of β subunits in pain signaling have been less studied and remain undefined. Although many questions in the field are still unresolved, future progress in GlyR research may soon open new exciting avenues into understanding and controlling chronic pain.
Collapse
Affiliation(s)
- Victoria P. San Martín
- Department of Physiology, Faculty of Biological Sciences, University of Concepcion, Concepcion, Chile
- Millennium Nucleus for the Study of Pain (MiNuSPain), Santiago, Chile
| | - Anggelo Sazo
- Department of Physiology, Faculty of Biological Sciences, University of Concepcion, Concepcion, Chile
- Millennium Nucleus for the Study of Pain (MiNuSPain), Santiago, Chile
| | - Elías Utreras
- Millennium Nucleus for the Study of Pain (MiNuSPain), Santiago, Chile
- Department of Biology, Faculty of Science, Universidad de Chile, Santiago, Chile
| | - Gustavo Moraga-Cid
- Department of Physiology, Faculty of Biological Sciences, University of Concepcion, Concepcion, Chile
- Millennium Nucleus for the Study of Pain (MiNuSPain), Santiago, Chile
| | - Gonzalo E. Yévenes
- Department of Physiology, Faculty of Biological Sciences, University of Concepcion, Concepcion, Chile
- Millennium Nucleus for the Study of Pain (MiNuSPain), Santiago, Chile
- *Correspondence: Gonzalo E. Yévenes,
| |
Collapse
|
36
|
Felsztyna I, Villarreal MA, García DA, Miguel V. Insect RDL Receptor Models for Virtual Screening: Impact of the Template Conformational State in Pentameric Ligand-Gated Ion Channels. ACS OMEGA 2022; 7:1988-2001. [PMID: 35071887 PMCID: PMC8771969 DOI: 10.1021/acsomega.1c05465] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 10/01/2021] [Accepted: 12/28/2021] [Indexed: 06/14/2023]
Abstract
The RDL receptor is one of the most relevant protein targets for insecticide molecules. It belongs to the pentameric ligand-gated ion channel (pLGIC) family. Given that the experimental structures of pLGICs are difficult to obtain, homology modeling has been extensively used for these proteins, particularly for the RDL receptor. However, no detailed assessments of the usefulness of homology models for virtual screening (VS) have been carried out for pLGICs. The aim of this study was to evaluate which are the determinant factors for a good VS performance using RDL homology models, specially analyzing the impact of the template conformational state. Fifteen RDL homology models were obtained based on different pLGIC templates representing the closed, open, and desensitized states. A retrospective VS process was performed on each model, and their performance in the prioritization of active ligands was assessed. In addition, the three best-performing models among each of the conformations were subjected to molecular dynamics simulations (MDS) in complex with a representative active ligand. The models showed variations in their VS performance parameters that were related to the structural properties of the binding site. VS performance tended to improve in more constricted binding cavities. The best performance was obtained with a model based on a template in the closed conformation. MDS confirmed that the closed model was the one that best represented the interactions with an active ligand. These results imply that different templates should be evaluated and the structural variations between their channel conformational states should be specially examined, providing guidelines for the application of homology modeling for VS in other proteins of the pLGIC family.
Collapse
Affiliation(s)
- Iván Felsztyna
- Facultad
de Ciencias Exactas, Físicas y Naturales, Departamento de Química.
Cátedra de Química Biológica, Universidad Nacional de Córdoba, Córdoba 5016, Argentina
- Instituto
de Investigaciones Biológicas y Tecnológicas (IIByT), CONICET-Universidad Nacional de Córdoba, Córdoba 5016, Argentina
| | - Marcos A. Villarreal
- Facultad
de Ciencias Químicas, Departamento de Química Teórica
y Computacional, Universidad Nacional de
Córdoba, Córdoba 5016, Argentina
- Instituto
de Investigaciones en Físico-Química de Córdoba
(INFIQC), CONICET-Universidad Nacional de
Córdoba, Córdoba 5016, Argentina
| | - Daniel A. García
- Facultad
de Ciencias Exactas, Físicas y Naturales, Departamento de Química.
Cátedra de Química Biológica, Universidad Nacional de Córdoba, Córdoba 5016, Argentina
- Instituto
de Investigaciones Biológicas y Tecnológicas (IIByT), CONICET-Universidad Nacional de Córdoba, Córdoba 5016, Argentina
| | - Virginia Miguel
- Facultad
de Ciencias Exactas, Físicas y Naturales, Departamento de Química.
Cátedra de Química Biológica, Universidad Nacional de Córdoba, Córdoba 5016, Argentina
- Instituto
de Investigaciones Biológicas y Tecnológicas (IIByT), CONICET-Universidad Nacional de Córdoba, Córdoba 5016, Argentina
| |
Collapse
|
37
|
Montgomery M, Rendine S, Zimmer CT, Elias J, Schaetzer J, Pitterna T, Benfatti F, Skaljac M, Bigot A. Structural Biology-Guided Design, Synthesis, and Biological Evaluation of Novel Insect Nicotinic Acetylcholine Receptor Orthosteric Modulators. J Med Chem 2022; 65:2297-2312. [PMID: 34986308 DOI: 10.1021/acs.jmedchem.1c01767] [Citation(s) in RCA: 17] [Impact Index Per Article: 8.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022]
Abstract
The development of novel and safe insecticides remains an important need for a growing world population to protect crops and animal and human health. New chemotypes modulating the insect nicotinic acetylcholine receptors have been recently brought to the agricultural market, yet with limited understanding of their molecular interactions at their target receptor. Herein, we disclose the first crystal structures of these insecticides, namely, sulfoxaflor, flupyradifurone, triflumezopyrim, flupyrimin, and the experimental compound, dicloromezotiaz, in a double-mutated acetylcholine-binding protein which mimics the insect-ion-channel orthosteric site. Enabled by these findings, we discovered novel pharmacophores with a related mode of action, and we describe herein their design, synthesis, and biological evaluation.
Collapse
Affiliation(s)
- Mark Montgomery
- Syngenta Crop Protection, Jealott's Hill International Research Centre, RG42 6EY Bracknell, Berkshire, U.K
| | - Stefano Rendine
- Syngenta Crop Protection AG, Schaffhauserstrasse 101, CH-4332 Stein, Switzerland
| | - Christoph T Zimmer
- Syngenta Crop Protection AG, Schaffhauserstrasse 101, CH-4332 Stein, Switzerland
| | - Jan Elias
- Syngenta Crop Protection AG, Rosentalstrasse 67, 4002 Basel, Switzerland
| | - Jürgen Schaetzer
- Syngenta Crop Protection AG, Schaffhauserstrasse 101, CH-4332 Stein, Switzerland
| | - Thomas Pitterna
- Syngenta Crop Protection AG, Schaffhauserstrasse 101, CH-4332 Stein, Switzerland
| | - Fides Benfatti
- Syngenta Crop Protection AG, Schaffhauserstrasse 101, CH-4332 Stein, Switzerland
| | - Marisa Skaljac
- Syngenta Crop Protection AG, Schaffhauserstrasse 101, CH-4332 Stein, Switzerland
| | - Aurélien Bigot
- Syngenta Crop Protection AG, Schaffhauserstrasse 101, CH-4332 Stein, Switzerland
| |
Collapse
|
38
|
Yin X, Yang GF, Niu DB, Chen J, Liao M, Cao HQ, Sheng CW. Identification and pharmacological characterization of histamine-gated chloride channels in the fall armyworm, Spodoptera frugiperda. INSECT BIOCHEMISTRY AND MOLECULAR BIOLOGY 2022; 140:103698. [PMID: 34848284 DOI: 10.1016/j.ibmb.2021.103698] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/12/2021] [Revised: 11/15/2021] [Accepted: 11/17/2021] [Indexed: 06/13/2023]
Abstract
Histamine-gated chloride channels (HACls) mediate fast inhibitory neurotransmission in invertebrate nervous systems and have important roles in light reception, color processing, temperature preference and light-dark cycle. The fall armyworm, Spodoptera frugiperda is a main destructive pest of grain and row crops. However, the pharmacological characterization of HACls in S. frugiperda remain unknown. In this study, we identified two cDNAs encoding SfHACl1 and SfHACl2 in S. frugiperda. They had similar expression patterns and were most abundantly expressed in the head of larvae and at the egg stage. Electrophysiological analysis with the two-electrode voltage clamp method showed that histamine (HA) and γ-aminobutyric acid (GABA) activated inward currents when SfHACls were singly or collectively expressed with different ratios in Xenopus laevis oocytes. These channels were ≥2000-fold more sensitive to HA than to GABA. They were anion-selective channels, which were highly dependent on changes in external chloride concentrations, but insensitive to changes in external sodium concentrations. The insecticides abamectin (ABM) and emamectin benzoate (EB) also activated these channels with the EC50 to SfHACl1 lower than that to SfHACl2. And the EC50s of ABM and EB to the co-expressed channels gradually increased with increase in the injection ratio of SfHACl2 cRNA. Homology models and docking simulations revealed that HA bound to the large amino-terminal extracellular domain of SfHACl1 and SfHACl2 by forming 4 and 2 hydrogen bonds, respectively. The docking simulations of ABM and EB had similar binding sites in the transmembrane regions. Overall, these findings indicated that HACls act as targets for macrolide, and this study provides theoretical guidance for further derivatization of abamectin insecticides.
Collapse
Affiliation(s)
- Xue Yin
- Anhui Province Key Laboratory of Integrated Pest Management on Crops, School of Plant Protection, Anhui Agricultural University, Hefei, 230036, PR China
| | - Guo-Feng Yang
- Anhui Province Key Laboratory of Integrated Pest Management on Crops, School of Plant Protection, Anhui Agricultural University, Hefei, 230036, PR China
| | - Duo-Bang Niu
- Anhui Province Key Laboratory of Integrated Pest Management on Crops, School of Plant Protection, Anhui Agricultural University, Hefei, 230036, PR China
| | - Jiao Chen
- Affiliated Hospital of Integrated Traditional Chinese and Western Medicine, Nanjing University of Chinese Medicine, Nanjing, 210028, PR China
| | - Min Liao
- Anhui Province Engineering Laboratory for Green Pesticide Development and Application, School of Plant Protection, Anhui Agricultural University, Hefei, 230036, PR China
| | - Hai-Qun Cao
- Anhui Province Key Laboratory of Integrated Pest Management on Crops, School of Plant Protection, Anhui Agricultural University, Hefei, 230036, PR China; Anhui Province Engineering Laboratory for Green Pesticide Development and Application, School of Plant Protection, Anhui Agricultural University, Hefei, 230036, PR China.
| | - Cheng-Wang Sheng
- Anhui Province Key Laboratory of Integrated Pest Management on Crops, School of Plant Protection, Anhui Agricultural University, Hefei, 230036, PR China; Anhui Province Engineering Laboratory for Green Pesticide Development and Application, School of Plant Protection, Anhui Agricultural University, Hefei, 230036, PR China.
| |
Collapse
|
39
|
Piro I, Eckes AL, Kasaragod VB, Sommer C, Harvey RJ, Schaefer N, Villmann C. Novel Functional Properties of Missense Mutations in the Glycine Receptor β Subunit in Startle Disease. Front Mol Neurosci 2021; 14:745275. [PMID: 34630038 PMCID: PMC8498107 DOI: 10.3389/fnmol.2021.745275] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/21/2021] [Accepted: 09/03/2021] [Indexed: 11/13/2022] Open
Abstract
Startle disease is a rare disorder associated with mutations in GLRA1 and GLRB, encoding glycine receptor (GlyR) α1 and β subunits, which enable fast synaptic inhibitory transmission in the spinal cord and brainstem. The GlyR β subunit is important for synaptic localization via interactions with gephyrin and contributes to agonist binding and ion channel conductance. Here, we have studied three GLRB missense mutations, Y252S, S321F, and A455P, identified in startle disease patients. For Y252S in M1 a disrupted stacking interaction with surrounding aromatic residues in M3 and M4 is suggested which is accompanied by an increased EC50 value. By contrast, S321F in M3 might stabilize stacking interactions with aromatic residues in M1 and M4. No significant differences in glycine potency or efficacy were observed for S321F. The A455P variant was not predicted to impact on subunit folding but surprisingly displayed increased maximal currents which were not accompanied by enhanced surface expression, suggesting that A455P is a gain-of-function mutation. All three GlyR β variants are trafficked effectively with the α1 subunit through intracellular compartments and inserted into the cellular membrane. In vivo, the GlyR β subunit is transported together with α1 and the scaffolding protein gephyrin to synaptic sites. The interaction of these proteins was studied using eGFP-gephyrin, forming cytosolic aggregates in non-neuronal cells. eGFP-gephyrin and β subunit co-expression resulted in the recruitment of both wild-type and mutant GlyR β subunits to gephyrin aggregates. However, a significantly lower number of GlyR β aggregates was observed for Y252S, while for mutants S321F and A455P, the area and the perimeter of GlyR β subunit aggregates was increased in comparison to wild-type β. Transfection of hippocampal neurons confirmed differences in GlyR-gephyrin clustering with Y252S and A455P, leading to a significant reduction in GlyR β-positive synapses. Although none of the mutations studied is directly located within the gephyrin-binding motif in the GlyR β M3-M4 loop, we suggest that structural changes within the GlyR β subunit result in differences in GlyR β-gephyrin interactions. Hence, we conclude that loss- or gain-of-function, or alterations in synaptic GlyR clustering may underlie disease pathology in startle disease patients carrying GLRB mutations.
Collapse
Affiliation(s)
- Inken Piro
- Department of Neurology, University Hospital Würzburg, Würzburg, Germany
| | - Anna-Lena Eckes
- Institute for Clinical Neurobiology, University Hospital, Julius-Maximilians-University Würzburg, Würzburg, Germany
| | - Vikram Babu Kasaragod
- Neurobiology Division, MRC Laboratory of Molecular Biology, Cambridge, United Kingdom
| | - Claudia Sommer
- Department of Neurology, University Hospital Würzburg, Würzburg, Germany
| | - Robert J. Harvey
- School of Health and Behavioural Sciences, University of the Sunshine Coast, Maroochydore, QLD, Australia
- Sunshine Coast Health Institute, Birtinya, QLD, Australia
| | - Natascha Schaefer
- Institute for Clinical Neurobiology, University Hospital, Julius-Maximilians-University Würzburg, Würzburg, Germany
| | - Carmen Villmann
- Institute for Clinical Neurobiology, University Hospital, Julius-Maximilians-University Würzburg, Würzburg, Germany
| |
Collapse
|
40
|
Han L, Shan Q. Different Behaviors of a Glycine Receptor Channel Pore Residue between Wild-Type-Mimicking and Disease-Type-Mimicking Formats. ACS Chem Neurosci 2021; 12:3397-3409. [PMID: 34460217 DOI: 10.1021/acschemneuro.1c00386] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/05/2023] Open
Abstract
The glycine receptor (GlyR) is a neurotransmitter-gated chloride channel that mediates fast inhibitory neurotransmission, predominantly in the spinal cord and brain stem. Mutations of the GlyR are the major cause of hereditary hyperekplexia. Site-specific cysteine substitution followed by labeling with a fluorophore has previously been used to explore the behaviors of the hyperekplexia-related 271 (19') residue of the GlyR. However, this manipulation dramatically compromises sensitivity toward the agonist glycine and alters the pharmacological effects of various agents in manners similar to those of the hyperekplexia-causing R19'Q/L mutations, raising the question whether what is reported by the substituted and modified residue faithfully reflects what actually happens to the wild-type (WT) residue. In this study, a mechanism-rescuing second-site mutation was introduced to create a WT-mimicking GlyR (with the 19' residue cysteine substitution and modification still in place), in which the sensitivity toward glycine and pharmacological effects of various agents were restored. Further experiments revealed stark differences in the behaviors upon the various pharmacological treatments and consequently the underlying mechanisms of the 19' residue between this WT-mimicking GlyR and the GlyR without the mechanism rescue, which is correspondingly defined as the disease-type (DT)-mimicking GlyR. The data presented in this study warn generally that caution is required when attempting to deduce the behaviors of a WT residue from data based on substituted or modified residues that alter protein structure and function. Extra measures, such as rescuing mechanisms via alternative means as presented in this study, are needed to mitigate this challenge.
Collapse
Affiliation(s)
- Lu Han
- Zhejiang Key Laboratory of Organ Development and Regeneration, College of Life and Environmental Sciences, Hangzhou Normal University, Hangzhou, Zhejiang 311121, China
| | - Qiang Shan
- Laboratory for Synaptic Plasticity, Shantou University Medical College, Shantou, Guangdong 515041, China
| |
Collapse
|
41
|
Yu H, Bai XC, Wang W. Characterization of the subunit composition and structure of adult human glycine receptors. Neuron 2021; 109:2707-2716.e6. [PMID: 34473954 DOI: 10.1016/j.neuron.2021.08.019] [Citation(s) in RCA: 29] [Impact Index Per Article: 9.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/09/2021] [Revised: 07/23/2021] [Accepted: 08/16/2021] [Indexed: 11/27/2022]
Abstract
The strychnine-sensitive pentameric glycine receptor (GlyR) mediates fast inhibitory neurotransmission in the mammalian nervous system. Only heteromeric GlyRs mediate synaptic transmission, as they contain the β subunit that permits clustering at the synapse through its interaction with scaffolding proteins. Here, we show that α2 and β subunits assemble with an unexpected 4:1 stoichiometry to produce GlyR with native electrophysiological properties. We determined structures in multiple functional states at 3.6-3.8 Å resolutions and show how 4:1 stoichiometry is consistent with the structural features of α2β GlyR. Furthermore, we show that one single β subunit in each GlyR gives rise to the characteristic electrophysiological properties of heteromeric GlyR, while more β subunits render GlyR non-conductive. A single β subunit ensures a univalent GlyR-scaffold linkage, which means the scaffold alone regulates the cluster properties.
Collapse
Affiliation(s)
- Hailong Yu
- Department of Biophysics, University of Texas Southwestern Medical Center, Dallas, TX 75390, USA
| | - Xiao-Chen Bai
- Department of Biophysics, University of Texas Southwestern Medical Center, Dallas, TX 75390, USA
| | - Weiwei Wang
- Department of Biophysics, University of Texas Southwestern Medical Center, Dallas, TX 75390, USA.
| |
Collapse
|
42
|
Sridhar A, Lummis SCR, Pasini D, Mehregan A, Brams M, Kambara K, Bertrand D, Lindahl E, Howard RJ, Ulens C. Regulation of a pentameric ligand-gated ion channel by a semiconserved cationic lipid-binding site. J Biol Chem 2021; 297:100899. [PMID: 34157288 PMCID: PMC8327344 DOI: 10.1016/j.jbc.2021.100899] [Citation(s) in RCA: 11] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/23/2021] [Revised: 06/10/2021] [Accepted: 06/18/2021] [Indexed: 02/08/2023] Open
Abstract
Pentameric ligand-gated ion channels (pLGICs) are crucial mediators of electrochemical signal transduction in various organisms from bacteria to humans. Lipids play an important role in regulating pLGIC function, yet the structural bases for specific pLGIC-lipid interactions remain poorly understood. The bacterial channel ELIC recapitulates several properties of eukaryotic pLGICs, including activation by the neurotransmitter GABA and binding and modulation by lipids, offering a simplified model system for structure-function relationship studies. In this study, functional effects of noncanonical amino acid substitution of a potential lipid-interacting residue (W206) at the top of the M1-helix, combined with detergent interactions observed in recent X-ray structures, are consistent with this region being the location of a lipid-binding site on the outward face of the ELIC transmembrane domain. Coarse-grained and atomistic molecular dynamics simulations revealed preferential binding of lipids containing a positive charge, particularly involving interactions with residue W206, consistent with cation-π binding. Polar contacts from other regions of the protein, particularly M3 residue Q264, further support lipid binding via headgroup ester linkages. Aromatic residues were identified at analogous sites in a handful of eukaryotic family members, including the human GABAA receptor ε subunit, suggesting conservation of relevant interactions in other evolutionary branches. Further mutagenesis experiments indicated that mutations at this site in ε-containing GABAA receptors can change the apparent affinity of the agonist response to GABA, suggesting a potential role of this site in channel gating. In conclusion, this work details type-specific lipid interactions, which adds to our growing understanding of how lipids modulate pLGICs.
Collapse
Affiliation(s)
- Akshay Sridhar
- Department of Applied Physics, Science for Life Laboratory, KTH Royal Institute of Technology, Solna, Sweden
| | - Sarah C R Lummis
- Department of Biochemistry, University of Cambridge, Cambridge, United Kingdom
| | - Diletta Pasini
- Laboratory of Structural Neurobiology, Department of Cellular and Molecular Medicine, Faculty of Medicine, KU Leuven, Leuven, Belgium
| | - Aujan Mehregan
- Laboratory of Structural Neurobiology, Department of Cellular and Molecular Medicine, Faculty of Medicine, KU Leuven, Leuven, Belgium
| | - Marijke Brams
- Laboratory of Structural Neurobiology, Department of Cellular and Molecular Medicine, Faculty of Medicine, KU Leuven, Leuven, Belgium
| | | | | | - Erik Lindahl
- Department of Applied Physics, Science for Life Laboratory, KTH Royal Institute of Technology, Solna, Sweden; Department of Biochemistry and Biophysics, Science for Life Laboratory, Stockholm University, Solna, Sweden
| | - Rebecca J Howard
- Department of Biochemistry and Biophysics, Science for Life Laboratory, Stockholm University, Solna, Sweden.
| | - Chris Ulens
- Laboratory of Structural Neurobiology, Department of Cellular and Molecular Medicine, Faculty of Medicine, KU Leuven, Leuven, Belgium.
| |
Collapse
|
43
|
Tong G, Baker MA, Shenvi RA. Change the channel: CysLoop receptor antagonists from nature. PEST MANAGEMENT SCIENCE 2021; 77:3650-3662. [PMID: 33135373 PMCID: PMC8087819 DOI: 10.1002/ps.6166] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/31/2020] [Revised: 10/27/2020] [Accepted: 11/01/2020] [Indexed: 05/04/2023]
Abstract
Vertebrate and invertebrate ligand-gated ion channels (LGICs) exhibit significant structural homology and often share ligands. As a result, ligands with activity against one class can be brought to bear against another, including for development as insecticides. Receptor selectivity, metabolism and distribution must then be optimized using chemical synthesis. Here we review natural products (NPs) that ligate and inhibit the Cys-loop family of LGICs, which benefit from the unique physicochemical properties of natural product space but often present a high synthetic burden. Recent advances in chemical synthesis, however, have opened practical entries into these complex structures, several of which are highlighted. © 2020 Society of Chemical Industry.
Collapse
Affiliation(s)
- Guanghu Tong
- Department of Chemistry, Scripps Research, La Jolla, CA, USA
| | - Meghan A Baker
- Department of Chemistry, Scripps Research, La Jolla, CA, USA
| | - Ryan A Shenvi
- Department of Chemistry, Scripps Research, La Jolla, CA, USA
| |
Collapse
|
44
|
Nakao T, Banba S. Important amino acids for function of the insect Rdl GABA receptor. PEST MANAGEMENT SCIENCE 2021; 77:3753-3762. [PMID: 33002317 DOI: 10.1002/ps.6121] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/24/2020] [Revised: 09/24/2020] [Accepted: 10/01/2020] [Indexed: 06/11/2023]
Abstract
BACKGROUND Insect Rdl GABA receptor is an important insecticide target. To design a novel insecticide, studies on the structures of homologous pentameric ligand-gated ion channels provide information about important amino acids that are necessary for the function of insect Rdl GABA receptors. RESULTS L9'A, T12'A, T13'A, T13'S, M15'S, and M15'N mutations in the Drosophila Rdl GABA receptor subunit caused the protein to spontaneously adopt the open state conformation. In contrast, the S16'A, S16'T, S17'A, and S17'H mutant homomers showed the same levels of agonist and antagonist sensitivity as the wild-type receptor. The G336M mutation in the Drosophila Rdl GABA receptor abolished the agonist activities of ivermectin and milbemectin, but the F339M mutation did not. Additionally, the F339M mutation caused spontaneous opening of the receptor. In the Drosophila Rdl model, the hydrophobic girdle plays an important role in stabilization of the closed state. Mutations which decrease hydrophobic interactions resulted in spontaneous opening, supporting the importance of the hydrophobic girdle for keeping the channel closed. Through a mutational study of transmembrane 3 (TM3) cytoplasmic domain and Rdl GABA receptor modeling, hydrophobic interactions between TM3 and TM4 and intersubunit interaction were demonstrated to be important for channel gating. Alternatively, the intrasubunit interaction between TM2 and TM3 domains were less important for channel gating in case of Drosophila Rdl GABA receptor. CONCLUSION This study demonstrates important amino acids critical to the function of the Drosophila Rdl GABA receptor based on the mutational studies and Drosophila Rdl GABA receptor modeling approach. © 2020 Society of Chemical Industry.
Collapse
Affiliation(s)
- Toshifumi Nakao
- Organic Chemistry G, Agrochemicals Research Center, Mitsui Chemicals Agro, Inc., Chiba, Japan
| | - Shinichi Banba
- Organic Chemistry G, Agrochemicals Research Center, Mitsui Chemicals Agro, Inc., Chiba, Japan
| |
Collapse
|
45
|
Elephants in the Dark: Insights and Incongruities in Pentameric Ligand-gated Ion Channel Models. J Mol Biol 2021; 433:167128. [PMID: 34224751 DOI: 10.1016/j.jmb.2021.167128] [Citation(s) in RCA: 14] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/14/2021] [Revised: 06/25/2021] [Accepted: 06/25/2021] [Indexed: 02/06/2023]
Abstract
The superfamily of pentameric ligand-gated ion channels (pLGICs) comprises key players in electrochemical signal transduction across evolution, including historic model systems for receptor allostery and targets for drug development. Accordingly, structural studies of these channels have steadily increased, and now approach 250 depositions in the protein data bank. This review contextualizes currently available structures in the pLGIC family, focusing on morphology, ligand binding, and gating in three model subfamilies: the prokaryotic channel GLIC, the cation-selective nicotinic acetylcholine receptor, and the anion-selective glycine receptor. Common themes include the challenging process of capturing and annotating channels in distinct functional states; partially conserved gating mechanisms, including remodeling at the extracellular/transmembrane-domain interface; and diversity beyond the protein level, arising from posttranslational modifications, ligands, lipids, and signaling partners. Interpreting pLGIC structures can be compared to describing an elephant in the dark, relying on touch alone to comprehend the many parts of a monumental beast: each structure represents a snapshot in time under specific experimental conditions, which must be integrated with further structure, function, and simulations data to build a comprehensive model, and understand how one channel may fundamentally differ from another.
Collapse
|
46
|
Zeilhofer HU, Werynska K, Gingras J, Yévenes GE. Glycine Receptors in Spinal Nociceptive Control-An Update. Biomolecules 2021; 11:846. [PMID: 34204137 PMCID: PMC8228028 DOI: 10.3390/biom11060846] [Citation(s) in RCA: 25] [Impact Index Per Article: 8.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/12/2021] [Revised: 06/03/2021] [Accepted: 06/03/2021] [Indexed: 12/12/2022] Open
Abstract
Diminished inhibitory control of spinal nociception is one of the major culprits of chronic pain states. Restoring proper synaptic inhibition is a well-established rational therapeutic approach explored by several pharmaceutical companies. A particular challenge arises from the need for site-specific intervention to avoid deleterious side effects such as sedation, addiction, or impaired motor control, which would arise from wide-range facilitation of inhibition. Specific targeting of glycinergic inhibition, which dominates in the spinal cord and parts of the hindbrain, may help reduce these side effects. Selective targeting of the α3 subtype of glycine receptors (GlyRs), which is highly enriched in the superficial layers of the spinal dorsal horn, a key site of nociceptive processing, may help to further narrow down pharmacological intervention on the nociceptive system and increase tolerability. This review provides an update on the physiological properties and functions of α3 subtype GlyRs and on the present state of related drug discovery programs.
Collapse
Affiliation(s)
- Hanns Ulrich Zeilhofer
- Institute of Pharmacology and Toxicology, University of Zurich, Winterthurerstrasse 190, CH-8057 Zürich, Switzerland;
- Institute of Pharmaceutical Sciences, Swiss Federal Institute of Technology (ETH) Zurich, Vladimir Prelog Weg, CH-8093 Zürich, Switzerland
- Drug Discovery Network Zurich, University of Zurich and ETH Zurich, Winterthurerstrasse 190, CH-8057 Zürich, Switzerland
| | - Karolina Werynska
- Institute of Pharmacology and Toxicology, University of Zurich, Winterthurerstrasse 190, CH-8057 Zürich, Switzerland;
| | - Jacinthe Gingras
- Homology Medicines Inc., 1 Patriots Park, Bedford, MA 01730, USA;
| | - Gonzalo E. Yévenes
- Department of Physiology, University of Concepción, Concepción 4070386, Chile;
- Millennium Nucleus for the Study of Pain (MiNuSPain), Santiago 8320000, Chile
| |
Collapse
|
47
|
An Inside Job: Molecular Determinants for Postsynaptic Localization of Nicotinic Acetylcholine Receptors. Molecules 2021; 26:molecules26113065. [PMID: 34063759 PMCID: PMC8196675 DOI: 10.3390/molecules26113065] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/30/2021] [Revised: 05/13/2021] [Accepted: 05/15/2021] [Indexed: 11/29/2022] Open
Abstract
Nicotinic acetylcholine receptors (nAChRs) mediate fast synaptic transmission at neuromuscular and autonomic ganglionic synapses in the peripheral nervous system. The postsynaptic localization of muscle ((α1)2β1γδ) and neuronal ((α3β4)2β4) nicotinic receptors at these synapses is mediated by interactions between the nAChR intracellular domains and cytoplasmic scaffolding proteins. Recent high resolution structures and functional studies provide new insights into the molecular determinants that mediate these interactions. Surprisingly, they reveal that the muscle nAChR binds 1–3 rapsyn scaffolding molecules, which dimerize and thereby form an interconnected lattice between receptors. Moreover, rapsyn binds two distinct sites on the nAChR subunit cytoplasmic loops; the MA-helix on one or more subunits and a motif specific to the β subunit. Binding at the latter site is regulated by agrin-induced phosphorylation of βY390, and increases the stoichiometry of rapsyn/AChR complexes. Similarly, the neuronal nAChR may be localized at ganglionic synapses by phosphorylation-dependent interactions with 14-3-3 adaptor proteins which bind specific motifs in each of the α3 subunit cytoplasmic loops. Thus, postsynaptic localization of nAChRs is mediated by regulated interactions with multiple scaffolding molecules, and the stoichiometry of these complexes likely helps regulate the number, density, and stability of receptors at the synapse.
Collapse
|
48
|
Crnjar A, Mesoy SM, Lummis SCR, Molteni C. A Single Mutation in the Outer Lipid-Facing Helix of a Pentameric Ligand-Gated Ion Channel Affects Channel Function Through a Radially-Propagating Mechanism. Front Mol Biosci 2021; 8:644720. [PMID: 33996899 PMCID: PMC8119899 DOI: 10.3389/fmolb.2021.644720] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/21/2020] [Accepted: 02/22/2021] [Indexed: 11/13/2022] Open
Abstract
Pentameric ligand-gated ion channels (pLGICs) mediate fast synaptic transmission and are crucial drug targets. Their gating mechanism is triggered by ligand binding in the extracellular domain that culminates in the opening of a hydrophobic gate in the transmembrane domain. This domain is made of four α-helices (M1 to M4). Recently the outer lipid-facing helix (M4) has been shown to be key to receptor function, however its role in channel opening is still poorly understood. It could act through its neighboring helices (M1/M3), or via the M4 tip interacting with the pivotal Cys-loop in the extracellular domain. Mutation of a single M4 tyrosine (Y441) to alanine renders one pLGIC-the 5-HT3A receptor-unable to function despite robust ligand binding. Using Y441A as a proxy for M4 function, we here predict likely paths of Y441 action using molecular dynamics, and test these predictions with functional assays of mutant receptors in HEK cells and Xenopus oocytes using fluorescent membrane potential sensitive dye and two-electrode voltage clamp respectively. We show that Y441 does not act via the M4 tip or Cys-loop, but instead connects radially through M1 to a residue near the ion channel hydrophobic gate on the pore-lining helix M2. This demonstrates the active role of the M4 helix in channel opening.
Collapse
Affiliation(s)
| | - Susanne M. Mesoy
- Department of Biochemistry, University of Cambridge, Cambridge, United Kingdom
| | - Sarah C. R. Lummis
- Department of Biochemistry, University of Cambridge, Cambridge, United Kingdom
| | - Carla Molteni
- Physics Department, King's College London, London, United Kingdom
| |
Collapse
|
49
|
Romero-Nava R, Alarcón-Aguilar FJ, Giacoman-Martínez A, Blancas-Flores G, Aguayo-Cerón KA, Ballinas-Verdugo MA, Sánchez-Muñoz F, Huang F, Villafaña-Rauda S, Almanza-Pérez JC. Glycine is a competitive antagonist of the TNF receptor mediating the expression of inflammatory cytokines in 3T3-L1 adipocytes. Inflamm Res 2021; 70:605-618. [PMID: 33877377 DOI: 10.1007/s00011-021-01462-1] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/03/2020] [Revised: 02/16/2021] [Accepted: 04/05/2021] [Indexed: 01/20/2023] Open
Abstract
OBJECTIVE To determine the involvement of TNF-α and glycine receptors in the inhibition of pro-inflammatory adipokines in 3T3-L1 cells. METHODS RT-PCR evidenced glycine receptors in 3T3-L1 adipocytes. 3T3-L1 cells were transfected with siRNA for the glycine (Glrb) and TNF1a (Tnfrsf1a) receptors and confirmed by confocal microscopy. Transfected cells were treated with glycine (10 mM). The expressions of TNF-α and IL-6 mRNA were measured by qRT-PCR, while concentrations were quantified by ELISA. RESULTS Glycine decreased the expression and concentration of TNF-α and IL-6; this effect did not occur in the absence of TNF-α receptor due to siRNA. In contrast, glycine produced only slight changes in the expression of TNF-α and IL-6 in the absence of the glycine receptor due to siRNA. A docking analysis confirmed the possibility of binding glycine to the TNF-α1a receptor. CONCLUSION These findings support the idea that glycine could partially inhibit the binding of TNF-α to its receptor and provide clues about the mechanisms by which glycine inhibits the secretion of pro-inflammatory adipokines in adipocytes through the TNF-α receptor.
Collapse
MESH Headings
- 3T3-L1 Cells
- Adipocytes/metabolism
- Adiponectin/genetics
- Animals
- Cytokines/genetics
- Cytokines/metabolism
- Gene Expression
- Glycine/pharmacology
- Mice
- NF-kappa B/antagonists & inhibitors
- NF-kappa B/metabolism
- RNA, Messenger/metabolism
- RNA, Small Interfering/genetics
- Receptors, Glycine/genetics
- Receptors, Tumor Necrosis Factor, Type I/antagonists & inhibitors
- Receptors, Tumor Necrosis Factor, Type I/genetics
- Receptors, Tumor Necrosis Factor, Type II/antagonists & inhibitors
- Receptors, Tumor Necrosis Factor, Type II/genetics
Collapse
Affiliation(s)
- Rodrigo Romero-Nava
- Laboratorio de Farmacología, Departamento de Ciencias de la Salud, DCBS, Universidad Autónoma Metropolitana-Iztapalapa (UAM-I), San Rafael Atlixco 186, Col. Vicentina. Iztapalapa, C.P. 09340, Mexico City, Mexico
- Departamento de Farmacología y Toxicología, Hospital Infantil de México Federico Gómez, Mexico City, Mexico
- Sección de Posgrado, Laboratorio de Señalización Intracelular, Escuela Superior de Medicina del Instituto Politécnico Nacional, Mexico City, Mexico
| | - Francisco J Alarcón-Aguilar
- Laboratorio de Farmacología, Departamento de Ciencias de la Salud, DCBS, Universidad Autónoma Metropolitana-Iztapalapa (UAM-I), San Rafael Atlixco 186, Col. Vicentina. Iztapalapa, C.P. 09340, Mexico City, Mexico
| | - Abraham Giacoman-Martínez
- Laboratorio de Farmacología, Departamento de Ciencias de la Salud, DCBS, Universidad Autónoma Metropolitana-Iztapalapa (UAM-I), San Rafael Atlixco 186, Col. Vicentina. Iztapalapa, C.P. 09340, Mexico City, Mexico
| | - Gerardo Blancas-Flores
- Laboratorio de Farmacología, Departamento de Ciencias de la Salud, DCBS, Universidad Autónoma Metropolitana-Iztapalapa (UAM-I), San Rafael Atlixco 186, Col. Vicentina. Iztapalapa, C.P. 09340, Mexico City, Mexico
| | - Karla A Aguayo-Cerón
- Sección de Posgrado, Laboratorio de Señalización Intracelular, Escuela Superior de Medicina del Instituto Politécnico Nacional, Mexico City, Mexico
| | - Martha A Ballinas-Verdugo
- Departamento de Inmunología, Instituto Nacional de Cardiología (Ignacio Chávez), Mexico City, Mexico
| | - Fausto Sánchez-Muñoz
- Departamento de Inmunología, Instituto Nacional de Cardiología (Ignacio Chávez), Mexico City, Mexico
| | - Fengyang Huang
- Departamento de Farmacología y Toxicología, Hospital Infantil de México Federico Gómez, Mexico City, Mexico
| | - Santiago Villafaña-Rauda
- Sección de Posgrado, Laboratorio de Señalización Intracelular, Escuela Superior de Medicina del Instituto Politécnico Nacional, Mexico City, Mexico
| | - Julio C Almanza-Pérez
- Laboratorio de Farmacología, Departamento de Ciencias de la Salud, DCBS, Universidad Autónoma Metropolitana-Iztapalapa (UAM-I), San Rafael Atlixco 186, Col. Vicentina. Iztapalapa, C.P. 09340, Mexico City, Mexico.
| |
Collapse
|
50
|
Signal transduction through Cys-loop receptors is mediated by the nonspecific bumping of closely apposed domains. Proc Natl Acad Sci U S A 2021; 118:2021016118. [PMID: 33785596 DOI: 10.1073/pnas.2021016118] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/18/2022] Open
Abstract
One of the most fundamental questions in the field of Cys-loop receptors (pentameric ligand-gated ion channels, pLGICs) is how the affinity for neurotransmitters and the conductive/nonconductive state of the transmembrane pore are correlated despite the ∼60-Å distance between the corresponding domains. Proposed mechanisms differ, but they all converge into the idea that interactions between wild-type side chains across the extracellular-transmembrane-domain (ECD-TMD) interface are crucial for this phenomenon. Indeed, the successful design of fully functional chimeras that combine intact ECD and TMD modules from different wild-type pLGICs has commonly been ascribed to the residual conservation of sequence that exists at the level of the interfacial loops even between evolutionarily distant parent channels. Here, using mutagenesis, patch-clamp electrophysiology, and radiolabeled-ligand binding experiments, we studied the effect of eliminating this residual conservation of sequence on ion-channel function and cell-surface expression. From our results, we conclude that proper state interconversion ("gating") does not require conservation of sequence-or even physicochemical properties-across the ECD-TMD interface. Wild-type ECD and TMD side chains undoubtedly interact with their surroundings, but the interactions between them-straddling the interface-do not seem to be more important for gating than those occurring elsewhere in the protein. We propose that gating of pLGICs requires, instead, that the overall structure of the interfacial loops be conserved, and that their relative orientation and distance be the appropriate ones for changes in one side to result in changes in the other, in a phenomenon akin to the nonspecific "bumping" of closely apposed domains.
Collapse
|