1
|
Luo Y, Sun L, Peng Y. The structural basis of the G protein-coupled receptor and ion channel axis. Curr Res Struct Biol 2025; 9:100165. [PMID: 40083915 PMCID: PMC11904507 DOI: 10.1016/j.crstbi.2025.100165] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/26/2024] [Revised: 01/25/2025] [Accepted: 02/17/2025] [Indexed: 03/16/2025] Open
Abstract
Sensory neurons play an essential role in recognizing and responding to detrimental, irritating, and inflammatory stimuli from our surroundings, such as pain, itch, cough, and neurogenic inflammation. The transduction of these physiological signals is chiefly mediated by G protein-coupled receptors (GPCRs) and ion channels. The binding of ligands to GPCRs triggers a signaling cascade, recruiting G proteins or β-arrestins, which subsequently interact with ion channels (e.g., GIRK and TRP channels). This interaction leads to the sensitization and activation of these channels, initiating the neuron's protective mechanisms. This review delves into the complex interplay between GPCRs and ion channels that underpin these physiological processes, with a particular focus on the role of structural biology in enhancing our comprehension. Through unraveling the intricacies of the GPCR-ion channel axis, we aim to shed light on the sophisticated intermolecular dynamics within these pivotal membrane protein families, ultimately guiding the development of precise therapeutic interventions.
Collapse
Affiliation(s)
- Yulin Luo
- iHuman Institute, ShanghaiTech University, Ren Building, 393 Middle Huaxia Road, Pudong, Shanghai, 201210, China
- School of Life Science and Technology, ShanghaiTech University, L Building, 393 Middle Huaxia Road, Pudong, Shanghai, 201210, China
| | - Liping Sun
- iHuman Institute, ShanghaiTech University, Ren Building, 393 Middle Huaxia Road, Pudong, Shanghai, 201210, China
| | - Yao Peng
- iHuman Institute, ShanghaiTech University, Ren Building, 393 Middle Huaxia Road, Pudong, Shanghai, 201210, China
| |
Collapse
|
2
|
Kaneda E, Kawai T, Okamura Y, Miyagawa S. Effects of moderate static magnetic fields on voltage-gated potassium ion channels in sympathetic neuron-like PC12 cells. Physiol Rep 2025; 13:e70236. [PMID: 40119575 PMCID: PMC11928678 DOI: 10.14814/phy2.70236] [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: 11/22/2024] [Revised: 01/16/2025] [Accepted: 01/24/2025] [Indexed: 03/24/2025] Open
Abstract
While exposure of moderate static magnetic fields (SMF) can alter neuronal excitability, the effects on sympathetic neurons remain underexplored. This study investigates the effects of moderate SMF on Kv channels in the plasma membrane of sympathetic neuron-like PC12 cells. The current density of Kv channels was significantly lower in the 18-h magnet-exposed group, with effects persisting even after the magnet was removed before patch-clamp measurements. The current density of outward current in the presence of TEA was not different between the two groups, indicating that magnetic field affects TEA-sensitive Kv channels. To further explore these changes, RNA sequencing was performed on samples from both the Sham and 18-h magnet-exposed groups, identifying 37 moderate SMF-sensitive genes. Changes in mRNA expression levels and KEGG analysis suggested that pathways involved in the inhibition of neuronal excitability, such as GABAB receptor activation and Kir3 channel opening, may be more likely to be activated. In conclusion, moderate SMF is strongly associated with reduced current density in PC12 cells, particularly affecting Kv channels. The present study provides fundamental information on the influence of long-term SMF exposure on the excitability of sympathetic neurons.
Collapse
Affiliation(s)
- Eri Kaneda
- Graduate School of Medicine, Osaka University, Suita, Osaka, Japan
| | - Takafumi Kawai
- Graduate School of Medicine, Osaka University, Suita, Osaka, Japan
| | - Yasushi Okamura
- Graduate School of Medicine, Osaka University, Suita, Osaka, Japan
- Graduate School of Frontier Bioscience, Osaka University, Suita, Osaka, Japan
| | - Shigeru Miyagawa
- Graduate School of Medicine, Osaka University, Suita, Osaka, Japan
| |
Collapse
|
3
|
Trovò L, Kouvaros S, Schwenk J, Fernandez-Fernandez D, Fritzius T, Rem PD, Früh S, Gassmann M, Fakler B, Bischofberger J, Bettler B. Synaptotagmin-11 facilitates assembly of a presynaptic signaling complex in post-Golgi cargo vesicles. EMBO Rep 2024; 25:2610-2634. [PMID: 38698221 PMCID: PMC11169412 DOI: 10.1038/s44319-024-00147-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/18/2023] [Revised: 04/11/2024] [Accepted: 04/12/2024] [Indexed: 05/05/2024] Open
Abstract
GABAB receptors (GBRs), the G protein-coupled receptors for GABA, regulate synaptic transmission throughout the brain. A main synaptic function of GBRs is the gating of Cav2.2-type Ca2+ channels. However, the cellular compartment where stable GBR/Cav2.2 signaling complexes form remains unknown. In this study, we demonstrate that the vesicular protein synaptotagmin-11 (Syt11) binds to both the auxiliary GBR subunit KCTD16 and Cav2.2 channels. Through these dual interactions, Syt11 recruits GBRs and Cav2.2 channels to post-Golgi vesicles, thus facilitating assembly of GBR/Cav2.2 signaling complexes. In addition, Syt11 stabilizes GBRs and Cav2.2 channels at the neuronal plasma membrane by inhibiting constitutive internalization. Neurons of Syt11 knockout mice exhibit deficits in presynaptic GBRs and Cav2.2 channels, reduced neurotransmitter release, and decreased GBR-mediated presynaptic inhibition, highlighting the critical role of Syt11 in the assembly and stable expression of GBR/Cav2.2 complexes. These findings support that Syt11 acts as a vesicular scaffold protein, aiding in the assembly of signaling complexes from low-abundance components within transport vesicles. This mechanism enables insertion of pre-assembled functional signaling units into the synaptic membrane.
Collapse
Affiliation(s)
- Luca Trovò
- Department of Biomedicine, University of Basel, Basel, Switzerland
| | | | - Jochen Schwenk
- Institute of Physiology, Faculty of Medicine, University of Freiburg, Freiburg, Germany
| | | | | | | | - Simon Früh
- Department of Biomedicine, University of Basel, Basel, Switzerland
| | - Martin Gassmann
- Department of Biomedicine, University of Basel, Basel, Switzerland
| | - Bernd Fakler
- Institute of Physiology, Faculty of Medicine, University of Freiburg, Freiburg, Germany
- CIBSS Center for Integrative Biological Signalling Studies, University of Freiburg, Freiburg, Germany
- Center for Basics in NeuroModulation, Freiburg, Germany
| | | | - Bernhard Bettler
- Department of Biomedicine, University of Basel, Basel, Switzerland.
| |
Collapse
|
4
|
Rizk R, Devost D, Pétrin D, Hébert TE. KCTD Proteins Have Redundant Functions in Controlling Cellular Growth. Int J Mol Sci 2024; 25:4993. [PMID: 38732215 PMCID: PMC11084553 DOI: 10.3390/ijms25094993] [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: 03/01/2024] [Revised: 04/23/2024] [Accepted: 04/29/2024] [Indexed: 05/13/2024] Open
Abstract
We explored the functional redundancy of three structurally related KCTD (Potassium Channel Tetramerization Domain) proteins, KCTD2, KCTD5, and KCTD17, by progressively knocking them out in HEK 293 cells using CRISPR/Cas9 genome editing. After validating the knockout, we assessed the effects of progressive knockout on cell growth and gene expression. We noted that the progressive effects of knockout of KCTD isoforms on cell growth were most pervasive when all three isoforms were deleted, suggesting some functions were conserved between them. This was also reflected in progressive changes in gene expression. Our previous work indicated that Gβ1 was involved in the transcriptional control of gene expression, so we compared the gene expression patterns between GNB1 and KCTD KO. Knockout of GNB1 led to numerous changes in the expression levels of other G protein subunit genes, while knockout of KCTD isoforms had the opposite effect, presumably because of their role in regulating levels of Gβ1. Our work demonstrates a unique relationship between KCTD proteins and Gβ1 and a global role for this subfamily of KCTD proteins in maintaining the ability of cells to survive and proliferate.
Collapse
Affiliation(s)
| | | | | | - Terence E. Hébert
- Department of Pharmacology and Therapeutics, McGill University, 3655 Promenade Sir-William-Osler, Room 1303, Montréal, QC H3G 1Y6, Canada; (R.R.); (D.D.); (D.P.)
| |
Collapse
|
5
|
Benke D, Bhat MA, Hleihil M. GABAB Receptors: Molecular Organization, Function, and Alternative Drug Development by Targeting Protein-Protein Interactions. THE RECEPTORS 2024:3-39. [DOI: 10.1007/978-3-031-67148-7_1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/03/2025]
|
6
|
The GABA and GABA-Receptor System in Inflammation, Anti-Tumor Immune Responses, and COVID-19. Biomedicines 2023; 11:biomedicines11020254. [PMID: 36830790 PMCID: PMC9953446 DOI: 10.3390/biomedicines11020254] [Citation(s) in RCA: 19] [Impact Index Per Article: 9.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/16/2022] [Revised: 01/16/2023] [Accepted: 01/16/2023] [Indexed: 01/20/2023] Open
Abstract
GABA and GABAA-receptors (GABAA-Rs) play major roles in neurodevelopment and neurotransmission in the central nervous system (CNS). There has been a growing appreciation that GABAA-Rs are also present on most immune cells. Studies in the fields of autoimmune disease, cancer, parasitology, and virology have observed that GABA-R ligands have anti-inflammatory actions on T cells and antigen-presenting cells (APCs), while also enhancing regulatory T cell (Treg) responses and shifting APCs toward anti-inflammatory phenotypes. These actions have enabled GABAA-R ligands to ameliorate autoimmune diseases, such as type 1 diabetes (T1D), multiple sclerosis (MS), and rheumatoid arthritis, as well as type 2 diabetes (T2D)-associated inflammation in preclinical models. Conversely, antagonism of GABAA-R activity promotes the pro-inflammatory responses of T cells and APCs, enhancing anti-tumor responses and reducing tumor burden in models of solid tumors. Lung epithelial cells also express GABA-Rs, whose activation helps maintain fluid homeostasis and promote recovery from injury. The ability of GABAA-R agonists to limit both excessive immune responses and lung epithelial cell injury may underlie recent findings that GABAA-R agonists reduce the severity of disease in mice infected with highly lethal coronaviruses (SARS-CoV-2 and MHV-1). These observations suggest that GABAA-R agonists may provide off-the-shelf therapies for COVID-19 caused by new SARS-CoV-2 variants, as well as novel beta-coronaviruses, which evade vaccine-induced immune responses and antiviral medications. We review these findings and further advance the notions that (1) immune cells possess GABAA-Rs to limit inflammation in the CNS, and (2) this natural "braking system" on inflammatory responses may be pharmacologically engaged to slow the progression of autoimmune diseases, reduce the severity of COVID-19, and perhaps limit neuroinflammation associated with long COVID.
Collapse
|
7
|
Martín-Belmonte A, Aguado C, Alfaro-Ruiz R, Moreno-Martínez AE, de la Ossa L, Aso E, Gómez-Acero L, Shigemoto R, Fukazawa Y, Ciruela F, Luján R. Nanoscale alterations in GABAB receptors and GIRK channel organization on the hippocampus of APP/PS1 mice. Alzheimers Res Ther 2022; 14:136. [PMID: 36131327 PMCID: PMC9490896 DOI: 10.1186/s13195-022-01078-5] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/08/2022] [Accepted: 09/13/2022] [Indexed: 11/10/2022]
Abstract
Alzheimer’s disease (AD) is characterized by a reorganization of brain activity determining network hyperexcitability and loss of synaptic plasticity. Precisely, a dysfunction in metabotropic GABAB receptor signalling through G protein-gated inwardly rectifying K+ (GIRK or Kir3) channels on the hippocampus has been postulated. Thus, we determined the impact of amyloid-β (Aβ) pathology in GIRK channel density, subcellular distribution, and its association with GABAB receptors in hippocampal CA1 pyramidal neurons from the APP/PS1 mouse model using quantitative SDS-digested freeze-fracture replica labelling (SDS-FRL) and proximity ligation in situ assay (P-LISA). In wild type mice, single SDS-FRL detection revealed a similar dendritic gradient for GIRK1 and GIRK2 in CA1 pyramidal cells, with higher densities in spines, and GIRK3 showed a lower and uniform distribution. Double SDS-FRL showed a co-clustering of GIRK2 and GIRK1 in post- and presynaptic compartments, but not for GIRK2 and GIRK3. Likewise, double GABAB1 and GIRK2 SDS-FRL detection displayed a high degree of co-clustering in nanodomains (40–50 nm) mostly in spines and axon terminals. In APP/PS1 mice, the density of GIRK2 and GIRK1, but not for GIRK3, was significantly reduced along the neuronal surface of CA1 pyramidal cells and in axon terminals contacting them. Importantly, GABAB1 and GIRK2 co-clustering was not present in APP/PS1 mice. Similarly, P-LISA experiments revealed a significant reduction in GABAB1 and GIRK2 interaction on the hippocampus of this animal model. Overall, our results provide compelling evidence showing a significant reduction on the cell surface density of pre- and postsynaptic GIRK1 and GIRK2, but not GIRK3, and a decline in GABAB receptors and GIRK2 channels co-clustering in hippocampal pyramidal neurons from APP/PS1 mice, thus suggesting that a disruption in the GABAB receptor–GIRK channel membrane assembly causes dysregulation in the GABAB signalling via GIRK channels in this AD animal model.
Collapse
|
8
|
Luo H, Marron Fernandez de Velasco E, Wickman K. Neuronal G protein-gated K + channels. Am J Physiol Cell Physiol 2022; 323:C439-C460. [PMID: 35704701 PMCID: PMC9362898 DOI: 10.1152/ajpcell.00102.2022] [Citation(s) in RCA: 28] [Impact Index Per Article: 9.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/08/2023]
Abstract
G protein-gated inwardly rectifying K+ (GIRK/Kir3) channels exert a critical inhibitory influence on neurons. Neuronal GIRK channels mediate the G protein-dependent, direct/postsynaptic inhibitory effect of many neurotransmitters including γ-aminobutyric acid (GABA), serotonin, dopamine, adenosine, somatostatin, and enkephalin. In addition to their complex regulation by G proteins, neuronal GIRK channel activity is sensitive to PIP2, phosphorylation, regulator of G protein signaling (RGS) proteins, intracellular Na+ and Ca2+, and cholesterol. The application of genetic and viral manipulations in rodent models, together with recent progress in the development of GIRK channel modulators, has increased our understanding of the physiological and behavioral impact of neuronal GIRK channels. Work in rodent models has also revealed that neuronal GIRK channel activity is modified, transiently or persistently, by various stimuli including exposure drugs of abuse, changes in neuronal activity patterns, and aversive experience. A growing body of preclinical and clinical evidence suggests that dysregulation of GIRK channel activity contributes to neurological diseases and disorders. The primary goals of this review are to highlight fundamental principles of neuronal GIRK channel biology, mechanisms of GIRK channel regulation and plasticity, the nascent landscape of GIRK channel pharmacology, and the potential relevance of GIRK channels to the pathophysiology and treatment of neurological diseases and disorders.
Collapse
Affiliation(s)
- Haichang Luo
- Department of Pharmacology, University of Minnesota, Minneapolis, MN, United States
| | | | - Kevin Wickman
- Department of Pharmacology, University of Minnesota, Minneapolis, MN, United States
| |
Collapse
|
9
|
Bhat MA, Esmaeili A, Neumann E, Balakrishnan K, Benke D. Targeting the Interaction of GABA B Receptors With CHOP After an Ischemic Insult Restores Receptor Expression and Inhibits Progressive Neuronal Death. Front Pharmacol 2022; 13:870861. [PMID: 35422706 PMCID: PMC9002115 DOI: 10.3389/fphar.2022.870861] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/07/2022] [Accepted: 03/14/2022] [Indexed: 01/01/2023] Open
Abstract
GABAB receptors control neuronal excitability via slow and prolonged inhibition in the central nervous system. One important function of GABAB receptors under physiological condition is to prevent neurons from shifting into an overexcitation state which can lead to excitotoxic death. However, under ischemic conditions, GABAB receptors are downregulated, fostering over-excitation and excitotoxicity. One mechanism downregulating GABAB receptors is mediated via the interaction with the endoplasmic reticulum (ER) stress-induced transcription factor CHOP. In this study, we investigated the hypothesis that preventing the interaction of CHOP with GABAB receptors after an ischemic insult restores normal expression of GABAB receptors and reduces neuronal death. For this, we designed an interfering peptide (R2-Pep) that restored the CHOP-induced downregulation of cell surface GABAB receptors in cultured cortical neurons subjected to oxygen and glucose deprivation (OGD). Administration of R2-Pep after OGD restored normal cell surface expression of GABAB receptors as well as GABAB receptor-mediated inhibition. As a result, R2-Pep reduced enhanced neuronal activity and inhibited progressive neuronal death in OGD stressed cultures. Thus, targeting diseases relevant protein-protein interactions might be a promising strategy for developing highly specific novel therapeutics.
Collapse
Affiliation(s)
- Musadiq A Bhat
- Institute of Pharmacology and Toxicology, University of Zurich, Zurich, Switzerland
| | - Abolghasem Esmaeili
- Department of Cell and Molecular Biology & Microbiology, Faculty of Biological Science and Technology, University of Isfahan, Isfahan, Iran
| | - Elena Neumann
- Institute of Pharmacology and Toxicology, University of Zurich, Zurich, Switzerland
| | - Karthik Balakrishnan
- Institute of Pharmacology and Toxicology, University of Zurich, Zurich, Switzerland
| | - Dietmar Benke
- Institute of Pharmacology and Toxicology, University of Zurich, Zurich, Switzerland.,Neuroscience Center Zurich, University of Zurich and ETH Zurich, Zurich, Switzerland.,Drug Discovery Network Zurich (DDNZ), Zurich, Switzerland
| |
Collapse
|
10
|
Ferré S, Ciruela F, Dessauer CW, González-Maeso J, Hébert TE, Jockers R, Logothetis DE, Pardo L. G protein-coupled receptor-effector macromolecular membrane assemblies (GEMMAs). Pharmacol Ther 2022; 231:107977. [PMID: 34480967 PMCID: PMC9375844 DOI: 10.1016/j.pharmthera.2021.107977] [Citation(s) in RCA: 40] [Impact Index Per Article: 13.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/20/2021] [Revised: 08/11/2021] [Accepted: 08/17/2021] [Indexed: 12/12/2022]
Abstract
G protein-coupled receptors (GPCRs) are the largest group of receptors involved in cellular signaling across the plasma membrane and a major class of drug targets. The canonical model for GPCR signaling involves three components - the GPCR, a heterotrimeric G protein and a proximal plasma membrane effector - that have been generally thought to be freely mobile molecules able to interact by 'collision coupling'. Here, we synthesize evidence that supports the existence of GPCR-effector macromolecular membrane assemblies (GEMMAs) comprised of specific GPCRs, G proteins, plasma membrane effector molecules and other associated transmembrane proteins that are pre-assembled prior to receptor activation by agonists, which then leads to subsequent rearrangement of the GEMMA components. The GEMMA concept offers an alternative and complementary model to the canonical collision-coupling model, allowing more efficient interactions between specific signaling components, as well as the integration of the concept of GPCR oligomerization as well as GPCR interactions with orphan receptors, truncated GPCRs and other membrane-localized GPCR-associated proteins. Collision-coupling and pre-assembled mechanisms are not exclusive and likely both operate in the cell, providing a spectrum of signaling modalities which explains the differential properties of a multitude of GPCRs in their different cellular environments. Here, we explore the unique pharmacological characteristics of individual GEMMAs, which could provide new opportunities to therapeutically modulate GPCR signaling.
Collapse
Affiliation(s)
- Sergi Ferré
- Integrative Neurobiology Section, National Institute on Drug Addiction, Intramural Research Program, NIH, DHHS, Baltimore, MD, USA.
| | - Francisco Ciruela
- Department of Pathology and Experimental Therapeutics, Faculty of Medicine and Health Sciences, Institute of Neurosciences, IDIBELL, University of Barcelona, L’Hospitalet de Llobregat, Spain
| | - Carmen W. Dessauer
- Department of Integrative Biology and Pharmacology, McGovern Medical School, University of Texas Health Science Center at Houston, Houston, Texas, USA
| | - Javier González-Maeso
- Department of Physiology and Biophysics, School of Medicine, Virginia Commonwealth University, Richmond, Virginia, USA
| | - Terence E. Hébert
- Department of Pharmacology and Therapeutics, McGill University, Montréal, Québec
| | - Ralf Jockers
- University of Paris, Institute Cochin, INSERM, CNRS, Paris, France
| | - Diomedes E. Logothetis
- Laboratory of Electrophysiology, Departments of Pharmaceutical Sciences, Chemistry and Chemical Biology and Center for Drug Discovery, School of Pharmacy at the Bouvé College of Health Sciences and College of Science, Northeastern University, Boston, Massachusetts, USA
| | - Leonardo Pardo
- Laboratory of Computational Medicine, Biostatistics Unit, Faculty of Medicine, Autonomous University of Barcelona, Bellaterra, Spain
| |
Collapse
|
11
|
Fritzius T, Stawarski M, Isogai S, Bettler B. Structural Basis of GABA B Receptor Regulation and Signaling. Curr Top Behav Neurosci 2022; 52:19-37. [PMID: 32812202 DOI: 10.1007/7854_2020_147] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 06/11/2023]
Abstract
GABAB receptors (GBRs), the G protein-coupled receptors for the inhibitory neurotransmitter γ-aminobutyric acid (GABA), activate Go/i-type G proteins that regulate adenylyl cyclase, Ca2+ channels, and K+ channels. GBR signaling to enzymes and ion channels influences neuronal activity, plasticity processes, and network activity throughout the brain. GBRs are obligatory heterodimers composed of GB1a or GB1b subunits with a GB2 subunit. Heterodimeric GB1a/2 and GB1b/2 receptors represent functional units that associate in a modular fashion with regulatory, trafficking, and effector proteins to generate receptors with distinct physiological functions. This review summarizes current knowledge on the structure, organization, and functions of multi-protein GBR complexes.
Collapse
Affiliation(s)
- Thorsten Fritzius
- Department of Biomedicine, Institute of Physiology, Pharmazentrum, University of Basel, Basel, Switzerland
| | - Michal Stawarski
- Department of Biomedicine, Institute of Physiology, Pharmazentrum, University of Basel, Basel, Switzerland
| | - Shin Isogai
- Biozentrum, Focal Area Structural Biology and Biophysics, University of Basel, Basel, Switzerland.
- Microbial Downstream Process Development, Lonza AG, Visp, Switzerland.
| | - Bernhard Bettler
- Department of Biomedicine, Institute of Physiology, Pharmazentrum, University of Basel, Basel, Switzerland.
| |
Collapse
|
12
|
Eszlari N, Bruncsics B, Millinghoffer A, Hullam G, Petschner P, Gonda X, Breen G, Antal P, Bagdy G, Deakin JFW, Juhasz G. Biology of Perseverative Negative Thinking: The Role of Timing and Folate Intake. Nutrients 2021; 13:4396. [PMID: 34959947 PMCID: PMC8703428 DOI: 10.3390/nu13124396] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/21/2021] [Revised: 12/02/2021] [Accepted: 12/06/2021] [Indexed: 11/20/2022] Open
Abstract
Past-oriented rumination and future-oriented worry are two aspects of perseverative negative thinking related to the neuroticism endophenotype and associated with depression and anxiety. Our present aim was to investigate the genomic background of these two aspects of perseverative negative thinking within separate groups of individuals with suboptimal versus optimal folate intake. We conducted a genome-wide association study in the UK Biobank database (n = 72,621) on the "rumination" and "worry" items of the Eysenck Personality Inventory Neuroticism scale in these separate groups. Optimal folate intake was related to lower worry, but unrelated to rumination. In contrast, genetic associations for worry did not implicate specific biological processes, while past-oriented rumination had a more specific genetic background, emphasizing its endophenotypic nature. Furthermore, biological pathways leading to rumination appeared to differ according to folate intake: purinergic signaling and circadian regulator gene ARNTL emerged in the whole sample, blastocyst development, DNA replication, and C-C chemokines in the suboptimal folate group, and prostaglandin response and K+ channel subunit gene KCNH3 in the optimal folate group. Our results point to possible benefits of folate in anxiety disorders, and to the importance of simultaneously taking into account genetic and environmental factors to determine personalized intervention in polygenic and multifactorial disorders.
Collapse
Affiliation(s)
- Nora Eszlari
- Department of Pharmacodynamics, Faculty of Pharmacy, Semmelweis University, Nagyvárad tér 4, H-1089 Budapest, Hungary; (P.P.); (G.B.); (G.J.)
- NAP-2-SE New Antidepressant Target Research Group, Hungarian Brain Research Program, Semmelweis University, Nagyvárad tér 4, H-1089 Budapest, Hungary; (A.M.); (X.G.)
| | - Bence Bruncsics
- Department of Measurement and Information Systems, Budapest University of Technology and Economics, Magyar Tudósok krt. 2, H-1521 Budapest, Hungary; (B.B.); (G.H.); (P.A.)
| | - Andras Millinghoffer
- NAP-2-SE New Antidepressant Target Research Group, Hungarian Brain Research Program, Semmelweis University, Nagyvárad tér 4, H-1089 Budapest, Hungary; (A.M.); (X.G.)
- Department of Measurement and Information Systems, Budapest University of Technology and Economics, Magyar Tudósok krt. 2, H-1521 Budapest, Hungary; (B.B.); (G.H.); (P.A.)
| | - Gabor Hullam
- Department of Measurement and Information Systems, Budapest University of Technology and Economics, Magyar Tudósok krt. 2, H-1521 Budapest, Hungary; (B.B.); (G.H.); (P.A.)
- MTA-SE Neuropsychopharmacology and Neurochemistry Research Group, Hungarian Academy of Sciences, Semmelweis University, Nagyvárad tér 4, H-1089 Budapest, Hungary
| | - Peter Petschner
- Department of Pharmacodynamics, Faculty of Pharmacy, Semmelweis University, Nagyvárad tér 4, H-1089 Budapest, Hungary; (P.P.); (G.B.); (G.J.)
- MTA-SE Neuropsychopharmacology and Neurochemistry Research Group, Hungarian Academy of Sciences, Semmelweis University, Nagyvárad tér 4, H-1089 Budapest, Hungary
- Bioinformatics Center, Institute for Chemical Research, Kyoto University, Gokasho, Uji, Kyoto 611-0011, Japan
| | - Xenia Gonda
- NAP-2-SE New Antidepressant Target Research Group, Hungarian Brain Research Program, Semmelweis University, Nagyvárad tér 4, H-1089 Budapest, Hungary; (A.M.); (X.G.)
- MTA-SE Neuropsychopharmacology and Neurochemistry Research Group, Hungarian Academy of Sciences, Semmelweis University, Nagyvárad tér 4, H-1089 Budapest, Hungary
- Department of Psychiatry and Psychotherapy, Semmelweis University, Gyulai Pál utca 2, H-1085 Budapest, Hungary
| | - Gerome Breen
- Social, Genetic and Developmental Psychiatry Centre, Institute of Psychiatry, Psychology and Neuroscience, King’s College London, Memory Lane, London SE5 8AF, UK;
- UK National Institute for Health Research (NIHR) Maudsley Biomedical Research Centre (BRC), London SE5 8AF, UK
| | - Peter Antal
- Department of Measurement and Information Systems, Budapest University of Technology and Economics, Magyar Tudósok krt. 2, H-1521 Budapest, Hungary; (B.B.); (G.H.); (P.A.)
| | - Gyorgy Bagdy
- Department of Pharmacodynamics, Faculty of Pharmacy, Semmelweis University, Nagyvárad tér 4, H-1089 Budapest, Hungary; (P.P.); (G.B.); (G.J.)
- NAP-2-SE New Antidepressant Target Research Group, Hungarian Brain Research Program, Semmelweis University, Nagyvárad tér 4, H-1089 Budapest, Hungary; (A.M.); (X.G.)
- MTA-SE Neuropsychopharmacology and Neurochemistry Research Group, Hungarian Academy of Sciences, Semmelweis University, Nagyvárad tér 4, H-1089 Budapest, Hungary
| | - John Francis William Deakin
- Division of Neuroscience and Experimental Psychology, Faculty of Biology, Medicine and Health, The University of Manchester, Oxford Road, Manchester M13 9PL, UK;
| | - Gabriella Juhasz
- Department of Pharmacodynamics, Faculty of Pharmacy, Semmelweis University, Nagyvárad tér 4, H-1089 Budapest, Hungary; (P.P.); (G.B.); (G.J.)
- MTA-SE Neuropsychopharmacology and Neurochemistry Research Group, Hungarian Academy of Sciences, Semmelweis University, Nagyvárad tér 4, H-1089 Budapest, Hungary
- SE-NAP 2 Genetic Brain Imaging Migraine Research Group, Hungarian Brain Research Program, Semmelweis University, Nagyvárad tér 4, H-1089 Budapest, Hungary
| |
Collapse
|
13
|
GABA B-Receptor Agonist-Based Immunotherapy for Type 1 Diabetes in NOD Mice. Biomedicines 2021; 9:biomedicines9010043. [PMID: 33418884 PMCID: PMC7825043 DOI: 10.3390/biomedicines9010043] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/07/2020] [Revised: 12/26/2020] [Accepted: 12/30/2020] [Indexed: 12/17/2022] Open
Abstract
Some immune system cells express type A and/or type B γ-aminobutyric acid receptors (GABAA-Rs and/or GABAB-Rs). Treatment with GABA, which activates both GABAA-Rs and GABAB-Rs), and/or a GABAA-R-specific agonist inhibits disease progression in mouse models of type 1 diabetes (T1D), multiple sclerosis, rheumatoid arthritis, and COVID-19. Little is known about the clinical potential of specifically modulating GABAB-Rs. Here, we tested lesogaberan, a peripherally restricted GABAB-R agonist, as an interventive therapy in diabetic NOD mice. Lesogaberan treatment temporarily restored normoglycemia in most newly diabetic NOD mice. Combined treatment with a suboptimal dose of lesogaberan and proinsulin/alum immunization in newly diabetic NOD mice or a low-dose anti-CD3 in severely hyperglycemic NOD mice greatly increased T1D remission rates relative to each monotherapy. Mice receiving combined lesogaberan and anti-CD3 displayed improved glucose tolerance and, unlike mice that received anti-CD3 alone, had some islets with many insulin+ cells, suggesting that lesogaberan helped to rapidly inhibit β-cell destruction. Hence, GABAB-R-specific agonists may provide adjunct therapies for T1D. Finally, the analysis of microarray and RNA-Seq databases suggested that the expression of GABAB-Rs and GABAA-Rs, as well as GABA production/secretion-related genes, may be a more common feature of immune cells than currently recognized.
Collapse
|
14
|
Mechanisms and Regulation of Neuronal GABA B Receptor-Dependent Signaling. Curr Top Behav Neurosci 2020; 52:39-79. [PMID: 32808092 DOI: 10.1007/7854_2020_129] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/26/2023]
Abstract
γ-Aminobutyric acid B receptors (GABABRs) are broadly expressed throughout the central nervous system where they play an important role in regulating neuronal excitability and synaptic transmission. GABABRs are G protein-coupled receptors that mediate slow and sustained inhibitory actions via modulation of several downstream effector enzymes and ion channels. GABABRs are obligate heterodimers that associate with diverse arrays of proteins to form modular complexes that carry out distinct physiological functions. GABABR-dependent signaling is fine-tuned and regulated through a multitude of mechanisms that are relevant to physiological and pathophysiological states. This review summarizes the current knowledge on GABABR signal transduction and discusses key factors that influence the strength and sensitivity of GABABR-dependent signaling in neurons.
Collapse
|
15
|
Eszlari N, Millinghoffer A, Petschner P, Gonda X, Baksa D, Pulay AJ, Réthelyi JM, Breen G, Deakin JFW, Antal P, Bagdy G, Juhasz G. Genome-wide association analysis reveals KCTD12 and miR-383-binding genes in the background of rumination. Transl Psychiatry 2019; 9:119. [PMID: 30886212 PMCID: PMC6423133 DOI: 10.1038/s41398-019-0454-1] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/21/2018] [Revised: 01/31/2019] [Accepted: 02/13/2019] [Indexed: 12/12/2022] Open
Abstract
Ruminative response style is a passive and repetitive way of responding to stress, associated with several disorders. Although twin and candidate gene studies have proven the genetic underpinnings of rumination, no genome-wide association study (GWAS) has been conducted yet. We performed a GWAS on ruminative response style and its two subtypes, brooding and reflection, among 1758 European adults recruited in the general population of Budapest, Hungary, and Manchester, United Kingdom. We evaluated single-nucleotide polymorphism (SNP)-based, gene-based and gene set-based tests, together with inferences on genes regulated by our most significant SNPs. While no genome-wide significant hit emerged at the SNP level, the association of rumination survived correction for multiple testing with KCTD12 at the gene level, and with the set of genes binding miR-383 at the gene set level. SNP-level results were concordant between the Budapest and Manchester subsamples for all three rumination phenotypes. SNP-level results and their links to brain expression levels based on external databases supported the role of KCTD12, SRGAP3, and SETD5 in rumination, CDH12 in brooding, and DPYSL5, MAPRE3, KCNK3, ATXN7L3B, and TPH2 in reflection, among others. The relatively low sample size is a limitation of our study. Results of the first GWAS on rumination identified genes previously implicated in psychiatric disorders underscoring the transdiagnostic nature of rumination, and pointed to the possible role of the dorsolateral prefrontal cortex, hippocampus, and cerebellum in this cognitive process.
Collapse
Affiliation(s)
- Nora Eszlari
- Department of Pharmacodynamics, Faculty of Pharmacy, Semmelweis University, Budapest, Hungary. .,NAP-2-SE New Antidepressant Target Research Group, Hungarian Brain Research Program, Semmelweis University, Budapest, Hungary.
| | - Andras Millinghoffer
- 0000 0001 0942 9821grid.11804.3cNAP-2-SE New Antidepressant Target Research Group, Hungarian Brain Research Program, Semmelweis University, Budapest, Hungary ,0000 0001 2180 0451grid.6759.dDepartment of Measurement and Information Systems, Budapest University of Technology and Economics, Budapest, Hungary
| | - Peter Petschner
- 0000 0001 0942 9821grid.11804.3cDepartment of Pharmacodynamics, Faculty of Pharmacy, Semmelweis University, Budapest, Hungary ,0000 0001 0942 9821grid.11804.3cMTA-SE Neuropsychopharmacology and Neurochemistry Research Group, Hungarian Academy of Sciences, Semmelweis University, Budapest, Hungary
| | - Xenia Gonda
- 0000 0001 0942 9821grid.11804.3cNAP-2-SE New Antidepressant Target Research Group, Hungarian Brain Research Program, Semmelweis University, Budapest, Hungary ,0000 0001 0942 9821grid.11804.3cMTA-SE Neuropsychopharmacology and Neurochemistry Research Group, Hungarian Academy of Sciences, Semmelweis University, Budapest, Hungary ,0000 0001 0942 9821grid.11804.3cDepartment of Psychiatry and Psychotherapy, Semmelweis University, Budapest, Hungary
| | - Daniel Baksa
- 0000 0001 0942 9821grid.11804.3cDepartment of Pharmacodynamics, Faculty of Pharmacy, Semmelweis University, Budapest, Hungary ,0000 0001 0942 9821grid.11804.3cSE-NAP 2 Genetic Brain Imaging Migraine Research Group, Hungarian Brain Research Program, Semmelweis University, Budapest, Hungary
| | - Attila J. Pulay
- 0000 0001 0942 9821grid.11804.3cDepartment of Psychiatry and Psychotherapy, Semmelweis University, Budapest, Hungary
| | - János M. Réthelyi
- 0000 0001 0942 9821grid.11804.3cDepartment of Psychiatry and Psychotherapy, Semmelweis University, Budapest, Hungary ,0000 0001 0942 9821grid.11804.3cNAP2 Molecular Psychiatry Research Group, Hungarian Brain Research Program, Semmelweis University, Budapest, Hungary
| | - Gerome Breen
- 0000 0001 2322 6764grid.13097.3cSocial, Genetic and Developmental Psychiatry Centre, King’s College London, London, UK
| | - John Francis William Deakin
- 0000000121662407grid.5379.8Division of Neuroscience and Experimental Psychology, Faculty of Biology, Medicine and Health, University of Manchester, Manchester, UK ,0000 0004 0417 0074grid.462482.eManchester Academic Health Sciences Centre, Manchester, UK ,0000 0004 0430 6955grid.450837.dGreater Manchester Mental Health NHS Foundation Trust, Prestwich, Manchester, M25 3BL UK
| | - Peter Antal
- 0000 0001 2180 0451grid.6759.dDepartment of Measurement and Information Systems, Budapest University of Technology and Economics, Budapest, Hungary
| | - Gyorgy Bagdy
- 0000 0001 0942 9821grid.11804.3cDepartment of Pharmacodynamics, Faculty of Pharmacy, Semmelweis University, Budapest, Hungary ,0000 0001 0942 9821grid.11804.3cNAP-2-SE New Antidepressant Target Research Group, Hungarian Brain Research Program, Semmelweis University, Budapest, Hungary ,0000 0001 0942 9821grid.11804.3cMTA-SE Neuropsychopharmacology and Neurochemistry Research Group, Hungarian Academy of Sciences, Semmelweis University, Budapest, Hungary
| | - Gabriella Juhasz
- 0000 0001 0942 9821grid.11804.3cDepartment of Pharmacodynamics, Faculty of Pharmacy, Semmelweis University, Budapest, Hungary ,0000 0001 0942 9821grid.11804.3cNAP-2-SE New Antidepressant Target Research Group, Hungarian Brain Research Program, Semmelweis University, Budapest, Hungary ,0000 0001 0942 9821grid.11804.3cMTA-SE Neuropsychopharmacology and Neurochemistry Research Group, Hungarian Academy of Sciences, Semmelweis University, Budapest, Hungary ,0000 0001 0942 9821grid.11804.3cSE-NAP 2 Genetic Brain Imaging Migraine Research Group, Hungarian Brain Research Program, Semmelweis University, Budapest, Hungary ,0000000121662407grid.5379.8Division of Neuroscience and Experimental Psychology, Faculty of Biology, Medicine and Health, University of Manchester, Manchester, UK ,0000 0004 0417 0074grid.462482.eManchester Academic Health Sciences Centre, Manchester, UK
| |
Collapse
|
16
|
Sleno R, Hébert TE. Shaky ground - The nature of metastable GPCR signalling complexes. Neuropharmacology 2019; 152:4-14. [PMID: 30659839 DOI: 10.1016/j.neuropharm.2019.01.018] [Citation(s) in RCA: 20] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/30/2018] [Revised: 12/20/2018] [Accepted: 01/16/2019] [Indexed: 01/19/2023]
Abstract
How G protein-coupled receptors (GPCR) interact with one another remains an area of active investigation. Obligate dimers of class C GPCRs such as metabotropic GABA and glutamate receptors are well accepted, although whether this is a general feature of other GPCRs is still strongly debated. In this review, we focus on the idea that GPCR dimers and oligomers are better imagined as parts of larger metastable signalling complexes. We discuss the nature of functional oligomeric entities, their stabilities and kinetic features and how structural and functional asymmetries of such metastable entities might have implications for drug discovery. This article is part of the Special Issue entitled 'Receptor heteromers and their allosteric receptor-receptor interactions'.
Collapse
Affiliation(s)
- Rory Sleno
- Marketed Pharmaceuticals and Medical Devices Bureau, Marketed Health Products Directorate, Health Products and Food Branch, Health Canada, Canada
| | - Terence E Hébert
- Department of Pharmacology and Therapeutics, McGill University, Canada.
| |
Collapse
|
17
|
Bukiya AN, Blank PS, Rosenhouse-Dantsker A. Cholesterol intake and statin use regulate neuronal G protein-gated inwardly rectifying potassium channels. J Lipid Res 2018; 60:19-29. [PMID: 30420402 DOI: 10.1194/jlr.m081240] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/18/2017] [Revised: 11/10/2018] [Indexed: 12/31/2022] Open
Abstract
Cholesterol, a critical component of the cellular plasma membrane, is essential for normal neuronal function. Cholesterol content is highest in the brain, where most cholesterol is synthesized de novo; HMG-CoA reductase controls the synthesis rate. Despite strict control, elevated blood cholesterol levels are common and are associated with various neurological disorders. G protein-gated inwardly rectifying potassium (GIRK) channels mediate the actions of inhibitory brain neurotransmitters. Loss of GIRK function enhances neuron excitability; gain of function reduces neuronal activity. However, the effect of dietary cholesterol or HMG-CoA reductase inhibition (i.e., statin therapy) on GIRK function remains unknown. Using a rat model, we compared the effects of a high-cholesterol versus normal diet both with and without atorvastatin, a widely prescribed HMG-CoA reductase inhibitor, on neuronal GIRK currents. The high-cholesterol diet increased hippocampal CA1 region cholesterol levels and correspondingly increased neuronal GIRK currents. Both phenomena were reversed by cholesterol depletion in vitro. Atorvastatin countered the high-cholesterol diet effects on neuronal cholesterol content and GIRK currents; these effects were reversed by cholesterol enrichment in vitro. Our findings suggest that high-cholesterol diet and atorvastatin therapy affect ion channel function in the brain by modulating neuronal cholesterol levels.
Collapse
Affiliation(s)
- Anna N Bukiya
- Department of Pharmacology, The University of Tennessee Health Science Center, Memphis, TN 38163
| | - Paul S Blank
- Section on Integrative Biophysics, Eunice Kennedy Shriver National Institute of Child Health and Human Development, National Institutes of Health, Bethesda, MD 20892
| | | |
Collapse
|
18
|
Sleno R, Hébert TE. The Dynamics of GPCR Oligomerization and Their Functional Consequences. INTERNATIONAL REVIEW OF CELL AND MOLECULAR BIOLOGY 2018; 338:141-171. [PMID: 29699691 DOI: 10.1016/bs.ircmb.2018.02.005] [Citation(s) in RCA: 29] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/20/2022]
Abstract
The functional importance of G protein-coupled receptor (GPCR) oligomerization remains controversial. Although obligate dimers of class C GPCRs are well accepted, the generalizability of this phenomenon is still strongly debated with respect to other classes of GPCRs. In this review, we focus on understanding the organization and dynamics between receptor equivalents and their signaling partners in oligomeric receptor complexes, with a view toward integrating disparate viewpoints into a unified understanding. We discuss the nature of functional oligomeric entities, and how asymmetries in receptor structure and function created by oligomers might have implications for receptor function as allosteric machines and for future drug discovery.
Collapse
|
19
|
Differential association of GABA B receptors with their effector ion channels in Purkinje cells. Brain Struct Funct 2017; 223:1565-1587. [PMID: 29177691 PMCID: PMC5869904 DOI: 10.1007/s00429-017-1568-y] [Citation(s) in RCA: 21] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/26/2017] [Accepted: 11/10/2017] [Indexed: 11/23/2022]
Abstract
Metabotropic GABAB receptors mediate slow inhibitory effects presynaptically and postsynaptically through the modulation of different effector signalling pathways. Here, we analysed the distribution of GABAB receptors using highly sensitive SDS-digested freeze-fracture replica labelling in mouse cerebellar Purkinje cells. Immunoreactivity for GABAB1 was observed on presynaptic and, more abundantly, on postsynaptic compartments, showing both scattered and clustered distribution patterns. Quantitative analysis of immunoparticles revealed a somato-dendritic gradient, with the density of immunoparticles increasing 26-fold from somata to dendritic spines. To understand the spatial relationship of GABAB receptors with two key effector ion channels, the G protein-gated inwardly rectifying K+ (GIRK/Kir3) channel and the voltage-dependent Ca2+ channel, biochemical and immunohistochemical approaches were performed. Co-immunoprecipitation analysis demonstrated that GABAB receptors co-assembled with GIRK and CaV2.1 channels in the cerebellum. Using double-labelling immunoelectron microscopic techniques, co-clustering between GABAB1 and GIRK2 was detected in dendritic spines, whereas they were mainly segregated in the dendritic shafts. In contrast, co-clustering of GABAB1 and CaV2.1 was detected in dendritic shafts but not spines. Presynaptically, although no significant co-clustering of GABAB1 and GIRK2 or CaV2.1 channels was detected, inter-cluster distance for GABAB1 and GIRK2 was significantly smaller in the active zone than in the dendritic shafts, and that for GABAB1 and CaV2.1 was significantly smaller in the active zone than in the dendritic shafts and spines. Thus, GABAB receptors are associated with GIRK and CaV2.1 channels in different subcellular compartments. These data provide a better framework for understanding the different roles played by GABAB receptors and their effector ion channels in the cerebellar network.
Collapse
|
20
|
Kahanovitch U, Berlin S, Dascal N. Collision coupling in the GABA
B
receptor–G protein–GIRK signaling cascade. FEBS Lett 2017; 591:2816-2825. [DOI: 10.1002/1873-3468.12756] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/10/2017] [Revised: 07/11/2017] [Accepted: 07/12/2017] [Indexed: 11/08/2022]
Affiliation(s)
- Uri Kahanovitch
- Department of Physiology and Pharmacology Sackler School of Medicine Tel Aviv University Israel
| | - Shai Berlin
- Department of Physiology and Pharmacology Sackler School of Medicine Tel Aviv University Israel
| | - Nathan Dascal
- Department of Physiology and Pharmacology Sackler School of Medicine Tel Aviv University Israel
- Sagol School of Neuroscience Tel Aviv University Israel
| |
Collapse
|
21
|
Contestabile A, Magara S, Cancedda L. The GABAergic Hypothesis for Cognitive Disabilities in Down Syndrome. Front Cell Neurosci 2017; 11:54. [PMID: 28326014 PMCID: PMC5339239 DOI: 10.3389/fncel.2017.00054] [Citation(s) in RCA: 83] [Impact Index Per Article: 10.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/21/2016] [Accepted: 02/14/2017] [Indexed: 12/04/2022] Open
Abstract
Down syndrome (DS) is a genetic disorder caused by the presence of a third copy of chromosome 21. DS affects multiple organs, but it invariably results in altered brain development and diverse degrees of intellectual disability. A large body of evidence has shown that synaptic deficits and memory impairment are largely determined by altered GABAergic signaling in trisomic mouse models of DS. These alterations arise during brain development while extending into adulthood, and include genesis of GABAergic neurons, variation of the inhibitory drive and modifications in the control of neural-network excitability. Accordingly, different pharmacological interventions targeting GABAergic signaling have proven promising preclinical approaches to rescue cognitive impairment in DS mouse models. In this review, we will discuss recent data regarding the complex scenario of GABAergic dysfunctions in the trisomic brain of DS mice and patients, and we will evaluate the state of current clinical research targeting GABAergic signaling in individuals with DS.
Collapse
Affiliation(s)
- Andrea Contestabile
- Department of Neuroscience and Brain Technologies, Istituto Italiano di Tecnologia (IIT) Genova, Italy
| | - Salvatore Magara
- Department of Neuroscience and Brain Technologies, Istituto Italiano di Tecnologia (IIT) Genova, Italy
| | - Laura Cancedda
- Department of Neuroscience and Brain Technologies, Istituto Italiano di Tecnologia (IIT)Genova, Italy; Dulbecco Telethon InstituteGenova, Italy
| |
Collapse
|
22
|
Bukiya AN, Durdagi S, Noskov S, Rosenhouse-Dantsker A. Cholesterol up-regulates neuronal G protein-gated inwardly rectifying potassium (GIRK) channel activity in the hippocampus. J Biol Chem 2017; 292:6135-6147. [PMID: 28213520 DOI: 10.1074/jbc.m116.753350] [Citation(s) in RCA: 30] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/11/2016] [Revised: 02/13/2017] [Indexed: 02/06/2023] Open
Abstract
Hypercholesterolemia is a well known risk factor for the development of neurodegenerative disease. However, the underlying mechanisms are mostly unknown. In recent years, it has become increasingly evident that cholesterol-driven effects on physiology and pathophysiology derive from its ability to alter the function of a variety of membrane proteins including ion channels. Yet, the effect of cholesterol on G protein-gated inwardly rectifying potassium (GIRK) channels expressed in the brain is unknown. GIRK channels mediate the actions of inhibitory brain neurotransmitters. As a result, loss of GIRK function can enhance neuron excitability, whereas gain of GIRK function can reduce neuronal activity. Here we show that in rats on a high-cholesterol diet, cholesterol levels in hippocampal neurons are increased. We also demonstrate that cholesterol plays a critical role in modulating neuronal GIRK currents. Specifically, cholesterol enrichment of rat hippocampal neurons resulted in enhanced channel activity. In accordance, elevated currents upon cholesterol enrichment were also observed in Xenopus oocytes expressing GIRK2 channels, the primary GIRK subunit expressed in the brain. Furthermore, using planar lipid bilayers, we show that although cholesterol did not affect the unitary conductance of GIRK2, it significantly enhanced the frequency of channel openings. Last, combining computational and functional approaches, we identified two putative cholesterol-binding sites in the transmembrane domain of GIRK2. These findings establish that cholesterol plays a critical role in modulating GIRK activity in the brain. Because up-regulation of GIRK function can reduce neuronal activity, our findings may lead to novel approaches for prevention and therapy of cholesterol-driven neurodegenerative disease.
Collapse
Affiliation(s)
- Anna N Bukiya
- the Department of Pharmacology, The University of Tennessee Health Science Center, Memphis, Tennessee 38103
| | - Serdar Durdagi
- the Centre for Molecular Simulation and Department of Biological Sciences, University of Calgary, Calgary, AB, T2N 1N4 Canada, and.,the Department of Biophysics, School of Medicine, Bahcesehir University, Istanbul 34353, Turkey
| | - Sergei Noskov
- the Centre for Molecular Simulation and Department of Biological Sciences, University of Calgary, Calgary, AB, T2N 1N4 Canada, and
| | | |
Collapse
|
23
|
Yakubovich D, Berlin S, Kahanovitch U, Rubinstein M, Farhy-Tselnicker I, Styr B, Keren-Raifman T, Dessauer CW, Dascal N. A Quantitative Model of the GIRK1/2 Channel Reveals That Its Basal and Evoked Activities Are Controlled by Unequal Stoichiometry of Gα and Gβγ. PLoS Comput Biol 2015; 11:e1004598. [PMID: 26544551 PMCID: PMC4636287 DOI: 10.1371/journal.pcbi.1004598] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/27/2015] [Accepted: 10/13/2015] [Indexed: 12/02/2022] Open
Abstract
G protein-gated K+ channels (GIRK; Kir3), activated by Gβγ subunits derived from Gi/o proteins, regulate heartbeat and neuronal excitability and plasticity. Both neurotransmitter-evoked (Ievoked) and neurotransmitter-independent basal (Ibasal) GIRK activities are physiologically important, but mechanisms of Ibasal and its relation to Ievoked are unclear. We have previously shown for heterologously expressed neuronal GIRK1/2, and now show for native GIRK in hippocampal neurons, that Ibasal and Ievoked are interrelated: the extent of activation by neurotransmitter (activation index, Ra) is inversely related to Ibasal. To unveil the underlying mechanisms, we have developed a quantitative model of GIRK1/2 function. We characterized single-channel and macroscopic GIRK1/2 currents, and surface densities of GIRK1/2 and Gβγ expressed in Xenopus oocytes. Based on experimental results, we constructed a mathematical model of GIRK1/2 activity under steady-state conditions before and after activation by neurotransmitter. Our model accurately recapitulates Ibasal and Ievoked in Xenopus oocytes, HEK293 cells and hippocampal neurons; correctly predicts the dose-dependent activation of GIRK1/2 by coexpressed Gβγ and fully accounts for the inverse Ibasal-Ra correlation. Modeling indicates that, under all conditions and at different channel expression levels, between 3 and 4 Gβγ dimers are available for each GIRK1/2 channel. In contrast, available Gαi/o decreases from ~2 to less than one Gα per channel as GIRK1/2's density increases. The persistent Gβγ/channel (but not Gα/channel) ratio support a strong association of GIRK1/2 with Gβγ, consistent with recruitment to the cell surface of Gβγ, but not Gα, by GIRK1/2. Our analysis suggests a maximal stoichiometry of 4 Gβγ but only 2 Gαi/o per one GIRK1/2 channel. The unique, unequal association of GIRK1/2 with G protein subunits, and the cooperative nature of GIRK gating by Gβγ, underlie the complex pattern of basal and agonist-evoked activities and allow GIRK1/2 to act as a sensitive bidirectional detector of both Gβγ and Gα. Many neurotransmitters and hormones inhibit the electric activity of excitable cells (such as cardiac cells and neurons) by activating a K+ channel, GIRK (G protein-gated Inwardly Rectifying K+ channel). GIRK channels also possess constitutive “basal” activity which contributes to regulation of neuronal and cardiac excitability and certain disorders, but the mechanism of this activity and its interrelation with the neurotransmitter-evoked activity are poorly understood. In this work we show that key features of basal and neurotransmitter-evoked activities are similar in cultured hippocampal neurons and in two model systems (mammalian HEK293 cells and Xenopus oocytes). Using experimental data of the neuronal GIRK1/2 channel function upon changes in GIRK and G protein concentrations, we constructed a mathematical model that quantitatively accounts for basal and evoked activity, and for the inverse correlation between the two. Our analysis suggests a novel and unexpected mechanism of interaction of GIRK1/2 with the G protein subunits, where the tetrameric GIRK channel can assemble with 4 molecules of the Gβγ subunits but only 2 molecules of Gα. GIRK is a prototypical effector of Gβγ, and the unequal stoichiometry of interaction with G protein subunits may have general implications for G protein signaling.
Collapse
Affiliation(s)
- Daniel Yakubovich
- Department of Physiology and Pharmacology and Sagol School of Neuroscience, Tel Aviv University, Tel Aviv, Israel
| | - Shai Berlin
- Department of Physiology and Pharmacology and Sagol School of Neuroscience, Tel Aviv University, Tel Aviv, Israel
| | - Uri Kahanovitch
- Department of Physiology and Pharmacology and Sagol School of Neuroscience, Tel Aviv University, Tel Aviv, Israel
| | - Moran Rubinstein
- Department of Physiology and Pharmacology and Sagol School of Neuroscience, Tel Aviv University, Tel Aviv, Israel
| | - Isabella Farhy-Tselnicker
- Department of Physiology and Pharmacology and Sagol School of Neuroscience, Tel Aviv University, Tel Aviv, Israel
| | - Boaz Styr
- Department of Physiology and Pharmacology and Sagol School of Neuroscience, Tel Aviv University, Tel Aviv, Israel
| | - Tal Keren-Raifman
- Department of Physiology and Pharmacology and Sagol School of Neuroscience, Tel Aviv University, Tel Aviv, Israel
| | - Carmen W. Dessauer
- Department of Integrative Biology and Pharmacology, University of Texas Health Science Center, Houston, Texas, United States of America
| | - Nathan Dascal
- Department of Physiology and Pharmacology and Sagol School of Neuroscience, Tel Aviv University, Tel Aviv, Israel
- * E-mail:
| |
Collapse
|
24
|
Raveh A, Turecek R, Bettler B. Mechanisms of fast desensitization of GABA(B) receptor-gated currents. ADVANCES IN PHARMACOLOGY (SAN DIEGO, CALIF.) 2015; 73:145-65. [PMID: 25637440 DOI: 10.1016/bs.apha.2014.11.004] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
GABA(B) receptors (GABA(B)Rs) regulate the excitability of most neurons in the central nervous system by modulating the activity of enzymes and ion channels. In the sustained presence of the neurotransmitter γ-aminobutyric acid, GABA(B)Rs exhibit a time-dependent decrease in the receptor response-a phenomenon referred to as homologous desensitization. Desensitization prevents excessive receptor influences on neuronal activity. Much work focused on the mechanisms of GABA(B)R desensitization that operate at the receptor and control receptor expression at the plasma membrane. Over the past few years, it became apparent that GABA(B)Rs additionally evolved mechanisms for faster desensitization. These mechanisms operate at the G protein rather than at the receptor and inhibit G protein signaling within seconds of agonist exposure. The mechanisms for fast desensitization are ideally suited to regulate receptor-activated ion channel responses, which influence neuronal activity on a faster timescale than effector enzymes. Here, we provide an update on the mechanisms for fast desensitization of GABA(B)R responses and discuss physiological and pathophysiological implications.
Collapse
Affiliation(s)
- Adi Raveh
- Department of Biomedicine, Institute of Physiology, Pharmazentrum, University of Basel, Basel, Switzerland
| | - Rostislav Turecek
- Department of Biomedicine, Institute of Physiology, Pharmazentrum, University of Basel, Basel, Switzerland; Department of Auditory Neuroscience, Institute of Experimental Medicine, ASCR, Prague, Czech Republic
| | - Bernhard Bettler
- Department of Biomedicine, Institute of Physiology, Pharmazentrum, University of Basel, Basel, Switzerland.
| |
Collapse
|
25
|
Activation of GABA(B) receptors potentiates inward rectifying potassium currents in satellite glial cells from rat trigeminal ganglia: in vivo patch-clamp analysis. Neuroscience 2014; 288:51-8. [PMID: 25542421 DOI: 10.1016/j.neuroscience.2014.12.024] [Citation(s) in RCA: 21] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/12/2014] [Revised: 12/15/2014] [Accepted: 12/15/2014] [Indexed: 11/23/2022]
Abstract
In a previous study, we demonstrated that inflammation suppressed inward rectifying K(+) (Kir) currents in satellite glial cells (SGCs) from the trigeminal ganglia (TRGs) and that this impairment of glial potassium homeostasis in the trigeminal ganglion (TRG) contributed to trigeminal pain. The aim of the present study was to investigate whether activation of GABAB receptors modulates the Kir current in SGCs using in vivo patch-clamp and immunohistochemical techniques. Immunohistochemically, we found that immunoreactivity for glial-specific Kir channel subunit Kir4.1 and the GABAB receptor was co-expressed in SGCs from the TRGs. In vivo whole-cell recordings were made using SGCs from the TRGs of urethane-anesthetized rats. Application of baclofen, a GABAB receptor agonist, significantly increased the mean peak amplitude of Kir currents in a concentration-dependent and reversible manner. Baclofen-induced potentiation of the Kir current was abolished by co-application of 3-amino-2-(4-chlorophenyl)-2-hydroxyprophylsulfonic acid (saclofen). In addition, baclofen significantly potentiated the density of the Ba(2+)-sensitive Kir current, and resulted in hyperpolarization of the mean membrane potential. These results suggest that activation of GABAB receptors potentiates the Kir current in SGCs and that GABA released from the TRG neuronal soma could contribute to buffering of extracellular K(+) concentrations following excitation of TRG neurons during the processing of sensory information, including the transmission of noxious stimuli.
Collapse
|
26
|
Borin M, Fogli Iseppe A, Pignatelli A, Belluzzi O. Inward rectifier potassium (Kir) current in dopaminergic periglomerular neurons of the mouse olfactory bulb. Front Cell Neurosci 2014; 8:223. [PMID: 25152712 PMCID: PMC4126183 DOI: 10.3389/fncel.2014.00223] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/27/2014] [Accepted: 07/21/2014] [Indexed: 11/23/2022] Open
Abstract
Dopaminergic (DA) periglomerular (PG) neurons are critically placed at the entry of the bulbar circuitry, directly in contact with both the terminals of olfactory sensory neurons and the apical dendrites of projection neurons; they are autorhythmic and are the target of numerous terminals releasing a variety of neurotransmitters. Despite the centrality of their position, suggesting a critical role in the sensory processing, their properties -and consequently their function- remain elusive. The current mediated by inward rectifier potassium (Kir) channels in DA-PG cells was recorded by adopting the perforated-patch configuration in thin slices; IKir could be distinguished from the hyperpolarization-activated current (I h ) by showing full activation in <10 ms, no inactivation, suppression by Ba(2+) in a typical voltage-dependent manner (IC50 208 μM) and reversal potential nearly coincident with EK. Ba(2+) (2 mM) induces a large depolarization of DA-PG cells, paralleled by an increase of the input resistance, leading to a block of the spontaneous activity, but the Kir current is not an essential component of the pacemaker machinery. The Kir current is negatively modulated by intracellular cAMP, as shown by a decrease of its amplitude induced by forskolin or 8Br-cAMP. We have also tested the neuromodulatory effects of the activation of several metabotropic receptors known to be present on these cells, showing that the current can be modulated by a multiplicity of pathways, whose activation in some case increases the amplitude of the current, as can be observed with agonists of D2, muscarinic, and GABAA receptors, whereas in other cases has the opposite effect, as it can be observed with agonists of α1 noradrenergic, 5-HT and histamine receptors. These characteristics of the Kir currents provide the basis for an unexpected plasticity of DA-PG cell function, making them potentially capable to reconfigure the bulbar network to allow a better flexibility.
Collapse
Affiliation(s)
| | | | | | - Ottorino Belluzzi
- Department of Life Sciences and Biotechnology, University of FerraraFerrara, Italy
| |
Collapse
|
27
|
Zylbergold P, Sleno R, Khan SM, Jacobi AM, Belhke MA, Hébert TE. Kir3 channel ontogeny - the role of Gβγ subunits in channel assembly and trafficking. Front Cell Neurosci 2014; 8:108. [PMID: 24782712 PMCID: PMC3995069 DOI: 10.3389/fncel.2014.00108] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/11/2014] [Accepted: 03/25/2014] [Indexed: 11/13/2022] Open
Abstract
The role of Gβγ subunits in Kir3 channel gating is well characterized. Here, we have studied the role of Gβγ dimers during their initial contact with Kir3 channels, prior to their insertion into the plasma membrane. We show that distinct Gβγ subunits play an important role in orchestrating and fine-tuning parts of the Kir3 channel life cycle. Gβ1γ2, apart from its role in channel opening that it shares with other Gβγ subunit combinations, may play a unique role in protecting maturing channels from degradation as they transit to the cell surface. Taken together, our data suggest that Gβ1γ2 prolongs the lifetime of the Kir3.1/Kir3.2 heterotetramer, although further studies would be required to shed more light on these early Gβγ effects on Kir3 maturation and trafficking.
Collapse
Affiliation(s)
- Peter Zylbergold
- Department of Pharmacology and Therapeutics, McGill University Montréal, QC, Canada
| | - Rory Sleno
- Department of Pharmacology and Therapeutics, McGill University Montréal, QC, Canada
| | - Shahriar M Khan
- Department of Pharmacology and Therapeutics, McGill University Montréal, QC, Canada
| | | | - Mark A Belhke
- Integrated DNA Technologies, Inc., Coralville IA, USA
| | - Terence E Hébert
- Department of Pharmacology and Therapeutics, McGill University Montréal, QC, Canada
| |
Collapse
|
28
|
Kan W, Adjobo-Hermans M, Burroughs M, Faibis G, Malik S, Tall GG, Smrcka AV. M3 muscarinic receptor interaction with phospholipase C β3 determines its signaling efficiency. J Biol Chem 2014; 289:11206-11218. [PMID: 24596086 DOI: 10.1074/jbc.m113.538546] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/06/2023] Open
Abstract
Phospholipase Cβ (PLCβ) enzymes are activated by G protein-coupled receptors through receptor-catalyzed guanine nucleotide exchange on Gαβγ heterotrimers containing Gq family G proteins. Here we report evidence for a direct interaction between M3 muscarinic receptor (M3R) and PLCβ3. Both expressed and endogenous M3R interacted with PLCβ in coimmunoprecipitation experiments. Stimulation of M3R with carbachol significantly increased this association. Expression of M3R in CHO cells promoted plasma membrane localization of YFP-PLCβ3. Deletion of the PLCβ3 C terminus or deletion of the PLCβ3 PDZ ligand inhibited coimmunoprecipitation with M3R and M3R-dependent PLCβ3 plasma membrane localization. Purified PLCβ3 bound directly to glutathione S-transferase (GST)-fused M3R intracellular loops 2 and 3 (M3Ri2 and M3Ri3) as well as M3R C terminus (M3R/H8-CT). PLCβ3 binding to M3Ri3 was inhibited when the PDZ ligand was removed. In assays using reconstituted purified components in vitro, M3Ri2, M3Ri3, and M3R/H8-CT potentiated Gαq-dependent but not Gβγ-dependent PLCβ3 activation. Disruption of key residues in M3Ri3N and of the PDZ ligand in PLCβ3 inhibited M3Ri3-mediated potentiation. We propose that the M3 muscarinic receptor maximizes the efficiency of PLCβ3 signaling beyond its canonical role as a guanine nucleotide exchange factor for Gα.
Collapse
Affiliation(s)
- Wei Kan
- Departments of Pharmacology and Physiology and University of Rochester School of Medicine and Dentistry, Rochester, New York 14642
| | - Merel Adjobo-Hermans
- Department of Biochemistry, Nijmegen Centre for Molecular Life Sciences, Radboud University Nijmegen Medical Centre, Geert Grooteplein 28, 6525 GA Nijmegen, The Netherlands
| | - Michael Burroughs
- Departments of Pharmacology and Physiology and University of Rochester School of Medicine and Dentistry, Rochester, New York 14642
| | - Guy Faibis
- Departments of Pharmacology and Physiology and University of Rochester School of Medicine and Dentistry, Rochester, New York 14642
| | - Sundeep Malik
- Departments of Pharmacology and Physiology and University of Rochester School of Medicine and Dentistry, Rochester, New York 14642
| | - Gregory G Tall
- Departments of Pharmacology and Physiology and University of Rochester School of Medicine and Dentistry, Rochester, New York 14642
| | - Alan V Smrcka
- Departments of Pharmacology and Physiology and University of Rochester School of Medicine and Dentistry, Rochester, New York 14642; Biochemistry and Biophysics and University of Rochester School of Medicine and Dentistry, Rochester, New York 14642; Aab Institute of Cardiovascular Research, University of Rochester School of Medicine and Dentistry, Rochester, New York 14642 and.
| |
Collapse
|
29
|
Babilon S, Mörl K, Beck-Sickinger AG. Towards improved receptor targeting: anterograde transport, internalization and postendocytic trafficking of neuropeptide Y receptors. Biol Chem 2013; 394:921-36. [DOI: 10.1515/hsz-2013-0123] [Citation(s) in RCA: 35] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/19/2013] [Accepted: 02/22/2013] [Indexed: 01/10/2023]
Abstract
Abstract
The neuropeptide Y system is known to be involved in the regulation of many central physiological and pathophysiological processes, such as energy homeostasis, obesity, cancer, mood disorders and epilepsy. Four Y receptor subtypes have been cloned from human tissue (hY1, hY2, hY4 and hY5) that form a multiligand/multireceptor system together with their three peptidic agonists (NPY, PYY and PP). Addressing this system for medical application requires on the one hand detailed information about the receptor-ligand interaction to design subtype-selective compounds. On the other hand comprehensive knowledge about alternative receptor signaling, as well as desensitization, localization and downregulation is crucial to circumvent the development of undesired side-effects and drug resistance. By bringing such knowledge together, highly potent and long-lasting drugs with minimized side-effects can be engineered. Here, current knowledge about Y receptor export, internalization, recycling, and degradation is summarized, with a focus on the human Y receptor subtypes, and is discussed in terms of its impact on therapeutic application.
Collapse
|
30
|
Murthy R, Kim J, Sun X, Giger RJ, Fink DJ, Mata M. Post-transcriptional regulation of GABAB receptor and GIRK1 channels by Nogo receptor 1. Mol Brain 2013; 6:30. [PMID: 23829864 PMCID: PMC3734105 DOI: 10.1186/1756-6606-6-30] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/02/2013] [Accepted: 07/01/2013] [Indexed: 11/10/2022] Open
Abstract
Background Type B GABA receptors (GABA Rs) play a critical role in synaptic transmission. We carried out studies to determine whether neuronal cell surface expression of GABAB-Rs might be regulated by the Nogo receptor 1 (NgR1). Results siRNA knock-down of NgR1 resulted in a selective increase of GABAB R1 and GABAB R2 protein without altering the expression of GABAA receptor or GAD65. The increase in GABAB receptor subunits was unaccompanied by a change in mRNA, but inhibition of mTOR by rapamycin blocked the increase in GABAB protein. NgR1 siRNA also caused an increase in G protein coupled inwardly rectifying potassium channel (GIRK1). The increase in GABAB receptor and GIRK1 channel proteins was in the plasma membrane, determined by cell surface biotinylation. In NgR1 knockout mice, the amount of GABAB R2 and GIRK1 in hippocampus-derived synaptosomes was increased. Conclusions Together these findings suggest that NgR1 mediated modulation of synaptic transmission may be accomplished, at least in part, through modulation of G protein coupled receptors and channels.
Collapse
Affiliation(s)
- Rachana Murthy
- Department of Neurology, University of Michigan and VA Ann Arbor Healthcare System-Neurology and GRECC, 5027 BSRB, 109 Zina Pitcher Place, Ann Arbor, MI 48109, USA
| | | | | | | | | | | |
Collapse
|
31
|
Richard-Lalonde M, Nagi K, Audet N, Sleno R, Amraei M, Hogue M, Balboni G, Schiller PW, Bouvier M, Hébert TE, Pineyro G. Conformational dynamics of Kir3.1/Kir3.2 channel activation via δ-opioid receptors. Mol Pharmacol 2013; 83:416-28. [PMID: 23175530 PMCID: PMC3558812 DOI: 10.1124/mol.112.081950] [Citation(s) in RCA: 44] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/19/2012] [Accepted: 11/21/2012] [Indexed: 01/09/2023] Open
Abstract
This study assessed how conformational information encoded by ligand binding to δ-opioid receptors (DORs) is transmitted to Kir3.1/Kir3.2 channels. Human embryonic kidney 293 cells were transfected with bioluminescence resonance energy transfer (BRET) donor/acceptor pairs that allowed us to evaluate independently reciprocal interactions among signaling partners. These and coimmunoprecipitation studies indicated that DORs, Gβγ, and Kir3 subunits constitutively interacted with one another. GαoA associated with DORs and Gβγ, but despite being part of the complex, no evidence of its direct association with the channel was obtained. DOR activation by different ligands left DOR-Kir3 interactions unmodified but modulated BRET between DOR-GαoA, DOR-Gβγ, GαoA-Gβγ, and Gβγ-Kir3 interfaces. Ligand-induced BRET changes assessing Gβγ-Kir3.1 subunit interaction 1) followed similar kinetics to those monitoring the GαoA-Gβγ interface, 2) displayed the same order of efficacy as those observed at the DOR-Gβγ interface, 3) were sensitive to pertussis toxin, and 4) were predictive of whether a ligand could evoke channel currents. Conformational changes at the Gβγ/Kir3 interface were lost when Kir3.1 subunits were replaced by a mutant lacking essential sites for Gβγ-mediated activation. Thus, conformational information encoded by agonist binding to the receptor is relayed to the channel via structural rearrangements that involve repositioning of Gβγ with respect to DORs, GαoA, and channel subunits. Further, the fact that BRET changes at the Gβγ-Kir3 interface are predictive of a ligand's ability to induce channel currents points to these conformational biosensors as screening tools for identifying GPCR ligands that induce Kir3 channel activation.
Collapse
|
32
|
Zylbergold P, Sleno R, Hébert TE. A novel, radiolabel-free pulse chase strategy to study Kir3 channel ontogeny. J Recept Signal Transduct Res 2013; 33:144-52. [DOI: 10.3109/10799893.2013.764898] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/26/2022]
|
33
|
Baloucoune GA, Chun L, Zhang W, Xu C, Huang S, Sun Q, Wang Y, Tu H, Liu J. GABAB receptor subunit GB1 at the cell surface independently activates ERK1/2 through IGF-1R transactivation. PLoS One 2012; 7:e39698. [PMID: 22761875 PMCID: PMC3386256 DOI: 10.1371/journal.pone.0039698] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/26/2012] [Accepted: 05/29/2012] [Indexed: 12/24/2022] Open
Abstract
Background Functional GABAB receptor is believed to require hetero-dimerization between GABAB1 (GB1) and GABAB2 (GB2) subunits. The GB1 extracellular domain is required for ligand binding, and the GB2 trans-membrane domain is responsible for coupling to G proteins. Atypical GABAB receptor responses observed in GB2-deficient mice suggested that GB1 may have activity in the absence of GB2. However the underlying mechanisms remain poorly characterized. Methodology/Principal Findings Here, by using cells overexpressing a GB1 mutant (GB1asa) with the ability to translocate to the cell surface in the absence of GB2, we show that GABAB receptor agonists, such as GABA and Baclofen, can induce ERK1/2 phosphorylation in the absence of GB2. Furthermore, we demonstrate that GB1asa induces ERK1/2 phosphorylation through Gi/o proteins and PLC dependent IGF-1R transactivation. Conclusions/Significance Our data suggest that GB1 may form a functional receptor at the cell surface in the absence of GB2.
Collapse
Affiliation(s)
- Guillaume A. Baloucoune
- Sino-France Laboratory for Drug Screening, Key Laboratory of Molecular Biophysics of Ministry of Education, College of Life Science and Technology, Huazhong University of Science and Technology, Wuhan, Hubei, China
| | - Lei Chun
- Sino-France Laboratory for Drug Screening, Key Laboratory of Molecular Biophysics of Ministry of Education, College of Life Science and Technology, Huazhong University of Science and Technology, Wuhan, Hubei, China
| | - Wenhua Zhang
- Sino-France Laboratory for Drug Screening, Key Laboratory of Molecular Biophysics of Ministry of Education, College of Life Science and Technology, Huazhong University of Science and Technology, Wuhan, Hubei, China
| | - Chanjuan Xu
- Sino-France Laboratory for Drug Screening, Key Laboratory of Molecular Biophysics of Ministry of Education, College of Life Science and Technology, Huazhong University of Science and Technology, Wuhan, Hubei, China
| | - Siluo Huang
- Sino-France Laboratory for Drug Screening, Key Laboratory of Molecular Biophysics of Ministry of Education, College of Life Science and Technology, Huazhong University of Science and Technology, Wuhan, Hubei, China
| | - Qian Sun
- Sino-France Laboratory for Drug Screening, Key Laboratory of Molecular Biophysics of Ministry of Education, College of Life Science and Technology, Huazhong University of Science and Technology, Wuhan, Hubei, China
| | - Yunyun Wang
- Sino-France Laboratory for Drug Screening, Key Laboratory of Molecular Biophysics of Ministry of Education, College of Life Science and Technology, Huazhong University of Science and Technology, Wuhan, Hubei, China
| | - Haijun Tu
- Sino-France Laboratory for Drug Screening, Key Laboratory of Molecular Biophysics of Ministry of Education, College of Life Science and Technology, Huazhong University of Science and Technology, Wuhan, Hubei, China
| | - Jianfeng Liu
- Sino-France Laboratory for Drug Screening, Key Laboratory of Molecular Biophysics of Ministry of Education, College of Life Science and Technology, Huazhong University of Science and Technology, Wuhan, Hubei, China
- * E-mail:
| |
Collapse
|
34
|
McQuail JA, Bañuelos C, LaSarge CL, Nicolle MM, Bizon JL. GABA(B) receptor GTP-binding is decreased in the prefrontal cortex but not the hippocampus of aged rats. Neurobiol Aging 2012; 33:1124.e1-12. [PMID: 22169202 PMCID: PMC3310948 DOI: 10.1016/j.neurobiolaging.2011.11.011] [Citation(s) in RCA: 31] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/14/2011] [Revised: 10/28/2011] [Accepted: 11/04/2011] [Indexed: 12/26/2022]
Abstract
Gamma aminobutyric acid (GABA)(B) receptors (GABA(B)Rs) have been linked to a wide range of physiological and cognitive processes and are of interest for treating a number of neurodegenerative and psychiatric disorders. As many of these diseases are associated with advanced age, it is important to understand how the normal aging process impacts GABA(B)R expression and signaling. Thus, we investigated GABA(B)R expression and function in the prefrontal cortex (PFC) and hippocampus of young and aged rats characterized in a spatial learning task. Baclofen-stimulated GTP-binding and GABA(B)R1 and GABA(B)R2 proteins were reduced in the prefrontal cortex of aged rats but these reductions were not associated with spatial learning abilities. In contrast, hippocampal GTP-binding was comparable between young and aged rats but reduced hippocampal GABA(B)R1 expression was observed in aged rats with spatial learning impairment. These data demonstrate marked regional differences in GABA(B)R complexes in the adult and aged brain and could have implications for both understanding the role of GABAergic processes in normal brain function and the development of putative interventions that target this system.
Collapse
Affiliation(s)
- Joseph A. McQuail
- Program in Neuroscience, Graduate School of Arts & Sciences, Wake Forest University, Winston-Salem, NC 27157
| | - Cristina Bañuelos
- Department of Neuroscience, College of Medicine, University of Florida, Gainesville, FL 32611
| | - Candi L. LaSarge
- Department of Anesthesia, Cincinnati Children’s Hospital, 3333 Burnet Avenue, Cincinnati, OH 45229
| | - Michelle M. Nicolle
- Program in Neuroscience, Graduate School of Arts & Sciences, Wake Forest University, Winston-Salem, NC 27157
- Department of Internal Medicine, Section of Gerontology, Wake Forest University, Winston-Salem, NC 27157
- Department of Physiology & Pharmacology, School of Medicine, Wake Forest University, Winston-Salem, NC 27157
| | - Jennifer L. Bizon
- Department of Neuroscience, College of Medicine, University of Florida, Gainesville, FL 32611
- McKnight Brain Institute, University of Florida, Gainesville, FL 32611
| |
Collapse
|
35
|
Dupré DJ, Hammad MM, Holland P, Wertman J. Role of chaperones in G protein coupled receptor signaling complex assembly. Subcell Biochem 2012; 63:23-42. [PMID: 23161131 DOI: 10.1007/978-94-007-4765-4_2] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022]
Abstract
G protein coupled receptors are involved in highly efficient and specific activation of signaling pathways. Yet, we do not fully understand the processes required to assemble the different partners of the GPCR signaling complex. In order to address this issue, we need to understand how receptors and their signaling -partners are synthesized, folded and regulated during quality control steps in order to generate functional proteins. Several molecular chaperones are involved in this process for most proteins, including GPCRs. Several membrane proteins require the assembly of different subunits to be functional. In recent years, GPCRs have been shown to form oligomers, which could be interpreted as subunits of a larger complex. Yet, those oligomers would not be functional without the association of other signaling partners; thus, there is a requirement for the specific assembly of the -different partners. In this chapter, we will cover some aspects of the current knowledge about how chaperones are involved in both the formation of GPCR oligomers and in the assembly of the receptors with their signaling complex components.
Collapse
Affiliation(s)
- Denis J Dupré
- Department of Pharmacology, Faculty of Medicine, Sir Charles Tupper Medical Building, Dalhousie University, 5850 College St, Halifax, NS, B3H 4R2, Canada,
| | | | | | | |
Collapse
|
36
|
Maurice P, Benleulmi-Chaachoua A, Jockers R. Differential assembly of GPCR signaling complexes determines signaling specificity. Subcell Biochem 2012; 63:225-40. [PMID: 23161141 DOI: 10.1007/978-94-007-4765-4_12] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/24/2022]
Abstract
Recent proteomic and biochemical evidence indicates that cellular -signaling is organized in protein modules. G protein-coupled receptors (GPCRs) are privileged entry points for extracellular signals that are transmitted through the plasma membrane into the cell. The adequate cellular response and signaling specificity is regulated by GPCR-associated protein modules. The composition of these modules is dynamic and might depend on receptor stimulation, the proteome of a given cellular context, the subcellular localization of receptor-associated modules, the formation of GPCR oligomers and the variation of expression levels of components of these modules under physiological, for example circadian rhythm, or pathological conditions. The current article will highlight the importance of GPCR-associated protein modules as a biochemical basis for signaling specificity.
Collapse
Affiliation(s)
- Pascal Maurice
- Inserm, U1016, Institut Cochin, 22 rue Méchain, 75014, Paris, France
| | | | | |
Collapse
|
37
|
Abstract
In almost 16 years since the word "dimer" was used in a publication to describe the organization of G protein-coupled receptors (GPCRs), a large number of studies have since weighed in on this notion. Are native, functional GPCRs monomers, dimers or as some would suggest even higher order structures? Here, we review some of the latest evidence regarding the organization of these receptors in both homo- and hetero-oligomeric formats, with a particular focus on β-adrenergic receptors. This is particularly important for understanding the allosteric nature of receptor/receptor interactions. It is likely that, over the course of evolution, mechanisms have come into play using all of the possible variations in receptor/receptor stoichiometry, depending on the cell and the physiological context in question. Finally, we provide some data that suggests that higher order structures of GPCRs, as with dimers themselves are probably assembled in the ER.
Collapse
|
38
|
Fernández-Alacid L, Watanabe M, Molnár E, Wickman K, Luján R. Developmental regulation of G protein-gated inwardly-rectifying K+ (GIRK/Kir3) channel subunits in the brain. Eur J Neurosci 2011; 34:1724-36. [PMID: 22098295 DOI: 10.1111/j.1460-9568.2011.07886.x] [Citation(s) in RCA: 52] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/15/2023]
Abstract
G protein-gated inwardly-rectifying K(+) (GIRK/family 3 of inwardly-rectifying K(+) ) channels are coupled to neurotransmitter action and can play important roles in modulating neuronal excitability. We investigated the temporal and spatial expression of GIRK1, GIRK2 and GIRK3 subunits in the developing and adult brain of mice and rats using biochemical, immunohistochemical and immunoelectron microscopic techniques. At all ages analysed, the overall distribution patterns of GIRK1-3 were very similar, with high expression levels in the neocortex, cerebellum, hippocampus and thalamus. Focusing on the hippocampus, histoblotting and immunohistochemistry showed that GIRK1-3 protein levels increased with age, and this was accompanied by a shift in the subcellular localization of the subunits. Early in development (postnatal day 5), GIRK subunits were predominantly localized to the endoplasmic reticulum in the pyramidal cells, but by postnatal day 60 they were mostly found along the plasma membrane. During development, GIRK1 and GIRK2 were found primarily at postsynaptic sites, whereas GIRK3 was predominantly detected at presynaptic sites. In addition, GIRK1 and GIRK2 expression on the spine plasma membrane showed identical proximal-to-distal gradients that differed from GIRK3 distribution. Furthermore, although GIRK1 was never found within the postsynaptic density (PSD), the level of GIRK2 in the PSD progressively increased and GIRK3 did not change in the PSD during development. Together, these findings shed new light on the developmental regulation and subcellular diversity of neuronal GIRK channels, and support the contention that distinct subpopulations of GIRK channels exert separable influences on neuronal excitability. The ability to selectively target specific subpopulations of GIRK channels may prove effective in the treatment of disorders of excitability.
Collapse
Affiliation(s)
- Laura Fernández-Alacid
- Departamento de Ciencias Médicas, Facultad de Medicina, Instituto de Investigación en Discapacidades Neurológicas (IDINE), Universidad de Castilla-La Mancha, Albacete, Spain
| | | | | | | | | |
Collapse
|
39
|
Vaniotis G, Allen BG, Hébert TE. Nuclear GPCRs in cardiomyocytes: an insider's view of β-adrenergic receptor signaling. Am J Physiol Heart Circ Physiol 2011; 301:H1754-64. [PMID: 21890692 DOI: 10.1152/ajpheart.00657.2011] [Citation(s) in RCA: 47] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/11/2023]
Abstract
In recent years, we have come to appreciate the complexity of G protein-coupled receptor signaling in general and β-adrenergic receptor (β-AR) signaling in particular. Starting originally from three β-AR subtypes expressed in cardiomyocytes with relatively simple, linear signaling cascades, it is now clear that there are large receptor-based networks which provide a rich and diverse set of responses depending on their complement of signaling partners and the physiological state. More recently, it has become clear that subcellular localization of these signaling complexes also enriches the diversity of phenotypic outcomes. Here, we review our understanding of the signaling repertoire controlled by nuclear β-AR subtypes as well our understanding of the novel roles for G proteins themselves in the nucleus, with a special focus, where possible, on their effects in cardiomyocytes. Finally, we discuss the potential pathological implications of alterations in nuclear β-AR signaling.
Collapse
|
40
|
Ciruela F, Fernández-Dueñas V, Sahlholm K, Fernández-Alacid L, Nicolau JC, Watanabe M, Luján R. Evidence for oligomerization between GABAB receptors and GIRK channels containing the GIRK1 and GIRK3 subunits. Eur J Neurosci 2010; 32:1265-77. [PMID: 20846323 DOI: 10.1111/j.1460-9568.2010.07356.x] [Citation(s) in RCA: 37] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/19/2022]
Abstract
The stimulation of inhibitory neurotransmitter receptors, such as γ-aminobutyric acid type B (GABA(B) ) receptors, activates G protein-gated inwardly-rectifying K(+) (GIRK) channels, which influence membrane excitability. There is now evidence suggesting that G protein-coupled receptors and G protein-gated inwardly-rectifying K(+) [GIRK/family 3 of inwardly-rectifying K(+) (Kir3)] channels do not diffuse freely within the plasma membrane, but instead there are direct protein-protein interactions between them. Here, we used bioluminescence resonance energy transfer, co-immunoprecipitation, confocal and electron microscopy techniques to investigate the oligomerization of GABA(B) receptors with GIRK channels containing the GIRK3 subunit, whose contribution to functional channels is still unresolved. Co-expression of GABA(B) receptors and GIRK channels in human embryonic kidney-293 cells in combination with co-immunoprecipitation experiments established that the metabotropic receptor forms stable complexes with GIRK channels. Using bioluminescence resonance energy transfer, we have shown that, in living cells under physiological conditions, GABA(B) receptors interact directly with GIRK1/GIRK3 heterotetramers. In addition, we have provided evidence that the receptor-effector complexes are also found in vivo and identified that the cerebellar granule cells are one neuron population where the interaction probably takes place. Altogether, our data show that signalling complexes containing GABA(B) receptors and GIRK channels are formed shortly after biosynthesis, probably in the endoplasmic reticulum and/or endoplasmic reticulum/Golgi apparatus complex, suggesting that this might be a general feature of receptor-effector ion channel signal transduction and supporting a channel-forming role for the GIRK3 subunit.
Collapse
Affiliation(s)
- Francisco Ciruela
- Unitat de Farmacologia (4102), Departament Patologia i Terapèutica Experimental, Facultat de Medicina-Bellvitge, Universitat de IDIBELL-Universitat de Barcelona, L'Hospitalet de Llobregat, Barcelona, Spain.
| | | | | | | | | | | | | |
Collapse
|
41
|
Zylbergold P, Ramakrishnan N, Hebert T. The role of G proteins in assembly and function of Kir3 inwardly rectifying potassium channels. Channels (Austin) 2010; 4:411-21. [PMID: 20855978 DOI: 10.4161/chan.4.5.13327] [Citation(s) in RCA: 21] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/21/2023] Open
Abstract
Kir3 channels (also known as GIRK channels) are important regulators of electrical excitability in both cardiomyocytes and neurons. Much is known regarding the assembly and function of these channels and the roles that their interacting proteins play in controlling these events. Further, they are one of the best studied effectors of heterotrimeric G proteins in general and Gβγ subunits in particular. However, our understanding of the roles of multiple Gβγ binding sites on Kir3 channels is still rudimentary. We discuss potential roles for Gβγ in channel assembly and trafficking in addition to their known role in cellular signaling.
Collapse
Affiliation(s)
- Peter Zylbergold
- Department of Pharmacology and Therapeutics, McGill University, Québec, Canada
| | | | | |
Collapse
|
42
|
Emerging roles for G protein-gated inwardly rectifying potassium (GIRK) channels in health and disease. Nat Rev Neurosci 2010; 11:301-15. [PMID: 20389305 DOI: 10.1038/nrn2834] [Citation(s) in RCA: 470] [Impact Index Per Article: 31.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
G protein-gated inwardly rectifying potassium (GIRK) channels hyperpolarize neurons in response to activation of many different G protein-coupled receptors and thus control the excitability of neurons through GIRK-mediated self-inhibition, slow synaptic potentials and volume transmission. GIRK channel function and trafficking are highly dependent on the channel subunit composition. Pharmacological investigations of GIRK channels and studies in animal models suggest that GIRK activity has an important role in physiological responses, including pain perception and memory modulation. Moreover, abnormal GIRK function has been implicated in altering neuronal excitability and cell death, which may be important in the pathophysiology of diseases such as epilepsy, Down's syndrome, Parkinson's disease and drug addiction. GIRK channels may therefore prove to be a valuable new therapeutic target.
Collapse
|
43
|
Audet M, Lagacé M, Silversides DW, Bouvier M. Protein-protein interactions monitored in cells from transgenic mice using bioluminescence resonance energy transfer. FASEB J 2010; 24:2829-38. [PMID: 20335229 DOI: 10.1096/fj.09-144816] [Citation(s) in RCA: 25] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/09/2023]
Abstract
Monitoring the dynamics of protein-protein interactions in their natural environment remains a challenge. Resonance energy transfer approaches represent a promising avenue to directly probe these interactions in real time. The present study aims at establishing a proof of principle that bioluminescence resonance energy transfer (BRET) can be used to study the regulation of protein-protein interaction in cells from transgenic animals. A transgenic mouse line coexpressing the beta(2)-adrenergic receptor fused to Renilla luciferase (beta(2)AR-Rluc) and beta arrestin-2 fused to a green fluorescent protein (GFP2-beta arr2) was generated. The fusion proteins were found to be functional in the transgenic animals and the beta(2)AR-Rluc maintained pharmacological properties, comparable to that of the native receptor. Sufficiently high luminescence signal was generated to allow detection of BRET in testis cells where the beta(2)AR-Rluc transgene was expressed at levels significantly higher than that of the endogenous receptor in this tissue but remain within physiological range when compared with other beta(2)AR-expressing tissues. Stimulation with a beta-adrenergic agonist led to a significant dose- and time-dependent increase in BRET, which reflected ligand-promoted recruitment of beta arr2 to the receptor. Our study demonstrates that BRET can be used to monitor the dynamic regulation of protein-protein interactions in cells derived from transgenic mice.
Collapse
Affiliation(s)
- Martin Audet
- Department of Biochemistry, Institute for Research in Immunology and Cancer, and Groupe de Recherche Universitaire sur le Médicament, Université de Montréal, Montréal, Québec, Canada
| | | | | | | |
Collapse
|
44
|
Direct interaction of GABAB receptors with M2 muscarinic receptors enhances muscarinic signaling. J Neurosci 2010; 29:15796-809. [PMID: 20016095 DOI: 10.1523/jneurosci.4103-09.2009] [Citation(s) in RCA: 31] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2022] Open
Abstract
Downregulation of G-protein-coupled receptors (GPCRs) provides an important mechanism for reducing neurotransmitter signaling during sustained stimulation. Chronic stimulation of M(2) muscarinic receptors (M(2)Rs) causes internalization of M(2)R and G-protein-activated inwardly rectifying potassium (GIRK) channels in neuronal PC12 cells, resulting in loss of function. Here, we show that coexpression of GABA(B) R2 receptors (GBR2s) rescues both surface expression and function of M(2)R, including M(2)R-induced activation of GIRKs and inhibition of cAMP production. GBR2 showed significant association with M(2)R at the plasma membrane but not other GPCRs (M(1)R, mu-opioid receptor), as detected by fluorescence resonance energy transfer measured with total internal reflection fluorescence microscopy. Unique regions of the proximal C-terminal domains of GBR2 and M(2)R mediate specific binding between M(2)R and GBR2. In the brain, GBR2, but not GBR1, biochemically coprecipitates with M(2)R and overlaps with M(2)R expression in cortical neurons. This novel heteromeric association between M(2)R and GBR2 provides a possible mechanism for altering muscarinic signaling in the brain and represents a previously unrecognized role for GBR2.
Collapse
|
45
|
|
46
|
Dai S, Hall DD, Hell JW. Supramolecular assemblies and localized regulation of voltage-gated ion channels. Physiol Rev 2009; 89:411-52. [PMID: 19342611 DOI: 10.1152/physrev.00029.2007] [Citation(s) in RCA: 266] [Impact Index Per Article: 16.6] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022] Open
Abstract
This review addresses the localized regulation of voltage-gated ion channels by phosphorylation. Comprehensive data on channel regulation by associated protein kinases, phosphatases, and related regulatory proteins are mainly available for voltage-gated Ca2+ channels, which form the main focus of this review. Other voltage-gated ion channels and especially Kv7.1-3 (KCNQ1-3), the large- and small-conductance Ca2+-activated K+ channels BK and SK2, and the inward-rectifying K+ channels Kir3 have also been studied to quite some extent and will be included. Regulation of the L-type Ca2+ channel Cav1.2 by PKA has been studied most thoroughly as it underlies the cardiac fight-or-flight response. A prototypical Cav1.2 signaling complex containing the beta2 adrenergic receptor, the heterotrimeric G protein Gs, adenylyl cyclase, and PKA has been identified that supports highly localized via cAMP. The type 2 ryanodine receptor as well as AMPA- and NMDA-type glutamate receptors are in close proximity to Cav1.2 in cardiomyocytes and neurons, respectively, yet independently anchor PKA, CaMKII, and the serine/threonine phosphatases PP1, PP2A, and PP2B, as is discussed in detail. Descriptions of the structural and functional aspects of the interactions of PKA, PKC, CaMKII, Src, and various phosphatases with Cav1.2 will include comparisons with analogous interactions with other channels such as the ryanodine receptor or ionotropic glutamate receptors. Regulation of Na+ and K+ channel phosphorylation complexes will be discussed in separate papers. This review is thus intended for readers interested in ion channel regulation or in localization of kinases, phosphatases, and their upstream regulators.
Collapse
Affiliation(s)
- Shuiping Dai
- Department of Pharmacology, University of Iowa, Iowa City, IA 52242-1109, USA
| | | | | |
Collapse
|
47
|
Intracellular trafficking and assembly of specific Kir3 channel/G protein complexes. Cell Signal 2009; 21:488-501. [DOI: 10.1016/j.cellsig.2008.11.011] [Citation(s) in RCA: 27] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/03/2008] [Revised: 11/13/2008] [Accepted: 11/15/2008] [Indexed: 12/27/2022]
|
48
|
Richer M, David M, Villeneuve LR, Trieu P, Ethier N, Pétrin D, Mamarbachi AM, Hébert TE. GABA-B(1) receptors are coupled to the ERK1/2 MAP kinase pathway in the absence of GABA-B(2) subunits. J Mol Neurosci 2008; 38:67-79. [PMID: 19052921 DOI: 10.1007/s12031-008-9163-6] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/16/2008] [Accepted: 11/06/2008] [Indexed: 01/22/2023]
Abstract
In the current model of gamma-aminobutyric acid (GABA) B receptor function, there is a requirement for GABA-B(1/2) heterodimerisation for targetting to the cell surface. However, different lines of evidence suggest that the GABA-B(1) subunit can form a functional receptor in the absence of GABA-B(2). We observed coupling of endogenous GABA-B(1) receptors in the DI-TNC1 glial cell line to the ERK pathway in response to baclofen even though these cells do not express GABA-B(2). GABA-B(1A) receptors were also able to mediate a rapid, transient, and dose-dependent activation of the ERK1/2 MAP kinase pathway when transfected alone into HEK 293 cells. The response was abolished by G(i/o) and MEK inhibition, potentiated by inhibitors of phospholipase C and protein kinase C and did not involve PI-3-kinase activity. Finally, using bioluminescence resonance energy transfer and co-immunoprecipitation, we show the existence of homodimeric GABA-B(1A) receptors in transfected HEK293 cells. Altogether, our observations show that GABA-B(1A) receptors are able to activate the ERK1/2 pathway despite the absence of surface targetting partner GABA-B(2) in both HEK 293 cells and the DI-TNC1 cell line.
Collapse
Affiliation(s)
- Maxime Richer
- Département de biochimie and Groupe de recherche universitaire sur le médicament (GRUM), Université de Montréal, Montréal, Québec, Canada
| | | | | | | | | | | | | | | |
Collapse
|
49
|
Membrane signalling complexes: implications for development of functionally selective ligands modulating heptahelical receptor signalling. Cell Signal 2008; 21:179-85. [PMID: 18790047 DOI: 10.1016/j.cellsig.2008.08.013] [Citation(s) in RCA: 23] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/11/2008] [Accepted: 08/24/2008] [Indexed: 11/24/2022]
Abstract
Technological development has considerably changed the way in which we evaluate drug efficacy and has led to a conceptual revolution in pharmacological theory. In particular, molecular resolution assays have revealed that heptahelical receptors may adopt multiple active conformations with unique signalling properties. It is therefore becoming widely accepted that ligand ability to stabilize receptor conformations with distinct signalling profiles may allow to direct the stimulus generated by an activated receptor towards a specific signalling pathway. This capacity to induce only a subset of the ensemble of responses regulated by a given receptor has been termed "functional selectivity" (or "stimulus trafficking"), and provides the bases for a highly specific regulation of receptor signalling. Concomitant with these observations, heptahelical receptors have been shown to associate with G proteins and effectors to form multimeric arrays. These complexes are constitutively formed during protein synthesis and are targeted to the cell surface as integral signalling units. Herein we summarize evidence supporting the existence of such constitutive signalling arrays and analyze the possibility that they may constitute viable targets for developing ligands with "functional selectivity".
Collapse
|
50
|
Doupnik CA. GPCR-Kir channel signaling complexes: defining rules of engagement. J Recept Signal Transduct Res 2008; 28:83-91. [PMID: 18437632 DOI: 10.1080/10799890801941970] [Citation(s) in RCA: 25] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/22/2022]
Abstract
Ion channels and G protein-coupled receptors (GPCRs) are integral transmembrane proteins vital to a multitude of cell signaling and physiological functions. Members of these large protein families are known to interact directly with various intracellular protein partners in a dynamic and isoform-dependent manner, ultimately shaping their life cycle and signal output. The family of G protein-gated inwardly rectifying potassium channels (Kir3 or GIRK) expressed in brain, heart, and endocrine tissues were recently shown to stably associate with several different GPCRs, forming the basis of a macromolecular ion channel-GPCR signaling complex. The molecular determinants that mediate and maintain GPCR-Kir3 channel complexes are currently not well understood. Recent findings and emerging hypotheses on the assembly and stability of multiprotein GPCR-Kir channel signaling complexes are discussed, highlighting distinct mechanisms used by different Kir channel families. These protein-protein interaction processes are crucial in determining both the synaptic response times and the extent of GPCR "cross-talk" in Kir3-mediated inhibitory synaptic transmission.
Collapse
Affiliation(s)
- Craig A Doupnik
- Department of Molecular Pharmacology and Physiology, University of South Florida College of Medicine, Tampa, Florida 33612, USA.
| |
Collapse
|