1
|
Chen X, Wang YJ, Mu TW. Proteostasis regulation of GABA A receptors in neuronal function and disease. Biomed Pharmacother 2025; 186:117992. [PMID: 40112516 PMCID: PMC12068001 DOI: 10.1016/j.biopha.2025.117992] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/10/2025] [Revised: 03/12/2025] [Accepted: 03/13/2025] [Indexed: 03/22/2025] Open
Abstract
The γ-aminobutyric acid type A receptors (GABAARs) are ligand-gated anion channels that mediate fast inhibitory neurotransmission in the mammalian central nervous system. GABAARs form heteropentameric assemblies comprising two α1, two β2, and one γ2 subunits as the most common subtype in mammalian brains. Proteostasis regulation of GABAARs involves subunit folding within the endoplasmic reticulum, assembling into heteropentamers, receptor trafficking to the cell surface, and degradation of terminally misfolded subunits. As GABAARs are surface proteins, their trafficking to the plasma membrane is critical for proper receptor function. Thus, variants in the genes encoding GABAARs that disrupt proteostasis result in various neurodevelopmental disorders, ranging from intellectual disability to idiopathic generalized epilepsy. This review summarizes recent progress about how the proteostasis network regulates protein folding, assembly, degradation, trafficking, and synaptic clustering of GABAARs. Additionally, emerging pharmacological approaches that restore proteostasis of pathogenic GABAAR variants are presented, providing a promising strategy to treat related neurological diseases.
Collapse
Affiliation(s)
- Xi Chen
- Department of Physiology and Biophysics, Case Western Reserve University School of Medicine, Cleveland, OH 44106, USA.
| | - Ya-Juan Wang
- Department of Physiology and Biophysics, Case Western Reserve University School of Medicine, Cleveland, OH 44106, USA.
| | - Ting-Wei Mu
- Department of Physiology and Biophysics, Case Western Reserve University School of Medicine, Cleveland, OH 44106, USA.
| |
Collapse
|
2
|
Katsuno Y, Jitsuki S, Ota W, Yamanoue T, Abe H, Takahashi T. Edonerpic maleate prevents epileptic seizure during recovery from brain damage by balancing excitatory and inhibitory inputs. Front Neural Circuits 2024; 18:1492043. [PMID: 39712974 PMCID: PMC11660091 DOI: 10.3389/fncir.2024.1492043] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/06/2024] [Accepted: 10/28/2024] [Indexed: 12/24/2024] Open
Abstract
Functional recovery from brain damage, such as stroke, is a plastic process in the brain. The excitatory glutamate α-amino-3-hydroxy-5-methyl-4-isoxazole propionic acid receptor (AMPAR) plays a crucial role in neuronal functions, and the synaptic trafficking of AMPAR is a fundamental mechanism underlying synaptic plasticity. We recently identified a collapsin response mediator protein 2 (CRMP2)-binding compound, edonerpic maleate, which augments rehabilitative training-dependent functional recovery from brain damage by facilitating experience-driven synaptic delivery of AMPARs. In animals recovering from cryogenic brain injury, a potential compensatory area adjacent to the injured region was observed, where the injection of CNQX, an AMPAR antagonist, significantly attenuated functional recovery. In the compensatory brain area of animals recovering from cryogenic injury, the administration of edonerpic maleate enhanced both excitatory and inhibitory synaptic inputs at pyramidal neurons. In contrast, recovered animals that did not receive the drug exhibited augmentation of only excitatory synaptic input. The threshold of picrotoxin-induced epileptic seizure in recovered animals without edonerpic maleate treatment was lower than in intact animals and recovered animals with edonerpic maleate. Thus, edonerpic maleate enhances motor function recovery from brain damage by balancing excitatory and inhibitory synaptic inputs, which helps prevent epileptic seizures during recovery.
Collapse
Affiliation(s)
- Yuki Katsuno
- Department of Physiology, Yokohama City University Graduate School of Medicine, Yokohama, Japan
| | - Susumu Jitsuki
- Department of Biochemistry, Mie University Graduate School of Medicine, Tsu, Japan
| | - Wataru Ota
- Department of Physiology, Yokohama City University Graduate School of Medicine, Yokohama, Japan
| | - Tomomi Yamanoue
- Department of Physiology, Yokohama City University Graduate School of Medicine, Yokohama, Japan
| | - Hiroki Abe
- Department of Physiology, Yokohama City University Graduate School of Medicine, Yokohama, Japan
| | - Takuya Takahashi
- Department of Physiology, Yokohama City University Graduate School of Medicine, Yokohama, Japan
| |
Collapse
|
3
|
Dilly GA, Blednov YA, Warden AS, Ezerskiy L, Fleischer C, Plotkin JD, Patil S, Osterndorff-Kahanek EA, Mayfield J, Mayfield RD, Homanics GE, Messing RO. Knockdown of Tlr3 in dorsal striatum reduces ethanol consumption and acute functional tolerance in male mice. Brain Behav Immun 2024; 118:437-448. [PMID: 38499210 PMCID: PMC11007683 DOI: 10.1016/j.bbi.2024.03.021] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/16/2023] [Revised: 03/05/2024] [Accepted: 03/15/2024] [Indexed: 03/20/2024] Open
Abstract
Systemic activation of toll-like receptor 3 (TLR3) signaling using poly(I:C), a TLR3 agonist, drives ethanol consumption in several rodent models, while global knockout of Tlr3 reduces drinking in C57BL/6J male mice. To determine if brain TLR3 pathways are involved in drinking behavior, we used CRISPR/Cas9 genome editing to generate a Tlr3 floxed (Tlr3F/F) mouse line. After sequence confirmation and functional validation of Tlr3 brain transcripts, we injected Tlr3F/F male mice with an adeno-associated virus expressing Cre recombinase (AAV5-CMV-Cre-GFP) to knockdown Tlr3 in the medial prefrontal cortex, nucleus accumbens, or dorsal striatum (DS). Only Tlr3 knockdown in the DS decreased two-bottle choice, every-other-day (2BC-EOD) ethanol consumption. DS-specific deletion of Tlr3 also increased intoxication and prevented acute functional tolerance to ethanol. In contrast, poly(I:C)-induced activation of TLR3 signaling decreased intoxication in male C57BL/6J mice, consistent with its ability to increase 2BC-EOD ethanol consumption in these mice. We also found that TLR3 was highly colocalized with DS neurons. AAV5-Cre transfection occurred predominantly in neurons, but there was minimal transfection in astrocytes and microglia. Collectively, our previous and current studies show that activating or inhibiting TLR3 signaling produces opposite effects on acute responses to ethanol and on ethanol consumption. While previous studies, however, used global knockout or systemic TLR3 activation (which alter peripheral and brain innate immune responses), the current results provide new evidence that brain TLR3 signaling regulates ethanol drinking. We propose that activation of TLR3 signaling in DS neurons increases ethanol consumption and that a striatal TLR3 pathway is a potential target to reduce excessive drinking.
Collapse
Affiliation(s)
- Geoffrey A Dilly
- Waggoner Center for Alcohol and Addiction Research, The University of Texas at Austin, Austin, TX 78712, United States; Institute for Neuroscience, The University of Texas at Austin, Austin, TX 78712, United States; Department of Neuroscience, The University of Texas at Austin, Austin, TX 78712, United States; Department of Neurology, Dell Medical School, The University of Texas at Austin, Austin, TX 78712, United States
| | - Yuri A Blednov
- Waggoner Center for Alcohol and Addiction Research, The University of Texas at Austin, Austin, TX 78712, United States
| | - Anna S Warden
- Waggoner Center for Alcohol and Addiction Research, The University of Texas at Austin, Austin, TX 78712, United States
| | - Lubov Ezerskiy
- Waggoner Center for Alcohol and Addiction Research, The University of Texas at Austin, Austin, TX 78712, United States; Department of Neuroscience, The University of Texas at Austin, Austin, TX 78712, United States
| | - Caleb Fleischer
- Waggoner Center for Alcohol and Addiction Research, The University of Texas at Austin, Austin, TX 78712, United States; Department of Neuroscience, The University of Texas at Austin, Austin, TX 78712, United States
| | - Jesse D Plotkin
- Waggoner Center for Alcohol and Addiction Research, The University of Texas at Austin, Austin, TX 78712, United States; Department of Neuroscience, The University of Texas at Austin, Austin, TX 78712, United States
| | - Shruti Patil
- Waggoner Center for Alcohol and Addiction Research, The University of Texas at Austin, Austin, TX 78712, United States
| | | | - Jody Mayfield
- Waggoner Center for Alcohol and Addiction Research, The University of Texas at Austin, Austin, TX 78712, United States
| | - R Dayne Mayfield
- Waggoner Center for Alcohol and Addiction Research, The University of Texas at Austin, Austin, TX 78712, United States; Department of Neuroscience, The University of Texas at Austin, Austin, TX 78712, United States
| | - Gregg E Homanics
- Departments of Anesthesiology & Perioperative Medicine, Neurobiology, and Pharmacology & Chemical Biology, University of Pittsburgh, Pittsburgh, PA 15261, United States
| | - Robert O Messing
- Waggoner Center for Alcohol and Addiction Research, The University of Texas at Austin, Austin, TX 78712, United States; Institute for Neuroscience, The University of Texas at Austin, Austin, TX 78712, United States; Department of Neuroscience, The University of Texas at Austin, Austin, TX 78712, United States; Department of Neurology, Dell Medical School, The University of Texas at Austin, Austin, TX 78712, United States.
| |
Collapse
|
4
|
Poliquin S, Nwosu G, Randhave K, Shen W, Flamm C, Kang JQ. Modulating Endoplasmic Reticulum Chaperones and Mutant Protein Degradation in GABRG2(Q390X) Associated with Genetic Epilepsy with Febrile Seizures Plus and Dravet Syndrome. Int J Mol Sci 2024; 25:4601. [PMID: 38731820 PMCID: PMC11083348 DOI: 10.3390/ijms25094601] [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: 03/04/2024] [Revised: 04/10/2024] [Accepted: 04/17/2024] [Indexed: 05/13/2024] Open
Abstract
A significant number of patients with genetic epilepsy do not obtain seizure freedom, despite developments in new antiseizure drugs, suggesting a need for novel therapeutic approaches. Many genetic epilepsies are associated with misfolded mutant proteins, including GABRG2(Q390X)-associated Dravet syndrome, which we have previously shown to result in intracellular accumulation of mutant GABAA receptor γ2(Q390X) subunit protein. Thus, a potentially promising therapeutic approach is modulation of proteostasis, such as increasing endoplasmic reticulum (ER)-associated degradation (ERAD). To that end, we have here identified an ERAD-associated E3 ubiquitin ligase, HRD1, among other ubiquitin ligases, as a strong modulator of wildtype and mutant γ2 subunit expression. Overexpressing HRD1 or knockdown of HRD1 dose-dependently reduced the γ2(Q390X) subunit. Additionally, we show that zonisamide (ZNS)-an antiseizure drug reported to upregulate HRD1-reduces seizures in the Gabrg2+/Q390X mouse. We propose that a possible mechanism for this effect is a partial rescue of surface trafficking of GABAA receptors, which are otherwise sequestered in the ER due to the dominant-negative effect of the γ2(Q390X) subunit. Furthermore, this partial rescue was not due to changes in ER chaperones BiP and calnexin, as total expression of these chaperones was unchanged in γ2(Q390X) models. Our results here suggest that leveraging the endogenous ERAD pathway may present a potential method to degrade neurotoxic mutant proteins like the γ2(Q390X) subunit. We also demonstrate a pharmacological means of regulating proteostasis, as ZNS alters protein trafficking, providing further support for the use of proteostasis regulators for the treatment of genetic epilepsies.
Collapse
Affiliation(s)
- Sarah Poliquin
- Neuroscience Graduate Program, Vanderbilt University, Nashville, TN 37232, USA;
- Vanderbilt Brain Institute, Vanderbilt University, Nashville, TN 37232, USA;
| | - Gerald Nwosu
- Vanderbilt Brain Institute, Vanderbilt University, Nashville, TN 37232, USA;
- Department of Neuroscience and Pharmacology, Meharry Medical College, Nashville, TN 37208, USA
- Department of Neurology, Vanderbilt University Medical Center, 465 21st Ave South, Nashville, TN 37232, USA; (K.R.); (W.S.); (C.F.)
| | - Karishma Randhave
- Department of Neurology, Vanderbilt University Medical Center, 465 21st Ave South, Nashville, TN 37232, USA; (K.R.); (W.S.); (C.F.)
| | - Wangzhen Shen
- Department of Neurology, Vanderbilt University Medical Center, 465 21st Ave South, Nashville, TN 37232, USA; (K.R.); (W.S.); (C.F.)
| | - Carson Flamm
- Department of Neurology, Vanderbilt University Medical Center, 465 21st Ave South, Nashville, TN 37232, USA; (K.R.); (W.S.); (C.F.)
| | - Jing-Qiong Kang
- Vanderbilt Brain Institute, Vanderbilt University, Nashville, TN 37232, USA;
- Department of Neurology, Vanderbilt University Medical Center, 465 21st Ave South, Nashville, TN 37232, USA; (K.R.); (W.S.); (C.F.)
- Department of Pharmacology, Vanderbilt University, Nashville, TN 37232, USA
- Vanderbilt Kennedy Center of Human Development, Vanderbilt University, Nashville, TN 37232, USA
| |
Collapse
|
5
|
Control of cell surface expression of GABA A receptors by a conserved region at the end of the N-terminal extracellular domain of receptor subunits. J Biol Chem 2022; 298:102590. [PMID: 36244453 PMCID: PMC9672411 DOI: 10.1016/j.jbc.2022.102590] [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: 02/25/2022] [Revised: 09/27/2022] [Accepted: 09/29/2022] [Indexed: 11/07/2022] Open
Abstract
Type A γ-aminobutyric acid receptors (GABAARs) represent a family of pentameric GABA-gated Cl-/HCO3- ion channels which mediate inhibitory transmission in the central nervous system. Cell surface expression of GABAARs, a prerequisite for their function, is dependent on the appropriate assembly of the receptor subunits and their transient interactions with molecular chaperones within the endoplasmic reticulum (ER) and Golgi apparatus. Here, we describe a highly conserved amino acid sequence within the extracellular N-terminal domain of the receptor subunits adjoining the first transmembrane domain as a region important for GABAAR processing within the ER. Modifications of this region in the α1, β3, and γ2 subunits using insertion or site-directed mutagenesis impaired GABAAR trafficking to the cell surface in heterologous cell systems although they had no effect on the subunit assembly. We found that mutated receptors accumulated in the ER where they were shown to associate with chaperones calnexin, BiP, and Grp94. However, their surface expression was increased when ER-associated degradation or proteosome function was inhibited, while modulation of ER calcium stores had little effect. When compared to the wt, mutated receptors showed decreased interaction with calnexin, similar binding to BiP, and increased association with Grp94. Structural modeling of calnexin interaction with the wt or mutated GABAAR revealed that disruption in structure caused by mutations in the conserved region adjoining the first transmembrane domain may impair calnexin binding. Thus, this previously uncharacterized region plays an important role in intracellular processing of GABAARs at least in part by stabilizing their interaction with calnexin.
Collapse
|
6
|
Li S, Huang H, Wei X, Ye L, Ma M, Ling M, Wu Y. The recycling of AMPA receptors/GABAa receptors is related to neuronal excitation/inhibition imbalance and may be regulated by KIF5A. ANNALS OF TRANSLATIONAL MEDICINE 2022; 10:1103. [PMID: 36388788 PMCID: PMC9652568 DOI: 10.21037/atm-22-4337] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/25/2022] [Accepted: 09/30/2022] [Indexed: 09/01/2023]
Abstract
BACKGROUND Excitation/inhibition imbalance (E/I imbalance), which involves an increase of alpha-amino-3-hydroxy-5-methyl-4-isoxazole (AMPA) receptors (AMPARs) and decrease of gamma-aminobutyric acid type A (GABA) type A receptors (GABAaRs) on the neuron surface, has been documented in the pathogenesis of seizures. Notably, it has been established that both the glutamate receptor subunit 2 (GluR2) of AMPARs and beta 2/3 subunits of GABAaRs (Gabrb2+3) participate in the recycling mechanism mediated by the kinesin heavy chain isoform 5A (KIF5A), which determines the number of neuron surface receptors. However, it remains unclear whether receptor recycling is involved in the pathogenesis of seizures. METHODS Twelve adult male Sprague-Dawley rats were randomly allocated to the normal control (Ctl) group (n=6) and the pentylenetetrazol (PTZ)-induced seizure (Sez) group (n=6). The rats in the Ctl group were treated with saline. The rats in the Sez group received an intraperitoneal injection of PTZ at an initial dose of 40 mg/kg. Primary cultured neurons were obtained from newborn rats (24-hour-old). The neurons were exposed to magnesium-free (Mg2+-free) extracellular fluid for 3 hours to establish the seizure model in vitro. We detected the electrophysiology of the seizure model, the expression levels of KIF5A, GluR2, and Gabrb2+3, the recycling ratio of GluR2 and Gabrb2+3, the interaction between KIF5A and GluR2, and the interaction between KIF5A and Gabrb2+3. RESULTS In the Sez group, the expression of GluR2 on the cell surface was increased and the expression of Gabrb2+3 on the cell surface was decreased. The amplitude and frequency of action potentials were significantly increased in the Mg2+-free group. The amplitude and decay time of AMPAR-mediated miniature excitatory postsynaptic currents were increased in the Mg2+-free group. The amplitude and decay time of miniature inhibitory postsynaptic currents were decreased in the Mg2+-free group. The recycling ratio of GluR2 was increased and the recycling ratio of Gabrb2+3 was decreased in the Mg2+-free group. The interaction between KIF5A and GluR2 was increased, and the interaction between KIF5A and Gabrb2+3 was decreased in the seizure model in vivo and in vitro. CONCLUSIONS The recycling of AMPA receptors/GABAa receptors is related to E/I imbalance and may be regulated by KIF5A.
Collapse
Affiliation(s)
- Sijun Li
- Department of Neurology, The First Affiliated Hospital of Guangxi Medical University, Nanning, China
| | - Hongmi Huang
- Department of Neurology, The First Affiliated Hospital of Guangxi Medical University, Nanning, China
| | - Xin Wei
- Department of Neurology, The First Affiliated Hospital of Guangxi Medical University, Nanning, China
| | - Lin Ye
- Department of Neurology, The First Affiliated Hospital of Guangxi Medical University, Nanning, China
| | - Meigang Ma
- Department of Neurology, The First Affiliated Hospital of Guangxi Medical University, Nanning, China
| | - Min Ling
- Department of Biotechnology, Guangxi Medical University, Nanning, China
| | - Yuan Wu
- Department of Neurology, The First Affiliated Hospital of Guangxi Medical University, Nanning, China
| |
Collapse
|
7
|
GABA A Receptor-Stabilizing Protein Ubqln1 Affects Hyperexcitability and Epileptogenesis after Traumatic Brain Injury and in a Model of In Vitro Epilepsy in Mice. Int J Mol Sci 2022; 23:ijms23073902. [PMID: 35409261 PMCID: PMC8999075 DOI: 10.3390/ijms23073902] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/28/2022] [Revised: 03/23/2022] [Accepted: 03/30/2022] [Indexed: 11/16/2022] Open
Abstract
Posttraumatic epilepsy (PTE) is a major public health concern and strongly contributes to human epilepsy cases worldwide. However, an effective treatment and prevention remains a matter of intense research. The present study provides new insights into the gamma aminobutyric acid A (GABAA)-stabilizing protein ubiquilin-1 (ubqln1) and its regulation in mouse models of traumatic brain injury (TBI) and in vitro epilepsy. We performed label-free quantification on isolated cortical GABAergic interneurons from GAD67-GFP mice that received unilateral TBI and discovered reduced expression of ubqln1 24 h post-TBI. To investigate the link between this regulation and the development of epileptiform activity, we further studied ubqln1 expression in hippocampal and cortical slices. Epileptiform events were evoked pharmacologically in acute brain slices by administration of picrotoxin (PTX, 50 μM) and kainic acid (KA, 500 nM) and recorded in the hippocampal CA1 subfield using Multi-electrode Arrays (MEA). Interestingly, quantitative Western blots revealed significant decreases in ubqln1 expression 1–7 h after seizure induction that could be restored by application of the non-selective monoamine oxidase inhibitor nialamide (NM, 10 μM). In picrotoxin-dependent dose–response relationships, NM administration alleviated the frequency and peak amplitude of seizure-like events (SLEs). These findings indicate a role of the monoamine transmitter systems and ubqln1 for cortical network activity during posttraumatic epileptogenesis.
Collapse
|
8
|
Gerson JE, Safren N, Fischer S, Patel R, Crowley EV, Welday JP, Windle AK, Barmada S, Paulson HL, Sharkey LM. Ubiquilin-2 differentially regulates polyglutamine disease proteins. Hum Mol Genet 2021; 29:2596-2610. [PMID: 32681165 DOI: 10.1093/hmg/ddaa152] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/13/2020] [Revised: 07/07/2020] [Accepted: 07/13/2020] [Indexed: 12/12/2022] Open
Abstract
Divergent protein context helps explain why polyglutamine expansion diseases differ clinically and pathologically. This heterogeneity may also extend to how polyglutamine disease proteins are handled by cellular pathways of proteostasis. Studies suggest, for example, that the ubiquitin-proteasome shuttle protein Ubiquilin-2 (UBQLN2) selectively interacts with specific polyglutamine disease proteins. Here we employ cellular models, primary neurons and mouse models to investigate the potential differential regulation by UBQLN2 of two polyglutamine disease proteins, huntingtin (HTT) and ataxin-3 (ATXN3). In cells, overexpressed UBQLN2 selectively lowered levels of full-length pathogenic HTT but not of HTT exon 1 fragment or full-length ATXN3. Consistent with these results, UBQLN2 specifically reduced accumulation of aggregated mutant HTT but not mutant ATXN3 in mouse models of Huntington's disease (HD) and spinocerebellar ataxia type 3 (SCA3), respectively. Normally a cytoplasmic protein, UBQLN2 translocated to the nuclei of neurons in HD mice but not in SCA3 mice. Remarkably, instead of reducing the accumulation of nuclear mutant ATXN3, UBQLN2 induced an accumulation of cytoplasmic ATXN3 aggregates in neurons of SCA3 mice. Together these results reveal a selective action of UBQLN2 toward polyglutamine disease proteins, indicating that polyglutamine expansion alone is insufficient to promote UBQLN2-mediated clearance of this class of disease proteins. Additional factors, including nuclear translocation of UBQLN2, may facilitate its action to clear intranuclear, aggregated disease proteins like HTT.
Collapse
Affiliation(s)
- Julia E Gerson
- Department of Neurology, University of Michigan, Ann Arbor, MI 48109-2200, USA
| | - Nathaniel Safren
- Department of Neurology, University of Michigan, Ann Arbor, MI 48109-2200, USA
| | - Svetlana Fischer
- Department of Neurology, University of Michigan, Ann Arbor, MI 48109-2200, USA
| | - Ronak Patel
- Department of Neurology, University of Michigan, Ann Arbor, MI 48109-2200, USA
| | - Emily V Crowley
- Department of Neurology, University of Michigan, Ann Arbor, MI 48109-2200, USA
| | - Jacqueline P Welday
- Department of Neurology, University of Michigan, Ann Arbor, MI 48109-2200, USA
| | - Alexandra K Windle
- Department of Neurology, University of Michigan, Ann Arbor, MI 48109-2200, USA
| | - Sami Barmada
- Department of Neurology, University of Michigan, Ann Arbor, MI 48109-2200, USA
| | - Henry L Paulson
- Department of Neurology, University of Michigan, Ann Arbor, MI 48109-2200, USA
| | - Lisa M Sharkey
- Department of Neurology, University of Michigan, Ann Arbor, MI 48109-2200, USA
| |
Collapse
|
9
|
Folci A, Mirabella F, Fossati M. Ubiquitin and Ubiquitin-Like Proteins in the Critical Equilibrium between Synapse Physiology and Intellectual Disability. eNeuro 2020; 7:ENEURO.0137-20.2020. [PMID: 32719102 PMCID: PMC7544190 DOI: 10.1523/eneuro.0137-20.2020] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/07/2020] [Revised: 06/08/2020] [Accepted: 06/17/2020] [Indexed: 01/04/2023] Open
Abstract
Posttranslational modifications (PTMs) represent a dynamic regulatory system that precisely modulates the functional organization of synapses. PTMs consist in target modifications by small chemical moieties or conjugation of lipids, sugars or polypeptides. Among them, ubiquitin and a large family of ubiquitin-like proteins (UBLs) share several features such as the structure of the small protein modifiers, the enzymatic cascades mediating the conjugation process, and the targeted aminoacidic residue. In the brain, ubiquitination and two UBLs, namely sumoylation and the recently discovered neddylation orchestrate fundamental processes including synapse formation, maturation and plasticity, and their alteration is thought to contribute to the development of neurological disorders. Remarkably, emerging evidence suggests that these pathways tightly interplay to modulate the function of several proteins that possess pivotal roles for brain homeostasis as well as failure of this crosstalk seems to be implicated in the development of brain pathologies. In this review, we outline the role of ubiquitination, sumoylation, neddylation, and their functional interplay in synapse physiology and discuss their implication in the molecular pathogenesis of intellectual disability (ID), a neurodevelopmental disorder that is frequently comorbid with a wide spectrum of brain pathologies. Finally, we propose a few outlooks that might contribute to better understand the complexity of these regulatory systems in regard to neuronal circuit pathophysiology.
Collapse
Affiliation(s)
- Alessandra Folci
- Humanitas Clinical and Research Center-IRCCS, via Manzoni 56, 20089, Rozzano (MI), Italy
| | - Filippo Mirabella
- Department of Biomedical Sciences, Humanitas University, Via Rita Levi Montalcini 4, 20090 Pieve 9 Emanuele - Milan, Italy
| | - Matteo Fossati
- Humanitas Clinical and Research Center-IRCCS, via Manzoni 56, 20089, Rozzano (MI), Italy
- CNR-Institute of Neuroscience, via Manzoni 56, 20089, Rozzano (MI), Italy
| |
Collapse
|
10
|
Kurlawala Z, Saurabh K, Dunaway R, Shah PP, Siskind LJ, Beverly LJ. Ubiquilin proteins regulate EGFR levels and activity in lung adenocarcinoma cells. J Cell Biochem 2020; 122:43-52. [PMID: 32720736 DOI: 10.1002/jcb.29830] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/18/2020] [Revised: 07/07/2020] [Accepted: 07/13/2020] [Indexed: 12/12/2022]
Abstract
Ubiquilin (UBQLN) proteins are involved in diverse cellular processes like endoplasmic reticulum-associated degradation, autophagy, apoptosis, and epithelial-to-mesenchymal transition. UBQLNs interact with a variety of substrates, including cell surface receptors, transcription factor regulators, proteasomal machinery proteins, and transmembrane proteins. In addition, previous work from our lab shows that UBQLN1 interacts with insulin-like growth factor receptor family members (IGF1R, IGF2R, and INSR) and this interaction regulates the activity and proteostasis of IGFR family members. We wondered whether UBQLN proteins could also bind and regulate additional receptor tyrosine kinases. Thus, we investigated a link between UBQLN and the oncogene epidermal growth factor receptor (EGFR) in lung adenocarcinoma cells. Loss of UBQLN1 occurs at high frequency in human lung cancer patient samples and we have shown that the loss of UBQLN1 is capable of altering processes involved in cell proliferation, migration, invasion, and epithelial-to-mesenchymal transition in lung adenocarcinoma cell lines. Here, we present data that loss of UBQLN1 resulted in increased turnover of total EGFR while increasing the relative amount of phosphorylated EGFR in lung adenocarcinoma cells, especially in the presence of its ligand EGF. Furthermore, the loss of UBQLN1 led to a more invasive cell phenotype as manifested by increased proliferation, migration, and speed of movement of these lung adenocarcinoma cells. Taken together, UBQLN1 regulates the expression and stability of EGFR in lung cancer cells.
Collapse
Affiliation(s)
- Zimple Kurlawala
- James Graham Brown Cancer Center, School of Medicine, University of Louisville, Louisville, Kentucky
| | - Kumar Saurabh
- James Graham Brown Cancer Center, School of Medicine, University of Louisville, Louisville, Kentucky
| | - Rain Dunaway
- School of Medicine, University of Louisville, Louisville, Kentucky
| | - Parag P Shah
- James Graham Brown Cancer Center, School of Medicine, University of Louisville, Louisville, Kentucky
| | - Leah J Siskind
- James Graham Brown Cancer Center, School of Medicine, University of Louisville, Louisville, Kentucky.,Department of Pharmacology and Toxicology, University of Louisville, Louisville, Kentucky
| | - Levi J Beverly
- James Graham Brown Cancer Center, School of Medicine, University of Louisville, Louisville, Kentucky.,Department of Pharmacology and Toxicology, University of Louisville, Louisville, Kentucky.,Division of Hematology and Oncology, School of Medicine, University of Louisville, Louisville, Kentucky
| |
Collapse
|
11
|
Jantrapirom S, Lo Piccolo L, Pruksakorn D, Potikanond S, Nimlamool W. Ubiquilin Networking in Cancers. Cancers (Basel) 2020; 12:E1586. [PMID: 32549375 PMCID: PMC7352256 DOI: 10.3390/cancers12061586] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/14/2020] [Revised: 06/11/2020] [Accepted: 06/11/2020] [Indexed: 12/13/2022] Open
Abstract
Ubiquilins or UBQLNs, members of the ubiquitin-like and ubiquitin-associated domain (UBL-UBA) protein family, serve as adaptors to coordinate the degradation of specific substrates via both proteasome and autophagy pathways. The UBQLN substrates reveal great diversity and impact a wide range of cellular functions. For decades, researchers have been attempting to uncover a puzzle and understand the role of UBQLNs in human cancers, particularly in the modulation of oncogene's stability and nucleotide excision repair. In this review, we summarize the UBQLNs' genetic variants that are associated with the most common cancers and also discuss their reliability as a prognostic marker. Moreover, we provide an overview of the UBQLNs networks that are relevant to cancers in different ways, including cell cycle, apoptosis, epithelial-mesenchymal transition, DNA repairs and miRNAs. Finally, we include a future prospective on novel ubiquilin-based cancer therapies.
Collapse
Affiliation(s)
- Salinee Jantrapirom
- Department of Pharmacology, Faculty of Medicine, Chiang Mai University, Muang, Chiang Mai 50200, Thailand; (S.J.); (S.P.)
| | - Luca Lo Piccolo
- Omics Center for Health Science, Faculty of Medicine, Chiang Mai University, Muang, Chiang Mai 50200, Thailand; (L.L.P.); (D.P.)
| | - Dumnoensun Pruksakorn
- Omics Center for Health Science, Faculty of Medicine, Chiang Mai University, Muang, Chiang Mai 50200, Thailand; (L.L.P.); (D.P.)
- Department of Orthopedics, Orthopedic Laboratory and Research Network Center (OLARN), Faculty of Medicine, Chiang Mai University, Chiang Mai 50200, Thailand
- Excellence Center in Osteology Research and Training Center (ORTC), Chiang Mai University, Chiang Mai 50200, Thailand
| | - Saranyapin Potikanond
- Department of Pharmacology, Faculty of Medicine, Chiang Mai University, Muang, Chiang Mai 50200, Thailand; (S.J.); (S.P.)
- Research Center of Pharmaceutical Nanotechnology, Chiang Mai University, Chiang Mai 50200, Thailand
| | - Wutigri Nimlamool
- Department of Pharmacology, Faculty of Medicine, Chiang Mai University, Muang, Chiang Mai 50200, Thailand; (S.J.); (S.P.)
- Research Center of Pharmaceutical Nanotechnology, Chiang Mai University, Chiang Mai 50200, Thailand
| |
Collapse
|
12
|
Lorenz-Guertin JM, Jacob TC. GABA type a receptor trafficking and the architecture of synaptic inhibition. Dev Neurobiol 2018; 78:238-270. [PMID: 28901728 PMCID: PMC6589839 DOI: 10.1002/dneu.22536] [Citation(s) in RCA: 38] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/01/2017] [Revised: 09/08/2017] [Accepted: 09/08/2017] [Indexed: 12/21/2022]
Abstract
Ubiquitous expression of GABA type A receptors (GABAA R) in the central nervous system establishes their central role in coordinating most aspects of neural function and development. Dysregulation of GABAergic neurotransmission manifests in a number of human health disorders and conditions that in certain cases can be alleviated by drugs targeting these receptors. Precise changes in the quantity or activity of GABAA Rs localized at the cell surface and at GABAergic postsynaptic sites directly impact the strength of inhibition. The molecular mechanisms constituting receptor trafficking to and from these compartments therefore dictate the efficacy of GABAA R function. Here we review the current understanding of how GABAA Rs traffic through biogenesis, plasma membrane transport, and degradation. Emphasis is placed on discussing novel GABAergic synaptic proteins, receptor and scaffolding post-translational modifications, activity-dependent changes in GABAA R confinement, and neuropeptide and neurosteroid mediated changes. We further highlight modern techniques currently advancing the knowledge of GABAA R trafficking and clinically relevant neurodevelopmental diseases connected to GABAergic dysfunction. © 2017 Wiley Periodicals, Inc. Develop Neurobiol 78: 238-270, 2018.
Collapse
Affiliation(s)
- Joshua M Lorenz-Guertin
- Department of Pharmacology and Chemical Biology, University of Pittsburgh School of Medicine, Pittsburgh, Pennsylvania, 15261
| | - Tija C Jacob
- Department of Pharmacology and Chemical Biology, University of Pittsburgh School of Medicine, Pittsburgh, Pennsylvania, 15261
| |
Collapse
|
13
|
Regulation of insulin-like growth factor receptors by Ubiquilin1. Biochem J 2017; 474:4105-4118. [PMID: 29054976 DOI: 10.1042/bcj20170620] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/07/2017] [Revised: 10/03/2017] [Accepted: 10/19/2017] [Indexed: 12/18/2022]
Abstract
Insulin-like growth factor-1 receptor (IGF1R) is a receptor tyrosine kinase that mediates growth, proliferation and survival. Dysregulation of IGF pathway contributes to the initiation, progression and metastasis of cancer and is also involved in diseases of glucose metabolism, such as diabetes. We have identified Ubiquilin1 (UBQLN1) as a novel interaction partner of IGF1R, IGF2R and insulin receptor (INSR). UBQLN family of proteins have been studied primarily in the context of protein quality control and in the field of neurodegenerative disorders. Our laboratory discovered a link between UBQLN1 function and tumorigenesis, such that UBQLN1 is lost and underexpressed in 50% of human lung adenocarcinoma cases. We demonstrate here that UBQLN1 regulates the expression and activity of IGF1R. Following loss of UBQLN1 in lung adenocarcinoma cells, there is accelerated loss of IGF1R. Despite decreased levels of total receptors, the ratio of active : total receptors is higher in cells that lack UBQLN1. UBQLN1 also regulates INSR and IGF2R post-stimulation with ligand. We conclude that UBQLN1 is essential for normal regulation of IGF receptors. UBQLN-1-deficient cells demonstrate increased cell viability compared with control when serum-starved and stimulation of IGF pathway in these cells increased their migratory potential by 3-fold. As the IGF pathway is involved in processes of normal growth, development, metabolism and cancer progression, understanding its regulation by Ubiquilin1 can be of tremendous value to many disciplines.
Collapse
|
14
|
Printsev I, Curiel D, Carraway KL. Membrane Protein Quantity Control at the Endoplasmic Reticulum. J Membr Biol 2017; 250:379-392. [PMID: 27743014 PMCID: PMC5392169 DOI: 10.1007/s00232-016-9931-0] [Citation(s) in RCA: 32] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/01/2016] [Accepted: 09/28/2016] [Indexed: 02/07/2023]
Abstract
The canonical function of the endoplasmic reticulum-associated degradation (ERAD) system is to enforce quality control among membrane-associated proteins by targeting misfolded secreted, intra-organellar, and intramembrane proteins for degradation. However, increasing evidence suggests that ERAD additionally functions in maintaining appropriate levels of a subset of membrane-associated proteins. In this 'quantity control' capacity, ERAD responds to environmental cues to regulate the proteasomal degradation of specific ERAD substrates according to cellular need. In this review, we discuss in detail seven proteins that are targeted by the ERAD quantity control system. Not surprisingly, ERAD-mediated protein degradation is a key regulatory feature of a variety of ER-resident proteins, including HMG-CoA reductase, cytochrome P450 3A4, IP3 receptor, and type II iodothyronine deiodinase. In addition, the ERAD quantity control system plays roles in maintaining the proper stoichiometry of multi-protein complexes by mediating the degradation of components that are produced in excess of the limiting subunit. Perhaps somewhat unexpectedly, recent evidence suggests that the ERAD quantity control system also contributes to the regulation of plasma membrane-localized signaling receptors, including the ErbB3 receptor tyrosine kinase and the GABA neurotransmitter receptors. For these substrates, a proportion of the newly synthesized yet properly folded receptors are diverted for degradation at the ER, and are unable to traffic to the plasma membrane. Given that receptor abundance or concentration within the plasma membrane plays key roles in determining signaling efficiency, these observations may point to a novel mechanism for modulating receptor-mediated cellular signaling.
Collapse
Affiliation(s)
- Ignat Printsev
- Department of Biochemistry and Molecular Medicine, and UC Davis Comprehensive Cancer Center, UC Davis School of Medicine, Research Building III, Room 1100B, 4645 2nd Avenue, Sacramento, CA, 95817, USA
| | - Daniel Curiel
- Department of Biochemistry and Molecular Medicine, and UC Davis Comprehensive Cancer Center, UC Davis School of Medicine, Research Building III, Room 1100B, 4645 2nd Avenue, Sacramento, CA, 95817, USA
| | - Kermit L Carraway
- Department of Biochemistry and Molecular Medicine, and UC Davis Comprehensive Cancer Center, UC Davis School of Medicine, Research Building III, Room 1100B, 4645 2nd Avenue, Sacramento, CA, 95817, USA.
| |
Collapse
|
15
|
Chen M, Wang J, Jiang J, Zheng X, Justice NJ, Wang K, Ran X, Li Y, Huo Q, Zhang J, Li H, Lu N, Wang Y, Zheng H, Long C, Yang L. APP modulates KCC2 expression and function in hippocampal GABAergic inhibition. eLife 2017; 6. [PMID: 28054918 PMCID: PMC5224924 DOI: 10.7554/elife.20142] [Citation(s) in RCA: 64] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/28/2016] [Accepted: 12/16/2016] [Indexed: 12/14/2022] Open
Abstract
Amyloid precursor protein (APP) is enriched at the synapse, but its synaptic function is still poorly understood. We previously showed that GABAergic short-term plasticity is impaired in App knock-out (App-/-) animals, but the precise mechanism by which APP regulates GABAergic synaptic transmission has remained elusive. Using electrophysiological, biochemical, moleculobiological, and pharmacological analysis, here we show that APP can physically interact with KCC2, a neuron-specific K+-Cl- cotransporter that is essential for Cl- homeostasis and fast GABAergic inhibition. APP deficiency results in significant reductions in both total and membrane KCC2 levels, leading to a depolarizing shift in the GABA reversal potential (EGABA). Simultaneous measurement of presynaptic action potentials and inhibitory postsynaptic currents (IPSCs) in hippocampal neurons reveals impaired unitary IPSC amplitudes attributable to a reduction in α1 subunit levels of GABAAR. Importantly, restoration of normal KCC2 expression and function in App-/- mice rescues EGABA, GABAAR α1 levels and GABAAR mediated phasic inhibition. We show that APP functions to limit tyrosine-phosphorylation and ubiquitination and thus subsequent degradation of KCC2, providing a mechanism by which APP influences KCC2 abundance. Together, these experiments elucidate a novel molecular pathway in which APP regulates, via protein-protein interaction with KCC2, GABAAR mediated inhibition in the hippocampus. DOI:http://dx.doi.org/10.7554/eLife.20142.001 Alzheimer’s disease is the most common form of dementia. One of the hallmarks of the disease is the formation of sticky protein clumps called amyloid plaques in the brain. These plaques are formed from specific fragments of a protein called APP. The intact form of APP is essential for synapses (the junctions across which neurons transmit signals) to form and work correctly. The hippocampus is one of the first brain regions to be affected in Alzheimer’s disease and is important for forming memories and emotions. In the hippocampus, GABAA receptors at synapses normally tightly regulate synaptic signaling by reducing the ability of the receiving neuron to respond, but this inhibition is disrupted in Alzheimer’s disease. Studies suggest that APP can affect how GABAA receptors transmit signals, but it is not known how it does so. One possibility is that APP regulates a protein called KCC2 that helps to maintain the inhibitory effect of GABAA receptors. To investigate this, Chen et al. genetically modified mice to lack the gene that produces APP. These mice had a lower level of KCC2 in their hippocampus than normal mice, and their GABAA receptors were less able to inhibit synaptic signaling. Further experiments demonstrated that APP physically interacts with KCC2 and maintains normal levels of the protein by preventing it from being chemically modified and degraded. Chen et al. also showed that treating mice that lack APP with specific compounds can restore KCC2 activity and return the behavior of synaptic GABAA receptors to normal. Future studies in mice (and eventually people) that exhibit symptoms of Alzheimer's disease will help to determine whether KCC2 is important in the development of the disease. If so, modifying the levels of the KCC2 protein in the brain could potentially help to slow down memory loss in Alzheimer’s disease. DOI:http://dx.doi.org/10.7554/eLife.20142.002
Collapse
Affiliation(s)
- Ming Chen
- School of Life Sciences, South China Normal University, Guangzhou, China
| | - Jinzhao Wang
- School of Life Sciences, South China Normal University, Guangzhou, China
| | - Jinxiang Jiang
- School of Psychology and Center for Studies of Psychological Application, South China Normal University, Guangzhou, China.,Brain Science Institute, South China Normal University, Guangzhou, China
| | - Xingzhi Zheng
- School of Life Sciences, South China Normal University, Guangzhou, China
| | - Nicholas J Justice
- Institute of Molecular Medicine, University of Texas Health Sciences Center, Houston, United States
| | - Kun Wang
- School of Life Sciences, South China Normal University, Guangzhou, China
| | - Xiangqian Ran
- School of Life Sciences, South China Normal University, Guangzhou, China
| | - Yi Li
- School of Life Sciences, South China Normal University, Guangzhou, China
| | - Qingwei Huo
- School of Life Sciences, South China Normal University, Guangzhou, China
| | - Jiajia Zhang
- School of Life Sciences, South China Normal University, Guangzhou, China
| | - Hongmei Li
- Huffington Center on Aging, Baylor College of Medicine, Houston, United States
| | - Nannan Lu
- School of Life Sciences, South China Normal University, Guangzhou, China
| | - Ying Wang
- School of Life Sciences, South China Normal University, Guangzhou, China
| | - Hui Zheng
- Huffington Center on Aging, Baylor College of Medicine, Houston, United States
| | - Cheng Long
- School of Life Sciences, South China Normal University, Guangzhou, China.,Brain Science Institute, South China Normal University, Guangzhou, China
| | - Li Yang
- School of Psychology and Center for Studies of Psychological Application, South China Normal University, Guangzhou, China.,Brain Science Institute, South China Normal University, Guangzhou, China.,Guangdong Key Laboratory of Mental Health and Cognitive Science, South China Normal University, Guangzhou, China
| |
Collapse
|
16
|
Mele M, Leal G, Duarte CB. Role of GABAAR trafficking in the plasticity of inhibitory synapses. J Neurochem 2016; 139:997-1018. [DOI: 10.1111/jnc.13742] [Citation(s) in RCA: 45] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/30/2016] [Revised: 07/12/2016] [Accepted: 07/13/2016] [Indexed: 12/22/2022]
Affiliation(s)
- Miranda Mele
- Center for Neuroscience and Cell Biology; University of Coimbra; Coimbra Portugal
| | - Graciano Leal
- Center for Neuroscience and Cell Biology; University of Coimbra; Coimbra Portugal
| | - Carlos B. Duarte
- Center for Neuroscience and Cell Biology; University of Coimbra; Coimbra Portugal
- Department of Life Sciences; University of Coimbra; Coimbra Portugal
| |
Collapse
|
17
|
Plic-1, a new target in repressing epileptic seizure by regulation of GABAAR function in patients and a rat model of epilepsy. Clin Sci (Lond) 2015; 129:1207-23. [PMID: 26415648 DOI: 10.1042/cs20150202] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/16/2015] [Accepted: 09/25/2015] [Indexed: 12/27/2022]
Abstract
Plic-1 regulates GABAAR expression at synaptic sites during epileptic seizure. Plic-1 prolongs the seizure latency and reduces the seizure severity in epileptic rats. Plic-1 affects the inhibitory function by changing the mIPSCs and evoked IPSCs of the phasic GABA-ergic synaptic current.
Collapse
|
18
|
Brady ML, Moon CE, Jacob TC. Using an α-bungarotoxin binding site tag to study GABA A receptor membrane localization and trafficking. J Vis Exp 2014. [PMID: 24747556 DOI: 10.3791/51365] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/20/2023] Open
Abstract
It is increasingly evident that neurotransmitter receptors, including ionotropic GABA A receptors (GABAAR), exhibit highly dynamic trafficking and cell surface mobility(1-7). To study receptor cell surface localization and endocytosis, the technique described here combines the use of fluorescent α-bungarotoxin with cells expressing constructs containing an α-bungarotoxin (Bgt) binding site (BBS). The BBS (WRYYESSLEPYPD) is based on the α subunit of the muscle nicotinic acetylcholine receptor, which binds Bgt with high affinity(8,9). Incorporation of the BBS site allows surface localization and measurements of receptor insertion or removal with application of exogenous fluorescent Bgt, as previously described in the tracking of GABAA and metabotropic GABAB receptors(2,10). In addition to the BBS site, we inserted a pH-sensitive GFP (pHGFP(11)) between amino acids 4 and 5 of the mature GABAAR subunit by standard molecular biology and PCR cloning strategies (see Figure 1)(12). The BBS is 3' of the pH-sensitive GFP reporter, separated by a 13-amino acid alanine/proline linker. For trafficking studies described in this publication that are based on fixed samples, the pHGFP serves as a reporter of total tagged GABAAR subunit protein levels, allowing normalization of the Bgt labeled receptor population to total receptor population. This minimizes cell to cell Bgt staining signal variability resulting from higher or lower baseline expression of the tagged GABAAR subunits. Furthermore the pHGFP tag enables easy identification of construct expressing cells for live or fixed imaging experiments.
Collapse
Affiliation(s)
- Megan L Brady
- Pharmacology & Chemical Biology Department, University of Pittsburgh School of Medicine
| | - Charles E Moon
- Pharmacology & Chemical Biology Department, University of Pittsburgh School of Medicine
| | - Tija C Jacob
- Pharmacology & Chemical Biology Department, University of Pittsburgh School of Medicine;
| |
Collapse
|
19
|
Zhou C, Huang Z, Ding L, Deel ME, Arain FM, Murray CR, Patel RS, Flanagan CD, Gallagher MJ. Altered cortical GABAA receptor composition, physiology, and endocytosis in a mouse model of a human genetic absence epilepsy syndrome. J Biol Chem 2013; 288:21458-21472. [PMID: 23744069 DOI: 10.1074/jbc.m112.444372] [Citation(s) in RCA: 41] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022] Open
Abstract
Patients with generalized epilepsy exhibit cerebral cortical disinhibition. Likewise, mutations in the inhibitory ligand-gated ion channels, GABAA receptors (GABAARs), cause generalized epilepsy syndromes in humans. Recently, we demonstrated that heterozygous knock-out (Hetα1KO) of the human epilepsy gene, the GABAAR α1 subunit, produced absence epilepsy in mice. Here, we determined the effects of Hetα1KO on the expression and physiology of GABAARs in the mouse cortex. We found that Hetα1KO caused modest reductions in the total and surface expression of the β2 subunit but did not alter β1 or β3 subunit expression, results consistent with a small reduction of GABAARs. Cortices partially compensated for Hetα1KO by increasing the fraction of residual α1 subunit on the cell surface and by increasing total and surface expression of α3, but not α2, subunits. Co-immunoprecipitation experiments revealed that Hetα1KO increased the fraction of α1 subunits, and decreased the fraction of α3 subunits, that associated in hybrid α1α3βγ receptors. Patch clamp electrophysiology studies showed that Hetα1KO layer VI cortical neurons exhibited reduced inhibitory postsynaptic current peak amplitudes, prolonged current rise and decay times, and altered responses to benzodiazepine agonists. Finally, application of inhibitors of dynamin-mediated endocytosis revealed that Hetα1KO reduced base-line GABAAR endocytosis, an effect that probably contributes to the observed changes in GABAAR expression. These findings demonstrate that Hetα1KO exerts two principle disinhibitory effects on cortical GABAAR-mediated inhibitory neurotransmission: 1) a modest reduction of GABAAR number and 2) a partial compensation with GABAAR isoforms that possess physiological properties different from those of the otherwise predominant α1βγ GABAARs.
Collapse
Affiliation(s)
- Chengwen Zhou
- From the Department of Neurology, Vanderbilt University, Nashville, Tennessee 37232
| | - Zhiling Huang
- From the Department of Neurology, Vanderbilt University, Nashville, Tennessee 37232
| | - Li Ding
- From the Department of Neurology, Vanderbilt University, Nashville, Tennessee 37232
| | - M Elizabeth Deel
- From the Department of Neurology, Vanderbilt University, Nashville, Tennessee 37232
| | - Fazal M Arain
- From the Department of Neurology, Vanderbilt University, Nashville, Tennessee 37232
| | - Clark R Murray
- From the Department of Neurology, Vanderbilt University, Nashville, Tennessee 37232
| | - Ronak S Patel
- From the Department of Neurology, Vanderbilt University, Nashville, Tennessee 37232
| | | | - Martin J Gallagher
- From the Department of Neurology, Vanderbilt University, Nashville, Tennessee 37232.
| |
Collapse
|
20
|
Takalo M, Haapasalo A, Natunen T, Viswanathan J, Kurkinen KM, Tanzi RE, Soininen H, Hiltunen M. Targeting ubiquilin-1 in Alzheimer's disease. Expert Opin Ther Targets 2013; 17:795-810. [PMID: 23600477 DOI: 10.1517/14728222.2013.791284] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/18/2023]
Abstract
INTRODUCTION Alzheimer's disease (AD) is a common neurodegenerative disorder affecting an increasing number of people worldwide as the population ages. Currently, there are no drugs available that could prevent AD pathogenesis or slow down its progression. Increasing evidence links ubiquilin-1, an ubiquitin-like protein, into the pathogenic mechanisms of AD and other neurodegenerative diseases. Ubiquilin-1 has been shown to play a key role in the regulation of the levels, subcellular targeting, aggregation and degradation of various neurodegenerative disease-associated proteins. These include the amyloid precursor protein and presenilins that are intimately involved in the mechanisms of AD. AREAS COVERED Here, the properties and diverse functions of ubiquilin-1 protein in the context of the pathogenesis of AD and other neurodegenerative disorders are discussed. This review recapitulates the available knowledge on the involvement of ubiquilin-1 in the genetic and molecular mechanisms in AD. Furthermore, the association of ubiquilin-1 with specific proteins and mechanisms involved in the pathogenesis of neurodegenerative diseases is described and the known ubiquilin-1-interacting proteins summarized. EXPERT OPINION The variety of ubiquilin-1-interacting proteins and its central role in the regulation of protein levels and degradation provides a number of novel candidates and approaches for future research and drug discovery.
Collapse
Affiliation(s)
- Mari Takalo
- Institute of Clinical Medicine-Neurology, University of Eastern Finland and Department of Neurology, Kuopio University Hospital, Kuopio, Finland
| | | | | | | | | | | | | | | |
Collapse
|
21
|
Ubiquitination of neurotransmitter receptors and postsynaptic scaffolding proteins. Neural Plast 2013; 2013:432057. [PMID: 23431475 PMCID: PMC3574743 DOI: 10.1155/2013/432057] [Citation(s) in RCA: 27] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/28/2012] [Accepted: 12/26/2012] [Indexed: 12/19/2022] Open
Abstract
The human brain is made up of an extensive network of neurons that communicate by forming specialized connections called synapses. The amount, location, and dynamic turnover of synaptic proteins, including neurotransmitter receptors and synaptic scaffolding molecules, are under complex regulation and play a crucial role in synaptic connectivity and plasticity, as well as in higher brain functions. An increasing number of studies have established ubiquitination and proteasome-mediated degradation as universal mechanisms in the control of synaptic protein homeostasis. In this paper, we focus on the role of the ubiquitin-proteasome system (UPS) in the turnover of major neurotransmitter receptors, including glutamatergic and nonglutamatergic receptors, as well as postsynaptic receptor-interacting proteins.
Collapse
|
22
|
El Ayadi A, Stieren ES, Barral JM, Boehning D. Ubiquilin-1 and protein quality control in Alzheimer disease. Prion 2013; 7:164-9. [PMID: 23360761 PMCID: PMC3609125 DOI: 10.4161/pri.23711] [Citation(s) in RCA: 20] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022] Open
Abstract
Single nucleotide polymorphisms in the ubiquilin-1 gene may confer risk for late-onset Alzheimer disease (AD). We have shown previously that ubiquilin-1 functions as a molecular chaperone for the amyloid precursor protein (APP) and that protein levels of ubiquilin-1 are decreased in the brains of AD patients. We have recently found that ubiquilin-1 regulates APP trafficking and subsequent secretase processing by stimulating non-degradative ubiquitination of a single lysine residue in the cytosolic domain of APP. Thus, ubiquilin-1 plays a central role in regulating APP biosynthesis, trafficking and ultimately toxicity. As ubiquilin-1 and other ubiquilin family members have now been implicated in the pathogenesis of numerous neurodegenerative diseases, these findings provide mechanistic insights into the central role of ubiquilin proteins in maintaining neuronal proteostasis.
Collapse
Affiliation(s)
- Amina El Ayadi
- Department of Neuroscience and Cell Biology, University of Texas Medical Branch, Galveston, TX USA
| | | | | | | |
Collapse
|
23
|
Benzodiazepine treatment induces subtype-specific changes in GABA(A) receptor trafficking and decreases synaptic inhibition. Proc Natl Acad Sci U S A 2012; 109:18595-600. [PMID: 23091016 DOI: 10.1073/pnas.1204994109] [Citation(s) in RCA: 66] [Impact Index Per Article: 5.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/18/2022] Open
Abstract
Benzodiazepines potentiate γ-aminobutyric acid type A receptor (GABA(A)R) activity and are widely prescribed to treat anxiety, insomnia, and seizure disorders. Unfortunately, clinical use of benzodiazepines (BZs) is severely limited by tolerance. The mechanisms leading to BZ tolerance are unknown. BZs bind at the interface between an α and γ subunit of GABA(A)Rs, preferentially enhancing synaptic receptors largely composed of α(1-3, 5), β3, and γ2 subunits. Using confocal imaging and patch-clamp approaches, we show that treatment with the BZ flurazepam decreases GABA(A)R surface levels and the efficacy of neuronal inhibition in hippocampal neurons. A dramatic decrease in surface and total levels of α2 subunit-containing GABA(A)Rs occurred within 24 h of flurazepam treatment, whereas GABA(A)Rs incorporating α1 subunits showed little alteration. The GABA(A)R surface depletion could be reversed by treatment with the BZ antagonist Ro 15-1788. Coincident with decreased GABA(A)R surface levels, flurazepam treatment reduced miniature inhibitory postsynaptic current amplitude, which returned to control levels with acute Ro 15-1788 treatment. GABA(A)R endocytosis and insertion rates were unchanged by flurazepam treatment. Treatment with leupeptin restored flurazepam lowered receptor surface levels, strongly suggesting that flurazepam increases lysosomal degradation of GABA(A)Rs. Together, these data suggest that flurazepam exposure enhances degradation of α2 subunit-containing GABA(A)Rs after their removal from the plasma membrane, leading to a reduction in inhibitory synapse size and number along with a decrease in the efficacy of synaptic inhibition. These reported subtype-specific changes in GABA(A)R trafficking provide significant mechanistic insight into the initial neuroadaptive responses occurring with BZ treatment.
Collapse
|
24
|
Rezvani K, Baalman K, Teng Y, Mee MP, Dawson SP, Wang H, De Biasi M, Mayer RJ. Proteasomal degradation of the metabotropic glutamate receptor 1α is mediated by Homer-3 via the proteasomal S8 ATPase: Signal transduction and synaptic transmission. J Neurochem 2012; 122:24-37. [PMID: 22486777 DOI: 10.1111/j.1471-4159.2012.07752.x] [Citation(s) in RCA: 20] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/29/2022]
Abstract
The metabotropic glutamate receptors (mGluRs) fine-tune the efficacy of synaptic transmission. This unique feature makes mGluRs potential targets for the treatment of various CNS disorders. There is ample evidence to show that the ubiquitin proteasome system mediates changes in synaptic strength leading to multiple forms of synaptic plasticity. The present study describes a novel interaction between post-synaptic adaptors, long Homer-3 proteins, and one of the 26S proteasome regulatory subunits, the S8 ATPase, that influences the degradation of the metabotropic glutamate receptor 1α (mGluR1α). We have shown that the two human long Homer-3 proteins specifically interact with human proteasomal S8 ATPase. We identified that mGluR1α and long Homer-3s immunoprecipitate with the 26S proteasome both in vitro and in vivo. We further found that the mGluR1α receptor can be ubiquitinated and degraded by the 26S proteasome and that Homer-3A facilitates this process. Furthermore, the siRNA mediated silencing of Homer-3 led to increased levels of total and plasma membrane-associated mGluR1α receptors. These results suggest that long Homer-3 proteins control the degradation of mGluR1α receptors by shuttling ubiquitinated mGluR-1α receptors to the 26S proteasome via the S8 ATPase which may modulate synaptic transmission.
Collapse
Affiliation(s)
- Khosrow Rezvani
- Division of Basic Biomedical Sciences, Sanford School of Medicine, The University of South Dakota, Vermillion, SD 57069, USA.
| | | | | | | | | | | | | | | |
Collapse
|
25
|
Hines RM, Davies PA, Moss SJ, Maguire J. Functional regulation of GABAA receptors in nervous system pathologies. Curr Opin Neurobiol 2011; 22:552-8. [PMID: 22036769 DOI: 10.1016/j.conb.2011.10.007] [Citation(s) in RCA: 95] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/21/2011] [Revised: 10/05/2011] [Accepted: 10/06/2011] [Indexed: 01/01/2023]
Abstract
Inhibitory neurotransmission is primarily governed by γ-aminobutyric acid (GABA) type A receptors (GABAARs). GABAARs are heteropentameric ligand-gated channels formed by the combination of 19 possible subunits. GABAAR subunits are subject to multiple types of regulation, impacting the localization, properties, and function of assembled receptors. GABAARs mediate both phasic (synaptic) and tonic (extrasynaptic) inhibition. While the regulatory mechanisms governing synaptic receptors have begun to be defined, little is known about the regulation of extrasynaptic receptors. We examine the contributions of GABAARs to the pathogenesis of neurodevelopmental disorders, schizophrenia, depression, epilepsy, and stroke, with particular focus on extrasynaptic GABAARs. We suggest that extrasynaptic GABAARs are attractive targets for the treatment of these disorders, and that research should be focused on delineating the mechanisms that regulate extrasynaptic GABAARs, promoting new therapeutic approaches.
Collapse
Affiliation(s)
- Rochelle M Hines
- Department of Neuroscience, Tufts University School of Medicine, 136 Harrison Ave, Boston, MA 02111, USA
| | | | | | | |
Collapse
|
26
|
Vithlani M, Terunuma M, Moss SJ. The dynamic modulation of GABA(A) receptor trafficking and its role in regulating the plasticity of inhibitory synapses. Physiol Rev 2011; 91:1009-22. [PMID: 21742794 DOI: 10.1152/physrev.00015.2010] [Citation(s) in RCA: 152] [Impact Index Per Article: 10.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022] Open
Abstract
Inhibition in the adult mammalian central nervous system (CNS) is mediated by γ-aminobutyric acid (GABA). The fast inhibitory actions of GABA are mediated by GABA type A receptors (GABA(A)Rs); they mediate both phasic and tonic inhibition in the brain and are the principle sites of action for anticonvulsant, anxiolytic, and sedative-hypnotic agents that include benzodiazepines, barbiturates, neurosteroids, and some general anesthetics. GABA(A)Rs are heteropentameric ligand-gated ion channels that are found concentrated at inhibitory postsynaptic sites where they mediate phasic inhibition and at extrasynaptic sites where they mediate tonic inhibition. The efficacy of inhibition and thus neuronal excitability is critically dependent on the accumulation of specific GABA(A)R subtypes at inhibitory synapses. Here we evaluate how neurons control the number of GABA(A)Rs on the neuronal plasma membrane together with their selective stabilization at synaptic sites. We then go on to examine the impact that these processes have on the strength of synaptic inhibition and behavior.
Collapse
Affiliation(s)
- Mansi Vithlani
- Department of Neuroscience, Tufts University School of Medicine, Boston, Massachusetts, USA
| | | | | |
Collapse
|
27
|
Sick E, Boukhari A, Deramaudt T, Rondé P, Bucher B, André P, Gies JP, Takeda K. Activation of CD47 receptors causes proliferation of human astrocytoma but not normal astrocytes via an Akt-dependent pathway. Glia 2011; 59:308-19. [PMID: 21125662 DOI: 10.1002/glia.21102] [Citation(s) in RCA: 50] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/18/2022]
Abstract
CD47 is a membrane receptor that plays pivotal roles in many pathophysiological processes, including infection, inflammation, cell spreading, proliferation, and apoptosis. We show that activation of CD47 increases proliferation of human U87 and U373 astrocytoma cells but not normal astrocytes. CD47 function-blocking antibodies inhibit proliferation of untreated U87 and U373 cells but not normal astrocytes, suggesting that CD47 may be constitutively activated in astrocytoma. CD47 expression levels were similar in our three cell types. CD47 couples to G-proteins in astrocytes and astrocytoma and especially to the Gβγ dimer. Downstream signaling following CD47 activation involves Gβγ dimer-dependent activation of the PI3K/Akt pathway in astrocytoma cells but not in normal astrocytes. This pathway is known to be deregulated in astrocytoma, leading to cell proliferation and enhanced survival signals. Putative PLIC-1 interaction with CD47 in astrocytoma cells but not astrocytes may contribute to the proliferative effect observed upon activation of CD47. Our data indicate that CD47 receptors have a stimulatory role in cell proliferation and demonstrate for the first time that CD47 signals via the PI3K/Akt pathway in cancerous cells but not normal cells.
Collapse
Affiliation(s)
- Emilie Sick
- Université de Strasbourg, CNRS UMR 7213-Pharmacologie, Faculté de Pharmacie, 74 rte du Rhin, Illkirch, France.
| | | | | | | | | | | | | | | |
Collapse
|
28
|
Allison JG, Das PM, Ma J, Inglis FM, Jones FE. The ERBB4 intracellular domain (4ICD) regulates NRG1-induced gene expression in hippocampal neurons. Neurosci Res 2011; 70:155-63. [PMID: 21352860 DOI: 10.1016/j.neures.2011.02.009] [Citation(s) in RCA: 13] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/15/2010] [Revised: 02/07/2011] [Accepted: 02/15/2011] [Indexed: 01/12/2023]
Abstract
The NRG1 growth factor and ERBB4 receptor have been identified as leading schizophrenia risk genes. Although NRG1 and ERBB4 have been shown to modulate neuronal functions involved in schizophrenia, including both GABAergic and glutamatergic synapses, the exact molecular mechanisms remain poorly understood. Here we investigated ERBB4 intracellular domain, 4ICD, transactivator function in rat hippocampal cultures by inhibiting γ-secretase mediated ERBB4 regulated intramembrane proteolysis (RIP). NRG1 stimulation resulted in a dramatic increase in the number of hippocampal cells displaying nuclear 4ICD which was abolished in cultures pretreated with the γ-secretase inhibitor compound E (CE). To identify NRG1-4ICD transactivated genes we compared global gene expression profiles of hippocampal cultures stimulated with NRG1 in the absence or presence of CE. In concordance with the contribution of NRG1-ERBB4 signaling to dendritic spine maturation and schizophrenia, global gene expression analysis followed by Ingenuity Pathway Analysis of the dataset identified NRG1-4ICD regulated genes significantly represented in semaphorin signaling and actin cytoskeletal plasticity and multiple genes with confirmed roles in dendritic spine morphogenesis. Using the power of global gene expression analysis our data provides a proof-of-concept supporting a role for non-canonical NRG1-4ICD signaling in the regulation of gene expression contributing to normal and schizophrenic neuronal function.
Collapse
Affiliation(s)
- June G Allison
- Department of Cell and Molecular Biology, Tulane University, 6400 Freret Street, New Orleans, LA 70118, USA
| | | | | | | | | |
Collapse
|
29
|
Abstract
Neurons are highly specialized cells whose connectivity at synapses subserves rapid information transfer in the brain. Proper information processing, learning, and memory storage in the brain requires continuous remodeling of synaptic networks. Such remodeling includes synapse formation, elimination, synaptic protein turnover, and changes in synaptic transmission. An emergent mechanism for regulating synapse function is posttranslational modification through the ubiquitin pathway at the postsynaptic membrane. Here, we discuss recent findings implicating ubiquitination and protein degradation in postsynaptic function and plasticity. We describe postsynaptic ubiquitination pathways and their role in brain development, neuronal physiology, and brain disorders.
Collapse
Affiliation(s)
- Angela M Mabb
- Department of Neurobiology, Duke University Medical Center, Durham, North Carolina 27710, USA
| | | |
Collapse
|
30
|
Adusei DC, Pacey LKK, Chen D, Hampson DR. Early developmental alterations in GABAergic protein expression in fragile X knockout mice. Neuropharmacology 2010; 59:167-71. [PMID: 20470805 DOI: 10.1016/j.neuropharm.2010.05.002] [Citation(s) in RCA: 107] [Impact Index Per Article: 7.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/08/2010] [Revised: 03/26/2010] [Accepted: 05/05/2010] [Indexed: 11/19/2022]
Abstract
Fragile X syndrome is the most common heritable form of mental retardation. It is caused by silencing of the Fmr1 gene and the absence of the encoded protein. The purpose of this study was to examine global protein expression levels of GABA(A) and GABA(B) receptors, and GABAergic enzymes and trafficking proteins in fragile X knockout mice during brain maturation. Quantitative western blotting of homogenates of forebrain revealed that the levels of GABA(A) beta1 and beta3, GABA(B)-R1, NKCC1, KCC2, gephyrin and ubiquilin were not significantly different from wild-type mice at any of the postnatal time points examined. In contrast, the GABA(A) receptor alpha1, beta2, and delta subunits, and the GABA enzymes GABA transaminase and succinic semialdehyde dehydrogenase were down-regulated during postnatal development, while GAD65 was up-regulated in the adult knockout mouse brain. The GABA(A) receptor alpha1 and beta2 subunits displayed a divergent pattern of developmental expression whereby alpha1 was reduced in the immature brain but regained a level of expression similar to wild-type mice by adulthood, while the expression of beta2 was similar to wild-types at postnatal day 5 but reduced at day 12 and in the adult brain. The GABA(A) receptor delta subunit and the GABA catabolic enzymes GABA transaminase and succinic semialdehyde dehydrogenase were simultaneously but transiently decreased only at postnatal day 12. Our results demonstrate that GABA(A) receptor subunits and GABA enzymes display complex patterns of changes during brain development suggesting that dynamic interactions may occur between GABA transmitter levels and GABA receptors in fragile X syndrome.
Collapse
MESH Headings
- 4-Aminobutyrate Transaminase/genetics
- 4-Aminobutyrate Transaminase/metabolism
- Adaptor Proteins, Signal Transducing
- Adaptor Proteins, Vesicular Transport/genetics
- Adaptor Proteins, Vesicular Transport/metabolism
- Age Factors
- Animals
- Animals, Newborn
- Autophagy-Related Proteins
- Carrier Proteins/genetics
- Carrier Proteins/metabolism
- Fragile X Mental Retardation Protein/genetics
- Gene Expression Regulation, Developmental/genetics
- Membrane Proteins/genetics
- Membrane Proteins/metabolism
- Mice
- Mice, Inbred C57BL
- Mice, Knockout
- Receptors, GABA-A/genetics
- Receptors, GABA-A/metabolism
- Receptors, GABA-B/genetics
- Receptors, GABA-B/metabolism
- Sodium-Potassium-Chloride Symporters/genetics
- Sodium-Potassium-Chloride Symporters/metabolism
- Solute Carrier Family 12, Member 2
- Symporters/genetics
- Symporters/metabolism
- gamma-Aminobutyric Acid/genetics
- gamma-Aminobutyric Acid/metabolism
- K Cl- Cotransporters
Collapse
Affiliation(s)
- Daniel C Adusei
- Department of Pharmaceutical Sciences, Leslie Dan Faculty of Pharmacy, University of Toronto, 144 College St., Toronto, Ontario, Canada M5S 3M2
| | | | | | | |
Collapse
|
31
|
Smith KR, Kittler JT. The cell biology of synaptic inhibition in health and disease. Curr Opin Neurobiol 2010; 20:550-6. [PMID: 20650630 DOI: 10.1016/j.conb.2010.06.001] [Citation(s) in RCA: 46] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/15/2010] [Revised: 06/08/2010] [Accepted: 06/10/2010] [Indexed: 11/24/2022]
Abstract
Fast synaptic inhibition is largely mediated by GABA(A) receptors (GABA(A)Rs), ligand-gated chloride channels that play an essential role in the control of cell and network activity in the brain. Recent work has demonstrated that the delivery, number and stability of GABA(A)Rs at inhibitory synapses play a key role in the dynamic regulation of inhibitory synaptic efficacy and plasticity. The regulatory pathways essential for the fine-tuning of synaptic inhibition have also emerged as key sites of vulnerability during pathological changes in cell excitability in disease states.
Collapse
Affiliation(s)
- Katharine R Smith
- Department of Neuroscience, Physiology and Pharmacology, University College London, Gower Street, London, WC1E 6BT, UK
| | | |
Collapse
|
32
|
Ding L, Feng HJ, Macdonald RL, Botzolakis EJ, Hu N, Gallagher MJ. GABA(A) receptor alpha1 subunit mutation A322D associated with autosomal dominant juvenile myoclonic epilepsy reduces the expression and alters the composition of wild type GABA(A) receptors. J Biol Chem 2010; 285:26390-405. [PMID: 20551311 DOI: 10.1074/jbc.m110.142299] [Citation(s) in RCA: 34] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/06/2022] Open
Abstract
A GABA(A) receptor (GABA(A)R) alpha1 subunit mutation, A322D (AD), causes an autosomal dominant form of juvenile myoclonic epilepsy (ADJME). Previous studies demonstrated that the mutation caused alpha1(AD) subunit misfolding and rapid degradation, reducing its total and surface expression substantially. Here, we determined the effects of the residual alpha1(AD) subunit expression on wild type GABA(A)R expression to determine whether the AD mutation conferred a dominant negative effect. We found that although the alpha1(AD) subunit did not substitute for wild type alpha1 subunits on the cell surface, it reduced the surface expression of alpha1beta2gamma2 and alpha3beta2gamma2 receptors by associating with the wild type subunits within the endoplasmic reticulum and preventing them from trafficking to the cell surface. The alpha1(AD) subunit reduced surface expression of alpha3beta2gamma2 receptors by a greater amount than alpha1beta2gamma2 receptors, thus altering cell surface GABA(A)R composition. When transfected into cultured cortical neurons, the alpha1(AD) subunit altered the time course of miniature inhibitory postsynaptic current kinetics and reduced miniature inhibitory postsynaptic current amplitudes. These findings demonstrated that, in addition to causing a heterozygous loss of function of alpha1(AD) subunits, this epilepsy mutation also elicited a modest dominant negative effect that likely shapes the epilepsy phenotype.
Collapse
Affiliation(s)
- Li Ding
- Department of Neurology, Vanderbilt University, Nashville, Tennessee 37232, USA
| | | | | | | | | | | |
Collapse
|
33
|
Thomson AM, Jovanovic JN. Mechanisms underlying synapse-specific clustering of GABA(A) receptors. Eur J Neurosci 2010; 31:2193-203. [PMID: 20550567 DOI: 10.1111/j.1460-9568.2010.07252.x] [Citation(s) in RCA: 28] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/30/2022]
Abstract
A principle that arises from a body of previous work is that each presynaptic terminal recognises its postsynaptic partner and that each postsynaptic site recognises the origin of the synaptic bouton innervating it. In response, the presynaptic terminal sequesters the proteins whose interactions result in the dynamic transmitter release pattern and chemical modulation appropriate for that connection. In parallel, the postsynaptic site sequesters, inserts or captures the receptors and postsynaptic density proteins appropriate for that type of synapse. The focus of this review is the selective clustering of GABA(A) receptors (GABA(A)R) at synapses made by each class of inhibitory interneurone. This provides a system in which the mechanisms underlying transynaptic recognition can be explored. There are many synaptic proteins, often with several isoforms created by post-translational modifications. Complex cascades of interactions between these proteins, on either side of the synaptic cleft, are essential for normal function, normal transmitter release and postsynaptic responsiveness. Interactions between presynaptic and postsynaptic proteins that have binding domains in the synaptic cleft are proposed here to result in a local cleft structure that captures and stabilises only the appropriate subtype of GABA(A)Rs, allowing others to drift away from that synapse, either to be captured by another synapse, or internalised.
Collapse
Affiliation(s)
- Alex M Thomson
- The School of Pharmacy, London University, 29-39 Brunswick Square, London WC1N 1AX, UK.
| | | |
Collapse
|
34
|
Smith KR, Oliver PL, Lumb MJ, Arancibia-Carcamo IL, Revilla-Sanchez R, Brandon NJ, Moss SJ, Kittler JT. Identification and characterisation of a Maf1/Macoco protein complex that interacts with GABAA receptors in neurons. Mol Cell Neurosci 2010; 44:330-41. [PMID: 20417281 DOI: 10.1016/j.mcn.2010.04.004] [Citation(s) in RCA: 13] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/04/2009] [Revised: 03/09/2010] [Accepted: 04/09/2010] [Indexed: 01/16/2023] Open
Abstract
The majority of fast inhibitory synaptic transmission in the mammalian nervous system is mediated by GABA(A) receptors (GABA(A)Rs). Here we report a novel interaction between the protein Maf1 and GABA(A)R beta-subunit intracellular domains. We find Maf1 to be highly expressed in brain and enriched in the hippocampus and cortex. In heterologous cells and neurons we show Maf1 co-localises with GABA(A)Rs in intracellular compartments and at the cell surface. In neurons, Maf1 is found localised in the cytoplasm in dendrites, partially overlapping with GABA(A)Rs and inhibitory synapses and in addition is enriched in the neuronal nucleus. We also report that Maf1 interacts with a novel coiled-coil domain containing protein that we have called Macoco (for Maf1 interacting coiled-coil protein). Like Maf1, Macoco can also be found localised to inhibitory synapses and directly interacts with GABA(A)Rs. Expressing Macoco in neurons increases surface GABA(A)R levels. Our results suggest that Maf1 and Macoco are novel GABA(A)R interacting proteins important for regulating GABA(A)R surface expression and GABA(A)R signalling in the brain.
Collapse
Affiliation(s)
- Katharine R Smith
- Department of Neuroscience, Physiology and Pharmacology, University College London, Gower Street, London, WC1E 6BT, UK
| | | | | | | | | | | | | | | |
Collapse
|
35
|
Araud T, Wonnacott S, Bertrand D. Associated proteins: The universal toolbox controlling ligand gated ion channel function. Biochem Pharmacol 2010; 80:160-9. [PMID: 20346921 DOI: 10.1016/j.bcp.2010.03.017] [Citation(s) in RCA: 14] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/30/2009] [Revised: 03/02/2010] [Accepted: 03/15/2010] [Indexed: 02/06/2023]
Abstract
Ligand gated ion channels are integral multimeric membrane proteins that can detect with high sensitivity the presence of a specific transmitter in the extracellular space and transduce this signal into an ion flux. While these receptors are widely expressed in the nervous system, their expression is not limited to neurons or their postsynaptic targets but extends to non-neuronal cells where they participate in many physiological responses. Cells have developed complex regulatory mechanisms allowing for the precise control and modulation of ligand gated ion channels. In this overview the roles of accessory subunits and associated proteins in these regulatory mechanisms are reviewed and their relevance illustrated by examples at different ligand gated ion channel types, with emphasis on nicotinic acetylcholine receptors. Dysfunction of ligand gated ion channels can result in neuromuscular, neurological or psychiatric disorders. A better understanding of the precise function of associated proteins and how they impact on ligand gated ion channels will provide new therapeutic opportunities for clinical intervention.
Collapse
|
36
|
Emerging role of Alzheimer's disease-associated ubiquilin-1 in protein aggregation. Biochem Soc Trans 2010; 38:150-5. [PMID: 20074050 DOI: 10.1042/bst0380150] [Citation(s) in RCA: 34] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/08/2023]
Abstract
Abnormal protein aggregation and intracellular or extracellular accumulation of misfolded and aggregated proteins are key events in the pathogenesis of different neurodegenerative diseases. Furthermore, endoplasmic reticulum stress and impairment of the ubiquitin-proteasome system probably contribute to neurodegeneration in these diseases. A characteristic feature of AD (Alzheimer's disease) is the abnormal accumulation of Abeta (amyloid beta-peptide) in the brain. Evidence shows that the AD-associated PS (presenilin) also forms aggregates under certain conditions and that another AD-associated protein, ubiquilin-1, controls protein aggregation and deposition of aggregated proteins. Here, we review the current knowledge of ubiquilin-1 and PS in protein aggregation and related events that potentially influence neurodegeneration.
Collapse
|
37
|
Saliba RS, Gu Z, Yan Z, Moss SJ. Blocking L-type voltage-gated Ca2+ channels with dihydropyridines reduces gamma-aminobutyric acid type A receptor expression and synaptic inhibition. J Biol Chem 2009; 284:32544-50. [PMID: 19778903 DOI: 10.1074/jbc.m109.040071] [Citation(s) in RCA: 42] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/06/2022] Open
Abstract
Gamma-aminobutyric acid type A receptors (GABA(A)Rs) are the major sites of fast inhibitory neurotransmission in the brain, and the numbers of these receptors at the cell surface can determine the strength of GABAergic neurotransmission. Chronic changes in neuronal activity lead to an adaptive modulation in the efficacy of GABAergic synaptic inhibition, brought about in part by changes in the number of synaptic GABA(A)Rs, a mechanism known as homeostatic synaptic plasticity. Reduction in the number of GABA(A)Rs in response to prolonged neuronal activity blockade is dependent on the ubiquitin-proteasome system. The underlying biochemical pathways linking chronic activity blockade to proteasome-dependent degradation of GABA(A)Rs are unknown. Here, we show that chronic blockade of L-type voltage-gated calcium channels (VGCCs) with nifedipine decreases the number of GABA(A)Rs at synaptic sites but not the overall number of inhibitory synapses. In parallel, blockade of L-type VGCCs decreases the amplitude but not the frequency of miniature inhibitory postsynaptic currents or expression of the glutamic acid decarboxylase GAD65. We further reveal that the activation of L-type VGCCs regulates the turnover of newly translated GABA(A)R subunits in a mechanism dependent upon the activity of the proteasome and thus regulates GABA(A)R insertion into the plasma membrane. Together, these observations suggest that activation of L-type VGCCs can regulate the abundance of synaptic GABA(A)Rs and the efficacy of synaptic inhibition, revealing a potential mechanism underlying the homeostatic adaptation of fast GABAergic inhibition to prolonged changes in activity.
Collapse
Affiliation(s)
- Richard S Saliba
- Department of Neuroscience, Tufts University, Boston, Massachusetts 02111, USA
| | | | | | | |
Collapse
|
38
|
Abstract
GABA(A) receptors (GABA(A)Rs), the principal sites of synaptic inhibition in the brain, are dynamic entities on the neuronal cell surface, but the role their membrane trafficking plays in shaping neuronal activity remains obscure. Here, we examined this by using mutant receptor beta3 subunits (beta3S408/9A), which have reduced binding to the clathrin adaptor protein-2, a critical regulator of GABA(A)R endocytosis. Neurons expressing beta3S408/9A subunits exhibited increases in the number and size of inhibitory synapses, together with enhanced inhibitory synaptic transmission due to reduced GABA(A)R endocytosis. Furthermore, neurons expressing beta3S408/9A subunits had deficits in the number of mature spines and reduced accumulation of postsynaptic density protein-95 at excitatory synapses. This deficit in spine maturity was reversed by pharmacological blockade of GABA(A)Rs. Therefore, regulating the efficacy of synaptic inhibition by modulating GABA(A)R membrane trafficking may play a critical role in regulating spine maturity with significant implications for synaptic plasticity together with behavior.
Collapse
|
39
|
UBXD4, a UBX-containing protein, regulates the cell surface number and stability of alpha3-containing nicotinic acetylcholine receptors. J Neurosci 2009; 29:6883-96. [PMID: 19474315 DOI: 10.1523/jneurosci.4723-08.2009] [Citation(s) in RCA: 38] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/11/2023] Open
Abstract
Adaptor proteins are likely to modulate spatially and temporally the trafficking of a number of membrane proteins, including neuronal nicotinic acetylcholine receptors (nAChRs). A yeast two-hybrid screen identified a novel UBX-containing protein, UBXD4, as one of the cytosolic proteins that interact directly with the alpha3 and alpha4 nAChR subunits. The function of UBX-containing proteins is largely unknown. Immunoprecipitation and confocal microscopy confirmed the interaction of UBXD4 with alpha3-containing nAChRs (alpha3* nAChRs) expressed in HEK293 cells, PC12 cells, and rat cortical neurons. Overexpression of UBXD4 in differentiated PC12 cells (dPC12) increased nAChR cell surface expression, especially that of the alpha3beta2 subtype. These findings were corroborated by electrophysiology, immunofluorescent staining, and biotinylation of surface receptors. Silencing of UBXD4 led to a significant reduction of alpha3* nAChRs in rat cortical neurons and dPC12 cells. Biochemical and immunofluorescence studies of endogenous UBXD4 showed that the protein is located in both the ER and cis-Golgi compartments. Our investigations also showed that the alpha3 subunit is ubiquitinated and that UBXD4 can interfere with its ubiquitination and consequent degradation by the proteasome. Our data suggest that UBXD4 modulates the distribution of alpha3* nAChRs between specialized intracellular compartments and the plasma membrane. This effect is achieved by controlling the stability of the alpha3 subunit and, consequently, the number of receptors at the cell surface.
Collapse
|
40
|
Ubiquitin-like and ubiquitin-associated domain proteins: significance in proteasomal degradation. Cell Mol Life Sci 2009; 66:2819-33. [PMID: 19468686 DOI: 10.1007/s00018-009-0048-9] [Citation(s) in RCA: 118] [Impact Index Per Article: 7.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/03/2009] [Revised: 04/14/2009] [Accepted: 04/29/2009] [Indexed: 10/20/2022]
Abstract
The ubiquitin-proteasome pathway of protein degradation is one of the major mechanisms that are involved in the maintenance of the proper levels of cellular proteins. The regulation of proteasomal degradation thus ensures proper cell functions. The family of proteins containing ubiquitin-like (UbL) and ubiquitin-associated (UBA) domains has been implicated in proteasomal degradation. UbL-UBA domain containing proteins associate with substrates destined for degradation as well as with subunits of the proteasome, thus regulating the proper turnover of proteins.
Collapse
|
41
|
Pless SA, Lynch JW. Distinct conformational changes in activated agonist-bound and agonist-free glycine receptor subunits. J Neurochem 2009; 108:1585-94. [DOI: 10.1111/j.1471-4159.2009.05930.x] [Citation(s) in RCA: 12] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/30/2022]
|