Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 167
Filter
Add more filters

Country/Region as subject
Publication year range
1.
J Biol Chem ; 298(5): 101933, 2022 05.
Article in English | MEDLINE | ID: mdl-35427648

ABSTRACT

Hyperammonemia is known to cause various neurological dysfunctions such as seizures and cognitive impairment. Several studies have suggested that hyperammonemia may also be linked to the development of Alzheimer's disease (AD). However, the direct evidence for a role of ammonia in the pathophysiology of AD remains to be discovered. Herein, we report that hyperammonemia increases the amount of mature amyloid precursor protein (mAPP) in astrocytes, the largest and most prevalent type of glial cells in the central nervous system that are capable of metabolizing glutamate and ammonia, and promotes amyloid beta (Aß) production. We demonstrate the accumulation of mAPP in astrocytes was primarily due to enhanced endocytosis of mAPP from the plasma membrane. A large proportion of internalized mAPP was targeted not to the lysosome, but to the endoplasmic reticulum, where processing enzymes ß-secretase BACE1 (beta-site APP cleaving enzyme 1) and γ-secretase presenilin-1 are expressed, and mAPP is cleaved to produce Aß. Finally, we show the ammonia-induced production of Aß in astrocytic endoplasmic reticulum was specific to Aß42, a principal component of senile plaques in AD patients. Our studies uncover a novel mechanism of Aß42 production in astrocytes and also provide the first evidence that ammonia induces the pathogenesis of AD by regulating astrocyte function.


Subject(s)
Alzheimer Disease , Ammonia , Amyloid beta-Peptides , Astrocytes , Hyperammonemia , Alzheimer Disease/physiopathology , Ammonia/metabolism , Amyloid Precursor Protein Secretases/genetics , Amyloid Precursor Protein Secretases/metabolism , Amyloid beta-Peptides/metabolism , Amyloid beta-Protein Precursor/metabolism , Aspartic Acid Endopeptidases/genetics , Aspartic Acid Endopeptidases/metabolism , Astrocytes/pathology , Endoplasmic Reticulum/metabolism , Humans , Hyperammonemia/metabolism
2.
Mol Psychiatry ; 27(3): 1729-1741, 2022 03.
Article in English | MEDLINE | ID: mdl-35169261

ABSTRACT

Intellectual disability (ID) is a common neurodevelopmental disorder that can arise from genetic mutations ranging from trisomy to single nucleotide polymorphism. Mutations in a growing number of single genes have been identified as causative in ID, including ARHGEF9. Evaluation of 41 ARHGEF9 patient reports shows ubiquitous inclusion of ID, along with other frequently reported symptoms of epilepsy, abnormal baseline EEG activity, behavioral symptoms, and sleep disturbances. ARHGEF9 codes for the Cdc42 Guanine Nucleotide Exchange Factor 9 collybistin (Cb), a known regulator of inhibitory synapse function via direct interaction with the adhesion molecule neuroligin-2 and the α2 subunit of GABAA receptors. We mutate the Cb binding motif within the large intracellular loop of α2 replacing it with the binding motif for gephyrin from the α1 subunit (Gabra2-1). The Gabra2-1 mutation causes a strong downregulation of Cb expression, particularly at cholecystokinin basket cell inhibitory synapses. Gabra2-1 mice have deficits in working and recognition memory, as well as hyperactivity, anxiety, and reduced social preference, recapitulating the frequently reported features of ARHGEF9 patients. Gabra2-1 mice also have spontaneous seizures during postnatal development which can lead to mortality, and baseline abnormalities in low-frequency wavelengths of the EEG. EEG abnormalities are vigilance state-specific and manifest as sleep disturbance including increased time in wake and a loss of free-running rhythmicity in the absence of light as zeitgeber. Gabra2-1 mice phenocopy multiple features of human ARHGEF9 mutation, and reveal α2 subunit-containing GABAA receptors as a druggable target for treatment of this complex ID syndrome.


Subject(s)
Intellectual Disability , Mutation , Receptors, GABA-A , Rho Guanine Nucleotide Exchange Factors , Animals , Humans , Intellectual Disability/genetics , Mice , Receptors, GABA-A/genetics , Receptors, GABA-A/metabolism , Rho Guanine Nucleotide Exchange Factors/genetics , Rho Guanine Nucleotide Exchange Factors/metabolism , Syndrome , gamma-Aminobutyric Acid/genetics , gamma-Aminobutyric Acid/metabolism
3.
Brain ; 145(3): 950-963, 2022 04 29.
Article in English | MEDLINE | ID: mdl-34528073

ABSTRACT

First-in-line benzodiazepine treatment fails to terminate seizures in about 30% of epilepsy patients, highlighting a need for novel anti-seizure strategies. It is emerging that impaired K+/Cl- cotransporter 2 (KCC2) activity leads to deficits in GABAergic inhibition and increased seizure vulnerability in patients. In neurons, the with-no-lysine (WNK) kinase-STE20/SPS1-related proline/alanine-rich (SPAK) kinase signalling pathway inhibits KCC2 activity via T1007 phosphorylation. Here, we exploit the selective WNK kinase inhibitor WNK463 to test the effects of pharmacological WNK inhibition on KCC2 function, GABAergic inhibition, and epileptiform activity. Immunoprecipitation and western blotting analysis revealed that WNK463 reduces KCC2-T1007 phosphorylation in vitro and in vivo. Using patch-clamp recordings in primary rat neurons, we further observed that WNK463 hyperpolarized the Cl- reversal potential, and enhanced KCC2-mediated Cl- extrusion. In the 4-aminopyridine slice model of acute seizures, WNK463 administration reduced the frequency and number of seizure-like events. In vivo, C57BL/6 mice that received intrahippocampal WNK463 experienced delayed onset of kainic acid-induced status epilepticus, less epileptiform EEG activity, and did not develop pharmaco-resistance to diazepam. Our findings demonstrate that acute WNK463 treatment potentiates KCC2 activity in neurons and limits seizure burden in two well-established models of seizures and epilepsy. In summary, our work suggests that agents which act to increase KCC2 activity may be useful adjunct therapeutics to alleviate diazepam-resistant status epilepticus.


Subject(s)
Epilepsy , Status Epilepticus , Symporters , Animals , Diazepam/metabolism , Diazepam/pharmacology , Hippocampus/metabolism , Humans , Lysine/metabolism , Mice , Mice, Inbred C57BL , Rats , Status Epilepticus/chemically induced , Status Epilepticus/drug therapy , Status Epilepticus/metabolism , Symporters/metabolism
4.
Proc Natl Acad Sci U S A ; 117(1): 677-688, 2020 01 07.
Article in English | MEDLINE | ID: mdl-31871190

ABSTRACT

A robust body of evidence supports the concept that phosphodiesterase 10A (PDE10A) activity in the basal ganglia orchestrates the control of coordinated movement in human subjects. Although human mutations in the PDE10A gene manifest in hyperkinetic movement disorders that phenocopy many features of early Huntington's disease, characterization of the maladapted molecular mechanisms and aberrant signaling processes that underpin these conditions remains scarce. Recessive mutations in the GAF-A domain have been shown to impair PDE10A function due to the loss of striatal PDE10A protein levels, but here we show that this paucity is caused by irregular intracellular trafficking and increased PDE10A degradation in the cytosolic compartment. In contrast to GAF-A mutants, dominant mutations in the GAF-B domain of PDE10A induce PDE10A misfolding, a common pathological phenotype in many neurodegenerative diseases. These data demonstrate that the function of striatal PDE10A is compromised in disorders where disease-associated mutations trigger a reduction in the fidelity of PDE compartmentalization.


Subject(s)
Cell Membrane/metabolism , Huntington Disease/genetics , Neurons/enzymology , Phosphoric Diester Hydrolases/genetics , Protein Domains/genetics , Animals , Autophagy/genetics , Corpus Striatum/cytology , Corpus Striatum/pathology , Cyclic AMP/metabolism , Embryo, Mammalian , HEK293 Cells , Humans , Huntington Disease/pathology , Hydrolysis , Isoenzymes/genetics , Isoenzymes/metabolism , Mutation , Neurons/cytology , Patch-Clamp Techniques , Phosphoric Diester Hydrolases/metabolism , Primary Cell Culture , Proteolysis , Rats , Recombinant Proteins/genetics , Recombinant Proteins/metabolism
5.
J Biol Chem ; 296: 100364, 2021.
Article in English | MEDLINE | ID: mdl-33539918

ABSTRACT

The K+/Cl- cotransporter KCC2 (SLC12A5) allows mature neurons in the CNS to maintain low intracellular Cl- levels that are critical in mediating fast hyperpolarizing synaptic inhibition via type A γ-aminobutyric acid receptors (GABAARs). In accordance with this, compromised KCC2 activity results in seizures, but whether such deficits directly contribute to the subsequent changes in neuronal structure and viability that lead to epileptogenesis remains to be assessed. Canonical hyperpolarizing GABAAR currents develop postnatally, which reflect a progressive increase in KCC2 expression levels and activity. To investigate the role that KCC2 plays in regulating neuronal viability and architecture, we have conditionally ablated KCC2 expression in developing and mature neurons. Decreasing KCC2 expression in mature neurons resulted in the rapid activation of the extrinsic apoptotic pathway. Intriguingly, direct pharmacological inhibition of KCC2 in mature neurons was sufficient to rapidly induce apoptosis, an effect that was not abrogated via blockade of neuronal depolarization using tetrodotoxin (TTX). In contrast, ablating KCC2 expression in immature neurons had no discernable effects on their subsequent development, arborization, or dendritic structure. However, removing KCC2 in immature neurons was sufficient to ablate the subsequent postnatal development of hyperpolarizing GABAAR currents. Collectively, our results demonstrate that KCC2 plays a critical role in neuronal survival by limiting apoptosis, and mature neurons are highly sensitive to the loss of KCC2 function. In contrast, KCC2 appears to play a minimal role in mediating neuronal development or architecture.


Subject(s)
Neurons/metabolism , Symporters/metabolism , Animals , Apoptosis , Chlorides/metabolism , Female , Male , Mice , Mice, Inbred C57BL , Mice, Knockout , Neurogenesis/drug effects , Neurons/physiology , Potassium/metabolism , Primary Cell Culture , Receptors, GABA/metabolism , Seizures , Symporters/physiology , gamma-Aminobutyric Acid/metabolism , K Cl- Cotransporters
6.
J Neurosci ; 40(14): 2808-2816, 2020 04 01.
Article in English | MEDLINE | ID: mdl-32111696

ABSTRACT

Addictive drugs usurp the brain's intrinsic mechanism for reward, leading to compulsive and destructive behaviors. In the ventral tegmental area (VTA), the center of the brain's reward circuit, GABAergic neurons control the excitability of dopamine (DA) projection neurons and are the site of initial psychostimulant-dependent changes in signaling. Previous work established that cocaine/methamphetamine exposure increases protein phosphatase 2A (PP2A) activity, which dephosphorylates the GABABR2 subunit, promotes internalization of the GABAB receptor (GABABR) and leads to smaller GABABR-activated G-protein-gated inwardly rectifying potassium (GIRK) currents in VTA GABA neurons. How the actions of PP2A become selective for a particular signaling pathway is poorly understood. Here, we demonstrate that PP2A can associate directly with a short peptide sequence in the C terminal domain of the GABABR1 subunit, and that GABABRs and PP2A are in close proximity in rodent neurons (mouse/rat; mixed sexes). We show that this PP2A-GABABR interaction can be regulated by intracellular Ca2+ Finally, a peptide that potentially reduces recruitment of PP2A to GABABRs and thereby limits receptor dephosphorylation increases the magnitude of baclofen-induced GIRK currents. Thus, limiting PP2A-dependent dephosphorylation of GABABRs may be a useful strategy to increase receptor signaling for treating diseases.SIGNIFICANCE STATEMENT Dysregulation of GABAB receptors (GABABRs) underlies altered neurotransmission in many neurological disorders. Protein phosphatase 2A (PP2A) is involved in dephosphorylating and subsequent internalization of GABABRs in models of addiction and depression. Here, we provide new evidence that PP2A B55 regulatory subunit interacts directly with a small region of the C-terminal domain of the GABABR1 subunit, and that this interaction is sensitive to intracellular Ca2+ We demonstrate that a short peptide corresponding to the PP2A interaction site on GABABR1 competes for PP2A binding, enhances phosphorylation GABABR2 S783, and affects functional signaling through GIRK channels. Our study highlights how targeting PP2A dependent dephosphorylation of GABABRs may provide a specific strategy to modulate GABABR signaling in disease conditions.


Subject(s)
Neurons/metabolism , Protein Phosphatase 2/metabolism , Receptors, GABA-B/metabolism , Signal Transduction/physiology , Animals , Brain/metabolism , Female , G Protein-Coupled Inwardly-Rectifying Potassium Channels/metabolism , HEK293 Cells , Humans , Male , Mice , Mice, Inbred C57BL , Phosphorylation , Rats , Synaptic Transmission/physiology
7.
J Biol Chem ; 295(35): 12330-12342, 2020 08 28.
Article in English | MEDLINE | ID: mdl-32620552

ABSTRACT

GABA type A receptors (GABAARs) mediate fast synaptic inhibition and are trafficked to functionally diverse synapses. However, the precise molecular mechanisms that regulate the synaptic targeting of these receptors are unclear. Whereas it has been previously shown that phosphorylation events in α4, ß, and γ subunits of GABAARs govern their function and trafficking, phosphorylation of other subunits has not yet been demonstrated. Here, we show that the α2 subunit of GABAARs is phosphorylated at Ser-359 and enables dynamic regulation of GABAAR binding to the scaffolding proteins gephyrin and collybistin. We initially identified Ser-359 phosphorylation by MS analysis, and additional experiments revealed that it is regulated by the activities of cAMP-dependent protein kinase (PKA) and the protein phosphatase 1 (PP1) and/or PP2A. GST-based pulldowns and coimmunoprecipitation experiments demonstrate preferential binding of both gephyrin and collybistin to WT and an S359A phosphonull variant, but not to an S359D phosphomimetic variant. Furthermore, the decreased capacity of the α2 S359D variant to bind collybistin and gephyrin decreased the density of synaptic α2-containing GABAAR clusters and caused an absence of α2 enrichment in the axon initial segment. These results suggest that PKA-mediated phosphorylation and PP1/PP2A-dependent dephosphorylation of the α2 subunit play a role in the dynamic regulation of GABAAR accumulation at inhibitory synapses, thereby regulating the strength of synaptic inhibition. The MS data have been deposited to ProteomeXchange, with the data set identifier PXD019597.


Subject(s)
Down-Regulation , Inhibitory Postsynaptic Potentials , Receptors, GABA-A/metabolism , Synapses/metabolism , Amino Acid Substitution , Animals , Cyclic AMP-Dependent Protein Kinases/genetics , Cyclic AMP-Dependent Protein Kinases/metabolism , Membrane Proteins/genetics , Membrane Proteins/metabolism , Mice , Mice, Transgenic , Mutation, Missense , Phosphorylation , Protein Phosphatase 2/genetics , Protein Phosphatase 2/metabolism , Rats , Rats, Wistar , Receptors, GABA-A/genetics , Rho Guanine Nucleotide Exchange Factors/genetics , Rho Guanine Nucleotide Exchange Factors/metabolism , Synapses/genetics
8.
Proc Natl Acad Sci U S A ; 115(40): 10166-10171, 2018 10 02.
Article in English | MEDLINE | ID: mdl-30224498

ABSTRACT

The type 2 K+/Cl- cotransporter (KCC2) allows neurons to maintain low intracellular levels of Cl-, a prerequisite for efficient synaptic inhibition. Reductions in KCC2 activity are evident in epilepsy; however, whether these deficits directly contribute to the underlying pathophysiology remains controversial. To address this issue, we created knock-in mice in which threonines 906 and 1007 within KCC2 have been mutated to alanines (KCC2-T906A/T1007A), which prevents its phospho-dependent inactivation. The respective mice appeared normal and did not show any overt phenotypes, and basal neuronal excitability was unaffected. KCC2-T906A/T1007A mice exhibited increased basal neuronal Cl- extrusion, without altering total or plasma membrane accumulation of KCC2. Critically, activity-induced deficits in synaptic inhibition were reduced in the mutant mice. Consistent with this, enhanced KCC2 was sufficient to limit chemoconvulsant-induced epileptiform activity. Furthermore, this increase in KCC2 function mitigated induction of aberrant high-frequency activity during seizures, highlighting depolarizing GABA as a key contributor to the pathological neuronal synchronization seen in epilepsy. Thus, our results demonstrate that potentiating KCC2 represents a therapeutic strategy to alleviate seizures.


Subject(s)
Epilepsy/metabolism , Neurons/metabolism , Seizures/metabolism , Symporters/metabolism , Synaptic Membranes/metabolism , gamma-Aminobutyric Acid/metabolism , Amino Acid Substitution , Animals , Epilepsy/genetics , Epilepsy/pathology , Gene Knock-In Techniques , Mice , Mutation, Missense , Neurons/pathology , Seizures/genetics , Seizures/pathology , Symporters/genetics , Synaptic Membranes/genetics , Synaptic Membranes/pathology , gamma-Aminobutyric Acid/genetics , K Cl- Cotransporters
9.
Proc Natl Acad Sci U S A ; 115(40): E9479-E9488, 2018 10 02.
Article in English | MEDLINE | ID: mdl-30228121

ABSTRACT

GABABR-dependent activation of G protein-gated inwardly rectifying potassium channels (GIRK or KIR3) provides a well-known source of inhibition in the brain, but the details on how this important inhibitory pathway affects neural circuits are lacking. We used sorting nexin 27 (SNX27), an endosomal adaptor protein that associates with GIRK2c and GIRK3 subunits, to probe the role of GIRK channels in reward circuits. A conditional knockout of SNX27 in both substantia nigra pars compacta and ventral tegmental area (VTA) dopamine neurons leads to markedly smaller GABABR- and dopamine D2R-activated GIRK currents, as well as to suprasensitivity to cocaine-induced locomotor sensitization. Expression of the SNX27-insensitive GIRK2a subunit in SNX27-deficient VTA dopamine neurons restored GIRK currents and GABABR-dependent inhibition of spike firing, while also resetting the mouse's sensitivity to cocaine-dependent sensitization. These results establish a link between slow inhibition mediated by GIRK channels in VTA dopamine neurons and cocaine addiction, revealing a therapeutic target for treating addiction.


Subject(s)
Cocaine-Related Disorders/metabolism , Cocaine/toxicity , Dopaminergic Neurons/metabolism , G Protein-Coupled Inwardly-Rectifying Potassium Channels/metabolism , Locomotion/drug effects , Animals , Cocaine-Related Disorders/genetics , Cocaine-Related Disorders/pathology , Dopaminergic Neurons/pathology , G Protein-Coupled Inwardly-Rectifying Potassium Channels/genetics , Mice , Mice, Knockout , Receptors, Dopamine D2/genetics , Receptors, Dopamine D2/metabolism , Sorting Nexins/genetics , Sorting Nexins/metabolism
10.
J Neurosci ; 39(19): 3611-3626, 2019 05 08.
Article in English | MEDLINE | ID: mdl-30846615

ABSTRACT

Developing cortical GABAergic interneurons rely on genetic programs, neuronal activity, and environmental cues to construct inhibitory circuits during early postnatal development. Disruption of these events can cause long-term changes in cortical inhibition and may be involved in neurological disorders associated with inhibitory circuit dysfunction. We hypothesized that tonic glutamate signaling in the neonatal cortex contributes to, and is necessary for, the maturation of cortical interneurons. To test this hypothesis, we used mice of both sexes to quantify extracellular glutamate concentrations in the cortex during development, measure ambient glutamate-mediated activation of developing cortical interneurons, and manipulate tonic glutamate signaling using subtype-specific NMDA receptor antagonists in vitro and in vivo We report that ambient glutamate levels are high (≈100 nm) in the neonatal cortex and decrease (to ≈50 nm) during the first weeks of life, coincident with increases in astrocytic glutamate uptake. Consistent with elevated ambient glutamate, putative parvalbumin-positive interneurons in the cortex (identified using G42:GAD1-eGFP reporter mice) exhibit a transient, tonic NMDA current at the end of the first postnatal week. GluN2C/GluN2D-containing NMDA receptors mediate the majority of this current and contribute to the resting membrane potential and intrinsic properties of developing putative parvalbumin interneurons. Pharmacological blockade of GluN2C/GluN2D-containing NMDA receptors in vivo during the period of tonic interneuron activation, but not later, leads to lasting decreases in interneuron morphological complexity and causes deficits in cortical inhibition later in life. These results demonstrate that dynamic ambient glutamate signaling contributes to cortical interneuron maturation via tonic activation of GluN2C/GluN2D-containing NMDA receptors.SIGNIFICANCE STATEMENT Inhibitory GABAergic interneurons make up 20% of cortical neurons and are critical to controlling cortical network activity. Dysfunction of cortical inhibition is associated with multiple neurological disorders, including epilepsy. Establishing inhibitory cortical networks requires in utero proliferation, differentiation, and migration of immature GABAergic interneurons, and subsequent postnatal morphological maturation and circuit integration. Here, we demonstrate that ambient glutamate provides tonic activation of immature, putative parvalbumin-positive GABAergic interneurons in the neonatal cortex via high-affinity NMDA receptors. When this activation is blocked, GABAergic interneuron maturation is disrupted, and cortical networks exhibit lasting abnormal hyperexcitability. We conclude that temporally precise activation of developing cortical interneurons by ambient glutamate is critically important for establishing normal cortical inhibition.


Subject(s)
Glutamic Acid/metabolism , Interneurons/metabolism , Receptors, N-Methyl-D-Aspartate/metabolism , Sensorimotor Cortex/metabolism , Animals , Animals, Newborn , Dose-Response Relationship, Drug , Excitatory Amino Acid Antagonists/pharmacology , Extracellular Fluid/drug effects , Extracellular Fluid/metabolism , Female , Interneurons/drug effects , Male , Mice , Mice, Inbred C57BL , Mice, Transgenic , Organ Culture Techniques , Receptors, N-Methyl-D-Aspartate/agonists , Receptors, N-Methyl-D-Aspartate/antagonists & inhibitors , Sensorimotor Cortex/drug effects
11.
J Biol Chem ; 294(32): 12220-12230, 2019 08 09.
Article in English | MEDLINE | ID: mdl-31239352

ABSTRACT

Neuroactive steroids (NASs) are synthesized within the brain and exert profound effects on behavior. These effects are primarily believed to arise from the activities of NASs as positive allosteric modulators (PAMs) of the GABA-type A receptor (GABAAR). NASs also activate a family of G protein-coupled receptors known as membrane progesterone receptors (mPRs). Here, using surface-biotinylation assays and electrophysiology techniques, we examined mPRs' role in mediating the effects of NAS on the efficacy of GABAergic inhibition. Selective mPR activation enhanced phosphorylation of Ser-408 and Ser-409 (Ser-408/9) within the GABAAR ß3 subunit, which depended on the activity of cAMP-dependent protein kinase A (PKA) and protein kinase C (PKC). mPR activation did not directly modify GABAAR activity and had no acute effects on phasic or tonic inhibition. Instead, mPR activation induced a sustained elevation in tonic current, which was blocked by PKA and PKC inhibition. Substitution of Ser-408/9 to alanine residues also prevented the effects of mPR activation on tonic current. Furthermore, this substitution abolished the effects of sustained NAS exposure on tonic inhibition. Interestingly, the allosteric effects of NAS on GABAergic inhibition were independent of Ser-408/9 in the ß3 subunit. Additionally, although allosteric effects of NAS on GABAergic inhibition were sensitive to a recently developed "NAS antagonist," the sustained effects of NAS on tonic inhibition were not. We conclude that metabotropic effects of NAS on GABAergic inhibition are mediated by mPR-dependent modulation of GABAAR phosphorylation. We propose that this mechanism may contribute to the varying behavioral effects of NAS.


Subject(s)
Neurosteroids/metabolism , Receptors, GABA-A/metabolism , Allosteric Regulation/drug effects , Animals , Cell Membrane/metabolism , Evoked Potentials/drug effects , GABA-A Receptor Antagonists/pharmacology , HEK293 Cells , Hippocampus/metabolism , Humans , Male , Mice , Mice, Inbred C57BL , Mutagenesis, Site-Directed , Neurosteroids/pharmacology , Phosphorylation/drug effects , Protein Kinase C/metabolism , Protein Subunits/chemistry , Protein Subunits/genetics , Protein Subunits/metabolism , Receptors, GABA-A/chemistry , Receptors, GABA-A/genetics , Receptors, Progesterone/agonists , Receptors, Progesterone/genetics , Receptors, Progesterone/metabolism
12.
Proc Natl Acad Sci U S A ; 114(44): 11763-11768, 2017 10 31.
Article in English | MEDLINE | ID: mdl-29078280

ABSTRACT

Estrogen plays a critical role in many physiological processes and exerts profound effects on behavior by regulating neuronal excitability. While estrogen has been established to exert effects on dendritic morphology and excitatory neurotransmission its role in regulating neuronal inhibition is poorly understood. Fast synaptic inhibition in the adult brain is mediated by specialized populations of γ-c aA receptors (GABAARs) that are selectively enriched at synapses, a process dependent upon their interaction with the inhibitory scaffold protein gephyrin. Here we have assessed the role that estradiol (E2) plays in regulating the dynamics of GABAARs and stability of inhibitory synapses. Treatment of cultured cortical neurons with E2 reduced the accumulation of GABAARs and gephyrin at inhibitory synapses. However, E2 exposure did not modify the expression of either the total or the plasma membrane GABAARs or gephyrin. Mechanistically, single-particle tracking revealed that E2 treatment selectively reduced the dwell time and thereby decreased the confinement of GABAARs at inhibitory synapses. Consistent with our cell biology measurements, we observed a significant reduction in amplitude of inhibitory synaptic currents in both cultured neurons and hippocampal slices exposed to E2, while their frequency was unaffected. Collectively, our results suggest that acute exposure of neurons to E2 leads to destabilization of GABAARs and gephyrin at inhibitory synapses, leading to reductions in the efficacy of GABAergic inhibition via a postsynaptic mechanism.


Subject(s)
Estradiol/pharmacology , Neural Inhibition/drug effects , Receptors, GABA-A/metabolism , Synapses/drug effects , Synapses/metabolism , Animals , Carrier Proteins/pharmacology , Cell Membrane/drug effects , Cell Membrane/metabolism , Cells, Cultured , Female , Hippocampus/drug effects , Hippocampus/metabolism , Male , Membrane Proteins/pharmacology , Mice , Mice, Inbred C57BL , Neurons/drug effects , Neurons/metabolism , Rats , Synaptic Transmission/drug effects
13.
J Lipid Res ; 59(5): 830-842, 2018 05.
Article in English | MEDLINE | ID: mdl-29563219

ABSTRACT

apoE is the primary lipid carrier within the CNS and the strongest genetic risk factor for late onset Alzheimer's disease (AD). apoE is primarily lipidated via ABCA1, and both are under transcriptional regulation by the nuclear liver X receptor (LXR). Considerable evidence from genetic (using ABCA1 overexpression) and pharmacological (using synthetic LXR agonists) studies in AD mouse models suggests that increased levels of lipidated apoE can improve cognitive performance and, in some strains, can reduce amyloid burden. However, direct synthetic LXR ligands have hepatotoxic side effects that limit their clinical use. Here, we describe a set of small molecules, previously annotated as antagonists of the purinergic receptor, P2X7, which enhance ABCA1 expression and activity as well as apoE secretion, and are not direct LXR ligands. Furthermore, P2X7 is not required for these molecules to induce ABCA1 upregulation and apoE secretion, demonstrating that the ABCA1 and apoE effects are mechanistically independent of P2X7 inhibition. Hence, we have identified novel dual activity compounds that upregulate ABCA1 across multiple CNS cell types, including human astrocytes, pericytes, and microglia, through an indirect LXR mechanism and that also independently inhibit P2X7 receptor activity.


Subject(s)
ATP Binding Cassette Transporter 1/agonists , Apolipoproteins E/agonists , Purinergic P2X Receptor Antagonists/pharmacology , Receptors, Purinergic P2X7/metabolism , Small Molecule Libraries/pharmacology , ATP Binding Cassette Transporter 1/metabolism , Adamantane/analogs & derivatives , Adamantane/chemistry , Adamantane/pharmacology , Animals , Apolipoproteins E/metabolism , Aziridines/chemistry , Aziridines/pharmacology , Benzamides/chemistry , Benzamides/pharmacology , Cells, Cultured , Humans , Mice , Mice, Knockout , Molecular Structure , Naphthoquinones/chemistry , Naphthoquinones/pharmacology , Purinergic P2X Receptor Antagonists/chemistry , Receptors, Purinergic P2X7/deficiency , Small Molecule Libraries/chemistry , Sulfonamides/chemistry , Sulfonamides/pharmacology , Up-Regulation/drug effects
14.
J Biol Chem ; 292(16): 6621-6632, 2017 04 21.
Article in English | MEDLINE | ID: mdl-28213518

ABSTRACT

The behavioral and anatomical deficits seen in fragile X syndrome (FXS) are widely believed to result from imbalances in the relative strengths of excitatory and inhibitory neurotransmission. Although modified neuronal excitability is thought to be of significance, the contribution that alterations in GABAergic inhibition play in the pathophysiology of FXS are ill defined. Slow sustained neuronal inhibition is mediated by γ-aminobutyric acid type B (GABAB) receptors, which are heterodimeric G-protein-coupled receptors constructed from R1a and R2 or R1b and R2 subunits. Via the activation of Gi/o, they limit cAMP accumulation, diminish neurotransmitter release, and induce neuronal hyperpolarization. Here we reveal that selective deficits in R1a subunit expression are seen in Fmr1 knock-out mice (KO) mice, a widely used animal model of FXS, but the levels of the respective mRNAs were unaffected. Similar trends of R1a expression were seen in a subset of FXS patients. GABAB receptors (GABABRs) exert powerful pre- and postsynaptic inhibitory effects on neurotransmission. R1a-containing GABABRs are believed to mediate presynaptic inhibition in principal neurons. In accordance with this result, deficits in the ability of GABABRs to suppress glutamate release were seen in Fmr1-KO mice. In contrast, the ability of GABABRs to suppress GABA release and induce postsynaptic hyperpolarization was unaffected. Significantly, this deficit contributes to the pathophysiology of FXS as the GABABR agonist (R)-baclofen rescued the imbalances between excitatory and inhibitory neurotransmission evident in Fmr1-KO mice. Collectively, our results provided evidence that selective deficits in the activity of presynaptic GABABRs contribute to the pathophysiology of FXS.


Subject(s)
Fragile X Mental Retardation Protein/genetics , Fragile X Syndrome/genetics , Receptors, GABA-B/metabolism , Aged , Aged, 80 and over , Animals , Behavior, Animal , Disease Models, Animal , Fragile X Syndrome/metabolism , Glutamic Acid/metabolism , Hippocampus/metabolism , Hippocampus/physiopathology , Humans , Male , Mice , Mice, Inbred C57BL , Mice, Knockout , Neurons/metabolism , Phosphorylation , RNA, Messenger/metabolism , Receptors, GABA-B/genetics , Serine/chemistry , Signal Transduction , Synaptic Transmission , gamma-Aminobutyric Acid/metabolism
15.
J Biol Chem ; 292(52): 21253-21263, 2017 12 29.
Article in English | MEDLINE | ID: mdl-29092909

ABSTRACT

K+/Cl- cotransporter 2 (KCC2) is selectively expressed in the adult nervous system and allows neurons to maintain low intracellular Cl- levels. Thus, KCC2 activity is an essential prerequisite for fast hyperpolarizing synaptic inhibition mediated by type A γ-aminobutyric acid (GABAA) receptors, which are Cl--permeable, ligand-gated ion channels. Consistent with this, deficits in the activity of KCC2 lead to epilepsy and are also implicated in neurodevelopmental disorders, neuropathic pain, and schizophrenia. Accordingly, there is significant interest in developing activators of KCC2 as therapeutic agents. To provide insights into the cellular processes that determine KCC2 activity, we have investigated the mechanism by which N-ethylmaleimide (NEM) enhances transporter activity using a combination of biochemical and electrophysiological approaches. Our results revealed that, within 15 min, NEM increased cell surface levels of KCC2 and modulated the phosphorylation of key regulatory residues within the large cytoplasmic domain of KCC2 in neurons. More specifically, NEM increased the phosphorylation of serine 940 (Ser-940), whereas it decreased phosphorylation of threonine 1007 (Thr-1007). NEM also reduced with no lysine (WNK) kinase phosphorylation of Ste20-related proline/alanine-rich kinase (SPAK), a kinase that directly phosphorylates KCC2 at residue Thr-1007. Mutational analysis revealed that Thr-1007 dephosphorylation mediated the effects of NEM on KCC2 activity. Collectively, our results suggest that compounds that either increase the surface stability of KCC2 or reduce Thr-1007 phosphorylation may be of use as enhancers of KCC2 activity.


Subject(s)
Ethylmaleimide/metabolism , Symporters/metabolism , Animals , Cell Membrane/metabolism , Embryo, Mammalian , Humans , Membrane Transport Modulators/metabolism , Neurons/metabolism , Phosphorylation/physiology , Rats , Rats, Sprague-Dawley , Receptors, GABA/metabolism , Symporters/physiology , K Cl- Cotransporters
16.
Physiol Rev ; 91(3): 1009-22, 2011 Jul.
Article in English | MEDLINE | ID: mdl-21742794

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.


Subject(s)
Neural Inhibition/physiology , Neuronal Plasticity/physiology , Receptors, GABA-A/metabolism , Synapses/physiology , Animals , Cell Membrane/metabolism , Endocytosis , Exocytosis , Humans , Mammals , Molecular Structure , Neurons/metabolism , Protein Processing, Post-Translational , Protein Transport , Receptors, GABA-A/chemistry
17.
Proc Natl Acad Sci U S A ; 112(48): 14805-10, 2015 Dec 01.
Article in English | MEDLINE | ID: mdl-26627235

ABSTRACT

Alterations in the efficacy of neuronal inhibition mediated by GABAA receptors (GABAARs) containing ß3 subunits are continually implicated in autism spectrum disorders (ASDs). In vitro, the plasma membrane stability of GABAARs is potentiated via phosphorylation of serine residues 408 and 409 (S408/9) in the ß3 subunit, an effect that is mimicked by their mutation to alanines. To assess if modifications in ß3 subunit expression contribute to ASDs, we have created a mouse in which S408/9 have been mutated to alanines (S408/9A). S408/9A homozygotes exhibited increased phasic, but decreased tonic, inhibition, events that correlated with alterations in the membrane stability and synaptic accumulation of the receptor subtypes that mediate these distinct forms of inhibition. S408/9A mice exhibited alterations in dendritic spine structure, increased repetitive behavior, and decreased social interaction, hallmarks of ASDs. ASDs are frequently comorbid with epilepsy, and consistent with this comorbidity, S408/9A mice exhibited a marked increase in sensitivity to seizures induced by the convulsant kainic acid. To assess the relevance of our studies using S408/9A mice for the pathophysiology of ASDs, we measured S408/9 phosphorylation in Fmr1 KO mice, a model of fragile X syndrome, the most common monogenetic cause of ASDs. Phosphorylation of S408/9 was selectively and significantly enhanced in Fmr1 KO mice. Collectively, our results suggest that alterations in phosphorylation and/or activity of ß3-containing GABAARs may directly contribute to the pathophysiology of ASDs.


Subject(s)
Autism Spectrum Disorder/genetics , Autism Spectrum Disorder/metabolism , Gene Expression Regulation , Receptors, GABA-A/genetics , Alanine/genetics , Animals , Behavior, Animal , Biotinylation , Cell Membrane/metabolism , Dendritic Spines/metabolism , Disease Models, Animal , Electroencephalography , Electrophysiological Phenomena , Epilepsy/complications , Fear , Gene Knock-In Techniques , Hippocampus/metabolism , Male , Mice , Mice, Inbred C57BL , Mutation , Phenotype , Phosphorylation , Serine/genetics , Social Behavior , Synapses/metabolism , gamma-Aminobutyric Acid/metabolism
18.
Proc Natl Acad Sci U S A ; 112(11): 3523-8, 2015 Mar 17.
Article in English | MEDLINE | ID: mdl-25733865

ABSTRACT

The K(+)/Cl(-) cotransporter (KCC2) allows adult neurons to maintain low intracellular Cl(-) levels, which are a prerequisite for efficient synaptic inhibition upon activation of γ-aminobutyric acid receptors. Deficits in KCC2 activity are implicated in epileptogenesis, but how increased neuronal activity leads to transporter inactivation is ill defined. In vitro, the activity of KCC2 is potentiated via phosphorylation of serine 940 (S940). Here we have examined the role this putative regulatory process plays in determining KCC2 activity during status epilepticus (SE) using knockin mice in which S940 is mutated to an alanine (S940A). In wild-type mice, SE induced by kainate resulted in dephosphorylation of S940 and KCC2 internalization. S940A homozygotes were viable and exhibited comparable basal levels of KCC2 expression and activity relative to WT mice. However, exposure of S940A mice to kainate induced lethality within 30 min of kainate injection and subsequent entrance into SE. We assessed the effect of the S940A mutation in cultured hippocampal neurons to explore the mechanisms underlying this phenotype. Under basal conditions, the mutation had no effect on neuronal Cl(-) extrusion. However, a selective deficit in KCC2 activity was seen in S940A neurons upon transient exposure to glutamate. Significantly, whereas the effects of glutamate on KCC2 function could be ameliorated in WT neurons with agents that enhance S940 phosphorylation, this positive modulation was lost in S940A neurons. Collectively our results suggest that phosphorylation of S940 plays a critical role in potentiating KCC2 activity to limit the development of SE.


Subject(s)
Status Epilepticus/metabolism , Status Epilepticus/pathology , Symporters/metabolism , Animals , Chlorides/metabolism , Endocytosis , Gene Knock-In Techniques , Glutamates/pharmacology , Mice , Mice, Neurologic Mutants , Mutant Proteins/metabolism , Mutation/genetics , Phosphorylation , Phosphoserine/metabolism , Protein Phosphatase 1/antagonists & inhibitors , Protein Phosphatase 1/metabolism , Symporters/genetics , gamma-Aminobutyric Acid/metabolism , K Cl- Cotransporters
19.
J Neurosci ; 36(11): 3106-14, 2016 Mar 16.
Article in English | MEDLINE | ID: mdl-26985023

ABSTRACT

Repeated exposure to psychostimulants induces locomotor sensitization and leads to persistent changes in the circuitry of the mesocorticolimbic dopamine (DA) system. G-protein-gated inwardly rectifying potassium (GIRK; also known as Kir3) channels mediate a slow IPSC and control the excitability of DA neurons. Repeated 5 d exposure to psychostimulants decreases the size of the GABAB receptor (GABABR)-activated GIRK currents (IBaclofen) in ventral tegmental area (VTA) DA neurons of mice, but the mechanism underlying this plasticity is poorly understood. Here, we show that methamphetamine-dependent attenuation of GABABR-GIRK currents in VTA DA neurons required activation of both D1R-like and D2R-like receptors. The methamphetamine-dependent decrease in GABABR-GIRK currents in VTA DA neurons did not depend on a mechanism of dephosphorylation of the GABAB R2 subunit found previously for other neurons in the reward pathway. Rather, the presence of the GIRK3 subunit appeared critical for the methamphetamine-dependent decrease of GABABR-GIRK current in VTA DA neurons. Together, these results highlight different regulatory mechanisms in the learning-evoked changes that occur in the VTA with repeated exposure to psychostimulants. SIGNIFICANCE STATEMENT: Exposure to addictive drugs such as psychostimulants produces persistent adaptations in inhibitory circuits within the mesolimbic dopamine system, suggesting that addictive behaviors are encoded by changes in the reward neural circuitry. One form of neuroadaptation that occurs with repeated exposure to psychostimulants is a decrease in slow inhibition, mediated by a GABAB receptor and a potassium channel. Here, we examine the subcellular mechanism that links psychostimulant exposure with changes in slow inhibition and reveal that one type of potassium channel subunit is important for mediating the effect of repeated psychostimulant exposure. Dissecting out the components of drug-dependent plasticity and uncovering novel protein targets in the reward circuit may lead to the development of new therapeutics for treating addiction.


Subject(s)
Dopamine Agents/pharmacology , Dopaminergic Neurons/drug effects , G Protein-Coupled Inwardly-Rectifying Potassium Channels/metabolism , Methamphetamine/pharmacology , Receptors, GABA-B/metabolism , Tyrosine 3-Monooxygenase/metabolism , Ventral Tegmental Area/cytology , Animals , Animals, Newborn , Baclofen/pharmacology , Female , G Protein-Coupled Inwardly-Rectifying Potassium Channels/genetics , GABA-B Receptor Agonists/pharmacology , Homeodomain Proteins/genetics , Homeodomain Proteins/metabolism , In Vitro Techniques , Male , Membrane Potentials/drug effects , Membrane Potentials/genetics , Mice , Mice, Inbred C57BL , Mice, Transgenic , Receptors, GABA-B/genetics , Time Factors , Transcription Factors/genetics , Transcription Factors/metabolism , Tyrosine 3-Monooxygenase/genetics
20.
J Biol Chem ; 291(23): 12394-407, 2016 Jun 03.
Article in English | MEDLINE | ID: mdl-27044742

ABSTRACT

The accumulation of γ-aminobutyric acid receptors (GABAARs) at the appropriate postsynaptic sites is critical for determining the efficacy of fast inhibitory neurotransmission. Although we know that the majority of synaptic GABAAR subtypes are assembled from α1-3, ß, and γ2 subunits, our understanding of how neurons facilitate their targeting to and stabilization at inhibitory synapses is rudimentary. To address these issues, we have created knock-in mice in which the pH-sensitive green fluorescent protein (GFP) and the Myc epitope were introduced to the extracellular domain of the mature receptor α2 subunit (pHα2). Using immunoaffinity purification and mass spectroscopy, we identified a stable complex of 174 proteins that were associated with pHα2, including other GABAAR subunits, and previously identified receptor-associated proteins such as gephyrin and collybistin. 149 of these proteins were novel GABAAR binding partners and included G-protein-coupled receptors and ion channel subunits, proteins that regulate trafficking and degradation, regulators of protein phosphorylation, GTPases, and a number of proteins that regulate their activity. Notably, members of the postsynaptic density family of proteins that are critical components of excitatory synapses were not associated with GABAARs. Crucially, we demonstrated for a subset of these novel proteins (including cullin1, ephexin, potassium channel tetramerization domain containing protein 12, mitofusin2, metabotropic glutamate receptor 5, p21-activated kinase 7, and Ras-related protein 5A) bind directly to the intracellular domains of GABAARs, validating our proteomic analysis. Thus, our experiments illustrate the complexity of the GABAAR proteome and enhance our understanding of the mechanisms neurons use to construct inhibitory synapses.


Subject(s)
Green Fluorescent Proteins/metabolism , Proteome/metabolism , Proteomics/methods , Receptors, GABA-A/metabolism , Synapses/metabolism , Animals , Blotting, Western , Electrophysiological Phenomena , Green Fluorescent Proteins/genetics , HEK293 Cells , Hippocampus/metabolism , Hippocampus/physiology , Humans , Hydrogen-Ion Concentration , Inhibitory Postsynaptic Potentials , Mass Spectrometry , Mice, 129 Strain , Mice, Inbred C57BL , Mice, Transgenic , Neurons/metabolism , Neurons/physiology , Proteome/genetics , Receptors, GABA-A/genetics , Synapses/physiology
SELECTION OF CITATIONS
SEARCH DETAIL