Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 83
Filter
2.
Nature ; 584(7820): 304-309, 2020 08.
Article in English | MEDLINE | ID: mdl-32581365

ABSTRACT

The human GABAB receptor-a member of the class C family of G-protein-coupled receptors (GPCRs)-mediates inhibitory neurotransmission and has been implicated in epilepsy, pain and addiction1. A unique GPCR that is known to require heterodimerization for function2-6, the GABAB receptor has two subunits, GABAB1 and GABAB2, that are structurally homologous but perform distinct and complementary functions. GABAB1 recognizes orthosteric ligands7,8, while GABAB2 couples with G proteins9-14. Each subunit is characterized by an extracellular Venus flytrap (VFT) module, a descending peptide linker, a seven-helix transmembrane domain and a cytoplasmic tail15. Although the VFT heterodimer structure has been resolved16, the structure of the full-length receptor and its transmembrane signalling mechanism remain unknown. Here we present a near full-length structure of the GABAB receptor, captured in an inactive state by cryo-electron microscopy. Our structure reveals several ligands that preassociate with the receptor, including two large endogenous phospholipids that are embedded within the transmembrane domains to maintain receptor integrity and modulate receptor function. We also identify a previously unknown heterodimer interface between transmembrane helices 3 and 5 of both subunits, which serves as a signature of the inactive conformation. A unique 'intersubunit latch' within this transmembrane interface maintains the inactive state, and its disruption leads to constitutive receptor activity.


Subject(s)
Cryoelectron Microscopy , Receptors, GABA-B/chemistry , Receptors, GABA-B/ultrastructure , Calcium/metabolism , Ethanolamines/chemistry , Ethanolamines/metabolism , Humans , Ligands , Models, Molecular , Phosphorylcholine/chemistry , Phosphorylcholine/metabolism , Protein Domains , Protein Multimerization , Protein Subunits/chemistry , Protein Subunits/metabolism , Receptors, GABA-B/metabolism , Structure-Activity Relationship
3.
J Neurosci ; 44(16)2024 Apr 17.
Article in English | MEDLINE | ID: mdl-38350999

ABSTRACT

Genome-wide association studies (GWAS) of electroencephalographic endophenotypes for alcohol use disorder (AUD) has identified noncoding polymorphisms within the KCNJ6 gene. KCNJ6 encodes GIRK2, a subunit of a G-protein-coupled inwardly rectifying potassium channel that regulates neuronal excitability. We studied the effect of upregulating KCNJ6 using an isogenic approach with human glutamatergic neurons derived from induced pluripotent stem cells (male and female donors). Using multielectrode arrays, population calcium imaging, single-cell patch-clamp electrophysiology, and mitochondrial stress tests, we find that elevated GIRK2 acts in concert with 7-21 d of ethanol exposure to inhibit neuronal activity, to counteract ethanol-induced increases in glutamate response, and to promote an increase intrinsic excitability. Furthermore, elevated GIRK2 prevented ethanol-induced changes in basal and activity-dependent mitochondrial respiration. These data support a role for GIRK2 in mitigating the effects of ethanol and a previously unknown connection to mitochondrial function in human glutamatergic neurons.


Subject(s)
Ethanol , G Protein-Coupled Inwardly-Rectifying Potassium Channels , Humans , Male , Female , Genome-Wide Association Study , Neurons , Respiration
4.
J Neurosci ; 43(45): 7587-7598, 2023 11 08.
Article in English | MEDLINE | ID: mdl-37940594

ABSTRACT

The human brain represents one of the most complex biological systems, containing billions of neurons interconnected through trillions of synapses. Inherent to the brain is a biochemical complexity involving ions, signaling molecules, and peptides that regulate neuronal activity and allow for short- and long-term adaptations. Large-scale and noninvasive imaging techniques, such as fMRI and EEG, have highlighted brain regions involved in specific functions and visualized connections between different brain areas. A major shortcoming, however, is the need for more information on specific cell types and neurotransmitters involved, as well as poor spatial and temporal resolution. Recent technologies have been advanced for neuronal circuit mapping and implemented in behaving model organisms to address this. Here, we highlight strategies for targeting specific neuronal subtypes, identifying, and releasing signaling molecules, controlling gene expression, and monitoring neuronal circuits in real-time in vivo Combined, these approaches allow us to establish direct causal links from genes and molecules to the systems level and ultimately to cognitive processes.


Subject(s)
Brain , Neurons , Humans , Brain/physiology , Neurons/physiology , Brain Mapping/methods , Synapses/physiology , Magnetic Resonance Imaging
5.
Mol Psychiatry ; 28(5): 1970-1982, 2023 05.
Article in English | MEDLINE | ID: mdl-34493831

ABSTRACT

Dopaminergic neurons are critical to movement, mood, addiction, and stress. Current techniques for generating dopaminergic neurons from human induced pluripotent stem cells (hiPSCs) yield heterogenous cell populations with variable purity and inconsistent reproducibility between donors, hiPSC clones, and experiments. Here, we report the rapid (5 weeks) and efficient (~90%) induction of induced dopaminergic neurons (iDANs) through transient overexpression of lineage-promoting transcription factors combined with stringent selection across five donors. We observe maturation-dependent increase in dopamine synthesis and electrophysiological properties consistent with midbrain dopaminergic neuron identity, such as slow-rising after- hyperpolarization potentials, an action potential duration of ~3 ms, tonic sub-threshold oscillatory activity, and spontaneous burst firing at a frequency of ~1.0-1.75 Hz. Transcriptome analysis reveals robust expression of genes involved in fetal midbrain dopaminergic neuron identity. Specifically expressed genes in iDANs, as well as those from isogenic induced GABAergic and glutamatergic neurons, were enriched in loci conferring heritability for cannabis use disorder, schizophrenia, and bipolar disorder; however, each neuronal subtype demonstrated subtype-specific heritability enrichments in biologically relevant pathways, and iDANs alone were uniquely enriched in autism spectrum disorder risk loci. Therefore, iDANs provide a critical tool for modeling midbrain dopaminergic neuron development and dysfunction in psychiatric disease.


Subject(s)
Autism Spectrum Disorder , Induced Pluripotent Stem Cells , Humans , Dopaminergic Neurons/metabolism , Autism Spectrum Disorder/metabolism , Reproducibility of Results , Induced Pluripotent Stem Cells/metabolism , Mesencephalon/metabolism
6.
Mol Psychiatry ; 28(2): 746-758, 2023 02.
Article in English | MEDLINE | ID: mdl-36207584

ABSTRACT

Synonymous and noncoding single nucleotide polymorphisms (SNPs) in the KCNJ6 gene, encoding G protein-gated inwardly rectifying potassium channel subunit 2 (GIRK2), have been linked with increased electroencephalographic frontal theta event-related oscillations (ERO) in subjects diagnosed with alcohol use disorder (AUD). To identify molecular and cellular mechanisms while retaining the appropriate genetic background, we generated induced excitatory glutamatergic neurons (iN) from iPSCs derived from four AUD-diagnosed subjects with KCNJ6 variants ("Affected: AF") and four control subjects without variants ("Unaffected: UN"). Neurons were analyzed for changes in gene expression, morphology, excitability and physiological properties. Single-cell RNA sequencing suggests that KCNJ6 AF variant neurons have altered patterns of synaptic transmission and cell projection morphogenesis. Results confirm that AF neurons express lower levels of GIRK2, have greater neurite area, and elevated excitability. Interestingly, exposure to intoxicating concentrations of ethanol induces GIRK2 expression and reverses functional effects in AF neurons. Ectopic overexpression of GIRK2 alone mimics the effect of ethanol to normalize induced excitability. We conclude that KCNJ6 variants decrease GIRK2 expression and increase excitability and that this effect can be minimized or reduced with ethanol.


Subject(s)
Alcoholism , G Protein-Coupled Inwardly-Rectifying Potassium Channels , Humans , G Protein-Coupled Inwardly-Rectifying Potassium Channels/genetics , G Protein-Coupled Inwardly-Rectifying Potassium Channels/metabolism , Ethanol/pharmacology , Ethanol/metabolism , Neurons/metabolism , Alcoholism/genetics , Alcoholism/metabolism , Electroencephalography
7.
Proc Natl Acad Sci U S A ; 116(17): 8370-8379, 2019 04 23.
Article in English | MEDLINE | ID: mdl-30971491

ABSTRACT

Metabotropic GABAB receptors mediate a significant fraction of inhibitory neurotransmission in the brain. Native GABAB receptor complexes contain the principal subunits GABAB1 and GABAB2, which form an obligate heterodimer, and auxiliary subunits, known as potassium channel tetramerization domain-containing proteins (KCTDs). KCTDs interact with GABAB receptors and modify the kinetics of GABAB receptor signaling. Little is known about the molecular mechanism governing the direct association and functional coupling of GABAB receptors with these auxiliary proteins. Here, we describe the high-resolution structure of the KCTD16 oligomerization domain in complex with part of the GABAB2 receptor. A single GABAB2 C-terminal peptide is bound to the interior of an open pentamer formed by the oligomerization domain of five KCTD16 subunits. Mutation of specific amino acids identified in the structure of the GABAB2-KCTD16 interface disrupted both the biochemical association and functional modulation of GABAB receptors and G protein-activated inwardly rectifying K+ channel (GIRK) channels. These interfacial residues are conserved among KCTDs, suggesting a common mode of KCTD interaction with GABAB receptors. Defining the binding interface of GABAB receptor and KCTD reveals a potential regulatory site for modulating GABAB-receptor function in the brain.


Subject(s)
Intracellular Signaling Peptides and Proteins , Nerve Tissue Proteins , Receptors, GABA-B , Binding Sites/genetics , Crystallography , Humans , Intracellular Signaling Peptides and Proteins/chemistry , Intracellular Signaling Peptides and Proteins/genetics , Intracellular Signaling Peptides and Proteins/metabolism , Models, Molecular , Nerve Tissue Proteins/chemistry , Nerve Tissue Proteins/genetics , Nerve Tissue Proteins/metabolism , Protein Binding/genetics , Receptors, GABA-B/chemistry , Receptors, GABA-B/genetics , Receptors, GABA-B/metabolism , Signal Transduction/genetics
8.
Angew Chem Int Ed Engl ; 61(34): e202206122, 2022 08 22.
Article in English | MEDLINE | ID: mdl-35723610

ABSTRACT

Neuropeptides are abundant signaling molecules in the central nervous system. Yet remarkably little is known about their spatiotemporal spread and biological activity. Here, we developed an integrated optical approach using Plasmonic nAnovesicles and cell-based neurotransmitter fluorescent engineered reporter (CNiFER), or PACE, to probe neuropeptide signaling in the mouse neocortex. Small volumes (fL to pL) of exogenously supplied somatostatin-14 (SST) can be rapidly released under near-infrared light stimulation from nanovesicles implanted in the brain and detected by SST2 CNiFERs with nM sensitivity. Our measurements reveal reduced but synchronized SST transmission within 130 µm, and markedly smaller and delayed transmission at longer distances. These measurements enabled a quantitative estimation of the SST loss rate due to peptide degradation and binding. PACE offers a new tool for determining the spatiotemporal scales of neuropeptide volume transmission and signaling in the brain.


Subject(s)
Neuropeptides , Animals , Brain/metabolism , Mice , Signal Transduction , Somatostatin/metabolism
9.
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
10.
Handb Exp Pharmacol ; 267: 1-49, 2021.
Article in English | MEDLINE | ID: mdl-34247281

ABSTRACT

K+ channels enable potassium to flow across the membrane with great selectivity. There are four K+ channel families: voltage-gated K (Kv), calcium-activated (KCa), inwardly rectifying K (Kir), and two-pore domain potassium (K2P) channels. All four K+ channels are formed by subunits assembling into a classic tetrameric (4x1P = 4P for the Kv, KCa, and Kir channels) or tetramer-like (2x2P = 4P for the K2P channels) architecture. These subunits can either be the same (homomers) or different (heteromers), conferring great diversity to these channels. They share a highly conserved selectivity filter within the pore but show different gating mechanisms adapted for their function. K+ channels play essential roles in controlling neuronal excitability by shaping action potentials, influencing the resting membrane potential, and responding to diverse physicochemical stimuli, such as a voltage change (Kv), intracellular calcium oscillations (KCa), cellular mediators (Kir), or temperature (K2P).


Subject(s)
Calcium Signaling , Action Potentials , Humans
11.
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
12.
Angew Chem Int Ed Engl ; 59(22): 8608-8615, 2020 05 25.
Article in English | MEDLINE | ID: mdl-32124529

ABSTRACT

Remote and minimally-invasive modulation of biological systems with light has transformed modern biology and neuroscience. However, light absorption and scattering significantly prevents penetration to deep brain regions. Herein, we describe the use of gold-coated mechanoresponsive nanovesicles, which consist of liposomes made from the artificial phospholipid Rad-PC-Rad as a tool for the delivery of bioactive molecules into brain tissue. Near-infrared picosecond laser pulses activated the gold-coating on the surface of nanovesicles, creating nanomechanical stress and leading to near-complete vesicle cargo release in sub-seconds. Compared to natural phospholipid liposomes, the photo-release was possible at 40 times lower laser energy. This high photosensitivity enables photorelease of molecules down to a depth of 4 mm in mouse brain. This promising tool provides a versatile platform to optically release functional molecules to modulate brain circuits.


Subject(s)
Brain/metabolism , Brain/radiation effects , Infrared Rays , Nanotechnology/methods , Animals , Biomechanical Phenomena , Gold/chemistry , Mice , Phospholipids/metabolism
13.
Alcohol Clin Exp Res ; 42(9): 1572-1590, 2018 09.
Article in English | MEDLINE | ID: mdl-29897633

ABSTRACT

Alcohol use disorder (AUD) affects millions of people and costs nearly 250 billion dollars annually. Few effective FDA-approved treatments exist, and more are needed. AUDs have a strong heritability, but only a few genes have been identified with a large effect size on disease phenotype. Genomewide association studies (GWASs) have identified common variants with low effect sizes, most of which are in noncoding regions of the genome. Animal models frequently fail to recapitulate key molecular features of neuropsychiatric disease due to the polygenic nature of the disease, partial conservation of coding regions, and significant disparity in noncoding regions. By contrast, human induced pluripotent stem cells (hiPSCs) derived from patients provide a powerful platform for evaluating genes identified by GWAS and modeling complex interactions in the human genome. hiPSCs can be differentiated into a wide variety of human cells, including neurons, glia, and hepatic cells, which are compatible with numerous functional assays and genome editing techniques. In this review, we focus on current applications and future directions of patient hiPSC-derived central nervous system cells for modeling AUDs in addition to highlighting successful applications of hiPSCs in polygenic neuropsychiatric diseases.


Subject(s)
Alcoholism/diagnosis , Alcoholism/genetics , Genetic Predisposition to Disease/genetics , Genome-Wide Association Study/trends , Induced Pluripotent Stem Cells/physiology , Alcoholism/therapy , Animals , Cells, Cultured , Genome-Wide Association Study/methods , Humans
14.
Proc Natl Acad Sci U S A ; 112(22): 7091-6, 2015 Jun 02.
Article in English | MEDLINE | ID: mdl-25964320

ABSTRACT

G protein-gated inwardly rectifying potassium (GIRK) channels are critical regulators of neuronal excitability and can be directly activated by ethanol. Constitutive deletion of the GIRK3 subunit has minimal phenotypic consequences, except in response to drugs of abuse. Here we investigated how the GIRK3 subunit contributes to the cellular and behavioral effects of ethanol, as well as to voluntary ethanol consumption. We found that constitutive deletion of GIRK3 in knockout (KO) mice selectively increased ethanol binge-like drinking, without affecting ethanol metabolism, sensitivity to ethanol intoxication, or continuous-access drinking. Virally mediated expression of GIRK3 in the ventral tegmental area (VTA) reversed the phenotype of GIRK3 KO mice and further decreased the intake of their wild-type counterparts. In addition, GIRK3 KO mice showed a blunted response of the mesolimbic dopaminergic (DA) pathway to ethanol, as assessed by ethanol-induced excitation of VTA neurons and DA release in the nucleus accumbens. These findings support the notion that the subunit composition of VTA GIRK channels is a critical determinant of DA neuron sensitivity to drugs of abuse. Furthermore, our study reveals the behavioral impact of this cellular effect, whereby the level of GIRK3 expression in the VTA tunes ethanol intake under binge-type conditions: the more GIRK3, the less ethanol drinking.


Subject(s)
Dopaminergic Neurons/metabolism , Ethanol/pharmacology , G Protein-Coupled Inwardly-Rectifying Potassium Channels/metabolism , Ion Channel Gating/physiology , Motivation/genetics , Analysis of Variance , Animals , Binge Drinking/genetics , DNA Primers/genetics , G Protein-Coupled Inwardly-Rectifying Potassium Channels/deficiency , In Situ Hybridization , Ion Channel Gating/genetics , Mice , Mice, Inbred C57BL , Mice, Knockout , Microdialysis , Reverse Transcriptase Polymerase Chain Reaction , Reward
15.
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
16.
Nat Methods ; 11(12): 1245-52, 2014 Dec.
Article in English | MEDLINE | ID: mdl-25344639

ABSTRACT

The neuronal coding of stimulus-to-action sequences is believed to involve the release of dopamine (DA) and norepinephrine (NE). The electrochemical similarity of these monoamines, however, confounds real-time measurements of their release. Here we report cell-based neurotransmitter fluorescent engineered reporters (CNiFERs) that use the specificity of G protein-coupled receptors (GPCRs) to discriminate nanomolar concentrations of DA and NE. CNiFERs were implanted into the frontal cortex of mice to measure the timing of neurotransmitter release during classical conditioning with the use of two-photon microscopy. The onset of DA release correlated with that of licking and shifted from the time of the reward toward that of the cue upon conditioning. In contrast, concurrent release of NE did not correlate with licking or the cue. This generation of CNiFERs provides unique tools to assess the release of monoamines. The molecular design of these CNiFERs may be generalized to realize CNiFERs for any molecule that activates a GPCR.


Subject(s)
Behavior, Animal , Cerebral Cortex/metabolism , Dopamine/metabolism , Fluorescent Dyes/chemistry , Norepinephrine/metabolism , Receptors, G-Protein-Coupled/metabolism , Animals , Cerebral Cortex/anatomy & histology , Fluorescence Resonance Energy Transfer , Learning/physiology , Mice , Synaptic Transmission/physiology
17.
Methods ; 101: 113-24, 2016 05 15.
Article in English | MEDLINE | ID: mdl-26626326

ABSTRACT

Since the discovery of somatic reprogramming, human induced pluripotent stem cells (hiPSCs) have been exploited to model a variety of neurological and psychiatric disorders. Because hiPSCs represent an almost limitless source of patient-derived neurons that retain the genetic variations thought to contribute to disease etiology, they have been heralded as a patient-specific platform for high throughput drug screening. However, the utility of current protocols for generating neurons from hiPSCs remains limited by protracted differentiation timelines and heterogeneity of the neuronal phenotypes produced. Neuronal induction via the forced expression of exogenous transcription factors rapidly induces defined populations of functional neurons from fibroblasts and hiPSCs. Here, we describe an adapted protocol that accelerates maturation of functional excitatory neurons from hiPSC-derived neural progenitor cells (NPCs) via lentiviral transduction of Neurogenin 2 (using both mNgn2 and hNGN2). This methodology, relying upon a robust and scalable starting population of hiPSC NPCs, should be readily amenable to scaling for hiPSC-based high-throughput drug screening.


Subject(s)
Basic Helix-Loop-Helix Transcription Factors/physiology , Induced Pluripotent Stem Cells/physiology , Nerve Tissue Proteins/physiology , Cell Culture Techniques , Cells, Cultured , Cellular Reprogramming , Gene Expression , Humans , Lentivirus , Neural Stem Cells/physiology , Transcriptional Activation
18.
J Neurosci ; 35(1): 352-65, 2015 Jan 07.
Article in English | MEDLINE | ID: mdl-25568127

ABSTRACT

In many cell types, differentiation requires an interplay between extrinsic signals and transcriptional changes mediated by repressive and activating histone modifications. Oligodendrocyte progenitors (OPCs) are electrically responsive cells receiving synaptic input. The differentiation of these cells into myelinating oligodendrocytes is characterized by temporal waves of gene repression followed by activation of myelin genes and progressive decline of electrical responsiveness. In this study, we used chromatin isolated from rat OPCs and immature oligodendrocytes, to characterize the genome-wide distribution of the repressive histone marks, H3K9me3 and H3K27me3, during differentiation. Although both marks were present at the OPC stage, only H3K9me3 marks (but not H3K27me3) were found to be increased during differentiation, at genes related to neuronal lineage and regulation of membrane excitability. Consistent with these findings, the levels and activity of H3K9 methyltransferases (H3K9 HMT), but not H3K27 HMT, increased more prominently upon exposure to oligodendrocyte differentiating stimuli and were detected in stage-specific repressive protein complexes containing the transcription factors SOX10 or YY1. Silencing H3K9 HMT, but not H3K27 HMT, impaired oligodendrocyte differentiation and functionally altered the response of oligodendrocytes to electrical stimulation. Together, these results identify repressive H3K9 methylation as critical for gene repression during oligodendrocyte differentiation.


Subject(s)
Cell Differentiation/physiology , Chromatin/metabolism , Histones/metabolism , Neurogenesis/physiology , Oligodendroglia/metabolism , Animals , Chromatin Immunoprecipitation/methods , Female , Male , Methylation , Mice , Mice, Inbred C57BL , Mice, Transgenic , Organ Culture Techniques , Pregnancy , Rats , Rats, Sprague-Dawley
19.
J Neurosci ; 35(18): 7131-42, 2015 May 06.
Article in English | MEDLINE | ID: mdl-25948263

ABSTRACT

G-protein-gated inwardly rectifying K(+) (GIRK/Kir3) channel activation underlies key physiological effects of opioids, including analgesia and dependence. GIRK channel activation has also been implicated in the opioid-induced inhibition of midbrain GABA neurons and consequent disinhibition of dopamine (DA) neurons in the ventral tegmental area (VTA). Drug-induced disinhibition of VTA DA neurons has been linked to reward-related behaviors and underlies opioid-induced motor activation. Here, we demonstrate that mouse VTA GABA neurons express a GIRK channel formed by GIRK1 and GIRK2 subunits. Nevertheless, neither constitutive genetic ablation of Girk1 or Girk2, nor the selective ablation of GIRK channels in GABA neurons, diminished morphine-induced motor activity in mice. Moreover, direct activation of GIRK channels in midbrain GABA neurons did not enhance motor activity. In contrast, genetic manipulations that selectively enhanced or suppressed GIRK channel function in midbrain DA neurons correlated with decreased and increased sensitivity, respectively, to the motor-stimulatory effect of systemic morphine. Collectively, these data support the contention that the unique GIRK channel subtype in VTA DA neurons, the GIRK2/GIRK3 heteromer, regulates the sensitivity of the mouse mesolimbic DA system to drugs with addictive potential.


Subject(s)
Analgesics, Opioid/pharmacology , Dopaminergic Neurons/physiology , G Protein-Coupled Inwardly-Rectifying Potassium Channels/physiology , GABAergic Neurons/physiology , Motor Activity/physiology , Animals , Dopaminergic Neurons/drug effects , Dose-Response Relationship, Drug , GABAergic Neurons/drug effects , Mice , Mice, Inbred C57BL , Mice, Knockout , Motor Activity/drug effects , Protein Subunits/physiology , Ventral Tegmental Area/drug effects , Ventral Tegmental Area/physiology
20.
Proc Natl Acad Sci U S A ; 110(45): 18309-14, 2013 Nov 05.
Article in English | MEDLINE | ID: mdl-24145411

ABSTRACT

Alcohol (ethanol) produces a wide range of pharmacological effects on the nervous system through its actions on ion channels. The molecular mechanism underlying ethanol modulation of ion channels is poorly understood. Here we used a unique method of alcohol-tagging to demonstrate that alcohol activation of a G-protein-gated inwardly rectifying potassium (GIRK or Kir3) channel is mediated by a defined alcohol pocket through changes in affinity for the membrane phospholipid signaling molecule phosphatidylinositol 4,5-bisphosphate. Surprisingly, hydrophobicity and size, but not the canonical hydroxyl, were important determinants of alcohol-dependent activation. Altering levels of G protein Gßγ subunits, conversely, did not affect alcohol-dependent activation, suggesting a fundamental distinction between receptor and alcohol gating of GIRK channels. The chemical properties of the alcohol pocket revealed here might extend to other alcohol-sensitive proteins, revealing a unique protein microdomain for targeting alcohol-selective therapeutics in the treatment of alcoholism and addiction.


Subject(s)
Ethanol/pharmacology , G Protein-Coupled Inwardly-Rectifying Potassium Channels/chemistry , G Protein-Coupled Inwardly-Rectifying Potassium Channels/metabolism , Models, Molecular , Protein Conformation , Analysis of Variance , Crystallization , HEK293 Cells , Humans , Mutagenesis , Patch-Clamp Techniques , Phosphatidylinositol 4,5-Diphosphate/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL