Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 59
Filter
Add more filters











Publication year range
1.
Mar Drugs ; 20(2)2022 Feb 15.
Article in English | MEDLINE | ID: mdl-35200669

ABSTRACT

(1) Background: G protein-coupled inward-rectifier potassium (GIRK) channels, especially neuronal GIRK1/2 channels, have been the focus of intense research interest for developing drugs against brain diseases. In this context, venom peptides that selectively activate GIRK channels can be seen as a new source for drug development. Here, we report on the identification and electrophysiological characterization of a novel activator of GIRK1/2 channels, AsKC11, found in the venom of the sea anemone Anemonia sulcata. (2) Methods: AsKC11 was purified from the sea anemone venom by reverse-phase chromatography and the sequence was identified by mass spectrometry. Using the two-electrode voltage-clamp technique, the activity of AsKC11 on GIRK1/2 channels was studied and its selectivity for other potassium channels was investigated. (3) Results: AsKC11, a Kunitz peptide found in the venom of A. sulcata, is the first peptide shown to directly activate neuronal GIRK1/2 channels independent from Gi/o protein activity, without affecting the inward-rectifier potassium channel (IRK1) and with only a minor effect on KV1.6 channels. Thus, AsKC11 is a novel activator of GIRK channels resulting in larger K+ currents because of an increased chord conductance. (4) Conclusions: These discoveries provide new insights into a novel class of GIRK activators.


Subject(s)
Cnidarian Venoms/chemistry , G Protein-Coupled Inwardly-Rectifying Potassium Channels/drug effects , Peptides/pharmacology , Animals , Chromatography, Reverse-Phase , Female , G Protein-Coupled Inwardly-Rectifying Potassium Channels/metabolism , Mass Spectrometry , Patch-Clamp Techniques , Peptides/chemistry , Peptides/isolation & purification , Xenopus laevis
2.
J Neurochem ; 160(2): 154-171, 2022 01.
Article in English | MEDLINE | ID: mdl-34738241

ABSTRACT

αO-Conotoxin GeXIVA is a 28 amino acid peptide derived from the venom of the marine snail Conus generalis. The presence of four cysteine residues in the structure of GeXIVA allows it to have three different disulfide isomers, that is, the globular, ribbon or bead isomer. All three isomers are active at α9α10 nicotinic acetylcholine receptors, with the bead isomer, GeXIVA[1,2], being the most potent and exhibiting analgesic activity in animal models of neuropathic pain. The original report of GeXIVA activity failed to observe any effect of the isomers on high voltage-activated (HVA) calcium channel currents in rat dorsal root ganglion (DRG) neurons. In this study, we report, for the first time, the activity of globular GeXIVA[1,3] at G protein-coupled GABAB receptors (GABAB R) inhibiting HVA N-type calcium (Cav2.2) channels and reducing membrane excitability in mouse DRG neurons. The inhibition of HVA Ba2+ currents and neuroexcitability by GeXIVA[1,3] was partially reversed by the selective GABAB R antagonist CGP 55845. In transfected HEK293T cells co-expressing human GABAB R1 and R2 subunits and Cav2.2 channels, both GeXIVA[1,3] and GeXIVA[1,4] inhibited depolarization-activated Ba2+ currents mediated by Cav2.2 channels, whereas GeXIVA[1,2] had no effect. The effects of three cyclized GeXIVA[1,4] ribbon isomers were also tested, with cGeXIVA GAG being the most potent at human GABAB R-coupled Cav2.2 channels. Interestingly, globular GeXIVA[1,3] also reversibly potentiated inwardly-rectifying K+ currents mediated by human GIRK1/2 channels co-expressed with GABAB R in HEK293T cells. This study highlights GABAB R as a potentially important receptor target for the activity of αO-conotoxin GeXIVA to mediate analgesia.


Subject(s)
Calcium Channels, N-Type/drug effects , Conotoxins/pharmacology , G Protein-Coupled Inwardly-Rectifying Potassium Channels/drug effects , Neurons/drug effects , Receptors, GABA-B/drug effects , Analgesics, Non-Narcotic/chemistry , Analgesics, Non-Narcotic/pharmacology , Animals , Calcium Channels, N-Type/metabolism , Conotoxins/chemistry , G Protein-Coupled Inwardly-Rectifying Potassium Channels/metabolism , Ganglia, Spinal/drug effects , HEK293 Cells , Humans , Male , Mice , Mice, Inbred C57BL , Protein Isoforms , Receptors, GABA-B/metabolism
3.
CNS Neurosci Ther ; 27(11): 1289-1299, 2021 11.
Article in English | MEDLINE | ID: mdl-34347369

ABSTRACT

BACKGROUND: Parkinson's disease (PD) is a neurodegenerative disease, and its pathogenesis is unclear. Previous studies mainly focus on the lesions of substantia nigra (SN) and striatum (Str) in PD. However, lesions are not limited. The olfactory bulb (OB), subventricular zone (SVZ), and hippocampus (Hippo) are also affected in PD. AIM: To reveal gene expression changes in the five brain regions (OB, SVZ, Str, SN, and Hippo), and to look for potential candidate genes and pathways that may be correlated with the pathogenesis of PD. MATERIALS AND METHODS: We established control group and 6-hydroxydopamine (6-OHDA) PD model group, and detected gene expressions in the five brain regions using RNA-seq and real-time quantitative polymerase chain reaction (RT-qPCR). We further analyzed the RNA-seq data by bioinformatics. RESULTS: We identified differentially expressed genes (DEGs) in all five brain regions. The DEGs were significantly enriched in the "dopaminergic synapse" and "retrograde endocannabinoid signaling," and Gi/o-GIRK is the shared cascade in the two pathways. We further identified Ephx2, Fam111a, and Gng2 as the potential candidate genes in the pathogenesis of PD for further studies. CONCLUSION: Our study suggested that gene expressions change in the five brain regions following exposure to 6-OHDA. The "dopaminergic synapse," "retrograde endocannabinoid signaling," and Gi/o-GIRK may be the key pathways and cascade of the synaptic damage in 6-OHDA PD rats. Ephx2, Fam111a, and Gng2 may play critical roles in the pathogenesis of PD.


Subject(s)
Brain Chemistry/genetics , Gene Expression Profiling , Oxidopamine , Parkinson Disease, Secondary/genetics , Transcriptome , Animals , Computational Biology , Dopaminergic Neurons , Endocannabinoids/metabolism , G Protein-Coupled Inwardly-Rectifying Potassium Channels/drug effects , G Protein-Coupled Inwardly-Rectifying Potassium Channels/metabolism , Gene Expression Regulation , Parkinson Disease, Secondary/chemically induced , Polymerase Chain Reaction , RNA-Seq , Rats , Rats, Sprague-Dawley
4.
Neuropharmacology ; 190: 108565, 2021 06 01.
Article in English | MEDLINE | ID: mdl-33891950

ABSTRACT

Arginine vasopressin (AVP) is a nonapeptide that serves as a neuromodulator in the brain and a hormone in the periphery that regulates water homeostasis and vasoconstriction. The subiculum is the major output region of the hippocampus and an integral component in the networks that processes sensory and motor cues to form a cognitive map encoding spatial, contextual, and emotional information. Whereas the subiculum expresses high densities of AVP-binding sites and AVP has been shown to increase the synaptic excitability of subicular pyramidal neurons, the underlying cellular and molecular mechanisms have not been determined. We found that activation of V1a receptors increased the excitability of subicular pyramidal neurons via activation of TRPV1 channels and depression of the GIRK channels. V1a receptor-induced excitation of subicular pyramidal neurons required the function of phospholipase Cß, but was independent of intracellular Ca2+ release. Protein kinase C was responsible for AVP-mediated depression of GIRK channels, whereas degradation of phosphatidylinositol 4,5-bisphosphate was involved in V1a receptor-elicited activation of TRPV1 channels. Our results may provide one of the cellular and molecular mechanisms to explain the physiological functions of AVP in the brain.


Subject(s)
G Protein-Coupled Inwardly-Rectifying Potassium Channels/metabolism , Hippocampus/metabolism , Pyramidal Cells/metabolism , Receptors, Vasopressin/metabolism , TRPV Cation Channels/metabolism , Action Potentials , Animals , Arginine Vasopressin/pharmacology , Calcium/metabolism , G Protein-Coupled Inwardly-Rectifying Potassium Channels/drug effects , Hippocampus/cytology , Hippocampus/drug effects , Membrane Potentials , Mice , Mice, Knockout , Patch-Clamp Techniques , Phosphatidylinositol 4,5-Diphosphate/metabolism , Phospholipase C beta/metabolism , Pyramidal Cells/drug effects , Receptors, Vasopressin/agonists , TRPV Cation Channels/drug effects , TRPV Cation Channels/genetics , Vasoconstrictor Agents/pharmacology
5.
Eur J Pharmacol ; 890: 173640, 2021 Jan 05.
Article in English | MEDLINE | ID: mdl-33045198

ABSTRACT

Opioids are the most powerful analgesics used clinically; however, severe side effects limit their long-term use. Various concepts involving biased intracellular signaling, partial agonism or multi-receptor targeting have been proposed to identify novel opioids with increased analgesic efficacy but reduced side effects. The search for such 'better opioids' implies screening of huge compound libraries and requires highly reliable, easy to perform and high throughput screening (HTS) assays. Here, we utilize an established membrane potential assay to monitor activation of G protein-coupled inwardly rectifying potassium (GIRK) channels, one of the main effectors of opioid receptor signaling, as readout to determine pharmacological profiles of opioids in a non-invasive manner. Specifically, in this study, we optimize assay conditions and extend the application of this assay to screen all four members of the opioid receptor family, stably expressed in AtT-20 and HEK293 cells. This ultra-sensitive system yielded EC50 values in the nano-molar range. We further validate this system for screening cells stably co-expressing two opioid receptors, which could be a valuable tool for investigating bi-functional ligands and studying interactions between receptors. Additionally, we demonstrate the utility of this assay to study antagonists as well as ligands with varying efficacies. Our results suggest that this assay could easily be up-scaled to HTS assay in order to efficiently study receptor activation and screen for novel opioids.


Subject(s)
GTP-Binding Proteins/drug effects , GTP-Binding Proteins/metabolism , High-Throughput Screening Assays/methods , Membrane Potentials/drug effects , Receptors, Opioid/metabolism , Signal Transduction/drug effects , Analgesics, Opioid/pharmacology , Animals , Cell Line, Tumor , Cell Separation , Flow Cytometry , Fluorescence , G Protein-Coupled Inwardly-Rectifying Potassium Channels/agonists , G Protein-Coupled Inwardly-Rectifying Potassium Channels/antagonists & inhibitors , G Protein-Coupled Inwardly-Rectifying Potassium Channels/drug effects , G Protein-Coupled Inwardly-Rectifying Potassium Channels/metabolism , GTP-Binding Proteins/agonists , GTP-Binding Proteins/antagonists & inhibitors , HEK293 Cells , Humans , Ligands , Mice
6.
J Neuroendocrinol ; 32(11): e12881, 2020 11.
Article in English | MEDLINE | ID: mdl-32803906

ABSTRACT

The secretion of prolactin from the pituitary is negatively controlled by tuberoinfundibular dopamine (TIDA) neurones. The electrical properties of TIDA cells have recently been identified as a modulatory target of neurotransmitters and hormones in the lactotrophic axis. The role of the GABAB receptor in this control has received little attention, yet is of particular interest because it may act as a TIDA neurone autoreceptor. Here, this issue was explored in a spontaneously active rat TIDA in vitro slice preparation using whole-cell recordings. Application of the GABAB receptor agonist, baclofen, dose-dependently slowed down or abolished the network oscillations typical of this preparation. Pharmacological manipulations identify the underlying mechanism as an outward current mediated by G-protein-coupled inwardly rectifying K+ -like channels. In addition to this postsynaptic modulation, we describe a presynaptic modulation where GABAB receptors restrain the release of glutamate and GABA onto TIDA neurones. Our data identify both pre- and postsynaptic modulation of TIDA neurones by GABAB receptors that may play a role in the neuronal network control of pituitary prolactin secretion and lactation.


Subject(s)
Dopaminergic Neurons/metabolism , Neuroendocrine Cells/metabolism , Receptors, GABA-B/metabolism , Receptors, Presynaptic/metabolism , Synapses/metabolism , Animals , Arcuate Nucleus of Hypothalamus/metabolism , Baclofen/pharmacology , Dopaminergic Neurons/drug effects , Dose-Response Relationship, Drug , Electrophysiological Phenomena , G Protein-Coupled Inwardly-Rectifying Potassium Channels/drug effects , G Protein-Coupled Inwardly-Rectifying Potassium Channels/metabolism , GABA Agonists/pharmacology , Male , Neuroendocrine Cells/drug effects , Patch-Clamp Techniques , Potassium Channels, Inwardly Rectifying/drug effects , Potassium Channels, Inwardly Rectifying/metabolism , Rats , Rats, Sprague-Dawley , Receptors, GABA-B/drug effects , Receptors, Presynaptic/drug effects , Synapses/drug effects
7.
Life Sci ; 240: 117068, 2020 Jan 01.
Article in English | MEDLINE | ID: mdl-31751583

ABSTRACT

AIMS: Bradycardia contributes to tachy-brady arrhythmias or sinus arrest during heart failure (HF). Sinoatrial node (SAN) adenosine A1 receptors (ADO A1Rs) are upregulated in HF, and adenosine is known to exert negative chronotropic effects on the SAN. Here, we investigated the role of A1R signaling at physiologically relevant ADO concentrations on HF SAN pacemaker cells. MAIN METHODS: Dogs with tachypacing-induced chronic HF and normal controls (CTL) were studied. SAN tissue was collected for A1R and GIRK mRNA quantification. SAN cells were isolated for perforated patch clamp recordings and firing rate (bpm), slope of slow diastolic depolarization (SDD), and maximum diastolic potential (MDP) were measured. Action potentials (APs) and currents were recorded before and after addition of 1 and 10 µM ADO. To assess contributions of A1R and G protein-coupled Inward Rectifier Potassium Current (GIRK) to ADO effects, APs were measured after the addition of DPCPX (selective A1R antagonist) or TPQ (selective GIRK blocker). KEY FINDINGS: A1R and GIRK mRNA expression were significantly increased in HF. In addition, ADO induced greater rate slowing and membrane hyperpolarization in HF vs CTL (p < 0.05). DPCPX prevented ADO-induced rate slowing in CTL and HF cells. The ADO-induced inward rectifying current, IKado, was observed significantly more frequently in HF than in CTL. TPQ prevented ADO-induced rate slowing in HF. SIGNIFICANCE: An increase in A1R and GIRK expression enhances IKAdo, causing hyperpolarization, and subsequent negative chronotropic effects in canine chronic HF at relevant [ADO]. GIRK blockade may be a useful strategy to mitigate bradycardia in HF.


Subject(s)
Adenosine A1 Receptor Agonists/pharmacology , Adenosine/pharmacology , G Protein-Coupled Inwardly-Rectifying Potassium Channels/agonists , Heart Failure/physiopathology , Heart Rate/drug effects , Receptor, Adenosine A1/metabolism , Sinoatrial Node/cytology , Sinoatrial Node/drug effects , Action Potentials/drug effects , Adenosine A1 Receptor Antagonists/pharmacology , Animals , Bee Venoms/pharmacology , Biological Clocks , Chronic Disease , Dogs , Female , G Protein-Coupled Inwardly-Rectifying Potassium Channels/antagonists & inhibitors , G Protein-Coupled Inwardly-Rectifying Potassium Channels/drug effects , In Vitro Techniques , Male , Patch-Clamp Techniques , Potassium Channel Blockers/pharmacology , Receptor, Adenosine A1/drug effects , Xanthines/pharmacology
8.
Brain Res ; 1710: 230-236, 2019 05 01.
Article in English | MEDLINE | ID: mdl-30584927

ABSTRACT

We previously reported that centrally acting non-narcotic antitussives, including tipepidine, inhibit G-protein-coupled inwardly rectifying potassium (GIRK) channel-activated currents of neurons. In addition, when administered at a cough suppressant dose, the drugs ameliorated the symptoms of various models of intractable brain disease in rodents. In the current study, we investigated whether tipepidine causes recovery from schizophrenia-like cognitive dysfunction, which was induced by MK-801 (0.2 mg/kg, i.p.) in mice. We also examined the effect of tipepidine and clozapine co-administration on the dysfunction. Moreover, we studied whether clozapine inhibits GIRK channel activated currents in single brain neurons using the patch-clamp technique. Tipepidine elicited recovery from MK-801-induced cognitive impairment in the novel objective recognition test and Y-maze test. Further, co-administration of tipepidine and clozapine, at subthreshold doses of each drug, improved MK-801-induced cognitive impairment in the novel objective recognition test. Clozapine (3 × 10-5 M) had a minor effect on baclofen-induced currents in dopamine neurons of the ventral tegmental area.


Subject(s)
Cognitive Dysfunction/drug therapy , G Protein-Coupled Inwardly-Rectifying Potassium Channels/drug effects , Piperidines/pharmacology , Animals , Antidepressive Agents/pharmacology , Antitussive Agents/pharmacology , Clozapine/pharmacology , Disease Models, Animal , Dizocilpine Maleate/pharmacology , Dopaminergic Neurons/drug effects , G Protein-Coupled Inwardly-Rectifying Potassium Channels/metabolism , Male , Mice , Patch-Clamp Techniques , Piperidines/metabolism , Rats , Rats, Wistar , Schizophrenia/drug therapy , Ventral Tegmental Area/drug effects
9.
Int J Neuropsychopharmacol ; 21(12): 1102-1108, 2018 12 01.
Article in English | MEDLINE | ID: mdl-29986044

ABSTRACT

Background: Previous evidence suggests that UNC9994 is a beta-arrestin2-selective agonist at the dopamine D2 receptor, lacking ability both to activate and antagonize G protein-dependent signaling. However, this has only been reported by one laboratory using a single assay. Methods: We used G protein-coupled inward rectifier potassium channel activation in Xenopus oocytes to investigate UNC9994-induced modulation of G protein-dependent signaling at dopamine D2 receptor and dopamine D3 receptor. Results: At dopamine D2 receptor, UNC9994 induced G protein-coupled inward rectifier potassium channel currents that were 15% of the maximal response to dopamine, with an EC50 of 185 nM. At dopamine D3 receptor, the ligand elicited 89% of the maximal dopamine response with an EC50 of 62 nM. Pertussis toxin abolished G protein-coupled inward rectifier potassium channel activation. Furthermore, UNC9994 antagonized dopamine-induced G protein-coupled inward rectifier potassium channel activation at dopamine D2 receptor. Conclusions: UNC9994 modulates G protein-coupled inward rectifier potassium channel channel activation via pertussis toxin-sensitive G proteins at dopamine D2 receptor and dopamine D3 receptor. These findings may have implications for the interpretation of data obtained with this ligand.


Subject(s)
Antipsychotic Agents/pharmacology , G Protein-Coupled Inwardly-Rectifying Potassium Channels/drug effects , Receptors, Dopamine D2 , Signal Transduction/drug effects , beta-Arrestin 2 , Animals , Humans , Ligands , Oocytes , Receptors, Dopamine D3 , Xenopus laevis
10.
J Physiol ; 596(10): 1833-1845, 2018 05 15.
Article in English | MEDLINE | ID: mdl-29063617

ABSTRACT

Ivermectin (IVM) is an antiparasitic drug that is used worldwide and rescues hundreds of millions of people from onchocerciasis and lymphatic filariasis. It was discovered by Satoshi Omura and William C. Campbell, to whom the 2015 Nobel Prize in Physiology or Medicine was awarded. It kills parasites by activating glutamate-gated Cl- channels, and it also targets several ligand-gated ion channels and receptors, including Cys-loop receptors, P2X4 receptors and fernesoid X receptors. Recently, we found that IVM also activates a novel target, the G-protein-gated inwardly rectifying K+ channel, and also identified the structural determinant for the activation. In this review, we aim to provide an update and summary of recent progress in the identification of IVM targets, as well as their modulation mechanisms, through molecular structures, chimeras and site-directed mutagenesis, and molecular docking and modelling studies.


Subject(s)
Antiparasitic Agents/pharmacology , Chloride Channels/physiology , G Protein-Coupled Inwardly-Rectifying Potassium Channels/physiology , Ion Channel Gating , Ivermectin/pharmacology , Receptors, Cytoplasmic and Nuclear/physiology , Receptors, Purinergic P2X4/physiology , Animals , Chloride Channels/drug effects , G Protein-Coupled Inwardly-Rectifying Potassium Channels/drug effects , Humans , Receptors, Cytoplasmic and Nuclear/drug effects , Receptors, Purinergic P2X4/drug effects
11.
Sci Rep ; 7(1): 14658, 2017 11 07.
Article in English | MEDLINE | ID: mdl-29116174

ABSTRACT

The hippocampus plays a critical role in learning and memory. Its correct performance relies on excitatory/inhibitory synaptic transmission balance. In early stages of Alzheimer's disease (AD), neuronal hyperexcitability leads to network dysfunction observed in cortical regions such as the hippocampus. G-protein-gated potassium (GirK) channels induce neurons to hyperpolarize, contribute to the resting membrane potential and could compensate any excesses of excitation. Here, we have studied the relationship between GirK channels and hippocampal function in a mouse model of early AD pathology. Intracerebroventricular injections of amyloid-ß (Aß 1-42) peptide-which have a causal role in AD pathogenesis-were performed to evaluate CA3-CA1 hippocampal synapse functionality in behaving mice. Aß increased the excitability of the CA3-CA1 synapse, impaired long-term potentiation (LTP) and hippocampal oscillatory activity, and induced deficits in novel object recognition (NOR) tests. Injection of ML297 alone, a selective GirK activator, was also translated in LTP and NOR deficits. However, increasing GirK activity rescued all hippocampal deficits induced by Aß due to the restoration of excitability values in the CA3-CA1 synapse. Our results show a synaptic mechanism, through GirK channel modulation, for the prevention of the hyperexcitability that causally contributes to synaptic, network, and cognitive deficits found in early AD pathogenesis.


Subject(s)
Alzheimer Disease/physiopathology , G Protein-Coupled Inwardly-Rectifying Potassium Channels/metabolism , Hippocampus/physiopathology , Alzheimer Disease/drug therapy , Alzheimer Disease/metabolism , Amyloid beta-Peptides/administration & dosage , Amyloid beta-Peptides/pharmacology , Animals , CA3 Region, Hippocampal/metabolism , CA3 Region, Hippocampal/physiopathology , Disease Models, Animal , G Protein-Coupled Inwardly-Rectifying Potassium Channels/drug effects , Hippocampus/metabolism , Injections, Intraventricular , Male , Mice , Mice, Inbred C57BL , Peptide Fragments/administration & dosage , Peptide Fragments/pharmacology , Phenylurea Compounds/pharmacology , Pyrazoles/pharmacology
12.
Bipolar Disord ; 19(7): 568-574, 2017 11.
Article in English | MEDLINE | ID: mdl-28895268

ABSTRACT

OBJECTIVES: Lithium (Li+ ) is one of the most widely used treatments for bipolar disorder (BD). However, the molecular and neuronal basis of BD, as well as the mechanisms of Li+ actions are poorly understood. Cellular and biochemical studies identified G proteins as being among the cellular targets for Li+ action, while genetic studies indicated an association with the KCNJ3 gene, which encodes the G protein-activated inwardly rectifying K+ (GIRK) channels. GIRK channels regulate neuronal excitability by mediating the inhibitory effects of multiple neurotransmitters and contribute to the resting potassium conductance. Here, we explored the effects of therapeutic dose of Li+ on neuronal excitability and the role of GIRK channels in Li+ actions. METHODS: Effects of Li+ on excitability were studied in hippocampal brain slices using whole-cell electrophysiological recordings. RESULTS: A therapeutic dose of Li+ (1 mM) dually regulated the function of GIRK channels in hippocampal slices. Li+ hyperpolarized the resting membrane potential of hippocampal CA1 pyramidal neurons and prolonged the latency to reach the action potential threshold and peak. These effects were abolished in the presence of tertiapin, a specific GIRK channel blocker, and at doses above the therapeutic window (2 mM). In contrast, Li+ reduced GIRK channel opening induced by GABAB receptor (GABAB R) activation, causing reduced hyperpolarization of the membrane potential, attenuated reduction of input resistance, and a smaller decrease of neuronal firing. CONCLUSIONS: A therapeutic dose of Li+ reduces the span of GIRK channel-mediated inhibition due to enhancement of basal GIRK currents and inhibition of GABAB R evoked responses, providing an important link between Li+ action, neuronal excitability, and cellular and genetic targets of BD.


Subject(s)
Antimanic Agents/pharmacology , Bipolar Disorder/drug therapy , G Protein-Coupled Inwardly-Rectifying Potassium Channels/drug effects , Hippocampus/drug effects , Lithium Compounds/pharmacology , Membrane Potentials/drug effects , Neural Inhibition/drug effects , Neurons/drug effects , Animals , Bipolar Disorder/metabolism , G Protein-Coupled Inwardly-Rectifying Potassium Channels/metabolism , Hippocampus/metabolism , Mice , Neurons/metabolism , Patch-Clamp Techniques
13.
J Neurosci ; 37(36): 8667-8677, 2017 09 06.
Article in English | MEDLINE | ID: mdl-28821664

ABSTRACT

Activation of somatic µ-opioid receptors (MORs) in hypothalamic proopiomelanocortin (POMC) neurons leads to the activation of G-protein-coupled inward rectifier potassium (GIRK) channels and hyperpolarization, but in response to continued signaling MORs undergo acute desensitization resulting in robust reduction in the peak GIRK current after minutes of agonist exposure. We hypothesized that the attenuation of the GIRK current would lead to a recovery of neuronal excitability whereby desensitization of the receptor would lead to a new steady state of POMC neuron activity reflecting the sustained GIRK current observed after the initial decline from peak with continued agonist exposure. However, electrophysiologic recordings and GCaMP6f Ca2+ imaging in POMC neurons in mouse brain slices indicate that maximal inhibition of cellular activity by these measures can be maintained after the GIRK current declines. Blockade of the GIRK current by Ba2+ or Tertiapin-Q did not disrupt the sustained inhibition of Ca2+ transients in the continued presence of agonist, indicating the activation of an effector other than GIRK channels. Use of an irreversible MOR antagonist and Furchgott analysis revealed a low receptor reserve for the activation of GIRK channels but a >90% receptor reserve for the inhibition of Ca2+ events. Altogether, the data show that somatodendritic MORs in POMC neurons inhibit neuronal activity through at least two effectors with distinct levels of receptor reserve and that differentially reflect receptor desensitization. Thus, in POMC cells, the decline in the GIRK current during prolonged MOR agonist exposure does not reflect an increase in cellular activity as expected.SIGNIFICANCE STATEMENT Desensitization of the µ-opioid receptor (MOR) is thought to underlie the development of cellular tolerance to opiate therapy. The present studies focused on MOR desensitization in hypothalamic proopiomelanocortin (POMC) neurons as these neurons produce the endogenous opioid ß-endorphin and are heavily regulated by opioids. Prolonged activation of somatic MORs in POMC neurons robustly inhibited action potential firing and Ca2+ activity despite desensitization of the MOR and reduced activation of a potassium current over the same time course. The data show that somatic MORs in POMC neurons couple to multiple effectors that have differential sensitivity to desensitization of the receptor. Thus, in these cells, the cellular consequence of MOR desensitization cannot be defined by the activity of a single effector system.


Subject(s)
Action Potentials/physiology , Hypothalamus/physiology , Neural Inhibition/physiology , Neurons/physiology , Pro-Opiomelanocortin/metabolism , Receptors, Opioid, mu/agonists , Receptors, Opioid, mu/metabolism , Animals , Central Nervous System Sensitization/drug effects , Central Nervous System Sensitization/physiology , Female , G Protein-Coupled Inwardly-Rectifying Potassium Channels/drug effects , G Protein-Coupled Inwardly-Rectifying Potassium Channels/physiology , Hypothalamus/drug effects , Ion Channel Gating/drug effects , Ion Channel Gating/physiology , Male , Mice , Mice, Inbred C57BL , Mice, Transgenic , Neural Inhibition/drug effects , Neurons/drug effects
14.
Naunyn Schmiedebergs Arch Pharmacol ; 390(5): 471-481, 2017 May.
Article in English | MEDLINE | ID: mdl-28160016

ABSTRACT

Nicotine abuse is associated with variety of diseases including arrhythmias, most often atrial fibrillation (AF). Altered inward rectifier potassium currents including acetylcholine-sensitive current I K(Ach) are known to be related to AF pathogenesis. Since relevant data are missing, we aimed to investigate I K(Ach) changes at clinically relevant concentrations of nicotine. Experiments were performed by the whole cell patch clamp technique at 23 ± 1 °C on isolated rat atrial myocytes. Nicotine was applied at following concentrations: 4, 40 and 400 nM; ethanol at 20 mM (∼0.09%). Nicotine at 40 and 400 nM significantly activated constitutively active component of I K(Ach) with the maximum effect at 40 nM (an increase by ∼100%); similar effect was observed at -110 and -50 mV. Changes at 4 nM nicotine were negligible on average. Coapplication of 40 nM nicotine and 20 mM ethanol (which is also known to activate this current) did not show cumulative effect. In the case of acetylcholine-induced component of I K(Ach), a dual effect of nicotine and its correlation with the current magnitude in control were apparent: the current was increased by nicotine in the cells showing small current in control and vice versa. The effect of 40 and 400 nM nicotine on acetylcholine-induced component of I K(Ach) was significantly different at -110 and -50 mV. We conclude that nicotine at clinically relevant concentrations significantly increased constitutively active component of I K(Ach) and showed a dual effect on its acetylcholine-induced component, similarly as ethanol. Synchronous application of nicotine and ethanol did not cause additive effect.


Subject(s)
Acetylcholine/pharmacology , G Protein-Coupled Inwardly-Rectifying Potassium Channels/agonists , G Protein-Coupled Inwardly-Rectifying Potassium Channels/drug effects , Heart Atria/drug effects , Myocytes, Cardiac/drug effects , Nicotine/toxicity , Animals , Arrhythmias, Cardiac/chemically induced , Arrhythmias, Cardiac/metabolism , Dose-Response Relationship, Drug , Ethanol/toxicity , Heart Atria/metabolism , In Vitro Techniques , Male , Membrane Potentials , Myocytes, Cardiac/metabolism , Rats, Wistar , Risk Assessment , Time Factors
15.
Trends Pharmacol Sci ; 38(4): 378-392, 2017 04.
Article in English | MEDLINE | ID: mdl-28188005

ABSTRACT

G protein-gated inwardly rectifying potassium (GIRK) channels are regulators of neuronal excitability in the brain. Knockout mice lacking GIRK channels display altered behavioral responses to multiple addictive drugs, implicating GIRK channels in addictive behaviors. Here, we review the effects of GIRK subunit deletions on the behavioral response to psychostimulants, such as cocaine and methamphetamine. Additionally, exposure of mice to psychostimulants produces alterations in the surface expression of GIRK channels in multiple types of neurons within the reward system of the brain. Thus, we compare the subcellular mechanisms by which drug exposure appears to alter GIRK expression in multiple cell types and provide an outlook on future studies examining the role of GIRK channels in addiction. A greater understanding of how GIRK channels are regulated by addictive drugs may enable the development of therapies to prevent or treat drug abuse.


Subject(s)
G Protein-Coupled Inwardly-Rectifying Potassium Channels/physiology , Substance-Related Disorders/drug therapy , Animals , Central Nervous System Stimulants/pharmacology , G Protein-Coupled Inwardly-Rectifying Potassium Channels/chemistry , G Protein-Coupled Inwardly-Rectifying Potassium Channels/drug effects , Humans , Long-Term Potentiation , Receptors, Dopamine D2/physiology , Receptors, GABA-B/physiology , Receptors, N-Methyl-D-Aspartate/physiology , Reward , Ventral Tegmental Area/physiology
16.
Neuropsychopharmacology ; 42(3): 707-715, 2017 02.
Article in English | MEDLINE | ID: mdl-27468917

ABSTRACT

The increase in dopamine (DA) neurotransmission stimulated by in vivo cocaine exposure is tempered by G protein-dependent inhibitory feedback mechanisms in DA neurons of the ventral tegmental area (VTA). G protein-gated inwardly rectifying K+ (GIRK/Kir3) channels mediate the direct inhibitory effect of GABAB receptor (GABABR) and D2 DA receptor (D2R) activation in VTA DA neurons. Here we examined the effect of the DA neuron-specific loss of GIRK channels on D2R-dependent regulation of VTA DA neuron excitability and on cocaine-induced, reward-related behaviors. Selective ablation of Girk2 in DA neurons did not alter the baseline excitability of VTA DA neurons but significantly reduced the magnitude of D2R-dependent inhibitory somatodendritic currents and blunted the impact of D2R activation on spontaneous activity and neuronal excitability. Mice lacking GIRK channels in DA neurons exhibited increased locomotor activation in response to acute cocaine administration and an altered locomotor sensitization profile, as well as increased responding for and intake of cocaine in an intravenous self-administration test. These mice, however, showed unaltered cocaine-induced conditioned place preference. Collectively, our data suggest that feedback inhibition to VTA DA neurons, mediated by GIRK channel activation, tempers the locomotor stimulatory effect of cocaine while also modulating the reinforcing effect of cocaine in an operant-based self-administration task.


Subject(s)
Behavior, Animal/drug effects , Cocaine/pharmacology , Dopaminergic Neurons/metabolism , G Protein-Coupled Inwardly-Rectifying Potassium Channels/metabolism , Learning/drug effects , Receptors, Dopamine D2/metabolism , Reward , Ventral Tegmental Area/metabolism , Animals , Dopaminergic Neurons/drug effects , G Protein-Coupled Inwardly-Rectifying Potassium Channels/drug effects , Mice , Mice, Transgenic , Receptors, Dopamine D2/drug effects , Ventral Tegmental Area/drug effects
17.
Ann Neurol ; 80(4): 511-21, 2016 10.
Article in English | MEDLINE | ID: mdl-27462820

ABSTRACT

OBJECTIVE: The Ts65Dn (Ts) mouse model of Down syndrome (DS) is exquisitely sensitive to an infantile spasms phenotype induced by γ-aminobutyric acidB receptor (GABAB R) agonists. The Ts mouse contains the core genomic triplication of the DS critical region, which includes 3 copies of the Kcnj6 gene that encodes the GABAB R-coupled G protein-coupled inward rectifying potassium channel subunit 2 (GIRK2) channel. We test the hypothesis that GIRK2 is necessary for the GABAB R agonist-induced infantile spasms phenotype in Ts. METHODS: We assessed the result of either genetic or pharmacological knockdown of the GIRK2 channel in Ts brain upon the GABAB R agonist-induced infantile spasms phenotype in the Ts mouse model of DS. As well, we examined GABAB R currents in hippocampal neurons prepared from GIRK2-trisomic Ts control mice and GIRK2-disomic Ts mice in which Kcnj6 had been genetically knocked down from 3 to 2 copies. RESULTS: The reduction of the copy number of Kcnj6 in Ts mice rescued the GABAB R agonist-induced infantile spasms phenotype. There was an increase in GABAB R-mediated GIRK2 currents in GIRK2-trisomic Ts mouse hippocampal neurons, which were normalized in the GIRK2-disomic Ts mice. Similarly, pharmacological knockdown of the GIRK2 channel in Ts brain using the GIRK antagonist tertiapin-Q also rescued the GABAB R agonist-induced infantile spasms phenotype in Ts mutants. INTERPRETATION: The GABAB R-coupled GIRK2 channel is necessary for the GABAB R agonist-induced infantile spasms phenotype in the Ts mouse and may represent a novel therapeutic target for the treatment of infantile spasms in DS. Ann Neurol 2016;80:511-521.


Subject(s)
G Protein-Coupled Inwardly-Rectifying Potassium Channels/metabolism , GABA-B Receptor Agonists/pharmacology , Potassium Channel Blockers/pharmacology , Receptors, GABA-B/metabolism , Spasms, Infantile/metabolism , Animals , Bee Venoms/pharmacology , Disease Models, Animal , Down Syndrome , Female , G Protein-Coupled Inwardly-Rectifying Potassium Channels/antagonists & inhibitors , G Protein-Coupled Inwardly-Rectifying Potassium Channels/drug effects , G Protein-Coupled Inwardly-Rectifying Potassium Channels/genetics , Hippocampus/metabolism , Hippocampus/physiopathology , Humans , Infant, Newborn , Male , Mice , Mice, Inbred C57BL , Mice, Transgenic , Neurons/metabolism , Neurons/physiology , Phenotype , Spasms, Infantile/chemically induced , Spasms, Infantile/genetics , Synaptic Potentials/physiology , Trisomy
18.
J Nat Prod ; 79(8): 1990-2004, 2016 08 26.
Article in English | MEDLINE | ID: mdl-27441737

ABSTRACT

GIRK channels are activated by a large number of G protein-coupled receptors and regulate the electrical activity of neurons, cardiac atrial myocytes, and ß-pancreatic cells. Abnormalities in GIRK channel function have been implicated in the pathophysiology of neuropathic pain, drug addiction, and cardiac arrhythmias. In the heart, GIRK channels are selectively expressed in the atrium, and their activation inhibits pacemaker activity, thereby slowing the heart rate. In the present study, 19 new diterpenes, falcatins A-S (1-19), and the known euphorprolitherin D (20) were isolated from Euphorbia falcata. The compounds were assayed on stable transfected HEK-hERG (Kv11.1) and HEK-GIRK1/4 (Kir3.1 and Kir3.4) cells. Blocking activity on GIRK channels was exerted by 13 compounds (61-83% at 10 µM), and, among them, five possessed low potency on the hERG channel (4-20% at 10 µM). These selective activities suggest that myrsinane-related diterpenes are potential lead compounds for the treatment of atrial fibrillation.


Subject(s)
Diterpenes , Euphorbia/chemistry , G Protein-Coupled Inwardly-Rectifying Potassium Channels/drug effects , Potassium Channel Blockers , Animals , Diterpenes/chemistry , Diterpenes/classification , Diterpenes/isolation & purification , Diterpenes/pharmacology , G Protein-Coupled Inwardly-Rectifying Potassium Channels/classification , Heart , Membrane Potentials/drug effects , Molecular Structure , Neurons/metabolism , Potassium Channel Blockers/pharmacology , Receptors, G-Protein-Coupled
19.
Naunyn Schmiedebergs Arch Pharmacol ; 389(10): 1049-58, 2016 Oct.
Article in English | MEDLINE | ID: mdl-27369777

ABSTRACT

Alcohol intoxication tends to induce arrhythmias, most often the atrial fibrillation. To elucidate arrhythmogenic mechanisms related to alcohol consumption, the effect of ethanol on main components of the ionic membrane current is investigated step by step. Considering limited knowledge, we aimed to examine the effect of clinically relevant concentrations of ethanol (0.8-80 mM) on acetylcholine-sensitive inward rectifier potassium current I K(Ach). Experiments were performed by the whole-cell patch clamp technique at 23 ± 1 °C on isolated rat and guinea-pig atrial myocytes, and on expressed human Kir3.1/3.4 channels. Ethanol induced changes of I K(Ach) in the whole range of concentrations applied; the effect was not voltage dependent. The constitutively active component of I K(Ach) was significantly increased by ethanol with the maximum effect (an increase by ∼100 %) between 8 and 20 mM. The changes were comparable in rat and guinea-pig atrial myocytes and also in expressed human Kir3.1/3.4 channels (i.e., structural correlate of I K(Ach)). In the case of the acetylcholine-induced component of I K(Ach), a dual ethanol effect was apparent with a striking heterogeneity of changes in individual cells. The effect correlated with the current magnitude in control: the current was increased by eth-anol in the cells showing small current in control and vice versa. The average effect peaked at 20 mM ethanol (an increase of the current by ∼20 %). Observed changes of action potential duration agreed well with the voltage clamp data. Ethanol significantly affected both components of I K(Ach) even in concentrations corresponding to light alcohol consumption.


Subject(s)
Acetylcholine/pharmacology , Arrhythmias, Cardiac/chemically induced , Ethanol/toxicity , G Protein-Coupled Inwardly-Rectifying Potassium Channels/drug effects , Heart Atria/drug effects , Myocytes, Cardiac/drug effects , Action Potentials , Animals , Arrhythmias, Cardiac/metabolism , Arrhythmias, Cardiac/physiopathology , CHO Cells , Computer Simulation , Cricetulus , Dose-Response Relationship, Drug , G Protein-Coupled Inwardly-Rectifying Potassium Channels/genetics , G Protein-Coupled Inwardly-Rectifying Potassium Channels/metabolism , Guinea Pigs , Heart Atria/metabolism , Heart Atria/physiopathology , Heart Rate/drug effects , Humans , Kinetics , Male , Models, Cardiovascular , Myocytes, Cardiac/metabolism , Rats, Wistar , Risk Assessment , Transfection
20.
Nat Med ; 22(3): 254-61, 2016 Mar.
Article in English | MEDLINE | ID: mdl-26808347

ABSTRACT

The lateral habenula (LHb) encodes aversive signals, and its aberrant activity contributes to depression-like symptoms. However, a limited understanding of the cellular mechanisms underlying LHb hyperactivity has precluded the development of pharmacological strategies to ameliorate depression-like phenotypes. Here we report that an aversive experience in mice, such as foot-shock exposure (FsE), induces LHb neuronal hyperactivity and depression-like symptoms. This occurs along with increased protein phosphatase 2A (PP2A) activity, a known regulator of GABAB receptor (GABABR) and G protein-gated inwardly rectifying potassium (GIRK) channel surface expression. Accordingly, FsE triggers GABAB1 and GIRK2 internalization, leading to rapid and persistent weakening of GABAB-activated GIRK-mediated (GABAB-GIRK) currents. Pharmacological inhibition of PP2A restores both GABAB-GIRK function and neuronal excitability. As a consequence, PP2A inhibition ameliorates depression-like symptoms after FsE and in a learned-helplessness model of depression. Thus, GABAB-GIRK plasticity in the LHb represents a cellular substrate for aversive experience. Furthermore, its reversal by PP2A inhibition may provide a novel therapeutic approach to alleviate symptoms of depression in disorders that are characterized by LHb hyperactivity.


Subject(s)
Behavior, Animal/drug effects , Bridged Bicyclo Compounds, Heterocyclic/pharmacology , Depression/metabolism , G Protein-Coupled Inwardly-Rectifying Potassium Channels/drug effects , Habenula/drug effects , Helplessness, Learned , Neuronal Plasticity/drug effects , Piperazines/pharmacology , Protein Phosphatase 2/antagonists & inhibitors , Receptors, GABA-B/drug effects , Stress, Psychological/metabolism , Animals , Disease Models, Animal , Electroshock , G Protein-Coupled Inwardly-Rectifying Potassium Channels/genetics , G Protein-Coupled Inwardly-Rectifying Potassium Channels/metabolism , Habenula/metabolism , Immunohistochemistry , Male , Mice , Mice, Inbred C57BL , Microscopy, Electron , Motor Activity , Patch-Clamp Techniques , Phenotype , Receptors, GABA-B/metabolism , Restraint, Physical , Reverse Transcriptase Polymerase Chain Reaction
SELECTION OF CITATIONS
SEARCH DETAIL