Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 10 de 10
Filter
Add more filters










Publication year range
1.
Exp Neurol ; 359: 114250, 2023 01.
Article in English | MEDLINE | ID: mdl-36240882

ABSTRACT

Orexin is a neuropeptide restrictedly synthesized in the hypothalamus, but extensively modulates the whole brain region activity including prefrontal cortex (PFC), and involved in the pathophysiology of psychiatric disorders. GABAergic interneurons in the mPFC are a promising pharmacological target for developing antidepressant therapies. Here, we examined the effects of the orexin on GABAergic transmission onto pyramidal neurons in the deep layers of the mPFC. We found that bath application of orexin dose-dependently increased the amplitude of evoked IPSCs (eIPSCs). Orexin increased the frequency but not the amplitude of miniature IPSCs (mIPSCs). Ca2+ influx through T-type voltage-gated Ca2+ channels is required for orexin-induced increases in GABA release. We also found orexin increases GABA release probability and the number of releasable vesicles. Orexin depolarizes somatostatin (Sst) interneurons without effects on the firing rate of action potentials (APs) of Sst interneurons. Orexin-induced depolarization of Sst interneurons is independent of extracellular Na+, Ca2+ and T-type Ca2+ channels, but requires inward rectifier K+ channels (Kirs). The present study suggests that orexin enhances GABAergic transmission onto mPFC pyramidal neurons through inhibiting Kirs on Sst interneurons, which further depolarizes interneurons leading to increase in Ca2+ influx via T-type Ca2+ channels. Our results may provide a cellular and molecular mechanism that helps explain the physiological functions of orexin in the brain.


Subject(s)
Potassium Channels, Inwardly Rectifying , Animals , Mice , gamma-Aminobutyric Acid/pharmacology , Interneurons/physiology , Orexins/pharmacology , Potassium Channels, Inwardly Rectifying/metabolism , Prefrontal Cortex/metabolism , Pyramidal Cells/physiology , Synaptic Transmission/physiology
2.
Angew Chem Int Ed Engl ; 61(13): e202112232, 2022 03 21.
Article in English | MEDLINE | ID: mdl-34985791

ABSTRACT

Cholesterol oligomers reside in multiple membrane protein X-ray crystal structures. Yet, there is no direct link between these oligomers and a biological function. Here we present the structural and functional details of a cholesterol dimer that stabilizes the inactivated state of an inward-rectifier potassium channel KirBac1.1. K+ efflux assays confirm that high cholesterol concentration reduces K+ conductance. We then determine the structure of the cholesterol-KirBac1.1 complex using Xplor-NIH simulated annealing calculations driven by solid-state NMR distance measurements. These calculations identified an α-α cholesterol dimer docked to a cleft formed by adjacent subunits of the homotetrameric protein. We compare these results to coarse grain molecular dynamics simulations. This is one of the first examples of a cholesterol oligomer performing a distinct biological function and structural characterization of a conserved promiscuous lipid binding region.


Subject(s)
Potassium Channels, Inwardly Rectifying , Cholesterol , Potassium/metabolism , Potassium Channels, Inwardly Rectifying/chemistry , Potassium Channels, Inwardly Rectifying/metabolism
3.
J Cereb Blood Flow Metab ; 41(6): 1313-1327, 2021 06.
Article in English | MEDLINE | ID: mdl-33050826

ABSTRACT

Traumatic brain injury (TBI) acutely impairs dynamic regulation of local cerebral blood flow, but long-term (>72 h) effects on functional hyperemia are unknown. Functional hyperemia depends on capillary endothelial cell inward rectifier potassium channels (Kir2.1) responding to potassium (K+) released during neuronal activity to produce a regenerative, hyperpolarizing electrical signal that propagates from capillaries to dilate upstream penetrating arterioles. We hypothesized that TBI causes widespread disruption of electrical signaling from capillaries-to-arterioles through impairment of Kir2.1 channel function. We randomized mice to TBI or control groups and allowed them to recover for 4 to 7 days post-injury. We measured in vivo cerebral hemodynamics and arteriolar responses to local stimulation of capillaries with 10 mM K+ using multiphoton laser scanning microscopy through a cranial window under urethane and α-chloralose anesthesia. Capillary angio-architecture was not significantly affected following injury. However, K+-induced hyperemia was significantly impaired. Electrophysiology recordings in freshly isolated capillary endothelial cells revealed diminished Ba2+-sensitive Kir2.1 currents, consistent with a reduction in channel function. In pressurized cerebral arteries isolated from TBI mice, K+ failed to elicit the vasodilation seen in controls. We conclude that disruption of endothelial Kir2.1 channel function impairs capillary-to-arteriole electrical signaling, contributing to altered cerebral hemodynamics after TBI.


Subject(s)
Arterioles/metabolism , Brain Injuries, Traumatic/physiopathology , Capillaries/metabolism , Cerebrovascular Circulation/physiology , Potassium Channels, Inwardly Rectifying/metabolism , Animals , Endothelial Cells/metabolism , Hemodynamics/physiology , Male , Mice , Mice, Inbred C57BL , Signal Transduction/physiology
4.
SLAS Discov ; 25(5): 420-433, 2020 06.
Article in English | MEDLINE | ID: mdl-32292089

ABSTRACT

K+ channels play a critical role in maintaining the normal electrical activity of excitable cells by setting the cell resting membrane potential and by determining the shape and duration of the action potential. In nonexcitable cells, K+ channels establish electrochemical gradients necessary for maintaining salt and volume homeostasis of body fluids. Inward rectifier K+ (Kir) channels typically conduct larger inward currents than outward currents, resulting in an inwardly rectifying current versus voltage relationship. This property of inward rectification results from the voltage-dependent block of the channels by intracellular polyvalent cations and makes these channels uniquely designed for maintaining the resting potential near the K+ equilibrium potential (EK). The Kir family of channels consist of seven subfamilies of channels (Kir1.x through Kir7.x) that include the classic inward rectifier (Kir2.x) channel, the G-protein-gated inward rectifier K+ (GIRK) (Kir3.x), and the adenosine triphosphate (ATP)-sensitive (KATP) (Kir 6.x) channels as well as the renal Kir1.1 (ROMK), Kir4.1, and Kir7.1 channels. These channels not only function to regulate electrical/electrolyte transport activity, but also serve as effector molecules for G-protein-coupled receptors (GPCRs) and as molecular sensors for cell metabolism. Of significance, Kir channels represent promising pharmacological targets for treating a number of clinical conditions, including cardiac arrhythmias, anxiety, chronic pain, and hypertension. This review provides a brief background on the structure, function, and pharmacology of Kir channels and then focuses on describing and evaluating current high-throughput screening (HTS) technologies, such as membrane potential-sensitive fluorescent dye assays, ion flux measurements, and automated patch clamp systems used for Kir channel drug discovery.


Subject(s)
Drug Evaluation, Preclinical , High-Throughput Screening Assays , Potassium Channel Blockers/isolation & purification , Potassium Channels/agonists , G Protein-Coupled Inwardly-Rectifying Potassium Channels/agonists , G Protein-Coupled Inwardly-Rectifying Potassium Channels/antagonists & inhibitors , Humans , Potassium Channel Blockers/therapeutic use , Potassium Channels/genetics , Potassium Channels, Inwardly Rectifying/agonists , Potassium Channels, Inwardly Rectifying/antagonists & inhibitors
5.
Eur J Pharmacol ; 815: 233-240, 2017 Nov 15.
Article in English | MEDLINE | ID: mdl-28935562

ABSTRACT

G protein-gated inward rectifier K+ (GIRK) channels are members of the super-family of proteins known as inward rectifier K+ (Kir) channels and are expressed throughout the peripheral and central nervous systems. Neuronal GIRK channels are the downstream targets of a number of neuromodulators including opioids, somatostatin, dopamine and cannabinoids. Previous studies have demonstrated that the ATP-sensitive K+ channel, another member of the Kir channel family, is regulated by sulfonamide drugs. Therefore, to determine if sulfonamides also modulate GIRK channels, we screened a library of arylsulfonamide compounds using a GIRK channel fluorescent assay that utilized pituitary AtT20 cells expressing GIRK channels along with the somatostatin type-2 and -5 receptors. Enhancement of the GIRK channel fluorescent signal by one compound, N-(2-methoxyphenyl) benzenesulfonamide (MPBS), was dependent on the activation of the channel by somatostatin. In whole-cell patch clamp experiments, application of MPBS both shifted the somatostatin concentration-response curve (EC50 = 3.5nM [control] vs.1.0nM [MPBS]) for GIRK channel activation and increased the maximum GIRK current measured with 100nM somatostatin. However, GIRK channel activation was not observed when MPBS was applied to the cells in the absence of somatostatin. While the MPBS structural analog 4-fluoro-N-(2-methoxyphenyl) benzenesulfonamide also augmented the somatostatin-induced GIRK fluorescent signal, no increase in the signal was observed with the sulfonamides tolbutamide, sulfapyridine and celecoxib. In conclusion, MPBS represents a novel prototypic GPCR-dependent regulator of neuronal GIRK channels.


Subject(s)
GTP-Binding Proteins/metabolism , Hydroxylamines/pharmacology , Ion Channel Gating/drug effects , Neurons/drug effects , Neurons/metabolism , Potassium Channels, Inwardly Rectifying/metabolism , Sulfonamides/pharmacology , Cell Line , Dose-Response Relationship, Drug , Humans , Benzenesulfonamides
6.
Pflugers Arch ; 468(11-12): 1931-1943, 2016 11.
Article in English | MEDLINE | ID: mdl-27838849

ABSTRACT

Inward rectifier K+ channels are important for maintaining normal electrical function in many cell types. The proper function of these channels requires the presence of membrane phosphoinositide 4,5-bisphosphate (PIP2). Stimulation of the Ca2+-sensing receptor CaR, a pleiotropic G protein-coupled receptor, activates both Gq/11, which decreases PIP2, and phosphatidylinositol 4-kinase (PI-4-K), which, conversely, increases PIP2. How membrane PIP2 levels are regulated by CaR activation and whether these changes modulate inward rectifier K+ are unknown. In this study, we found that activation of CaR by the allosteric agonist, NPSR568, increased inward rectifier K+ current (I K1) in guinea pig ventricular myocytes and currents mediated by Kir2.1 channels exogenously expressed in HEK293T cells with a similar sensitivity. Moreover, using the fluorescent PIP2 reporter tubby-R332H-cYFP to monitor PIP2 levels, we found that CaR activation in HEK293T cells increased membrane PIP2 concentrations. Pharmacological studies showed that both phospholipase C (PLC) and PI-4-K are activated by CaR stimulation with the latter played a dominant role in regulating membrane PIP2 and, thus, Kir currents. These results provide the first direct evidence that CaR activation upregulates currents through inward rectifier K+ channels by accelerating PIP2 synthesis. The regulation of I K1 plays a critical role in the stability of the electrical properties of many excitable cells, including cardiac myocytes and neurons. Further, synthetic allosteric modulators that increase CaR activity have been used to treat hyperparathyroidism, and negative CaR modulators are of potential importance in the treatment of osteoporosis. Thus, our results provide further insight into the roles played by CaR in the cardiovascular system and are potentially valuable for heart disease treatment and drug safety.


Subject(s)
1-Phosphatidylinositol 4-Kinase/metabolism , Action Potentials , Phosphatidylinositol 4,5-Diphosphate/metabolism , Potassium Channels, Inwardly Rectifying/metabolism , Receptors, Calcium-Sensing/metabolism , Animals , Cells, Cultured , Guinea Pigs , HEK293 Cells , Humans , Myocytes, Cardiac/metabolism , Myocytes, Cardiac/physiology , Type C Phospholipases/metabolism
7.
Neuropharmacology ; 109: 18-28, 2016 10.
Article in English | MEDLINE | ID: mdl-27236080

ABSTRACT

The overexpression of Kir3.2, a subunit of the G protein-gated inwardly rectifying K(+) channel, is implicated in some of the neurological phenotypes of Down syndrome (DS). Chemical compounds that block Kir3.2 are expected to improve the symptoms of DS. The purpose of this study is to develop a cell-based screening system to identify Kir3.2 blockers and then investigate the mode of action of the blocker. Chemical screening was carried out using a K(+) transporter-deficient yeast strain that expressed a constitutively active Kir3.2 mutant. The mode of action of an effective blocker was electrophysiologically analyzed using Kir channels expressed in Xenopus oocytes. Proflavine was identified to inhibit the growth of Kir3.2-transformant cells and Kir3.2 activity in a concentration-dependent manner. The current inhibition was strong when membrane potentials (Vm) was above equilibrium potential of K(+) (EK). When Vm was below EK, the blockage apparently depended on the difference between Vm and [K(+)]. Furthermore, the inhibition became stronger by lowering extracellular [K(+)]. These results indicated that the yeast strain serves as a screening system to isolate Kir3.2 blockers and proflavine is a prototype of a pore blocker of Kir3.2.


Subject(s)
G Protein-Coupled Inwardly-Rectifying Potassium Channels/antagonists & inhibitors , G Protein-Coupled Inwardly-Rectifying Potassium Channels/physiology , Growth Inhibitors/pharmacology , Potassium Channel Blockers/pharmacology , Proflavine/pharmacology , Animals , Cell Proliferation/drug effects , Cell Proliferation/physiology , Dose-Response Relationship, Drug , Female , Growth Inhibitors/chemistry , Mice , Potassium Channel Blockers/chemistry , Proflavine/chemistry , Xenopus laevis
8.
Perfusion ; 29(4): 321-326, 2014 Jul.
Article in English | MEDLINE | ID: mdl-24395682

ABSTRACT

BACKGROUND: Selective inhibition of inward rectifier K+ channels could abolish the protection mediated by ischemic preconditioning, but the roles of these channels in ischemic postconditioning have not been well characterized. Our study aims to evaluate the effect of inward rectifier K+ channels on the protection induced by ischemic postconditioning. METHODS: Langendorff-perfused rat hearts (n=8 per group) were split into four groups: postconditioning hearts (IPO group); ischemic postconditioning with BaCl2 hearts (PB group); ischemic postconditioning with zacopride hearts (PZ group); and without ischemic postconditioning (CON group). After suffering 30 minutes of global ischemia, groups IPO, PB and PZ went through 10 seconds of ischemic postconditioning with three different perfusates: respectively, Krebs-Henseleit buffer (IPO group); 20 µmol/L BaCl2 (antagonist of the channel, PB group); 1 µmol/L zacopride (agonist of the channel, PZ group). RESULTS: At the end of reperfusion, the myocardial performance was better preserved in the PZ group than the other three groups. The PB group showed no significant differences from the CON group. CONCLUSIONS: Our study has shown that the IK1 channel agonist zacopride is associated with the enhancement of ischemic postconditioning.

9.
Biochim Biophys Acta ; 1838(2): 521-31, 2014 Feb.
Article in English | MEDLINE | ID: mdl-24028827

ABSTRACT

A variety of extracellular stimuli regulate cellular responses via membrane receptors. A well-known group of seven-transmembrane domain-containing proteins referred to as G protein-coupled receptors, directly couple with the intracellular GTP-binding proteins (G proteins) across cell membranes and trigger various cellular responses by regulating the activity of several enzymes as well as ion channels. Many specific populations of ion channels are directly controlled by G proteins; however, indirect modulation of some channels by G protein-dependent phosphorylation events and lipid metabolism is also observed. G protein-mediated diverse modifications affect the ion channel activities and spatio-temporally regulate membrane potentials as well as of intracellular Ca(2+) concentrations in both excitatory and non-excitatory cells. This article is part of a Special Issue entitled: Reciprocal influences between cell cytoskeleton and membrane channels, receptors and transporters. Guest Editor: Jean Claude Hervé.


Subject(s)
GTP-Binding Proteins/metabolism , Ion Channels/metabolism , Animals , Humans , Signal Transduction
10.
Yonsei Medical Journal ; : 459-471, 2008.
Article in English | WPRIM (Western Pacific) | ID: wpr-79505

ABSTRACT

PURPOSE: This study examined the expression and function of inward rectifier K+ channels in cultured rat hepatic stellate cells (HSC). MATERIALS AND METHODS: The expression of inward rectifier K+ channels was measured using real-time RT-PCR, and electrophysiological properties were determined using the gramicidin-perforated patch-clamp technique. RESULTS: The dominant inward rectifier K+ channel subtypes were K(ir)2.1 and K(ir)6.1. These dominant K+ channel subtypes decreased significantly during the primary culture throughout activation process. HSC can be classified into two subgroups: one with an inward-rectifying K+ current (type 1) and the other without (type 2). The inward current was blocked by Ba2+ (100micrometer) and enhanced by high K+ (140mM), more prominently in type 1 HSC. There was a correlation between the amplitude of the Ba2+-sensitive current and the membrane potential. In addition, Ba2+ (300micrometer) depolarized the membrane potential. After the culture period, the amplitude of the inward current decreased and the membrane potential became depolarized. CONCLUSION: HSC express inward rectifier K+ channels, which physiologically regulate membrane potential and decrease during the activation process. These results will potentially help determine properties of the inward rectifier K+ channels in HSC as well as their roles in the activation process.


Subject(s)
Animals , Male , Rats , Barium/pharmacology , Blotting, Western , Cells, Cultured , Electrophysiology , Liver/cytology , Membrane Potentials/drug effects , Potassium/pharmacology , Potassium Channels, Inwardly Rectifying/genetics , Rats, Sprague-Dawley , Reverse Transcriptase Polymerase Chain Reaction
SELECTION OF CITATIONS
SEARCH DETAIL
...